1
|
Summers RJ, Teachey DT, Hunger SP. How I treat ETP-ALL in children. Blood 2025; 145:43-52. [PMID: 38364183 DOI: 10.1182/blood.2023023155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
ABSTRACT Early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) is a unique subtype of immature T-cell ALL that was initially associated with a dramatically inferior prognosis compared with non-ETP T-cell ALL (Not-ETP) when it was first described in 2009. Analyses of larger patient cohorts treated with more contemporary regimens, however, have shown minimal survival differences between ETP and Not-ETP. In this manuscript, we use representative cases to explore therapeutic advances and address common clinical questions regarding the management of children, adolescents, and young adults with ETP-ALL. We describe our recommended treatment approach for a child or adolescent with newly diagnosed ETP-ALL, with an emphasis on the prognostic significance of induction failure and detectable minimal residual disease and the role of hematopoietic stem cell transplant in first remission. We discuss the interplay between the ETP immunophenotype and genomic markers of immaturity in T-cell ALL. Finally, we review novel therapeutic approaches that should be considered when managing relapsed or refractory ETP-ALL.
Collapse
Affiliation(s)
- Ryan J Summers
- Department of Pediatrics, Emory University, Atlanta, GA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA
| | - David T Teachey
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Stephen P Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
2
|
Burattin FV, Vadalà R, Panepuccia M, Ranzani V, Crosti M, Colombo FA, Ruberti C, Erba E, Prati D, Nittoli T, Montini G, Ronchi A, Pugni L, Mosca F, Ricciardi S, Abrignani S, Pietrasanta C, Marasca F, Bodega B. LINE1 modulate human T cell function by regulating protein synthesis during the life span. SCIENCE ADVANCES 2024; 10:eado2134. [PMID: 39383231 PMCID: PMC11463280 DOI: 10.1126/sciadv.ado2134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 09/04/2024] [Indexed: 10/11/2024]
Abstract
The molecular mechanisms responsible for the heightened reactivity of quiescent T cells in human early life remain largely elusive. Our previous research identified that quiescent adult naïve CD4+ T cells express LINE1 (long interspersed nuclear elements 1) spliced in previously unknown isoforms, and their down-regulation marks the transition to activation. Here, we unveil that neonatal naïve T cell quiescence is characterized by enhanced energy production and protein synthesis. This phenotype is associated with the absence of LINE1 expression attributed to tonic T cell receptor/mTOR complex 1 (mTORC1) signaling and (polypyrimidine tract-binding protein 1 (PTBP1)-mediated LINE1 splicing suppression. The absence of LINE1 expression primes these cells for rapid execution of the activation program by directly regulating protein synthesis. LINE1 expression progressively increases in childhood and adults, peaking in elderly individuals, and, by decreasing protein synthesis, contributes to immune senescence in aging. Our study proposes LINE1 as a critical player of human T cell function across the human life span.
Collapse
Affiliation(s)
- Filippo V. Burattin
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Rebecca Vadalà
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Michele Panepuccia
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
- SEMM, European School of Molecular Medicine, Milan 20139, Italy
| | - Valeria Ranzani
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
| | - Mariacristina Crosti
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
| | - Federico A. Colombo
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Cristina Ruberti
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Elisa Erba
- Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Teresa Nittoli
- Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Giovanni Montini
- Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan 20122, Italy
| | - Andrea Ronchi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Lorenza Pugni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Fabio Mosca
- Department of Clinical Sciences and Community Health, University of Milan, Milan 20122, Italy
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Sara Ricciardi
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Sergio Abrignani
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan 20122, Italy
| | - Carlo Pietrasanta
- Department of Clinical Sciences and Community Health, University of Milan, Milan 20122, Italy
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Federica Marasca
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
| | - Beatrice Bodega
- Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi” (INGM), Milan 20122, Italy
- Department of Biosciences, University of Milan, Milan 20133, Italy
| |
Collapse
|
3
|
Deep D, Gudjonson H, Brown CC, Rose SA, Sharma R, Paucar Iza YA, Hong S, Hemmers S, Schizas M, Wang ZM, Chen Y, Wesemann DR, Pascual V, Pe’er D, Rudensky AY. Precursor central memory versus effector cell fate and naïve CD4+ T cell heterogeneity. J Exp Med 2024; 221:e20231193. [PMID: 39321257 PMCID: PMC11448869 DOI: 10.1084/jem.20231193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/08/2024] [Accepted: 08/12/2024] [Indexed: 09/27/2024] Open
Abstract
Upon antigenic stimulation, naïve CD4+ T cells can give rise to phenotypically distinct effector T helper cells and long-lived memory T cells. We computationally reconstructed the in vivo trajectory of CD4+ T cell differentiation during a type I inflammatory immune response and identified two distinct differentiation paths for effector and precursor central memory T cells arising directly from naïve CD4+ T cells. Unexpectedly, our studies revealed heterogeneity among naïve CD4+ T cells, which are typically considered homogeneous save for their diverse T cell receptor usage. Specifically, a previously unappreciated population of naïve CD4+ T cells sensing environmental type I IFN exhibited distinct activation thresholds, suggesting that naïve CD4+ T cell differentiation potential may be influenced by environmental cues. This population was expanded in human viral infection and type I IFN response-lined autoimmunity. Understanding the relevance of naïve T cell heterogeneity to beneficial and maladaptive T cell responses may have therapeutic implications for adoptive T cell therapies in cancer immunotherapy and vaccination.
Collapse
Affiliation(s)
- Deeksha Deep
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, The Rockefeller University and Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA
| | - Herman Gudjonson
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chrysothemis C. Brown
- Immuno-Oncology, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel A. Rose
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roshan Sharma
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yoselin A. Paucar Iza
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA
| | - Seunghee Hong
- Drukier Institute for Children’s Health at Weill Cornell Medicine, New York, NY, USA
| | - Saskia Hemmers
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Michail Schizas
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhong-Min Wang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Gerstner Sloan Kettering Graduate School in Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Discovery, Genentech Inc., South San Francisco, CA, USA
| | - Yuezhou Chen
- Department of Medicine, Division of Allergy and Immunology, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Duane R. Wesemann
- Department of Medicine, Division of Allergy and Immunology, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Virginia Pascual
- Drukier Institute for Children’s Health at Weill Cornell Medicine, New York, NY, USA
| | - Dana Pe’er
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Alexander Y. Rudensky
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
4
|
Baldwin I, Robey EA. Adjusting to self in the thymus: CD4 versus CD8 lineage commitment and regulatory T cell development. J Exp Med 2024; 221:e20230896. [PMID: 38980291 PMCID: PMC11232887 DOI: 10.1084/jem.20230896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/22/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
During thymic development, thymocytes adjust their TCR response based on the strength of their reactivity to self-peptide MHC complexes. This tuning process allows thymocytes with a range of self-reactivities to survive positive selection and contribute to a diverse T cell pool. In this review, we will discuss recent advances in our understanding of how thymocytes tune their responsiveness during positive selection, and we present a "sequential selection" model to explain how MHC specificity influences lineage choice. We also discuss recent evidence for cell type diversity in the medulla and discuss how this heterogeneity may contribute to medullary niches for negative selection and regulatory T cell development.
Collapse
Affiliation(s)
- Isabel Baldwin
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ellen A. Robey
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
5
|
Lo WL, Huseby ES. The partitioning of TCR repertoires by thymic selection. J Exp Med 2024; 221:e20230897. [PMID: 39167074 PMCID: PMC11338286 DOI: 10.1084/jem.20230897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/22/2024] [Accepted: 07/09/2024] [Indexed: 08/23/2024] Open
Abstract
αβ T cells are critical components of the adaptive immune system; they maintain tissue and immune homeostasis during health, provide sterilizing immunity after pathogen infection, and are capable of eliminating transformed tumor cells. Fundamental to these distinct functions is the ligand specificity of the unique antigen receptor expressed on each mature T cell (TCR), which endows lymphocytes with the ability to behave in a cell-autonomous, disease context-specific manner. Clone-specific behavioral properties are initially established during T cell development when thymocytes use TCR recognition of major histocompatibility complex (MHC) and MHC-like ligands to instruct survival versus death and to differentiate into a plethora of inflammatory and regulatory T cell lineages. Here, we review the ligand specificity of the preselection thymocyte repertoire and argue that developmental stage-specific alterations in TCR signaling control cross-reactivity and foreign versus self-specificity of T cell sublineages.
Collapse
Affiliation(s)
- Wan-Lin Lo
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Eric S Huseby
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
6
|
Adegoke AO, Thangavelu G, Chou TF, Petersen MI, Kakugawa K, May JF, Joannou K, Wang Q, Ellestad KK, Boon L, Bretscher PA, Cheroutre H, Kronenberg M, Baldwin TA, Anderson CC. Internal regulation between constitutively expressed T cell co-inhibitory receptors BTLA and CD5 and tolerance in recent thymic emigrants. Open Biol 2024; 14:240178. [PMID: 39471840 PMCID: PMC11521602 DOI: 10.1098/rsob.240178] [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: 06/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 11/01/2024] Open
Abstract
Immunologic self-tolerance involves signals from co-inhibitory receptors. Several T cell co-inhibitors, including PD-1, are expressed upon activation, whereas CD5 and BTLA are expressed constitutively. The relationship between constitutively expressed co-inhibitors and when they are needed is unknown. Deletion of Btla demonstrated BTLA regulates CD5 expression. Loss of BTLA signals, but not signalling by its ligand, HVEM, leads to increased CD5 expression. Higher CD5 expression set during thymic selection is associated with increased self-recognition, suggesting that BTLA might be needed early to establish self-tolerance. We found that BTLA and PD-1 were needed post-thymic selection in recent thymic emigrants (RTE). RTE lacking BTLA caused a CD4 T cell and MHC class II dependent multi-organ autoimmune disease. Together, our findings identify a negative regulatory pathway between two constitutively expressed co-inhibitors, calibrating their expression. Expression of constitutive and induced co-inhibitory receptors is needed early to establish tolerance in the periphery for RTE.
Collapse
Affiliation(s)
| | - Govindarajan Thangavelu
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Alberta Diabetes and Transplant Institutes, University of Alberta, Edmonton, AB, Canada
| | - Ting-Fang Chou
- La Jolla Institute for Immunology, La Jolla, CA92037, USA
| | - Marcos I. Petersen
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Kiyokazu Kakugawa
- Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro, Tsurumi-ku, Yokohama230-0045, Japan
| | - Julia F. May
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Kevin Joannou
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Qingyang Wang
- La Jolla Institute for Immunology, La Jolla, CA92037, USA
| | - Kristofor K. Ellestad
- Alberta Diabetes and Transplant Institutes, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | | | - Peter A. Bretscher
- Department of Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Hilde Cheroutre
- La Jolla Institute for Immunology, La Jolla, CA92037, USA
- Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro, Tsurumi-ku, Yokohama230-0045, Japan
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA92037, USA
- Department of Molecular Biology, University of California San Diego, La Jolla, CA92093, USA
| | - Troy A. Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Colin C. Anderson
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Alberta Diabetes and Transplant Institutes, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
7
|
Lang HP, Osum KC, Friedenberg SG. A review of CD4 + T cell differentiation and diversity in dogs. Vet Immunol Immunopathol 2024; 275:110816. [PMID: 39173398 PMCID: PMC11421293 DOI: 10.1016/j.vetimm.2024.110816] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
CD4+ T cells are an integral component of the adaptive immune response, carrying out many functions to combat a diverse range of pathogenic challenges. These cells exhibit remarkable plasticity, differentiating into specialized subsets such as T helper type 1 (TH1), TH2, TH9, TH17, TH22, regulatory T cells (Tregs), and follicular T helper (TFH) cells. Each subset is capable of addressing a distinct immunological need ranging from pathogen eradication to regulation of immune homeostasis. As the immune response subsides, CD4+ T cells rest down into long-lived memory phenotypes-including central memory (TCM), effector memory (TEM), resident memory (TRM), and terminally differentiated effector memory cells (TEMRA) that are localized to facilitate a swift and potent response upon antigen re-encounter. This capacity for long-term immunological memory and rapid reactivation upon secondary exposure highlights the role CD4+ T cells play in sustaining both adaptive defense mechanisms and maintenance. Decades of mouse, human, and to a lesser extent, pig T cell research has provided the framework for understanding the role of CD4+ T cells in immune responses, but these model systems do not always mimic each other. Although our understanding of pig immunology is not as extensive as mouse or human research, we have gained valuable insight by studying this model. More akin to pigs, our understanding of CD4+ T cells in dogs is much less complete. This disparity exists in part because canine immunologists depend on paradigms from mouse and human studies to characterize CD4+ T cells in dogs, with a fraction of available lineage-defining antibody markers. Despite this, every major CD4+ T cell subset has been described to some extent in dogs. These subsets have been studied in various contexts, including in vitro stimulation, homeostatic conditions, and across a range of disease states. Canine CD4+ T cells have been categorized according to lineage-defining characteristics, trafficking patterns, and what cytokines they produce upon stimulation. This review addresses our current understanding of canine CD4+ T cells from a comparative perspective by highlighting both the similarities and differences from mouse, human, and pig CD4+ T cell biology. We also discuss knowledge gaps in our current understanding of CD4+ T cells in dogs that could provide direction for future studies in the field.
Collapse
Affiliation(s)
- Haeree P Lang
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
| | - Kevin C Osum
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA.
| | - Steven G Friedenberg
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
| |
Collapse
|
8
|
Park CS, Guan J, Rhee P, Gonzalez F, Lee HS, Park JH, Coscoy L, Robey EA, Shastri N, Sadegh-Nasseri S. Fam49b dampens TCR signal strength to regulate survival of positively selected thymocytes and peripheral T cells. eLife 2024; 13:e76940. [PMID: 39158947 PMCID: PMC11333044 DOI: 10.7554/elife.76940] [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: 01/10/2022] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
The fate of developing T cells is determined by the strength of T cell receptor (TCR) signal they receive in the thymus. This process is finely regulated through the tuning of positive and negative regulators in thymocytes. The Family with sequence similarity 49 member B (Fam49b) protein is a newly discovered negative regulator of TCR signaling that has been shown to suppress Rac-1 activity in vitro in cultured T cell lines. However, the contribution of Fam49b to the thymic development of T cells is unknown. To investigate this important issue, we generated a novel mouse line deficient in Fam49b (Fam49b-KO). We observed that Fam49b-KO double positive (DP) thymocytes underwent excessive negative selection, whereas the positive selection stage was unaffected. Fam49b deficiency impaired the survival of single positive thymocytes and peripheral T cells. This altered development process resulted in significant reductions in CD4 and CD8 single-positive thymocytes as well as peripheral T cells. Interestingly, a large proportion of the TCRγδ+ and CD8αα+TCRαβ+ gut intraepithelial T lymphocytes were absent in Fam49b-KO mice. Our results demonstrate that Fam49b dampens thymocytes TCR signaling in order to escape negative selection during development, uncovering the function of Fam49b as a critical regulator of the selection process to ensure normal thymocyte development and peripheral T cells survival.
Collapse
Affiliation(s)
- Chan-Su Park
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National UniversityCheongjuRepublic of Korea
| | - Jian Guan
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Peter Rhee
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Federico Gonzalez
- Department of Nutritional Sciences and Toxicology, University of California,BerkeleyBerkeleyUnited States
| | - Hee-sung Lee
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National UniversityCheongjuRepublic of Korea
| | - Ji-hyun Park
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National UniversityCheongjuRepublic of Korea
| | - Laurent Coscoy
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Ellen A Robey
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Nilabh Shastri
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | | |
Collapse
|
9
|
Tai TS, Yang HY, Chuang WC, Huang YW, Ho IC, Tsai CC, Chuang YT. ScRNA-Seq Analyses Define the Role of GATA3 in iNKT Cell Effector Lineage Differentiation. Cells 2024; 13:1073. [PMID: 38920701 PMCID: PMC11201670 DOI: 10.3390/cells13121073] [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: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
While the transcription factor GATA-3 is well-established for its crucial role in T cell development, its specific influence on invariant natural killer T (iNKT) cells remains relatively unexplored. Using flow cytometry and single-cell transcriptomic analysis, we demonstrated that GATA-3 deficiency in mice leads to the absence of iNKT2 and iNKT17 cell subsets, as well as an altered distribution of iNKT1 cells. Thymic iNKT cells lacking GATA-3 exhibited diminished expression of PLZF and T-bet, key transcription factors involved in iNKT cell differentiation, and reduced production of Th2, Th17, and cytotoxic effector molecules. Single-cell transcriptomics revealed a comprehensive absence of iNKT17 cells, a substantial reduction in iNKT2 cells, and an increase in iNKT1 cells in GATA-3-deficient thymi. Differential expression analysis highlighted the regulatory role of GATA-3 in T cell activation signaling and altered expression of genes critical for iNKT cell differentiation, such as Icos, Cd127, Eomes, and Zbtb16. Notably, restoration of Icos, but not Cd127, expression could rescue iNKT cell development in GATA-3-deficient mice. In conclusion, our study demonstrates the pivotal role of GATA-3 in orchestrating iNKT cell effector lineage differentiation through the regulation of T cell activation pathways and Icos expression, providing insights into the molecular mechanisms governing iNKT cell development and function.
Collapse
Affiliation(s)
- Tzong-Shyuan Tai
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Huang-Yu Yang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Advanced Immunology Laboratory, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Wan-Chu Chuang
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Yu-Wen Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - I-Cheng Ho
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, 60 Fenwood Road, Boston, MA 02115, USA;
- Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Ching-Chung Tsai
- Department of Pediatrics, E-Da Hospital, I-Shou University, Kaohsiung City 82445, Taiwan
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Ya-Ting Chuang
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| |
Collapse
|
10
|
Kumari S, Singh B, Kureel AK, Saini S, Prakash S, Chauhan A, Kumar P, Singh K, Rai AK. Benzo[a]pyrene exposure causes exonal switch resulting in reduced surface CD5 expression in an AHR-dependent manner. Immunol Lett 2024; 267:106858. [PMID: 38631465 DOI: 10.1016/j.imlet.2024.106858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024]
Abstract
The function of CD5 protein in T cells is well documented, but regulation of its surface-level expression has yet to be fully understood. However, variation in its surface expression is associated with various immunopathological conditions and haematological malignancies. Briefly, expression of an alternate exon E1B of a human endogenous retroviruses (HERV) origin directly downregulates the conventional transcript variant (E1A), as its expression leads to the retention of the resultant protein at the intracellular level (cCD5). A separate promoter governs the expression of E1B and may be influenced by different transcription factors. Hence, we performed in silico transcription factor binding site (TFBS) analysis of the 3 kb upstream region from TSS of exon E1B and found five putative DREs (Dioxin Response elements) with good similarity scores. Further, we observed the upregulation in E1B expression after the exposure of BaP (a dioxin) and the reduction of E1A expression and their respective protein, i.e. sCD5 and cCD5. The binding of AHR at the predicted DRE sites was confirmed by ChIP qPCR and AHR specific inhibitor and gene silencing studies suggested the involvement of AHR in exonal switch. This study indicates that the polycyclic aromatic hydrocarbon decreases the sCD5 expression by upregulating alternative exon expression, which may adversely affect the overall T cell functions.
Collapse
Affiliation(s)
- Smita Kumari
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Bharat Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Amit Kumar Kureel
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Sheetal Saini
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Satya Prakash
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Aditi Chauhan
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, U.P. 226014, India
| | - Prabin Kumar
- Department of Transplant Immunology and Immunogenetics, All India Institute of medical Sciences (A.I.I.M.S.), New Delhi, 110029, India
| | - Kulwant Singh
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, U.P. 226014, India
| | - Ambak Kumar Rai
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India.
| |
Collapse
|
11
|
Steier Z, Kim EJY, Aylard DA, Robey EA. The CD4 Versus CD8 T Cell Fate Decision: A Multiomics-Informed Perspective. Annu Rev Immunol 2024; 42:235-258. [PMID: 38271641 DOI: 10.1146/annurev-immunol-083122-040929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
The choice of developing thymocytes to become CD8+ cytotoxic or CD4+ helper T cells has been intensely studied, but many of the underlying mechanisms remain to be elucidated. Recent multiomics approaches have provided much higher resolution analysis of gene expression in developing thymocytes than was previously achievable, thereby offering a fresh perspective on this question. Focusing on our recent studies using CITE-seq (cellular indexing of transcriptomes and epitopes) analyses of mouse thymocytes, we present a detailed timeline of RNA and protein expression changes during CD8 versus CD4 T cell differentiation. We also revisit our current understanding of the links between T cell receptor signaling and expression of the lineage-defining transcription factors ThPOK and RUNX3. Finally, we propose a sequential selection model to explain the tight linkage between MHC-I versus MHC-II recognition and T cell lineage choice. This model incorporates key aspects of previously proposed kinetic signaling, instructive, and stochastic/selection models.
Collapse
Affiliation(s)
- Zoë Steier
- Department of Bioengineering and Center for Computational Biology, University of California, Berkeley, California, USA
- Graduate Program in Bioengineering, University of California, Berkeley, and University of California, San Francisco, Berkeley and San Francisco, California, USA
- Current affiliation: Institute for Medical Engineering and Science, Massachusetts Institute of Technology; Broad Institute of MIT and Harvard; and Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Esther Jeong Yoon Kim
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, California, USA;
| | - Dominik A Aylard
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, California, USA;
| | - Ellen A Robey
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, California, USA;
| |
Collapse
|
12
|
Wang T, Guo J, Liping Li, Jin Q, Zhang F, Hou B, Zhang Y, Zhou X. The histone lysine methyltransferase MLL1 regulates the activation and functional specialization of regulatory T cells. Cell Rep 2024; 43:114222. [PMID: 38735046 DOI: 10.1016/j.celrep.2024.114222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/19/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024] Open
Abstract
The activation and specialization of regulatory T cells (Tregs) are crucial for maintaining immune self-tolerance; however, the regulation of these processes by histone modifications is not fully understood. Here, we show that T cell-specific deletion of the lysine methyltransferase MLL1 results in a spontaneous lymphocyte proliferation phenotype in aged mice without disturbing the development of conventional T cells and Tregs. Treg-specific MLL1 ablation leads to a systemic autoimmune disease associated with Treg dysfunction. Moreover, RNA sequencing demonstrates that the induction of multiple genes involved in Treg activation, functional specialization, and tissue immigration is defective in MLL1-deficient Tregs. This dysregulation is associated with defects in H3K4 trimethylation at these genes' transcription start sites. Finally, using a T-bet fate-mapping mouse system, we determine that MLL1 is required to establish stable Th1-type Tregs. Thus, MLL1 is essential in optimal Treg function by providing a coordinated chromatin context for activation and specialization.
Collapse
Affiliation(s)
- Ting Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science (CAS), Beijing 100101, China; Department of Savaid Medical School, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Guo
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science (CAS), Beijing 100101, China
| | - Liping Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science (CAS), Beijing 100101, China; Department of Savaid Medical School, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Qiuzhu Jin
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science (CAS), Beijing 100101, China; Department of Savaid Medical School, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Fuping Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science (CAS), Beijing 100101, China; Department of Savaid Medical School, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Baidong Hou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, China
| | - Yan Zhang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Xuyu Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science (CAS), Beijing 100101, China; Department of Savaid Medical School, University of the Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
13
|
Mélique S, Vadel A, Rouquié N, Yang C, Bories C, Cotineau C, Saoudi A, Fazilleau N, Lesourne R. THEMIS promotes T cell development and maintenance by rising the signaling threshold of the inhibitory receptor BTLA. Proc Natl Acad Sci U S A 2024; 121:e2318773121. [PMID: 38713628 PMCID: PMC11098085 DOI: 10.1073/pnas.2318773121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/12/2024] [Indexed: 05/09/2024] Open
Abstract
The current paradigm about the function of T cell immune checkpoints is that these receptors switch on inhibitory signals upon cognate ligand interaction. We here revisit this simple switch model and provide evidence that the T cell lineage protein THEMIS enhances the signaling threshold at which the immune checkpoint BTLA (B- and T-lymphocyte attenuator) represses T cell responses. THEMIS is recruited to the cytoplasmic domain of BTLA and blocks its signaling capacity by promoting/stabilizing the oxidation of the catalytic cysteine of the tyrosine phosphatase SHP-1. In contrast, THEMIS has no detectable effect on signaling pathways regulated by PD-1 (Programmed cell death protein 1), which depend mainly on the tyrosine phosphatase SHP-2. BTLA inhibitory signaling is tuned according to the THEMIS expression level, making CD8+ T cells more resistant to BTLA-mediated inhibition than CD4+ T cells. In the absence of THEMIS, the signaling capacity of BTLA is exacerbated, which results in the attenuation of signals driven by the T cell antigen receptor and by receptors for IL-2 and IL-15, consequently hampering thymocyte positive selection and peripheral CD8+ T cell maintenance. By characterizing the pivotal role of THEMIS in restricting the transmission of BTLA signals, our study suggests that immune checkpoint operability is conditioned by intracellular signal attenuators.
Collapse
Affiliation(s)
- Suzanne Mélique
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Aurélie Vadel
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Nelly Rouquié
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Cui Yang
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Cyrielle Bories
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Coline Cotineau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Abdelhadi Saoudi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Nicolas Fazilleau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Renaud Lesourne
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| |
Collapse
|
14
|
Kumar A, Ye C, Nkansah A, Decoville T, Fogo GM, Sajjakulnukit P, Reynolds MB, Zhang L, Quaye O, Seo YA, Sanderson TH, Lyssiotis CA, Chang CH. Iron regulates the quiescence of naive CD4 T cells by controlling mitochondria and cellular metabolism. Proc Natl Acad Sci U S A 2024; 121:e2318420121. [PMID: 38621136 PMCID: PMC11047099 DOI: 10.1073/pnas.2318420121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/14/2024] [Indexed: 04/17/2024] Open
Abstract
In response to an immune challenge, naive T cells undergo a transition from a quiescent to an activated state acquiring the effector function. Concurrently, these T cells reprogram cellular metabolism, which is regulated by iron. We and others have shown that iron homeostasis controls proliferation and mitochondrial function, but the underlying mechanisms are poorly understood. Given that iron derived from heme makes up a large portion of the cellular iron pool, we investigated iron homeostasis in T cells using mice with a T cell-specific deletion of the heme exporter, FLVCR1 [referred to as knockout (KO)]. Our finding revealed that maintaining heme and iron homeostasis is essential to keep naive T cells in a quiescent state. KO naive CD4 T cells exhibited an iron-overloaded phenotype, with increased spontaneous proliferation and hyperactive mitochondria. This was evidenced by reduced IL-7R and IL-15R levels but increased CD5 and Nur77 expression. Upon activation, however, KO CD4 T cells have defects in proliferation, IL-2 production, and mitochondrial functions. Iron-overloaded CD4 T cells failed to induce mitochondrial iron and exhibited more fragmented mitochondria after activation, making them susceptible to ferroptosis. Iron overload also led to inefficient glycolysis and glutaminolysis but heightened activity in the hexosamine biosynthetic pathway. Overall, these findings highlight the essential role of iron in controlling mitochondrial function and cellular metabolism in naive CD4 T cells, critical for maintaining their quiescent state.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Chenxian Ye
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Afia Nkansah
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, AccraG4522, Ghana
| | - Thomas Decoville
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Garrett M. Fogo
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI48109
| | - Peter Sajjakulnukit
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI48109
| | - Mack B. Reynolds
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Li Zhang
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI48109
| | - Osbourne Quaye
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, AccraG4522, Ghana
| | - Young-Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI48109
| | - Thomas H. Sanderson
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI48109
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Costas A. Lyssiotis
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI48109
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI48109
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Cheong-Hee Chang
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| |
Collapse
|
15
|
Ham SD, Abraham MN, Deutschman CS, Taylor MD. Single-cell RNA sequencing reveals Immune Education promotes T cell survival in mice subjected to the cecal ligation and puncture sepsis model. Front Immunol 2024; 15:1366955. [PMID: 38562928 PMCID: PMC10982361 DOI: 10.3389/fimmu.2024.1366955] [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: 01/07/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Background Individual T cell responses vary significantly based on the microenvironment present at the time of immune response and on prior induced T cell memory. While the cecal ligation and puncture (CLP) model is the most commonly used murine sepsis model, the contribution of diverse T cell responses has not been explored. We defined T cell subset responses to CLP using single-cell RNA sequencing and examined the effects of prior induced T cell memory (Immune Education) on these responses. We hypothesized that Immune Education prior to CLP would alter T cell responses at the single cell level at a single, early post-CLP time point. Methods Splenic T cells were isolated from C57BL/6 mice. Four cohorts were studied: Control, Immune-Educated, CLP, and Immune-Educated CLP. At age 8 weeks, Immune-Educated and Immune-Educated CLP mice received anti-CD3ϵ antibody; Control and CLP mice were administered an isotype control. CLP (two punctures with a 22-gauge needle) was performed at 12-13 weeks of life. Mice were sacrificed at baseline or 24-hours post-CLP. Unsupervised clustering of the transcriptome library identified six distinct T cell subsets: quiescent naïve CD4+, primed naïve CD4+, memory CD4+, naïve CD8+, activated CD8+, and CD8+ cytotoxic T cell subsets. T cell subset specific gene set enrichment analysis and Hurdle analysis for differentially expressed genes (DEGs) were performed. Results T cell responses to CLP were not uniform - subsets of activated and suppressed T cells were identified. Immune Education augmented specific T cell subsets and led to genomic signatures favoring T cell survival in unoperated and CLP mice. Additionally, the combination of Immune Education and CLP effected the expression of genes related to T cell activity in ways that differed from CLP alone. Validating our finding that IL7R pathway markers were upregulated in Immune-Educated CLP mice, we found that Immune Education increased T cell surface IL7R expression in post-CLP mice. Conclusion Immune Education enhanced the expression of genes associated with T cell survival in unoperated and CLP mice. Induction of memory T cell compartments via Immune Education combined with CLP may increase the model's concordance to human sepsis.
Collapse
Affiliation(s)
- Steven D. Ham
- The Division of Critical Care Medicine, Department of Pediatrics, Cohen Children’s Medical Center/Northwell Health, New Hyde Park, NY, United States
- Sepsis Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Mabel N. Abraham
- The Division of Critical Care Medicine, Department of Pediatrics, Cohen Children’s Medical Center/Northwell Health, New Hyde Park, NY, United States
- Sepsis Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Clifford S. Deutschman
- The Division of Critical Care Medicine, Department of Pediatrics, Cohen Children’s Medical Center/Northwell Health, New Hyde Park, NY, United States
- Sepsis Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Matthew D. Taylor
- The Division of Critical Care Medicine, Department of Pediatrics, Cohen Children’s Medical Center/Northwell Health, New Hyde Park, NY, United States
- Sepsis Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| |
Collapse
|
16
|
Herr LA, Fiala GJ, Sagar, Schaffer AM, Hummel JF, Zintchenko M, Raute K, Velasco Cárdenas RMH, Heizmann B, Ebert K, Fehrenbach K, Janowska I, Chan S, Tanriver Y, Minguet S, Schamel WW. Kidins220 and Aiolos promote thymic iNKT cell development by reducing TCR signals. SCIENCE ADVANCES 2024; 10:eadj2802. [PMID: 38489359 PMCID: PMC10942104 DOI: 10.1126/sciadv.adj2802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024]
Abstract
Development of T cells is controlled by the signal strength of the TCR. The scaffold protein kinase D-interacting substrate of 220 kilodalton (Kidins220) binds to the TCR; however, its role in T cell development was unknown. Here, we show that T cell-specific Kidins220 knockout (T-KO) mice have strongly reduced invariant natural killer T (iNKT) cell numbers and modest decreases in conventional T cells. Enhanced apoptosis due to increased TCR signaling in T-KO iNKT thymocytes of developmental stages 2 and 3 shows that Kidins220 down-regulates TCR signaling at these stages. scRNA-seq indicated that the transcription factor Aiolos is down-regulated in Kidins220-deficient iNKT cells. Analysis of an Aiolos KO demonstrated that Aiolos is a downstream effector of Kidins220 during iNKT cell development. In the periphery, T-KO iNKT cells show reduced TCR signaling upon stimulation with α-galactosylceramide, suggesting that Kidins220 promotes TCR signaling in peripheral iNKT cells. Thus, Kidins220 reduces or promotes signaling dependent on the iNKT cell developmental stage.
Collapse
Affiliation(s)
- Laurenz A. Herr
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Gina J. Fiala
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Sagar
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna-Maria Schaffer
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Jonas F. Hummel
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
| | - Marina Zintchenko
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Katrin Raute
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Rubí M.-H. Velasco Cárdenas
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Beate Heizmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Karolina Ebert
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
| | - Kerstin Fehrenbach
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Iga Janowska
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Susan Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Yakup Tanriver
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
- Department of Medicine IV: Nephrology and Primary Care, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susana Minguet
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Wolfgang W. Schamel
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| |
Collapse
|
17
|
Caruso B, Weeder BR, Thompson RF, Moran AE. PD-1 Limits IL-2 Production and Thymic Regulatory T Cell Development. Immunohorizons 2024; 8:281-294. [PMID: 38551395 PMCID: PMC10985057 DOI: 10.4049/immunohorizons.2300079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 04/02/2024] Open
Abstract
Inhibitory proteins, such as programmed cell death protein 1 (PD-1), have been studied extensively in peripheral T cell responses to foreign Ags, self-Ags, and neoantigens. Notably, these proteins are first expressed during T cell development in the thymus. Reports suggest that PD-1 limits regulatory T cell (Treg) development, but the mechanism by which PD-1 exerts this function remains unknown. The present study expands the evaluation of murine PD-1 and its ligands in the thymus, demonstrating that some of the highest expressers of PD-1 and programmed death-ligand 1 are agonist selected cells. Surprisingly, we reveal a selective role for PD-1 in regulating the developmental niche only for Tregs because other agonist selected cell populations, such as NK T cells, remain unchanged. We also ruled out PD-1 as a regulator of proliferation or cell death of agonist selected Tregs and further demonstrated that PD-1-deficient Tregs have reduced TCR signaling. Unexpectedly, the data suggest that PD-1-deficient thymocytes produce elevated levels of IL-2, a Treg niche-limiting cytokine. Collectively, these data suggest a novel role for PD-1 in regulating IL-2 production and the concurrent agonist selection of murine thymic Tregs. This observation has implications for the use of checkpoint blockade in the context of cancer and infection.
Collapse
Affiliation(s)
- Breanna Caruso
- Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
| | - Benjamin R. Weeder
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Reid F. Thompson
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR
- Department of Radiation Medicine, Oregon Health and Science University, Portland, OR
- Veterans Affairs Portland Health Care System, Portland,OR
| | - Amy E. Moran
- Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| |
Collapse
|
18
|
Alotaibi FM, Min WP, Koropatnick J. CD5 blockade, a novel immune checkpoint inhibitor, enhances T cell anti-tumour immunity and delays tumour growth in mice harbouring poorly immunogenic 4T1 breast tumour homografts. Front Immunol 2024; 15:1256766. [PMID: 38487537 PMCID: PMC10937348 DOI: 10.3389/fimmu.2024.1256766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 02/07/2024] [Indexed: 03/17/2024] Open
Abstract
CD5 is a member of the scavenger receptor cysteine-rich superfamily that is expressed on T cells and a subset of B cells (B1a) cell and can regulate the T cell receptor signaling pathway. Blocking CD5 function may have therapeutic potential in treatment of cancer by enhancing cytotoxic T lymphocyte recognition and ablation of tumour cells. The effect of administering an anti-CD5 antibody to block or reduce CD5 function as an immune checkpoint blockade to enhance T cell anti-tumour activation and function in vivo has not been explored. Here we challenged mice with poorly immunogenic 4T1 breast tumour cells and tested whether treatment with anti-CD5 monoclonal antibodies (MAb) in vivo could enhance non-malignant T cell anti-tumour immunity and reduce tumour growth. Treatment with anti-CD5 MAb resulted in an increased fraction of CD8+ T cells compared to CD4+ T cell in draining lymph nodes and the tumour microenvironment. In addition, it increased activation and effector function of T cells isolated from spleens, draining lymph nodes, and 4T1 tumours. Furthermore, tumour growth was delayed in mice treated with anti-CD5 MAb. These data suggest that use of anti-CD5 MAb as an immune checkpoint blockade can both enhance activation of T cells in response to poorly immunogenic antigens and reduce tumour growth in vivo. Exploration of anti-CD5 therapies in treatment of cancer, alone and in combination with other immune therapeutic drugs, is warranted.
Collapse
Affiliation(s)
- Faizah M. Alotaibi
- College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Alahsa, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Wei-Ping Min
- Department of Oncology, The University of Western Ontario, London, ON, Canada
| | - James Koropatnick
- Department of Oncology, The University of Western Ontario, London, ON, Canada
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
- Cancer Research Laboratory Program, London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
19
|
Jin Y, Yuan H, Mehta I, Ezenwa O, Morel PA. Alternatively Spliced Variants of Murine CD247 Influence T Cell Development and Activation, Revealing the Importance of the CD3ζ C-Terminal Region. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:541-550. [PMID: 38117282 PMCID: PMC10872740 DOI: 10.4049/jimmunol.2300511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023]
Abstract
CD247, also known as CD3ζ, is a crucial signaling molecule that transduces signals delivered by TCR through its three ITAMs. CD3ζ is required for successful thymocyte development. Three additional alternatively spliced variants of murine CD247 have been described, that is, CD3ι, CD3θ, and CD3η, that differ from CD3ζ in the C terminus such that the third ITAM is lost. Previous studies demonstrated defects in T cell development in mice expressing CD3η, but the TCR signaling pathways affected by CD3η and the impacts of the CD3ι and CD3θ on T cell development were not explored. In this study, we used a retrovirus-mediated gene transfer technique to express these three isoforms individually and examined the roles of them on T cell development and activation. Rag1-/- mice reconstituted with CD3θ-expressing bone marrow failed to develop mature T cells. CD3ι-expressing T cells exhibited similar development and activation as cells expressing CD3ζ. In contrast, thymic development was severely impaired in CD3η-reconstituted mice. Single-positive but not double-positive CD3η-expressing thymocytes had reduced TCR expression, and CD5 expression was decreased at the double-positive stage, suggesting a defect in positive selection. Peripheral CD3η-expressing T cells had expanded CD44hi populations and upregulation of exhaustion markers seen by flow cytometry and RNA sequencing analysis. Analysis of early signaling events demonstrated significantly reduced activation of both the PLCγ1 and Akt/mTOR signaling pathways. There was also a reduction in the frequency of activation of CD3η-expressing T cells. These studies reveal the importance of the CD3ζ C-terminal region in T cell development and activation.
Collapse
Affiliation(s)
- Ye Jin
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Huijuan Yuan
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh PA
| | - Isha Mehta
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ogechukwu Ezenwa
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Penelope A Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
20
|
Santamaria JC, Vuillier S, Galindo-Albarrán AO, Castan S, Detraves C, Joffre OP, Romagnoli P, van Meerwijk JPM. The type 1 diabetes susceptibility locus Idd5 favours robust neonatal development of highly autoreactive regulatory T cells in the NOD mouse. Front Immunol 2024; 15:1358459. [PMID: 38404576 PMCID: PMC10884962 DOI: 10.3389/fimmu.2024.1358459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/18/2024] [Indexed: 02/27/2024] Open
Abstract
Regulatory T lymphocytes expressing the transcription factor Foxp3 (Tregs) play an important role in the prevention of autoimmune diseases and other immunopathologies. Aberrations in Treg-mediated immunosuppression are therefore thought to be involved in the development of autoimmune pathologies, but few have been documented. Recent reports indicated a central role for Tregs developing during the neonatal period in the prevention of autoimmune pathology. We therefore investigated the development of Tregs in neonatal NOD mice, an important animal model for autoimmune type 1 diabetes. Surprisingly, we found that, as compared with seven other commonly studied inbred mouse strains, in neonatal NOD mice, exceptionally large proportions of developing Tregs express high levels of GITR and PD-1. The latter phenotype was previously associated with high Treg autoreactivity in C57BL/6 mice, which we here confirm for NOD animals. The proportions of newly developing GITRhighPD-1+ Tregs rapidly drop during the first week of age. A genome-wide genetic screen indicated the involvement of several diabetes susceptibility loci in this trait. Analysis of a congenic mouse strain confirmed that Idd5 contributes to the genetic control of GITRhighPD-1+ Treg development in neonates. Our data thus demonstrate an intriguing and paradoxical correlation between an idiosyncrasy in Treg development in NOD mice and their susceptibility to type 1 diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Joost P. M. van Meerwijk
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Institut National de la santé et de la recherche médicale (Inserm) UMR1291 – Centre national de la recherche scientifique (CNRS) UMR5051 – University Toulouse III, Toulouse, France
| |
Collapse
|
21
|
Eggert J, Zinzow-Kramer WM, Hu Y, Kolawole EM, Tsai YL, Weiss A, Evavold BD, Salaita K, Scharer CD, Au-Yeung BB. Cbl-b mitigates the responsiveness of naive CD8 + T cells that experience extensive tonic T cell receptor signaling. Sci Signal 2024; 17:eadh0439. [PMID: 38319998 PMCID: PMC10897907 DOI: 10.1126/scisignal.adh0439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 01/18/2024] [Indexed: 02/08/2024]
Abstract
Naive T cells experience tonic T cell receptor (TCR) signaling in response to self-antigens presented by major histocompatibility complex (MHC) in secondary lymphoid organs. We investigated how relatively weak or strong tonic TCR signals influence naive CD8+ T cell responses to stimulation with foreign antigens. The heterogeneous expression of Nur77-GFP, a transgenic reporter of tonic TCR signaling, in naive CD8+ T cells suggests variable intensities or durations of tonic TCR signaling. Although the expression of genes associated with acutely stimulated T cells was increased in Nur77-GFPHI cells, these cells were hyporesponsive to agonist TCR stimulation compared with Nur77-GFPLO cells. This hyporesponsiveness manifested as diminished activation marker expression and decreased secretion of IFN-γ and IL-2. The protein abundance of the ubiquitin ligase Cbl-b, a negative regulator of TCR signaling, was greater in Nur77-GFPHI cells than in Nur77-GFPLO cells, and Cbl-b deficiency partially restored the responsiveness of Nur77-GFPHI cells. Our data suggest that the cumulative effects of previously experienced tonic TCR signaling recalibrate naive CD8+ T cell responsiveness. These changes include gene expression changes and negative regulation partially dependent on Cbl-b. This cell-intrinsic negative feedback loop may enable the immune system to restrain naive CD8+ T cells with higher self-reactivity.
Collapse
Affiliation(s)
- Joel Eggert
- Division of Immunology, Lowance Center for Human Immunology, Department of Medicine, Emory University; Atlanta, 30322, USA
| | - Wendy M. Zinzow-Kramer
- Division of Immunology, Lowance Center for Human Immunology, Department of Medicine, Emory University; Atlanta, 30322, USA
| | - Yuesong Hu
- Department of Chemistry, Emory University; Atlanta, 30322, USA
| | - Elizabeth M. Kolawole
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, 84112, USA
| | - Yuan-Li Tsai
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Departments of Medicine and of Microbiology and Immunology, University of California, San Francisco; San Francisco, 94143, USA
| | - Arthur Weiss
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Departments of Medicine and of Microbiology and Immunology, University of California, San Francisco; San Francisco, 94143, USA
| | - Brian D. Evavold
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, 84112, USA
| | - Khalid Salaita
- Department of Chemistry, Emory University; Atlanta, 30322, USA
| | | | - Byron B. Au-Yeung
- Division of Immunology, Lowance Center for Human Immunology, Department of Medicine, Emory University; Atlanta, 30322, USA
| |
Collapse
|
22
|
Jamaleddine H, Rogers D, Perreault G, Postat J, Patel D, Mandl JN, Khadra A. Chronic infection control relies on T cells with lower foreign antigen binding strength generated by N-nucleotide diversity. PLoS Biol 2024; 22:e3002465. [PMID: 38300945 PMCID: PMC10833529 DOI: 10.1371/journal.pbio.3002465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 12/08/2023] [Indexed: 02/03/2024] Open
Abstract
The breadth of pathogens to which T cells can respond is determined by the T cell receptors (TCRs) present in an individual's repertoire. Although more than 90% of the sequence diversity among TCRs is generated by terminal deoxynucleotidyl transferase (TdT)-mediated N-nucleotide addition during V(D)J recombination, the benefit of TdT-altered TCRs remains unclear. Here, we computationally and experimentally investigated whether TCRs with higher N-nucleotide diversity via TdT make distinct contributions to acute or chronic pathogen control specifically through the inclusion of TCRs with lower antigen binding strengths (i.e., lower reactivity to peptide-major histocompatibility complex (pMHC)). When T cells with high pMHC reactivity have a greater propensity to become functionally exhausted than those of low pMHC reactivity, our computational model predicts a shift toward T cells with low pMHC reactivity over time during chronic, but not acute, infections. This TCR-affinity shift is critical, as the elimination of T cells with lower pMHC reactivity in silico substantially increased the time to clear a chronic infection, while acute infection control remained largely unchanged. Corroborating an affinity-centric benefit for TCR diversification via TdT, we found evidence that TdT-deficient TCR repertoires possess fewer T cells with weaker pMHC binding strengths in vivo and showed that TdT-deficient mice infected with a chronic, but not an acute, viral pathogen led to protracted viral clearance. In contrast, in the case of a chronic fungal pathogen where T cells fail to clear the infection, both our computational model and experimental data showed that TdT-diversified TCR repertoires conferred no additional protection to the hosts. Taken together, our in silico and in vivo data suggest that TdT-mediated TCR diversity is of particular benefit for the eventual resolution of prolonged pathogen replication through the inclusion of TCRs with lower foreign antigen binding strengths.
Collapse
Affiliation(s)
| | - Dakota Rogers
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, Montreal, Quebec, Canada
| | - Geneviève Perreault
- McGill University Research Centre on Complex Traits, Montreal, Quebec, Canada
| | - Jérémy Postat
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, Montreal, Quebec, Canada
| | - Dhanesh Patel
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, Montreal, Quebec, Canada
| | - Judith N. Mandl
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
23
|
Perez C, Plaza-Rojas L, Boucher JC, Nagy MZ, Kostenko E, Prajapati K, Burke B, Reyes MD, Austin AL, Zhang S, Le PT, Guevara-Patino JA. NKG2D receptor signaling shapes T cell thymic education. J Leukoc Biol 2024; 115:306-321. [PMID: 37949818 DOI: 10.1093/jleuko/qiad130] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 08/11/2023] [Accepted: 09/30/2023] [Indexed: 11/12/2023] Open
Abstract
The role of natural killer group 2D (NKG2D) in peripheral T cells as a costimulatory receptor is well established. However, its contribution to T cell thymic education and functional imprint is unknown. Here, we report significant changes in development, receptor signaling, transcriptional program, and function in T cells from mice lacking NKG2D signaling. In C57BL/6 (B6) and OT-I mice, we found that NKG2D deficiency results in Vβ chain usage changes and stagnation of the double-positive stage in thymic T cell development. We found that the expression of CD5 and CD45 in thymocytes from NKG2D deficient mice were reduced, indicating a direct influence of NKG2D on the strength of T cell receptor (TCR) signaling during the developmental stage of T cells. Depicting the functional consequences of NKG2D, peripheral OT-I NKG2D-deficient cells were unresponsive to ovalbumin peptide stimulation. Paradoxically, while αCD3/CD28 agonist antibodies led to phenotypic T cell activation, their ability to produce cytokines remained severely compromised. We found that OT-I NKG2D-deficient cells activate STAT5 in response to interleukin-15 but were unable to phosphorylate ERK or S6 upon TCR engagement, underpinning a defect in TCR signaling. Finally, we showed that NKG2D is expressed in mouse and human thymic T cells at the double-negative stage, suggesting an evolutionarily conserved function during T cell development. The data presented in this study indicate that NKG2D impacts thymic T cell development at a fundamental level by reducing the TCR threshold and affecting the functional imprint of the thymic progeny. In summary, understanding the impact of NKG2D on thymic T cell development and TCR signaling contributes to our knowledge of immune system regulation, immune dysregulation, and the design of immunotherapies.
Collapse
Affiliation(s)
- Cynthia Perez
- Department of Cancer Biology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
| | - Lourdes Plaza-Rojas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, United States
| | - Justin C Boucher
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, United States
| | - Mate Z Nagy
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, United States
| | - Elena Kostenko
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, United States
| | - Kushal Prajapati
- Department of Cancer Biology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
| | - Brianna Burke
- Department of Cancer Biology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
| | - Michael Delos Reyes
- Department of Cancer Biology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
| | - Anna L Austin
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, United States
| | - Shubin Zhang
- Department of Cancer Biology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
- Department of Microbiology and Immunology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
| | - Phong T Le
- Department of Cancer Biology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
- Department of Microbiology and Immunology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
| | - José A Guevara-Patino
- Department of Cancer Biology, Loyola University Chicago, 2160 S. First Ave, Maywood, IL 60153, United States
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, United States
| |
Collapse
|
24
|
Zamorano B, Atik H, Brooks WH, Milhes J, Renaudineau Y. Infections and B1 Cells. INFECTION AND AUTOIMMUNITY 2024:91-114. [DOI: 10.1016/b978-0-323-99130-8.00019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
25
|
Lui VG, Hoenig M, Cabrera-Martinez B, Baxter RM, Garcia-Perez JE, Bailey O, Acharya A, Lundquist K, Capera J, Matusewicz P, Hartl FA, D’Abramo M, Alba J, Jacobsen EM, Niewolik D, Lorenz M, Pannicke U, Schulz AS, Debatin KM, Schamel WW, Minguet S, Gumbart JC, Dustin ML, Cambier JC, Schwarz K, Hsieh EW. A partial human LCK defect causes a T cell immunodeficiency with intestinal inflammation. J Exp Med 2024; 221:e20230927. [PMID: 37962568 PMCID: PMC10644909 DOI: 10.1084/jem.20230927] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/09/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Lymphocyte-specific protein tyrosine kinase (LCK) is essential for T cell antigen receptor (TCR)-mediated signal transduction. Here, we report two siblings homozygous for a novel LCK variant (c.1318C>T; P440S) characterized by T cell lymphopenia with skewed memory phenotype, infant-onset recurrent infections, failure to thrive, and protracted diarrhea. The patients' T cells show residual TCR signal transduction and proliferation following anti-CD3/CD28 and phytohemagglutinin (PHA) stimulation. We demonstrate in mouse models that complete (Lck-/-) versus partial (LckP440S/P440S) loss-of-function LCK causes disease with differing phenotypes. While both Lck-/- and LckP440S/P440S mice exhibit arrested thymic T cell development and profound T cell lymphopenia, only LckP440S/P440S mice show residual T cell proliferation, cytokine production, and intestinal inflammation. Furthermore, the intestinal disease in the LckP440S/P440S mice is prevented by CD4+ T cell depletion or regulatory T cell transfer. These findings demonstrate that P440S LCK spares sufficient T cell function to allow the maturation of some conventional T cells but not regulatory T cells-leading to intestinal inflammation.
Collapse
Affiliation(s)
- Victor G. Lui
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Manfred Hoenig
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Berenice Cabrera-Martinez
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ryan M. Baxter
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Josselyn E. Garcia-Perez
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Olivia Bailey
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Atanu Acharya
- School of Physics, Georgia Institute of Technology, Atlanta, GA, USA
- BioInspired Syracuse and Department of Chemistry, Syracuse University, Syracuse, NY, USA
| | - Karl Lundquist
- School of Physics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jesusa Capera
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Paul Matusewicz
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University, Freiburg, Germany
| | - Frederike A. Hartl
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University, Freiburg, Germany
| | - Marco D’Abramo
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Josephine Alba
- Department of Biology, Université de Fribourg, Fribourg, Switzerland
| | | | - Doris Niewolik
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Myriam Lorenz
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Ulrich Pannicke
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Ansgar S. Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | | | - Wolfgang W. Schamel
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University, Freiburg, Germany
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University, Freiburg, Germany
| | - James C. Gumbart
- School of Physics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael L. Dustin
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - John C. Cambier
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Human Immunology and Immunotherapy Initiative, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Klaus Schwarz
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg-Hessen, Ulm, Germany
| | - Elena W.Y. Hsieh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Human Immunology and Immunotherapy Initiative, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Department of Pediatrics, Section of Allergy and Immunology, Children’s Hospital Colorado, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| |
Collapse
|
26
|
Kowarsch F, Maurer-Granofszky M, Weijler L, Wödlinger M, Reiter M, Schumich A, Feuerstein T, Sala S, Nováková M, Faggin G, Gaipa G, Hrusak O, Buldini B, Dworzak MN. FCM marker importance for MRD assessment in T-cell acute lymphoblastic leukemia: An AIEOP-BFM-ALL-FLOW study group report. Cytometry A 2024; 105:24-35. [PMID: 37776305 DOI: 10.1002/cyto.a.24805] [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: 11/21/2022] [Revised: 08/07/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
T-lineage acute lymphoblastic leukemia (T-ALL) accounts for about 15% of pediatric and about 25% of adult ALL cases. Minimal/measurable residual disease (MRD) assessed by flow cytometry (FCM) is an important prognostic indicator for risk stratification. In order to assess the MRD a limited number of antibodies directed against the most discriminative antigens must be selected. We propose a pipeline for evaluating the influence of different markers for cell population classification in FCM data. We use linear support vector machine, fitted to each sample individually to avoid issues with patient and laboratory variations. The best separating hyperplane direction as well as the influence of omitting specific markers is considered. Ninety-one bone marrow samples of 43 pediatric T-ALL patients from five reference laboratories were analyzed by FCM regarding marker importance for blast cell identification using combinations of eight different markers. For all laboratories, CD48 and CD99 were among the top three markers with strongest contribution to the optimal hyperplane, measured by median separating hyperplane coefficient size for all samples per center and time point (diagnosis, Day 15, Day 33). Based on the available limited set tested (CD3, CD4, CD5, CD7, CD8, CD45, CD48, CD99), our findings prove that CD48 and CD99 are useful markers for MRD monitoring in T-ALL. The proposed pipeline can be applied for evaluation of other marker combinations in the future.
Collapse
Affiliation(s)
- Florian Kowarsch
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Margarita Maurer-Granofszky
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik GmbH, Vienna, Austria
| | - Lisa Weijler
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Matthias Wödlinger
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Michael Reiter
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Angela Schumich
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Tamar Feuerstein
- The Rina Zaizov Division of Pediatric Hematology-Oncology, Schneider's Children's Medical Center, Petah Tikva, Israel
| | - Simona Sala
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Michaela Nováková
- Department of Pediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Giovanni Faggin
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, Padova, Italy
| | - Giuseppe Gaipa
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Ondrej Hrusak
- Department of Pediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Barbara Buldini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, Padova, Italy
- Advanced Diagnostics and Target Discovery in ALL, Fondazione istituto di Ricerca pediatrica Città della Speranza, Padova, Italy
| | - Michael N Dworzak
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik GmbH, Vienna, Austria
| |
Collapse
|
27
|
Textor J, Buytenhuijs F, Rogers D, Gauthier ÈM, Sultan S, Wortel IMN, Kalies K, Fähnrich A, Pagel R, Melichar HJ, Westermann J, Mandl JN. Machine learning analysis of the T cell receptor repertoire identifies sequence features of self-reactivity. Cell Syst 2023; 14:1059-1073.e5. [PMID: 38061355 DOI: 10.1016/j.cels.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/01/2023] [Accepted: 11/09/2023] [Indexed: 12/23/2023]
Abstract
The T cell receptor (TCR) determines specificity and affinity for both foreign and self-peptides presented by the major histocompatibility complex (MHC). Although the strength of TCR interactions with self-pMHC impacts T cell function, it has been challenging to identify TCR sequence features that predict T cell fate. To discern patterns distinguishing TCRs from naive CD4+ T cells with low versus high self-reactivity, we used data from 42 mice to train a machine learning (ML) algorithm that identifies population-level differences between TCRβ sequence sets. This approach revealed that weakly self-reactive T cell populations were enriched for longer CDR3β regions and acidic amino acids. We tested our ML predictions of self-reactivity using retrogenic mice with fixed TCRβ sequences. Extrapolating our analyses to independent datasets, we predicted high self-reactivity for regulatory T cells and slightly reduced self-reactivity for T cells responding to chronic infections. Our analyses suggest a potential trade-off between TCR repertoire diversity and self-reactivity. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Johannes Textor
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, the Netherlands; Medical BioSciences, Radboudumc, Nijmegen 6525 GA, the Netherlands.
| | - Franka Buytenhuijs
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, the Netherlands
| | - Dakota Rogers
- Department of Physiology, McGill University, Montreal, QC H3G 0B1, Canada; McGill Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada
| | - Ève Mallet Gauthier
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Shabaz Sultan
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, the Netherlands; Medical BioSciences, Radboudumc, Nijmegen 6525 GA, the Netherlands
| | - Inge M N Wortel
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, the Netherlands; Medical BioSciences, Radboudumc, Nijmegen 6525 GA, the Netherlands
| | - Kathrin Kalies
- Institut für Anatomie, Universität zu Lübeck, 23562 Lübeck, Germany
| | - Anke Fähnrich
- Institut für Anatomie, Universität zu Lübeck, 23562 Lübeck, Germany
| | - René Pagel
- Institut für Anatomie, Universität zu Lübeck, 23562 Lübeck, Germany
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada; Department of Medicine, Université de Montréal, Montréal, QC H1T 2M4, Canada; Department of Microbiology & Immunology, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | | | - Judith N Mandl
- Department of Physiology, McGill University, Montreal, QC H3G 0B1, Canada; Department of Microbiology & Immunology, McGill University, Montreal, QC H3A 1A3, Canada; McGill Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada.
| |
Collapse
|
28
|
Wood BL, Devidas M, Summers RJ, Chen Z, Asselin B, Rabin KR, Zweidler-McKay PA, Winick NJ, Borowitz MJ, Carroll WL, Raetz EA, Loh ML, Hunger SP, Dunsmore KP, Teachey DT, Winter SS. Prognostic significance of ETP phenotype and minimal residual disease in T-ALL: a Children's Oncology Group study. Blood 2023; 142:2069-2078. [PMID: 37556734 PMCID: PMC10862241 DOI: 10.1182/blood.2023020678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023] Open
Abstract
The early thymic precursor (ETP) immunophenotype was previously reported to confer poor outcome in T-cell acute lymphoblastic leukemia (T-ALL). Between 2009 and 2014, 1256 newly diagnosed children and young adults enrolled in Children's Oncology Group (COG) AALL0434 were assessed for ETP status and minimal residual disease (MRD) using flow cytometry at a central reference laboratory. The subject phenotypes were categorized as ETP (n = 145; 11.5%), near-ETP (n = 209; 16.7%), or non-ETP (n = 902; 71.8%). Despite higher rates of induction failure for ETP (6.2%) and near-ETP (6.2%) than non-ETP (1.2%; P < .0001), all 3 groups showed excellent 5-year event-free survival (EFS) and overall survival (OS): ETP (80.4% ± 3.9% and 86.8 ± 3.4%, respectively), near-ETP (81.1% ± 3.3% and 89.6% ± 2.6%, respectively), and non-ETP (85.3% ± 1.4% and 90.0% ± 1.2%, respectively; P = .1679 and P = .3297, respectively). There was no difference in EFS or OS for subjects with a day-29 MRD <0.01% vs 0.01% to 0.1%. However, day-29 MRD ≥0.1% was associated with inferior EFS and OS for patients with near-ETP and non-ETP, but not for those with ETP. For subjects with day-29 MRD ≥1%, end-consolidation MRD ≥0.01% was a striking predictor of inferior EFS (80.9% ± 4.1% vs 52.4% ± 8.1%, respectively; P = .0001). When considered as a single variable, subjects with all 3 T-ALL phenotypes had similar outcomes and subjects with persistent postinduction disease had inferior outcomes, regardless of their ETP phenotype. This clinical trial was registered at AALL0434 as #NCT00408005.
Collapse
Affiliation(s)
- Brent L. Wood
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, Saint Jude Children's Research Hospital, Memphis, TN
| | - Ryan J. Summers
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA
- Department of Pediatrics, Emory University, Atlanta, GA
| | - Zhiguo Chen
- Department of Biostatistics, University of Florida, Gainesville, FL
| | - Barbara Asselin
- Department of Pediatrics, University of Rochester, Rochester, NY
| | - Karen R. Rabin
- Pediatric Hematology/Oncology, Baylor College of Medicine/Dan L Duncan Comprehensive Cancer Center, Houston, TX
| | | | - Naomi J. Winick
- Pediatric Hematology and Oncology, UT Southwestern/Simmons Cancer Center-Dallas, Dallas, TX
| | - Michael J. Borowitz
- Department of Pathology, Johns Hopkins University/Sidney Kimmel Cancer Center, Baltimore, MD
| | - William L. Carroll
- Department of Pediatrics and Pathology, Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, Hassenfeld Children's Center, New York, NY
| | - Elizabeth A. Raetz
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Mignon L. Loh
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA
- Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA
| | - Stephen P. Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Kimberly P. Dunsmore
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - David T. Teachey
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Stuart S. Winter
- Cancer and Blood Disorders Program, Children’s Minnesota, Minneapolis, MN
| |
Collapse
|
29
|
Tang J, Zhao X. Chimeric antigen receptor T cells march into T cell malignancies. J Cancer Res Clin Oncol 2023; 149:13459-13475. [PMID: 37468610 DOI: 10.1007/s00432-023-05148-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023]
Abstract
T cell malignancies represent a diverse collection of leukemia/lymphoma conditions in humans arising from aberrant T cells. Such malignancies are often associated with poor clinical prognoses, cancer relapse, as well as progressive resistance to anti-cancer treatments. While chimeric antigen receptor (CAR) T cell immunotherapy has emerged as a revolutionary treatment strategy that is highly effective for treating B cell malignancies, its application as a treatment for T cell malignancies remains to be better explored. Furthermore, the effectiveness of CAR-T treatment in T cell malignancies is significantly influenced by the quality of contamination-free CAR-T cells during the manufacturing process, as well as by multiple characteristics of such malignancies, including the sharing of antigens across normal and malignant T cells, fratricide, and T cell aplasia. In this review, we provide a detailed account of the current developments in the clinical application of CAR-T therapy to treat T cell malignancies, offer strategies for addressing current challenges, and outline a roadmap toward its effective implementation as a broad treatment option for this condition.
Collapse
Affiliation(s)
- Jie Tang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xudong Zhao
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Smid AI, Garforth SJ, Obaid MS, Bollons HR, James JR. Pre-T cell receptor localization and trafficking are independent of its signaling. J Cell Biol 2023; 222:e202212106. [PMID: 37516909 PMCID: PMC10373305 DOI: 10.1083/jcb.202212106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/08/2023] [Accepted: 07/06/2023] [Indexed: 07/31/2023] Open
Abstract
Expression of the pre-T cell receptor (preTCR) is an important checkpoint during the development of T cells, an essential cell type of our adaptive immune system. The preTCR complex is only transiently expressed and rapidly internalized in developing T cells and is thought to signal in a ligand-independent manner. However, identifying a mechanistic basis for these unique features of the preTCR compared with the final TCR complex has been confounded by the concomitant signaling that is normally present. Thus, we have reconstituted preTCR expression in non-immune cells to uncouple receptor trafficking dynamics from its associated signaling. We find that all the defining features of the preTCR are intrinsic properties of the receptor itself, driven by exposure of an extracellular hydrophobic region, and are not the consequence of receptor activation. Finally, we show that transitory preTCR cell surface expression can sustain tonic signaling in the absence of ligand binding, suggesting how the preTCR can nonetheless drive αβTCR lineage commitment.
Collapse
Affiliation(s)
- Andrei I. Smid
- Molecular Immunity Unit, Department of Medicine, Medical Research Council–Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Sam J. Garforth
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Maryam S. Obaid
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Hannah R. Bollons
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - John R. James
- Molecular Immunity Unit, Department of Medicine, Medical Research Council–Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
31
|
Gao Y, Li W, Wang Z, Zhang C, He Y, Liu X, Tang K, Zhang W, Long Q, Liu Y, Zhang J, Zhang B, Zhang L. SEL1L preserves CD8 + T-cell survival and homeostasis by fine-tuning PERK signaling and the IL-15 receptor-mediated mTORC1 axis. Cell Mol Immunol 2023; 20:1232-1250. [PMID: 37644166 PMCID: PMC10541435 DOI: 10.1038/s41423-023-01078-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023] Open
Abstract
SEL1L-mediated endoplasmic reticulum-associated degradation (ERAD) plays critical roles in controlling protein homeostasis by degrading misfolded or terminal unfolded proteins. However, it remains unclear how SEL1L regulates peripheral T-cell survival and homeostasis. Herein, we found that SEL1L deficiency led to a greatly reduced frequency and number of mature T cells, which was further validated by adoptive transfer experiments or bone marrow chimera experiments, accompanied by the induction of multiple forms of cell death. Furthermore, SEL1L deficiency selectively disrupted naïve CD8+ T-cell homeostasis, as indicated by the severe loss of the naïve T-cell subset but an increase in the memory T-cell subset. We also found that SEL1L deficiency fueled mTORC1/c-MYC activation and induced a metabolic shift, which was largely attributable to enhanced expression of the IL-15 receptor α and β chains. Mechanistically, single-cell transcriptomic profiling and biochemical analyses further revealed that Sel1l-/- CD8+ T cells harbored excessive ER stress, particularly aberrant activation of the PERK-ATF4-CHOP-Bim pathway, which was alleviated by supplementing IL-7 or IL-15. Importantly, PERK inhibition greatly resolved the survival defects of Sel1l-/- CD8+ T cells. In addition, IRE1α deficiency decreased mTORC1 signaling in Sel1l-/- naïve CD8+ T cells by downregulating the IL-15 receptor α chain. Altogether, these observations suggest that the ERAD adaptor molecule SEL1L acts as an important checkpoint for preserving the survival and homeostasis of peripheral T cells by regulating the PERK signaling cascade and IL-15 receptor-mediated mTORC1 axis.
Collapse
Affiliation(s)
- Yafeng Gao
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Wenhui Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Zhenghao Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shaanxi, China
| | - Yaping He
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaowei Liu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Kexin Tang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiguo Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Qiaoming Long
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, The Institute for Advanced Studies, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jinping Zhang
- Institute of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shaanxi, China.
| | - Lianjun Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
- Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
32
|
Berton RR, McGonagil PW, Jensen IJ, Ybarra TK, Bishop GA, Harty JT, Griffith TS, Badovinac VP. Sepsis leads to lasting changes in phenotype and function of naïve CD8 T cells. PLoS Pathog 2023; 19:e1011720. [PMID: 37824591 PMCID: PMC10597476 DOI: 10.1371/journal.ppat.1011720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 10/24/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Sepsis, an amplified immune response to systemic infection, is characterized by a transient cytokine storm followed by chronic immune dysfunction. Consequently, sepsis survivors are highly susceptible to newly introduced infections, suggesting sepsis can influence the function and composition of the naïve CD8 T cell pool and resulting pathogen-induced primary CD8 T cell responses. Here, we explored the extent to which sepsis induces phenotypic and functional changes within the naïve CD8 T cell pool. To interrogate this, the cecal ligation and puncture (CLP) mouse model of polymicrobial sepsis was used. In normal, non-septic mice, we show type-I interferon (IFN I)-mediated signaling plays an important role in driving the phenotypic and functional heterogeneity in the naïve CD8 T cell compartment leading to increased representation of Ly6C+ naïve CD8 T cells. In response to viral infection after sepsis resolution, naïve Ly6C+ CD8 T cells generated more primary effector and memory CD8 T cells with slower conversion to a central memory CD8 T cell phenotype (Tcm) than Ly6C- naïve CD8 T cells. Importantly, as a potent inducer of cytokine storm and IFN I production, sepsis leads to increased representation of Ly6C+ naïve CD8 T cells that maintained their heightened ability to respond (i.e., effector and memory CD8 T cell accumulation and cytokine production) to primary LCMV infection. Lastly, longitudinal analyses of peripheral blood samples obtained from septic patients revealed profound changes in CD8 T cell subset composition and frequency compared to healthy controls. Thus, sepsis has the capacity to alter the composition of naïve CD8 T cells, directly influencing primary CD8 T cell responses to newly introduced infections.
Collapse
Affiliation(s)
- Roger R. Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Patrick W. McGonagil
- Department of Surgery, University of Iowa, Iowa City, Iowa, United States of America
| | - Isaac J. Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York City, New York, United States of America
| | - Tiffany K. Ybarra
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Gail A. Bishop
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - John T. Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota, United States of America
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
33
|
David NA, Lee RD, LaRue RS, Joo S, Farrar MA. Nuclear corepressors NCOR1 and NCOR2 entrain thymocyte signaling, selection, and emigration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559810. [PMID: 37808728 PMCID: PMC10557688 DOI: 10.1101/2023.09.27.559810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
T cell development proceeds via discrete stages that require both gene induction and gene repression. Transcription factors direct gene repression by associating with corepressor complexes containing chromatin-remodeling enzymes; the corepressors NCOR1 and NCOR2 recruit histone deacetylases to these complexes to silence transcription of target genes. Earlier work identified the importance of NCOR1 in promoting the survival of positively-selected thymocytes. Here, we used flow cytometry and single-cell RNA sequencing to identify a broader role for NCOR1 and NCOR2 in regulating thymocyte development. Using Cd4-cre mice, we found that conditional deletion of NCOR2 had no effect on thymocyte development, whereas conditional deletion of NCOR1 had a modest effect. In contrast, Cd4-cre x Ncor1f/f x Ncor2f/f mice exhibited a significant block in thymocyte development at the DP to SP transition. Combined NCOR1/2 deletion resulted in increased signaling through the T cell receptor, ultimately resulting in elevated BIM expression and increased negative selection. The NF-κB, NUR77, and MAPK signaling pathways were also upregulated in the absence of NCOR1/2, contributing to altered CD4/CD8 lineage commitment, TCR rearrangement, and thymocyte emigration. Taken together, our data identify multiple critical roles for the combined action of NCOR1 and NCOR2 over the course of thymocyte development.
Collapse
Affiliation(s)
- Natalie A David
- Center for Immunology, Masonic Cancer Center, Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Robin D Lee
- Center for Immunology, Masonic Cancer Center, Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Rebecca S LaRue
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455
| | - Sookyong Joo
- Center for Immunology, Masonic Cancer Center, Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Michael A Farrar
- Center for Immunology, Masonic Cancer Center, Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
34
|
Dart RJ, Zlatareva I, Vantourout P, Theodoridis E, Amar A, Kannambath S, East P, Recaldin T, Mansfield JC, Lamb CA, Parkes M, Irving PM, Prescott NJ, Hayday AC. Conserved γδ T cell selection by BTNL proteins limits progression of human inflammatory bowel disease. Science 2023; 381:eadh0301. [PMID: 37708268 PMCID: PMC7615126 DOI: 10.1126/science.adh0301] [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: 02/09/2023] [Accepted: 07/19/2023] [Indexed: 09/16/2023]
Abstract
Murine intraepithelial γδ T cells include distinct tissue-protective cells selected by epithelial butyrophilin-like (BTNL) heteromers. To determine whether this biology is conserved in humans, we characterized the colonic γδ T cell compartment, identifying a diverse repertoire that includes a phenotypically distinct subset coexpressing T cell receptor Vγ4 and the epithelium-binding integrin CD103. This subset was disproportionately diminished and dysregulated in inflammatory bowel disease, whereas on-treatment CD103+γδ T cell restoration was associated with sustained inflammatory bowel disease remission. Moreover, CD103+Vγ4+cell dysregulation and loss were also displayed by humans with germline BTNL3/BTNL8 hypomorphism, which we identified as a risk factor for penetrating Crohn's disease (CD). Thus, BTNL-dependent selection and/or maintenance of distinct tissue-intrinsic γδ T cells appears to be an evolutionarily conserved axis limiting the progression of a complex, multifactorial, tissue-damaging disease of increasing global incidence.
Collapse
Affiliation(s)
- Robin J Dart
- Peter Gorer Dept of Immunobiology, King’s College London at Guy’s Hospital Campus, London, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
- Department of Gastroenterology, Guy’s and St Thomas’ Foundation Trust, London, UK
| | - Iva Zlatareva
- Peter Gorer Dept of Immunobiology, King’s College London at Guy’s Hospital Campus, London, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Pierre Vantourout
- Peter Gorer Dept of Immunobiology, King’s College London at Guy’s Hospital Campus, London, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Efstathios Theodoridis
- Peter Gorer Dept of Immunobiology, King’s College London at Guy’s Hospital Campus, London, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Ariella Amar
- Department of Medical and Molecular Genetics, King’s College London, London, UK
| | | | - Philip East
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | | | - John C Mansfield
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Christopher A Lamb
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Miles Parkes
- Department of Medicine, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
| | - Peter M Irving
- Department of Gastroenterology, Guy’s and St Thomas’ Foundation Trust, London, UK
| | - Natalie J Prescott
- Department of Medical and Molecular Genetics, King’s College London, London, UK
| | - Adrian C Hayday
- Peter Gorer Dept of Immunobiology, King’s College London at Guy’s Hospital Campus, London, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
35
|
Hiratsuka T, Ito S, Sakai R, Yokose T, Endo T, Daigo Y, Miyagi Y, Tsuruyama T. Proteome analysis of CD5-positive diffuse large B cell lymphoma FFPE tissue reveals downregulation of DDX3X, DNAJB1, and B cell receptor signaling pathway proteins including BTK and Immunoglobulins. Clin Proteomics 2023; 20:36. [PMID: 37705009 PMCID: PMC10498596 DOI: 10.1186/s12014-023-09422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/25/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND The molecular pathology of diffuse large B cell lymphoma (DLBCL) has been extensively studied. Among DLBCL subtypes, the prognosis of CD5-positive DLBCL is worse than that of CD5-negative DLBCL, considering the central nervous system relapse and poor response to R-CHOP therapy. However, the molecular mechanisms underlying the tumorigenesis and progression of CD5-positive DLBCL remain unknown. METHODS To identify molecular markers that can be targeted for treating DLBCL, a proteomic study was performed using liquid chromatography-mass spectrometry with chemically pretreated formalin-fixed paraffin-embedded specimens from CD5-positive (n = 5) and CD5-negative DLBCL patients (n = 6). RESULTS Twenty-one proteins showed significant downregulation in CD5-positive DLBCL compared to CD5-negative DLBCL. Principal component analysis of protein expression profiling in CD5-positive and CD5-negative DLBCL revealed that DNAJB1, DDX3X, and BTK, which is one of the B cell phenotypic proteins, were the most significantly downregulated proteins and served as biomarkers that distinguished both groups. Additionally, a set of immunoglobulins, including IgG4, exhibited significant downregulation. Immunohistochemistry analysis for BTK demonstrated reduced staining in CD5-positive DLBCL compared to CD5-negative DLBCL. CONCLUSIONS In conclusion, DNAJB1 and DDX3X, BTK, and a set of immunoglobulins are promising biomarkers. Probably, the suppression of BCR signaling is the unique phenotype of CD5-positive DLBCL. This formalin-fixed paraffin-embedded (FFPE)-based profiling may help to develop novel therapeutic molecularly targeted drugs for treating DLBCL.
Collapse
Affiliation(s)
- Takuya Hiratsuka
- Department of Drug Discovery Medicine, Pathology Division, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rika Sakai
- Department of Oncology, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tatsuya Endo
- Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan
| | - Yataro Daigo
- Department of Medical Oncology, Cancer Center, and Center for Advanced Medicine Against Cancer, Shiga University of Medical Science, Otsu, Japan
- Center for Antibody and Vaccine Therapy, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tatsuaki Tsuruyama
- Department of Drug Discovery Medicine, Pathology Division, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan.
- Tazuke-Kofukai Medical Institute Kitano Hospital, Ogimachi, Osaka, Japan.
| |
Collapse
|
36
|
Frech M, Danzer H, Uchil P, Azizov V, Schmid E, Schälter F, Dürholz K, Mauro D, Rauber S, Muñoz L, Taher L, Ciccia F, Schober K, Irla M, Sarter K, Schett G, Zaiss MM. Butyrophilin 2a2 (Btn2a2) expression on thymic epithelial cells promotes central T cell tolerance and prevents autoimmune disease. J Autoimmun 2023; 139:103071. [PMID: 37356345 DOI: 10.1016/j.jaut.2023.103071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 06/27/2023]
Abstract
Butyrophilins are surface receptors belonging to the immunoglobulin superfamily. While several members of the butyrophilin family have been implicated in the development of unconventional T cells, butyrophilin 2a2 (Btn2a2) has been shown to inhibit conventional T cell activation. Here, we demonstrate that in steady state, the primary source of Btn2a2 are thymic epithelial cells (TEC). Absence of Btn2a2 alters thymic T cell maturation and bypasses central tolerance mechanisms. Furthermore, Btn2a2-/- mice develop spontaneous autoimmunity resembling human primary Sjögren's Syndrome (pSS), including formation of tertiary lymphoid structures (TLS) in target organs. Ligation of Btn2a2 on developing thymocytes is associated with reduced TCR signaling and CD5 levels, while absence of Btn2a2 results in increased TCR signaling and CD5 levels. These results define a novel role for Btn2a2 in promoting central tolerance by modulating TCR signaling strength and indicate a potential mechanism of pSS development.
Collapse
Affiliation(s)
- Michael Frech
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Heike Danzer
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Pooja Uchil
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Vugar Azizov
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Eva Schmid
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Fabian Schälter
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kerstin Dürholz
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Daniele Mauro
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria
| | - Simon Rauber
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luis Muñoz
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Leila Taher
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria
| | - Francesco Ciccia
- Dipartimento di Medicina di Precisione, University Della Campania L. Vanvitelli, Naples, Italy
| | - Kilian Schober
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Magali Irla
- CNRS, INSERM, Centre D'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
| | - Kerstin Sarter
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mario M Zaiss
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universiät Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
37
|
Kumari S, Sahu S, Singh B, Gupta S, Kureel AK, Srivastava A, Rikhari D, Srivastava S, Rai AK. HIF-1α regulates the expression of the non-conventional isoform of the cd5 gene in T cells under the hypoxic condition: A potential mechanism for CD5 neg/low phenotype of infiltrating cells in solid tumors. Cell Immunol 2023; 391-392:104755. [PMID: 37544247 DOI: 10.1016/j.cellimm.2023.104755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/08/2023]
Abstract
CD5, a T-cell receptor (TCR) negative regulator, is reduced on the surface of CD8+ lymphocytes in the tumor microenvironment (TME). Reduced surface CD5 expression (sCD5) occurs due to the preferential transcription of HERV-E derived exon E1B, i.e., anon-conventional formofthe cd5gene instead of its conventional exon E1A. A tumor employs several mechanisms to evade anti-tumor response, and hypoxia is one such mechanism that prevails in the TME and modulates the infiltrated T lymphocytes. We identified hypoxia response elements (HREs) upstream of E1B. We showed binding of HIF-1α onto these HREs and increased E1B mRNA expression in hypoxic T cells. This results in decreased sCD5 expression and increased cytoplasmic accumulation in T cells. We also validated our study in a solid tumor, i.e., colorectal cancer (CRC) patient samples. This hypoxia-driven mechanism reduces the surface CD5 expression on infiltrated T-cells in solid tumors.
Collapse
Affiliation(s)
- Smita Kumari
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Srishti Sahu
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Bharat Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Swarnima Gupta
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Amit Kumar Kureel
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Ankit Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Deeksha Rikhari
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Sameer Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Ambak Kumar Rai
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India.
| |
Collapse
|
38
|
Kim YJ, Rho KN, Jeong S, Lee GW, Kim HO, Cho HJ, Bae WK, Oh IJ, Lee SW, Cho JH. CD5 Expression Dynamically Changes During the Differentiation of Human CD8 + T Cells Predicting Clinical Response to Immunotherapy. Immune Netw 2023; 23:e35. [PMID: 37670812 PMCID: PMC10475823 DOI: 10.4110/in.2023.23.e35] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/07/2023] Open
Abstract
Defining the molecular dynamics associated with T cell differentiation enhances our understanding of T cell biology and opens up new possibilities for clinical implications. In this study, we investigated the dynamics of CD5 expression in CD8+ T cell differentiation and explored its potential clinical uses. Using PBMCs from 29 healthy donors, we observed a stepwise decrease in CD5 expression as CD8+ T cells progressed through the differentiation stages. Interestingly, we found that CD5 expression was initially upregulated in response to T cell receptor stimulation, but diminished as the cells underwent proliferation, potentially explaining the differentiation-associated CD5 downregulation. Based on the proliferation-dependent downregulation of CD5, we hypothesized that relative CD5 expression could serve as a marker to distinguish the heterogeneous CD8+ T cell population based on their proliferation history. In support of this, we demonstrated that effector memory CD8+ T cells with higher CD5 expression exhibited phenotypic and functional characteristics resembling less differentiated cells compared to those with lower CD5 expression. Furthermore, in the retrospective analysis of PBMCs from 30 non-small cell lung cancer patients, we found that patients with higher CD5 expression in effector memory T cells displayed CD8+ T cells with a phenotype closer to the less differentiated cells, leading to favorable clinical outcomes in response to immune checkpoint inhibitor (ICI) therapy. These findings highlight the dynamics of CD5 expression as an indicator of CD8+ T cell differentiation status, and have implications for the development of predictive biomarker for ICI therapy.
Collapse
Affiliation(s)
- Young Ju Kim
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Kyung Na Rho
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Saei Jeong
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Gil-Woo Lee
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
| | | | - Hyun-Ju Cho
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Hwasun 58128, Korea
| | - Woo Kyun Bae
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Hwasun 58128, Korea
| | - In-Jae Oh
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Hwasun 58128, Korea
| | - Sung-Woo Lee
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Jae-Ho Cho
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun 58128, Korea
| |
Collapse
|
39
|
Afroz S, Bartolo L, Su LF. Pre-existing T Cell Memory to Novel Pathogens. Immunohorizons 2023; 7:543-553. [PMID: 37436166 PMCID: PMC10587503 DOI: 10.4049/immunohorizons.2200003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
Immunological experiences lead to the development of specific T and B cell memory, which readies the host for a later pathogen rechallenge. Currently, immunological memory is best understood as a linear process whereby memory responses are generated by and directed against the same pathogen. However, numerous studies have identified memory cells that target pathogens in unexposed individuals. How "pre-existing memory" forms and impacts the outcome of infection remains unclear. In this review, we discuss differences in the composition of baseline T cell repertoire in mice and humans, factors that influence pre-existing immune states, and recent literature on their functional significance. We summarize current knowledge on the roles of pre-existing T cells in homeostasis and perturbation and their impacts on health and disease.
Collapse
Affiliation(s)
- Sumbul Afroz
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA
| | - Laurent Bartolo
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA
| | - Laura F. Su
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| |
Collapse
|
40
|
Vicente MM, Alves I, Fernandes Â, Dias AM, Santos-Pereira B, Pérez-Anton E, Santos S, Yang T, Correia A, Münster-Kühnel A, Almeida ARM, Ravens S, Rabinovich GA, Vilanova M, Sousa AE, Pinho SS. Mannosylated glycans impair normal T-cell development by reprogramming commitment and repertoire diversity. Cell Mol Immunol 2023:10.1038/s41423-023-01052-7. [PMID: 37344746 PMCID: PMC10387478 DOI: 10.1038/s41423-023-01052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 05/29/2023] [Indexed: 06/23/2023] Open
Abstract
T-cell development ensures the formation of diverse repertoires of T-cell receptors (TCRs) that recognize a variety of antigens. Glycosylation is a major posttranslational modification present in virtually all cells, including T-lymphocytes, that regulates activity/functions. Although these structures are known to be involved in TCR-selection in DP thymocytes, it is unclear how glycans regulate other thymic development processes and how they influence susceptibility to disease. Here, we discovered stage-specific glycome compositions during T-cell development in human and murine thymocytes, as well as dynamic alterations. After restricting the N-glycosylation profile of thymocytes to high-mannose structures, using specific glycoengineered mice (Rag1CreMgat1fl/fl), we showed remarkable defects in key developmental checkpoints, including ß-selection, regulatory T-cell generation and γδT-cell development, associated with increased susceptibility to colon and kidney inflammation and infection. We further demonstrated that a single N-glycan antenna (modeled in Rag1CreMgat2fl/fl mice) is the sine-qua-non condition to ensure normal development. In conclusion, we revealed that mannosylated thymocytes lead to a dysregulation in T-cell development that is associated with inflammation susceptibility.
Collapse
Affiliation(s)
- Manuel M Vicente
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Graduate Program in Areas of Basic and Applied Biology (GABBA), ICBAS, University of Porto, Porto, Portugal
| | - Inês Alves
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Ângela Fernandes
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Ana M Dias
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Beatriz Santos-Pereira
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Elena Pérez-Anton
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Sofia Santos
- Nephrology Department, Centro Hospitalar e Universitário do Porto, Porto, Portugal
| | - Tao Yang
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Alexandra Correia
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Anja Münster-Kühnel
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Afonso R M Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires, Argentina
- Laboratorio de Inmuno-oncología Translacional, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Manuel Vilanova
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Ana E Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Salomé S Pinho
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
- Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
41
|
Istomine R, Al-Aubodah TA, Alvarez F, Smith JA, Wagner C, Piccirillo CA. The eIF4EBP-eIF4E axis regulates CD4 + T cell differentiation through modulation of T cell activation and metabolism. iScience 2023; 26:106683. [PMID: 37187701 PMCID: PMC10176268 DOI: 10.1016/j.isci.2023.106683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/27/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
CD4+ T cells are critical for adaptive immunity, differentiating into distinct effector and regulatory subsets. Although the transcriptional programs underlying their differentiation are known, recent research has highlighted the importance of mRNA translation in determining protein abundance. We previously conducted genome-wide analysis of translation in CD4+ T cells revealing distinct translational signatures distinguishing these subsets, identifying eIF4E as a central differentially translated transcript. As eIF4E is vital for eukaryotic translation, we examined how altered eIF4E activity affected T cell function using mice lacking eIF4E-binding proteins (BP-/-). BP-/- effector T cells showed elevated Th1 responses ex vivo and upon viral challenge with enhanced Th1 differentiation observed in vitro. This was accompanied by increased TCR activation and elevated glycolytic activity. This study highlights how regulating T cell-intrinsic eIF4E activity can influence T cell activation and differentiation, suggesting the eIF4EBP-eIF4E axis as a potential therapeutic target for controlling aberrant T cell responses.
Collapse
Affiliation(s)
- Roman Istomine
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC H4A 3J1, Canada
| | - Tho-Alfakar Al-Aubodah
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC H4A 3J1, Canada
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC H4A 3J1, Canada
| | - Jacob A. Smith
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carston Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC H4A 3J1, Canada
- Corresponding author
| |
Collapse
|
42
|
Lee V, Rodriguez DM, Ganci NK, Zeng S, Ai J, Chao JL, Walker MT, Miller CH, Klawon DEJ, Schoenbach MH, Kennedy DE, Maienschein-Cline M, Socci ND, Clark MR, Savage PA. The endogenous repertoire harbors self-reactive CD4 + T cell clones that adopt a follicular helper T cell-like phenotype at steady state. Nat Immunol 2023; 24:487-500. [PMID: 36759711 PMCID: PMC9992328 DOI: 10.1038/s41590-023-01425-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/04/2023] [Indexed: 02/11/2023]
Abstract
The T cell repertoire of healthy mice and humans harbors self-reactive CD4+ conventional T (Tconv) cells capable of inducing autoimmunity. Using T cell receptor profiling paired with in vivo clonal analysis of T cell differentiation, we identified Tconv cell clones that are recurrently enriched in non-lymphoid organs following ablation of Foxp3+ regulatory T (Treg) cells. A subset of these clones was highly proliferative in the lymphoid organs at steady state and exhibited overt reactivity to self-ligands displayed by dendritic cells, yet were not purged by clonal deletion. These clones spontaneously adopted numerous hallmarks of follicular helper T (TFH) cells, including expression of Bcl6 and PD-1, exhibited an elevated propensity to localize within B cell follicles at steady state, and produced interferon-γ in non-lymphoid organs following sustained Treg cell depletion. Our work identifies a naturally occurring population of self-reactive TFH-like cells and delineates a previously unappreciated fate for self-specific Tconv cells.
Collapse
Affiliation(s)
- Victoria Lee
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Donald M Rodriguez
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Nicole K Ganci
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Sharon Zeng
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Junting Ai
- Section of Rheumatology, Department of Medicine and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Jaime L Chao
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Matthew T Walker
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Christine H Miller
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - David E J Klawon
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | | | - Domenick E Kennedy
- Section of Rheumatology, Department of Medicine and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
- Drug Discovery Science and Technology, AbbVie, North Chicago, IL, USA
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Nicholas D Socci
- Bioinformatics Core, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Marcus R Clark
- Section of Rheumatology, Department of Medicine and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Peter A Savage
- Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
43
|
He M, Roussak K, Ma F, Borcherding N, Garin V, White M, Schutt C, Jensen TI, Zhao Y, Iberg CA, Shah K, Bhatia H, Korenfeld D, Dinkel S, Gray J, Antonova AU, Ferris S, Donermeyer D, Arlehamn CL, Gubin MM, Luo J, Gorvel L, Pellegrini M, Sette A, Tung T, Bak R, Modlin RL, Fields RC, Schreiber RD, Allen PM, Klechevsky E. CD5 expression by dendritic cells directs T cell immunity and sustains immunotherapy responses. Science 2023; 379:eabg2752. [PMID: 36795805 PMCID: PMC10424698 DOI: 10.1126/science.abg2752] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 01/17/2023] [Indexed: 02/18/2023]
Abstract
The induction of proinflammatory T cells by dendritic cell (DC) subtypes is critical for antitumor responses and effective immune checkpoint blockade (ICB) therapy. Here, we show that human CD1c+CD5+ DCs are reduced in melanoma-affected lymph nodes, with CD5 expression on DCs correlating with patient survival. Activating CD5 on DCs enhanced T cell priming and improved survival after ICB therapy. CD5+ DC numbers increased during ICB therapy, and low interleukin-6 (IL-6) concentrations promoted their de novo differentiation. Mechanistically, CD5 expression by DCs was required to generate optimally protective CD5hi T helper and CD8+ T cells; further, deletion of CD5 from T cells dampened tumor elimination in response to ICB therapy in vivo. Thus, CD5+ DCs are an essential component of optimal ICB therapy.
Collapse
Affiliation(s)
- Mingyu He
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kate Roussak
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Feiyang Ma
- Molecular Cell and Developmental Biology at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vince Garin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mike White
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Charles Schutt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Trine I. Jensen
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Yun Zhao
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Courtney A. Iberg
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kairav Shah
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Himanshi Bhatia
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel Korenfeld
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sabrina Dinkel
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Judah Gray
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stephen Ferris
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Donermeyer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cecilia Lindestam Arlehamn
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Matthew M. Gubin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jingqin Luo
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laurent Gorvel
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matteo Pellegrini
- Molecular Cell and Developmental Biology at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Thomas Tung
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rasmus Bak
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
- Aarhus Institute of Advanced Studies (AIAS), Aarhus University, 8000 Aarhus C, Denmark
| | - Robert L. Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Ryan C. Fields
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paul M. Allen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eynav Klechevsky
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
44
|
Lee SW, Lee GW, Kim HO, Cho JH. Shaping Heterogeneity of Naive CD8 + T Cell Pools. Immune Netw 2023; 23:e2. [PMID: 36911807 PMCID: PMC9995989 DOI: 10.4110/in.2023.23.e2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/12/2023] [Accepted: 02/12/2023] [Indexed: 03/07/2023] Open
Abstract
Immune diversification helps protect the host against a myriad of pathogens. CD8+ T cells are essential adaptive immune cells that inhibit the spread of pathogens by inducing apoptosis in infected host cells, ultimately ensuring complete elimination of infectious pathogens and suppressing disease development. Accordingly, numerous studies have been conducted to elucidate the mechanisms underlying CD8+ T cell activation, proliferation, and differentiation into effector and memory cells, and to identify various intrinsic and extrinsic factors regulating these processes. The current knowledge accumulated through these studies has led to a huge breakthrough in understanding the existence of heterogeneity in CD8+ T cell populations during immune response and the principles underlying this heterogeneity. As the heterogeneity in effector/memory phases has been extensively reviewed elsewhere, in the current review, we focus on CD8+ T cells in a "naïve" state, introducing recent studies dealing with the heterogeneity of naive CD8+ T cells and discussing the factors that contribute to such heterogeneity. We also discuss how this heterogeneity contributes to establishing the immense complexity of antigen-specific CD8+ T cell response.
Collapse
Affiliation(s)
- Sung-Woo Lee
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea.,Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Gil-Woo Lee
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea.,Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
| | | | - Jae-Ho Cho
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Korea.,Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea.,BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun 58128, Korea
| |
Collapse
|
45
|
Beckmann D, Langnaese K, Gottfried A, Hradsky J, Tedford K, Tiwari N, Thomas U, Fischer KD, Korthals M. Ca 2+ Homeostasis by Plasma Membrane Ca 2+ ATPase (PMCA) 1 Is Essential for the Development of DP Thymocytes. Int J Mol Sci 2023; 24:ijms24021442. [PMID: 36674959 PMCID: PMC9865543 DOI: 10.3390/ijms24021442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
The strength of Ca2+ signaling is a hallmark of T cell activation, yet the role of Ca2+ homeostasis in developing T cells before expressing a mature T cell receptor is poorly understood. We aimed to unveil specific functions of the two plasma membrane Ca2+ ATPases expressed in T cells, PMCA1 and PMCA4. On a transcriptional and protein level we found that PMCA4 was expressed at low levels in CD4-CD8- double negative (DN) thymocytes and was even downregulated in subsequent stages while PMCA1 was present throughout development and upregulated in CD4+CD8+ double positive (DP) thymocytes. Mice with a targeted deletion of Pmca1 in DN3 thymocytes had an almost complete block of DP thymocyte development with an accumulation of DN4 thymocytes but severely reduced numbers of CD8+ immature single positive (ISP) thymocytes. The DN4 thymocytes of these mice showed strongly elevated basal cytosolic Ca2+ levels and a pre-mature CD5 expression, but in contrast to the DP thymocytes they were only mildly prone to apoptosis. Surprisingly, mice with a germline deletion of Pmca4 did not show any signs of altered progression through the developmental thymocyte stages, nor altered Ca2+ homeostasis throughout this process. PMCA1 is, therefore, non-redundant in keeping cellular Ca2+ levels low in the early thymocyte development required for the DN to DP transition.
Collapse
Affiliation(s)
- David Beckmann
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Kristina Langnaese
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Anna Gottfried
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Johannes Hradsky
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Kerry Tedford
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Nikhil Tiwari
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39120 Magdeburg, Germany
| | - Ulrich Thomas
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39120 Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
- Correspondence:
| | - Mark Korthals
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
46
|
This S, Rogers D, Mallet Gauthier È, Mandl JN, Melichar HJ. What's self got to do with it: Sources of heterogeneity among naive T cells. Semin Immunol 2023; 65:101702. [PMID: 36463711 DOI: 10.1016/j.smim.2022.101702] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/04/2022]
Abstract
There is a long-standing assumption that naive CD4+ and CD8+ T cells are largely homogeneous populations despite the extraordinary diversity of their T cell receptors (TCR). The self-immunopeptidome plays a key role in the selection of the naive T cell repertoire in the thymus, and self-peptides are also an important driver of differences between individual naive T cells with regard to their subsequent functional contributions to an immune response. Accumulating evidence suggests that as early as the β-selection stage of T cell development, when only one of the recombined chains of the mature TCR is expressed, signaling thresholds may be established for positive selection of immature thymocytes. Stochastic encounters subsequently made with self-ligands during positive selection in the thymus imprint functional biases that a T cell will carry with it throughout its lifetime, although ongoing interactions with self in the periphery ensure a level of plasticity in the gene expression wiring of naive T cells. Identifying the sources of heterogeneity in the naive T cell population and which functional attributes of T cells can be modulated through post-thymic interventions versus those that are fixed during T cell development, could enable us to better select or generate T cells with particular traits to improve the efficacy of T cell therapies.
Collapse
Affiliation(s)
- Sébastien This
- Department of Microbiology, Infectious Disease, and Immunology, Université de Montréal, Montreal, Canada; Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Canada
| | - Dakota Rogers
- Department of Physiology and McGill Research Centre on Complex Traits, McGill University, Montreal, Canada
| | - Ève Mallet Gauthier
- Department of Microbiology, Infectious Disease, and Immunology, Université de Montréal, Montreal, Canada; Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Canada
| | - Judith N Mandl
- Department of Physiology and McGill Research Centre on Complex Traits, McGill University, Montreal, Canada.
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada.
| |
Collapse
|
47
|
Werlen G, Li ML, Tottone L, da Silva-Diz V, Su X, Herranz D, Jacinto E. Dietary glucosamine overcomes the defects in αβ-T cell ontogeny caused by the loss of de novo hexosamine biosynthesis. Nat Commun 2022; 13:7404. [PMID: 36456551 PMCID: PMC9715696 DOI: 10.1038/s41467-022-35014-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
T cell development requires the coordinated rearrangement of T cell receptor (TCR) gene segments and the expression of either αβ or γδ TCR. However, whether and how de novo synthesis of nutrients contributes to thymocyte commitment to either lineage remains unclear. Here, we find that T cell-specific deficiency in glutamine:fructose-6-phosphate aminotransferase 1 (GFAT1), the rate-limiting enzyme of the de novo hexosamine biosynthesis pathway (dn-HBP), attenuates hexosamine levels, blunts N-glycosylation of TCRβ chains, reduces surface expression of key developmental receptors, thus impairing αβ-T cell ontogeny. GFAT1 deficiency triggers defects in N-glycans, increases the unfolded protein response, and elevates γδ-T cell numbers despite reducing γδ-TCR diversity. Enhancing TCR expression or PI3K/Akt signaling does not reverse developmental defects. Instead, dietary supplementation with the salvage metabolite, glucosamine, and an α-ketoglutarate analogue partially restores αβ-T cell development in GFAT1T-/- mice, while fully rescuing it in ex vivo fetal thymic organ cultures. Thus, dn-HBP fulfils, while salvage nutrients partially satisfy, the elevated demand for hexosamines during early T cell development.
Collapse
Affiliation(s)
- Guy Werlen
- grid.430387.b0000 0004 1936 8796Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State Univ. of New Jersey, Piscataway, NJ 08854 USA
| | - Mei-Ling Li
- grid.430387.b0000 0004 1936 8796Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State Univ. of New Jersey, Piscataway, NJ 08854 USA
| | - Luca Tottone
- grid.430387.b0000 0004 1936 8796Dept. of Pharmacology and Pediatrics, Robert Wood Johnson Medical School, and Rutgers Cancer Institute of New Jersey, Rutgers, The State Univ. of New Jersey, New Brunswick, NJ 08901 USA ,grid.26790.3a0000 0004 1936 8606Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, FL Miami, 33136 USA
| | - Victoria da Silva-Diz
- grid.430387.b0000 0004 1936 8796Dept. of Pharmacology and Pediatrics, Robert Wood Johnson Medical School, and Rutgers Cancer Institute of New Jersey, Rutgers, The State Univ. of New Jersey, New Brunswick, NJ 08901 USA
| | - Xiaoyang Su
- grid.430387.b0000 0004 1936 8796Dept. of Medicine, Div. of Endocrinology, Child Health Inst. of New Jersey, Rutgers, The State Univ. of New Jersey, New Brunswick, NJ 08901 USA
| | - Daniel Herranz
- grid.430387.b0000 0004 1936 8796Dept. of Pharmacology and Pediatrics, Robert Wood Johnson Medical School, and Rutgers Cancer Institute of New Jersey, Rutgers, The State Univ. of New Jersey, New Brunswick, NJ 08901 USA
| | - Estela Jacinto
- grid.430387.b0000 0004 1936 8796Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State Univ. of New Jersey, Piscataway, NJ 08854 USA
| |
Collapse
|
48
|
Kim CY, Parrish HL, Kuhns MS. The TCR Cα Domain Regulates Responses to Self-pMHC Class II. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2033-2041. [PMID: 36426940 PMCID: PMC9643626 DOI: 10.4049/jimmunol.2200377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/05/2022] [Indexed: 12/31/2022]
Abstract
T cells play a central role in adaptive immunity by recognizing peptide Ags presented by MHC molecules (pMHC) via their clonotypic TCRs. αβTCRs are heterodimers, consisting of TCRα and TCRβ subunits that are composed of variable (Vα, Vβ) and constant (Cα, Cβ) domains. Whereas the Vα, Vβ, and Cβ domains adopt typical Ig folds in the extracellular space, the Cα domain lacks a top β sheet and instead has two loosely associated top strands (C- and F-strands) on its surface. Previous results suggest that this unique Ig-like fold mediates homotypic TCR interactions and influences signaling in vitro. To better understand why evolution has selected this unique structure, we asked, what is the fitness cost for development and function of mouse CD4+ T cells bearing a mutation in the Cα C-strand? In both TCR retrogenic and transgenic mice we observed increased single-positive thymocytes bearing mutant TCRs compared with those expressing wild-type TCRs. Furthermore, our analysis of mutant TCR transgenic mice revealed an increase in naive CD4+ T cells experiencing strong tonic TCR signals, increased homeostatic survival, and increased recruitment of responders to cognate pMHC class II upon immunization compared with the wild-type. The mutation did not, however, overtly impact CD4+ T cell proliferation or differentiation after immunization. We interpret these data as evidence that the unique Cα domain has evolved to fine-tune TCR signaling, particularly in response to weak interactions with self-pMHC class II.
Collapse
Affiliation(s)
- Caleb Y. Kim
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ
| | - Heather L. Parrish
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ
| | - Michael S. Kuhns
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ
- Genetics Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ
- The BIO-5 Institute, The University of Arizona, Tucson, AZ
- The University of Arizona Cancer Center, Tucson, AZ
- The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, AZ
| |
Collapse
|
49
|
Xu L, Wei C, Chen Y, Wu Y, Shou X, Chen W, Lu D, Sun H, Li W, Yu B, Wang X, Zhang X, Yu Y, Lei Z, Tang R, Zhu J, Li Y, Lu L, Zhou H, Zhou S, Su C, Chen X. IL-33 induces thymic involution-associated naive T cell aging and impairs host control of severe infection. Nat Commun 2022; 13:6881. [PMID: 36371464 PMCID: PMC9653498 DOI: 10.1038/s41467-022-34660-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022] Open
Abstract
Severe infection commonly results in immunosuppression, which leads to impaired pathogen clearance or increased secondary infection in both humans and animals. However, the exact mechanisms remain poorly understood. Here, we demonstrate that IL-33 results in immunosuppression by inducing thymic involution-associated naive T cell dysfunction with aberrant expression of aging-associated genes and impairs host control of infection in mouse disease models of schistosomiasis or sepsis. Furthermore, we illustrate that IL-33 triggers the excessive generation of medullary thymic epithelial cell (mTEC) IV (thymic tuft cells) in a Pou2f3-dependent manner, as a consequence, disturbs mTEC/cortical TEC (cTEC) compartment and causes thymic involution during severe infection. More importantly, IL-33 deficiency, the anti-IL-33 neutralizing antibody treatment, or IL-33 receptor ST2 deficient thymus transplantation rescues T cell immunity to better control infection in mice. Our findings not only uncover a link between severe infection-induced IL-33 and thymic involution-mediated naive T cell aging, but also suggest that targeting IL-33 or ST2 is a promising strategy to rejuvenate T cell immunity to better control severe infection.
Collapse
Affiliation(s)
- Lei Xu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Respiratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Clinical Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 P. R. China
| | - Chuan Wei
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Ying Chen
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Yue Wu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Xiaoli Shou
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Wenjie Chen
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Di Lu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Haoran Sun
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Wei Li
- grid.89957.3a0000 0000 9255 8984Department of Clinical Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 P. R. China
| | - Beibei Yu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Xiaowei Wang
- grid.452511.6Department of Blood Transfusion, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008 P. R. China
| | - Xiaojun Zhang
- grid.452511.6Imaging Center, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008 P. R. China
| | - Yanxiong Yu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Zhigang Lei
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Rui Tang
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Jifeng Zhu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Yalin Li
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Linrong Lu
- grid.13402.340000 0004 1759 700XInstitute of Immunology, School of Medicine, Zhejiang University, Hangzhou, 310058 P. R. China
| | - Hong Zhou
- grid.186775.a0000 0000 9490 772XDepartment of Cell Biology, School of Life Sciences, Anhui Medical University, Hefei, 230032 P. R. China
| | - Sha Zhou
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Chuan Su
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Xiaojun Chen
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| |
Collapse
|
50
|
Baumgarth N. Breaking the Paradigm: Selection of Self-Reactive Natural Antibodies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1621-1623. [PMID: 36253068 PMCID: PMC9586457 DOI: 10.4049/jimmunol.2200406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
This Pillars of Immunology article is a commentary on “Positive Selection of Natural Autoreactive B Cells,” a pivotal article written by K. Hayakawa, M. Asano, S. A. Shinton, M. Gui, D. Allman, C. L. Stewart, J. Silver, and R. R. Hardy, and published in Science, in 1999. https://doi.org/10.1126/science.285.5424.113.
Collapse
Affiliation(s)
- Nicole Baumgarth
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, CA
| |
Collapse
|