1
|
Gober JG, Capietto AH, Hoshyar R, Darwish M, Vandlen R, Linehan JL, Delamarre L, ElSohly AM. MHC2-SCALE enhances identification of immunogenic neoantigens. iScience 2025; 28:112212. [PMID: 40235585 PMCID: PMC11999303 DOI: 10.1016/j.isci.2025.112212] [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: 09/25/2024] [Revised: 12/13/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Recent studies suggest that CD4+ T cells can exert potent anti-tumor effects and improve immunotherapy efficacy by aiding CD8+ T cells. However, characterizing the mechanism of CD4+ T cells' anti-tumor activity has been challenging due to inaccurate major histocompatibility complex class II (MHC-II) peptide prediction algorithms and the lack of high-quality reagents for immune monitoring. To address this, we developed MHC2-substitution of CLIP and analytical LCMS evaluation (MHC2-SCALE), a streamlined approach combining affinity optimized class II-associated invariant chain peptide (CLIP) exchange technology, high throughput 2D-LCMS analysis, and rapid generation of peptide-bound MHC-II monomers for subsequent multimer assembly. We validated MHC-II peptide candidates predicted by the immune epitope database (IEDB) algorithm, as well as uncovered many true and immunogenic MHC-II binders that were not predicted by IEDB. Thus, MHC2-SCALE expands the opportunities for discovering, tracking, and phenotyping antigen-specific CD4+ T cells in preclinical and clinical settings, thereby improving therapies for cancer, autoimmunity, or infectious diseases.
Collapse
Affiliation(s)
- Joshua G. Gober
- Department of Protein Chemistry, Genentech Inc, South San Francisco, CA, USA
| | | | - Reyhane Hoshyar
- Cancer Immunology Department, Genentech Inc, South San Francisco, CA, USA
| | - Martine Darwish
- Department of Protein Chemistry, Genentech Inc, South San Francisco, CA, USA
| | - Richard Vandlen
- Department of Protein Chemistry, Genentech Inc, South San Francisco, CA, USA
| | | | - Lélia Delamarre
- Cancer Immunology Department, Genentech Inc, South San Francisco, CA, USA
| | - Adel M. ElSohly
- Department of Protein Chemistry, Genentech Inc, South San Francisco, CA, USA
- Department of Immunology Discovery, Genentech Inc, South San Francisco, CA, USA
| |
Collapse
|
2
|
Edmans MD, Connelley TK, Morgan S, Pediongco TJ, Jayaraman S, Juno JA, Meehan BS, Dewar PM, Maze EA, Roos EO, Paudyal B, Mak JYW, Liu L, Fairlie DP, Wang H, Corbett AJ, McCluskey J, Benedictus L, Tchilian E, Klenerman P, Eckle SBG. MAIT cell-MR1 reactivity is highly conserved across multiple divergent species. J Biol Chem 2024; 300:107338. [PMID: 38705391 PMCID: PMC11190491 DOI: 10.1016/j.jbc.2024.107338] [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: 09/10/2023] [Revised: 04/03/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are a subset of unconventional T cells that recognize small molecule metabolites presented by major histocompatibility complex class I related protein 1 (MR1), via an αβ T cell receptor (TCR). MAIT TCRs feature an essentially invariant TCR α-chain, which is highly conserved between mammals. Similarly, MR1 is the most highly conserved major histocompatibility complex-I-like molecule. This extreme conservation, including the mode of interaction between the MAIT TCR and MR1, has been shown to allow for species-mismatched reactivities unique in T cell biology, thereby allowing the use of selected species-mismatched MR1-antigen (MR1-Ag) tetramers in comparative immunology studies. However, the pattern of cross-reactivity of species-mismatched MR1-Ag tetramers in identifying MAIT cells in diverse species has not been formally assessed. We developed novel cattle and pig MR1-Ag tetramers and utilized these alongside previously developed human, mouse, and pig-tailed macaque MR1-Ag tetramers to characterize cross-species tetramer reactivities. MR1-Ag tetramers from each species identified T cell populations in distantly related species with specificity that was comparable to species-matched MR1-Ag tetramers. However, there were subtle differences in staining characteristics with practical implications for the accurate identification of MAIT cells. Pig MR1 is sufficiently conserved across species that pig MR1-Ag tetramers identified MAIT cells from the other species. However, MAIT cells in pigs were at the limits of phenotypic detection. In the absence of sheep MR1-Ag tetramers, a MAIT cell population in sheep blood was identified phenotypically, utilizing species-mismatched MR1-Ag tetramers. Collectively, our results validate the use and define the limitations of species-mismatched MR1-Ag tetramers in comparative immunology studies.
Collapse
Affiliation(s)
- Matthew D Edmans
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom; Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom.
| | - Timothy K Connelley
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Roslin, United Kingdom
| | - Sophie Morgan
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Troi J Pediongco
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Siddharth Jayaraman
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Roslin, United Kingdom
| | - Jennifer A Juno
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Bronwyn S Meehan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Phoebe M Dewar
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Emmanuel A Maze
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Eduard O Roos
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Basudev Paudyal
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Jeffrey Y W Mak
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ligong Liu
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Huimeng Wang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia; State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Alexandra J Corbett
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lindert Benedictus
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Roslin, United Kingdom; Faculty of Veterinary Medicine, Department of Population Health Sciences, Utrecht University, Utrecht, The Netherlands
| | - Elma Tchilian
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Sidonia B G Eckle
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Smith JA, Yuen BTK, Purtha W, Balolong JM, Phipps JD, Crawford F, Bluestone JA, Kappler JW, Anderson MS. Aire mediates tolerance to insulin through thymic trimming of high-affinity T cell clones. Proc Natl Acad Sci U S A 2024; 121:e2320268121. [PMID: 38709934 PMCID: PMC11098115 DOI: 10.1073/pnas.2320268121] [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: 11/27/2023] [Accepted: 03/16/2024] [Indexed: 05/08/2024] Open
Abstract
Insulin is a central autoantigen in the pathogenesis of T1D, and thymic epithelial cell expression of insulin under the control of the Autoimmune Regulator (Aire) is thought to be a key component of maintaining tolerance to insulin. In spite of this general working model, direct detection of this thymic selection on insulin-specific T cells has been somewhat elusive. Here, we used a combination of highly sensitive T cell receptor transgenic models for detecting thymic selection and sorting and sequencing of Insulin-specific CD4+ T cells from Aire-deficient mice as a strategy to further define their selection. This analysis revealed a number of unique t cell receptor (TCR) clones in Aire-deficient hosts with high affinity for insulin/major histocompatibility complex (MHC) ligands. We then modeled the thymic selection of one of these clones in Aire-deficient versus wild-type hosts and found that this model clone could escape thymic negative selection in the absence of thymic Aire. Together, these results suggest that thymic expression of insulin plays a key role in trimming and removing high-affinity insulin-specific T cells from the repertoire to help promote tolerance.
Collapse
Affiliation(s)
- Jennifer A. Smith
- Diabetes Center, University of California San Francisco, San Francisco, CA94143
| | - Benjamin T. K. Yuen
- Diabetes Center, University of California San Francisco, San Francisco, CA94143
| | - Whitney Purtha
- Diabetes Center, University of California San Francisco, San Francisco, CA94143
| | - Jared M. Balolong
- Diabetes Center, University of California San Francisco, San Francisco, CA94143
| | - Jonah D. Phipps
- Diabetes Center, University of California San Francisco, San Francisco, CA94143
| | - Frances Crawford
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO80206
| | - Jeffrey A. Bluestone
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, CA94143
| | - John W. Kappler
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO80206
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO80045
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Mark S. Anderson
- Diabetes Center, University of California San Francisco, San Francisco, CA94143
| |
Collapse
|
4
|
Ishina IA, Kurbatskaia IN, Mamedov AE, Shramova EI, Deyev SM, Nurbaeva KS, Rubtsov YP, Belogurov AA, Gabibov AG, Zakharova MY. Genetically engineered CD80-pMHC-harboring extracellular vesicles for antigen-specific CD4 + T-cell engagement. Front Bioeng Biotechnol 2024; 11:1341685. [PMID: 38304104 PMCID: PMC10833362 DOI: 10.3389/fbioe.2023.1341685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/29/2023] [Indexed: 02/03/2024] Open
Abstract
The identification of low-frequency antigen-specific CD4+ T cells is crucial for effective immunomonitoring across various diseases. However, this task still encounters experimental challenges necessitating the implementation of enrichment procedures. While existing antigen-specific expansion technologies predominantly concentrate on the enrichment of CD8+ T cells, advancements in methods targeting CD4+ T cells have been limited. In this study, we report a technique that harnesses antigen-presenting extracellular vesicles (EVs) for stimulation and expansion of antigen-specific CD4+ T cells. EVs are derived from a genetically modified HeLa cell line designed to emulate professional antigen-presenting cells (APCs) by expressing key costimulatory molecules CD80 and specific peptide-MHC-II complexes (pMHCs). Our results demonstrate the beneficial potent stimulatory capacity of EVs in activating both immortalized and isolated human CD4+ T cells from peripheral blood mononuclear cells (PBMCs). Our technique successfully expands low-frequency influenza-specific CD4+ T cells from healthy individuals. In summary, the elaborated methodology represents a streamlined and efficient approach for the detection and expansion of antigen-specific CD4+ T cells, presenting a valuable alternative to existing antigen-specific T-cell expansion protocols.
Collapse
Affiliation(s)
- Irina A. Ishina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Inna N. Kurbatskaia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Azad E. Mamedov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Elena I. Shramova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Sergey M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Biomarker Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Sechenov First Moscow State Medical University, Sechenov University, Moscow, Russia
| | | | - Yury P. Rubtsov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- N. N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation (NN Blokhin NMRCO), Moscow, Russia
| | - Alexey A. Belogurov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Department of Biological Chemistry, Evdokimov Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Alexander G. Gabibov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Department of Life Sciences, Higher School of Economics, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Maria Y. Zakharova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
5
|
McElwee MK, Dileepan T, Mahmud SA, Jenkins MK. The CD4+ T cell repertoire specific for citrullinated peptides shows evidence of immune tolerance. J Exp Med 2023; 220:e20230209. [PMID: 37831103 PMCID: PMC10570851 DOI: 10.1084/jem.20230209] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/27/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Rheumatoid arthritis occurs most often in people who express HLA-DR molecules containing a five aa "shared epitope" in the β chain. These MHCII molecules preferentially bind citrullinated peptides formed by posttranslational modification of arginine. Citrullinated peptide:HLA-DR complexes may act as arthritis-initiating neo-antigens for CD4+ T cells. Here, we used fluorophore-conjugated HLA-DR tetramers containing citrullinated peptides from human cartilage intermediate layer protein, fibrinogen, vimentin, or enolase 1 to track cognate CD4+ T cells. Immunization of HLA-DR transgenic mice with citrullinated peptides from vimentin or enolase 1 failed to cause any expansion of tetramer-binding cells, whereas immunization with citrullinated peptides from cartilage intermediate layer protein or fibrinogen elicited some expansion. The expanded tetramer-binding populations, however, had lower T helper 1 and higher regulatory T cell frequencies than populations elicited by viral peptides. These results indicate that HLA-DR-bound citrullinated peptides are not neo-antigens and induce varying degrees of immune tolerance that could pose a barrier to rheumatoid arthritis.
Collapse
Affiliation(s)
- Matthew K. McElwee
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Rheumatic and Autoimmune Diseases, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Thamotharampillai Dileepan
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Shawn A. Mahmud
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Pediatric Rheumatology, Allergy and Immunology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Marc K. Jenkins
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
6
|
Emami S, Rojas Converso T, Persson JJ, Johansson-Lindbom B. Insertion of an immunodominant T helper cell epitope within the Group A Streptococcus M protein promotes an IFN-γ-dependent shift from a non-protective to a protective immune response. Front Immunol 2023; 14:1241485. [PMID: 37654501 PMCID: PMC10465795 DOI: 10.3389/fimmu.2023.1241485] [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: 06/16/2023] [Accepted: 07/28/2023] [Indexed: 09/02/2023] Open
Abstract
The common pathogen Group A Streptococcus (GAS, Streptococcus pyogenes) is an extracellular bacterium that is associated with a multitude of infectious syndromes spanning a wide range of severity. The surface-exposed M protein is a major GAS virulence factor that is also target for protective antibody responses. In this study, we use a murine immunization model to investigate aspects of the cellular and molecular foundation for protective adaptive immune responses generated against GAS. We show that a wild type M1 GAS strain induces a non-protective antibody response, while an isogenic strain carrying the immunodominant 2W T helper cell epitope within the M protein elicits an immune response that is protective against the parental non-recombinant M1 GAS strain. Although the two strains induce total anti-GAS IgG levels of similar magnitude, only the 2W-carrying strain promotes elevated titers of the complement-fixing IgG2c subclass. Protection is dependent on IFN-γ, and IFN-γ-deficient mice show a specific reduction in IgG2c levels. Our findings suggest that inclusion of the 2W T cell epitope in the M protein confers essential qualitative alterations in the adaptive immune response against GAS, and that sparsity in IFN-γ-promoting Th cell epitopes in the M protein may constitute an immune evasion mechanism, evolved to allow the pathogen to avoid attack by complement-fixing antibodies.
Collapse
|
7
|
Pircher H, Pinschewer DD, Boehm T. MHC I tetramer staining tends to overestimate the number of functionally relevant self-reactive CD8 T cells in the preimmune repertoire. Eur J Immunol 2023; 53:e2350402. [PMID: 37179469 DOI: 10.1002/eji.202350402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/19/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Previous studies that used peptide-MHC (pMHC) tetramers (tet) to identify self-specific T cells have questioned the effectiveness of thymic-negative selection. Here, we used pMHCI tet to enumerate CD8 T cells specific for the immunodominant gp33 epitope of lymphocytic choriomeningitis virus glycoprotein (GP) in mice transgenically engineered to express high levels of GP as a self-antigen in the thymus. In GP-transgenic mice (GP+ ), monoclonal P14 TCR+ CD8 T cells that express a GP-specific TCR could not be detected by gp33/Db -tet staining, indicative of their complete intrathymic deletion. By contrast, in the same GP+ mice, substantial numbers of polyclonal CD8 T cells identifiable by gp33/Db -tet were present. The gp33-tet staining profiles of polyclonal T cells from GP+ and GP-negative (GP- ) mice were overlapping, but mean fluorescence intensities were ∼15% lower in cells from GP+ mice. Remarkably, the gp33-tet+ T cells in GP+ mice failed to clonally expand after lymphocytic choriomeningitis virus infection, whereas those of GP- mice did so. In Nur77GFP -reporter mice, dose-dependent responses to gp33 peptide-induced TCR stimulation revealed that gp33-tet+ T cells with high ligand sensitivity are lacking in GP+ mice. Hence, pMHCI tet staining identifies self-specific CD8 T cells but tends to overestimate the number of truly self-reactive cells.
Collapse
Affiliation(s)
- Hanspeter Pircher
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Daniel D Pinschewer
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Thomas Boehm
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
8
|
Jones MC, Castonguay C, Nanaware PP, Weaver GC, Stadinski B, Kugler-Umana OA, Huseby ES, Stern LJ, McKinstry KK, Strutt TM, Devarajan P, Swain SL. CD4 Effector TCR Avidity for Peptide on APC Determines the Level of Memory Generated. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1950-1961. [PMID: 37093656 PMCID: PMC10247507 DOI: 10.4049/jimmunol.2200337] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 03/30/2023] [Indexed: 04/25/2023]
Abstract
Initial TCR affinity for peptide Ag is known to impact the generation of memory; however, its contributions later, when effectors must again recognize Ag at 5-8 d postinfection to become memory, is unclear. We examined whether the effector TCR affinity for peptide at this "effector checkpoint" dictates the extent of memory and degree of protection against rechallenge. We made an influenza A virus nucleoprotein (NP)-specific TCR transgenic mouse strain, FluNP, and generated NP-peptide variants that are presented by MHC class II to bind to the FluNP TCR over a broad range of avidity. To evaluate the impact of avidity in vivo, we primed naive donor FluNP in influenza A virus-infected host mice, purified donor effectors at the checkpoint, and cotransferred them with the range of peptides pulsed on activated APCs into second uninfected hosts. Higher-avidity peptides yielded higher numbers of FluNP memory cells in spleen and most dramatically in lung and draining lymph nodes and induced better protection against lethal influenza infection. Avidity determined memory cell number, not cytokine profile, and already impacted donor cell number within several days of transfer. We previously found that autocrine IL-2 production at the checkpoint prevents default effector apoptosis and supports memory formation. Here, we find that peptide avidity determines the level of IL-2 produced by these effectors and that IL-2Rα expression by the APCs enhances memory formation, suggesting that transpresentation of IL-2 by APCs further amplifies IL-2 availability. Secondary memory generation was also avidity dependent. We propose that this regulatory pathway selects CD4 effectors of highest affinity to progress to memory.
Collapse
Affiliation(s)
- Michael C. Jones
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Catherine Castonguay
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Padma P. Nanaware
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Grant C. Weaver
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Brian Stadinski
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Olivia A. Kugler-Umana
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Eric S. Huseby
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lawrence J. Stern
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Karl Kai McKinstry
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL. 32827,USA
| | - Tara M. Strutt
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL. 32827,USA
| | - Priyadharshini Devarajan
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Susan L. Swain
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
9
|
Abstract
Specialized subpopulations of CD4+ T cells survey major histocompatibility complex class II-peptide complexes to control phagosomal infections, help B cells, regulate tissue homeostasis and repair or perform immune regulation. Memory CD4+ T cells are positioned throughout the body and not only protect the tissues from reinfection and cancer, but also participate in allergy, autoimmunity, graft rejection and chronic inflammation. Here we provide updates on our understanding of the longevity, functional heterogeneity, differentiation, plasticity, migration and human immunodeficiency virus reservoirs as well as key technological advances that are facilitating the characterization of memory CD4+ T cell biology.
Collapse
Affiliation(s)
- Marco Künzli
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - David Masopust
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
10
|
Rollins MR, Raynor JF, Miller EA, Butler JZ, Spartz EJ, Lahr WS, You Y, Burrack AL, Moriarity BS, Webber BR, Stromnes IM. Germline T cell receptor exchange results in physiological T cell development and function. Nat Commun 2023; 14:528. [PMID: 36726009 PMCID: PMC9892040 DOI: 10.1038/s41467-023-36180-1] [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: 08/27/2021] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
T cell receptor (TCR) transgenic mice represent an invaluable tool to study antigen-specific immune responses. In the pre-existing models, a monoclonal TCR is driven by a non-physiologic promoter and randomly integrated into the genome. Here, we create a highly efficient methodology to develop T cell receptor exchange (TRex) mice, in which TCRs, specific to the self/tumor antigen mesothelin (Msln), are integrated into the Trac locus, with concomitant Msln disruption to circumvent T cell tolerance. We show that high affinity TRex thymocytes undergo all sequential stages of maturation, express the exogenous TCR at DN4, require MHC class I for positive selection and undergo negative selection only when both Msln alleles are present. By comparison of TCRs with the same specificity but varying affinity, we show that Trac targeting improves functional sensitivity of a lower affinity TCR and confers resistance to T cell functional loss. By generating P14 TRex mice with the same specificity as the widely used LCMV-P14 TCR transgenic mouse, we demonstrate increased avidity of Trac-targeted TCRs over transgenic TCRs, while preserving physiologic T cell development. Together, our results support that the TRex methodology is an advanced tool to study physiological antigen-specific T cell behavior.
Collapse
Affiliation(s)
- Meagan R Rollins
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Jackson F Raynor
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Ebony A Miller
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Jonah Z Butler
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Ellen J Spartz
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, UCLA Health, Los Angeles, CA, USA
| | - Walker S Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Yun You
- Mouse Genetics Laboratory, University of Minnesota, Minneapolis, MN, USA
| | - Adam L Burrack
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Ingunn M Stromnes
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
11
|
Rushdi MN, Pan V, Li K, Choi HK, Travaglino S, Hong J, Griffitts F, Agnihotri P, Mariuzza RA, Ke Y, Zhu C. Cooperative binding of T cell receptor and CD4 to peptide-MHC enhances antigen sensitivity. Nat Commun 2022; 13:7055. [PMID: 36396644 PMCID: PMC9671906 DOI: 10.1038/s41467-022-34587-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 10/29/2022] [Indexed: 11/19/2022] Open
Abstract
Antigen recognition by the T cell receptor (TCR) of CD4+ T cells can be greatly enhanced by the coreceptor CD4. Yet, understanding of the molecular mechanism is hindered by the ultra-low affinity of CD4 binding to class-II peptide-major histocompatibility complexes (pMHC). Here we show, using two-dimensional (2D) mechanical-based assays, that the affinity of CD4-pMHC interaction is 3-4 logs lower than that of cognate TCR-pMHC interactions, and it is more susceptible to increased dissociation by forces (slip bond). In contrast, CD4 binds TCR-pre-bound pMHC at 3-6 logs higher affinity, forming TCR-pMHC-CD4 tri-molecular bonds that are prolonged by force (catch bond), and modulated by protein mobility on the cell membrane, indicating profound TCR-CD4 cooperativity. Consistent with a tri-crystal structure, using DNA origami as a molecular ruler to titrate spacing between TCR and CD4 we show that 7-nm proximity optimizes TCR-pMHC-CD4 tri-molecular bond formation with pMHC. Our results thus provide deep mechanistic insight into CD4 enhancement of TCR antigen recognition.
Collapse
Affiliation(s)
- Muaz Nik Rushdi
- grid.213917.f0000 0001 2097 4943Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA ,grid.419673.e0000 0000 9545 2456Present Address: Medtronic CO., Minneapolis, MN USA
| | - Victor Pan
- grid.213917.f0000 0001 2097 4943Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA ,grid.510306.10000 0004 5907 6472Present Address: Intellia Therapeutics, Cambridge, MA USA
| | - Kaitao Li
- grid.213917.f0000 0001 2097 4943Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA
| | - Hyun-Kyu Choi
- grid.213917.f0000 0001 2097 4943Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA
| | - Stefano Travaglino
- grid.213917.f0000 0001 2097 4943Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA
| | - Jinsung Hong
- grid.213917.f0000 0001 2097 4943Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA ,grid.417587.80000 0001 2243 3366Present Address: Food and Drug Administration, Silver Spring, MD USA
| | - Fletcher Griffitts
- grid.213917.f0000 0001 2097 4943Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA USA
| | - Pragati Agnihotri
- grid.440664.40000 0001 0313 4029W. M. Keck Laboratory for Structural Biology, Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD USA ,grid.164295.d0000 0001 0941 7177Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD USA ,grid.281126.e0000 0004 0612 4549Present Address: Advanced Bioscience Laboratories, Rockville, MD USA
| | - Roy A. Mariuzza
- grid.440664.40000 0001 0313 4029W. M. Keck Laboratory for Structural Biology, Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD USA ,grid.164295.d0000 0001 0941 7177Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD USA
| | - Yonggang Ke
- grid.213917.f0000 0001 2097 4943Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502Medical Scientist Training Program, Emory University School of Medicine, Atlanta, GA USA
| | - Cheng Zhu
- grid.213917.f0000 0001 2097 4943Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA USA
| |
Collapse
|
12
|
Huisman W, de Gier M, Hageman L, Shomuradova AS, Leboux DA, Amsen D, Falkenburg JF, Jedema I. Amino acids at position 5 in the peptide/MHC binding region of a public virus-specific TCR are completely inter-changeable without loss of function. Eur J Immunol 2022; 52:1819-1828. [PMID: 36189878 PMCID: PMC9828479 DOI: 10.1002/eji.202249975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/25/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023]
Abstract
Anti-viral T-cell responses are usually directed against a limited set of antigens, but often contain many T cells expressing different T-cell receptors (TCRs). Identical TCRs found within virus-specific T-cell populations in different individuals are known as public TCRs, but also TCRs highly-similar to these public TCRs, with only minor variations in amino acids on specific positions in the Complementary Determining Regions (CDRs), are frequently found. However, the degree of freedom at these positions was not clear. In this study, we used the HLA-A*02:01-restricted EBV-LMP2FLY -specific public TCR as model and modified the highly-variable position 5 of the CDR3β sequence with all 20 amino acids. Our results demonstrate that amino acids at this particular position in the CDR3β region of this TCR are completely inter-changeable, without loss of TCR function. We show that the inability to find certain variants in individuals is explained by their lower recombination probability rather than by steric hindrance.
Collapse
Affiliation(s)
- Wesley Huisman
- Department of HematologyLeiden University Medical CenterThe Netherlands,Department of HematopoiesisSanquin Research and Landsteiner Laboratory for Blood Cell ResearchAmsterdamThe Netherlands
| | - Melanie de Gier
- Department of HematologyLeiden University Medical CenterThe Netherlands
| | - Lois Hageman
- Department of HematologyLeiden University Medical CenterThe Netherlands
| | - Alina S. Shomuradova
- Laboratory for Transplantation ImmunologyNational Research Center for HematologyMoscowRussia
| | | | - Derk Amsen
- Department of HematopoiesisSanquin Research and Landsteiner Laboratory for Blood Cell ResearchAmsterdamThe Netherlands
| | | | - Inge Jedema
- Department of HematologyLeiden University Medical CenterThe Netherlands
| |
Collapse
|
13
|
CD4 expression in effector T cells depends on DNA demethylation over a developmentally established stimulus-responsive element. Nat Commun 2022; 13:1477. [PMID: 35304452 PMCID: PMC8933563 DOI: 10.1038/s41467-022-28914-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 02/16/2022] [Indexed: 12/17/2022] Open
Abstract
The epigenetic patterns that are established during early thymic development might determine mature T cell physiology and function, but the molecular basis and topography of the genetic elements involved are not fully known. Here we show, using the Cd4 locus as a paradigm for early developmental programming, that DNA demethylation during thymic development licenses a novel stimulus-responsive element that is critical for the maintenance of Cd4 gene expression in effector T cells. We document the importance of maintaining high CD4 expression during parasitic infection and show that by driving transcription, this stimulus-responsive element allows for the maintenance of histone H3K4me3 levels during T cell replication, which is critical for preventing de novo DNA methylation at the Cd4 promoter. A failure to undergo epigenetic programming during development leads to gene silencing during effector T cell replication. Our study thus provides evidence of early developmental events shaping the functional fitness of mature effector T cells.
Collapse
|
14
|
Pan YG, Su LF. Identification of Human Antigen-Specific T Cells Using Class II MHC Tetramer Staining and Enrichment. Methods Mol Biol 2022; 2574:31-40. [PMID: 36087197 DOI: 10.1007/978-1-0716-2712-9_3] [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] [Indexed: 06/15/2023]
Abstract
The development of peptide-major histocompatibility complex tetramers has enabled direct characterization and enumeration of antigen-specific T cells. However, the weaker interaction between class II tetramers and CD4+ T cells increases the challenge of using tetramers to analyze CD4+ T cell responses. Here, we provide an optimized class II tetramer staining protocol with a magnetic-bead enrichment strategy for the detection and functional analyses of human antigen-specific CD4+ T cells. This approach enables direct sampling of lymphocytes that recognize specific peptide-MHC complexes, including rare pathogen-specific CD4+ T cells from unexposed individuals.
Collapse
Affiliation(s)
- Yi-Gen Pan
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura F Su
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA.
- Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Lindeman I, Sollid LM. Single-cell approaches to dissect adaptive immune responses involved in autoimmunity: the case of celiac disease. Mucosal Immunol 2022; 15:51-63. [PMID: 34531547 DOI: 10.1038/s41385-021-00452-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 02/04/2023]
Abstract
Single-cell analysis is a powerful technology that has found widespread use in recent years. For diseases with involvement of adaptive immunity, single-cell analysis of antigen-specific T cells and B cells is particularly informative. In autoimmune diseases, the adaptive immune system is obviously at play, yet the ability to identify the culprit T and B cells recognizing disease-relevant antigen can be difficult. Celiac disease, a widespread disorder with autoimmune components, is unique in that disease-relevant antigens for both T cells and B cells are well defined. Furthermore, the celiac disease gut lesion is readily accessible allowing for sampling of tissue-resident cells. Thus, disease-relevant T cells and B cells from the gut and blood can be studied at the level of single cells. Here we review single-cell studies providing information on such adaptive immune cells and outline some future perspectives in the area of single-cell analysis in autoimmune diseases.
Collapse
Affiliation(s)
- Ida Lindeman
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway.,Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Ludvig M Sollid
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway. .,Department of Immunology, Oslo University Hospital, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
16
|
van Zelm MC, McKenzie CI, Varese N, Rolland JM, O’Hehir RE. Advances in allergen-specific immune cell measurements for improved detection of allergic sensitization and immunotherapy responses. Allergy 2021; 76:3374-3382. [PMID: 34355403 DOI: 10.1111/all.15036] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/02/2021] [Indexed: 01/10/2023]
Abstract
Over the past two decades, precision medicine has advanced diagnostics and treatment of allergic diseases. Component-resolved analysis of allergen sensitization facilitates stratification of patients. Furthermore, new formulations of allergen immunotherapy (AIT) products can more effectively deliver the relevant components. Molecular insights from the identification of allergen component sensitization and clinical outcomes of treatment with new AIT formulations can now be utilized for a deeper understanding of the nature of the pathogenic immune response in allergy and how this can be corrected by AIT. Fundamental in these processes are the allergen-specific B and T cells. Within the large B- and T-cell compartments, only those that specifically recognize the allergen with their immunoglobulin (Ig) or T-cell receptor (TCR), respectively, are of clinical relevance. With peripheral blood allergen-specific B- and T-cell frequencies below 1%, bulk cell analysis is typically insufficiently sensitive. We here review the latest technologies to detect allergen-specific B and T cells, as well as new developments in utilizing these tools for diagnostics and therapy monitoring to advance precision medicine for allergic diseases.
Collapse
Affiliation(s)
- Menno C. van Zelm
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Craig I. McKenzie
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
| | - Nirupama Varese
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Jennifer M. Rolland
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Robyn E. O’Hehir
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| |
Collapse
|
17
|
Greaves SA, Ravindran A, Santos RG, Chen L, Falta MT, Wang Y, Mitchell AM, Atif SM, Mack DG, Tinega AN, Maier LA, Dai S, Pinilla C, Grunewald J, Fontenot AP. CD4+ T cells in the lungs of acute sarcoidosis patients recognize an Aspergillus nidulans epitope. J Exp Med 2021; 218:212583. [PMID: 34410304 PMCID: PMC8383815 DOI: 10.1084/jem.20210785] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/18/2021] [Accepted: 07/22/2021] [Indexed: 11/05/2022] Open
Abstract
Löfgren’s syndrome (LS) is an acute form of sarcoidosis characterized by a genetic association with HLA-DRB1*03 (HLA-DR3) and an accumulation of CD4+ T cells of unknown specificity in the bronchoalveolar lavage (BAL). Here, we screened related LS-specific TCRs for antigen specificity and identified a peptide derived from NAD-dependent histone deacetylase hst4 (NDPD) of Aspergillus nidulans that stimulated these CD4+ T cells in an HLA-DR3–restricted manner. Using ELISPOT analysis, a greater number of IFN-γ– and IL-2–secreting T cells in the BAL of DR3+ LS subjects compared with DR3+ control subjects was observed in response to the NDPD peptide. Finally, increased IgG antibody responses to A. nidulans NDPD were detected in the serum of DR3+ LS subjects. Thus, our findings identify a ligand for CD4+ T cells derived from the lungs of LS patients and suggest a role of A. nidulans in the etiology of LS.
Collapse
Affiliation(s)
- Sarah A Greaves
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Avinash Ravindran
- Department of Medicine, Solna, Karolinska University Hospital, Stockholm, Sweden.,Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Radleigh G Santos
- Department of Mathematics, Nova Southeastern University, Ft. Lauderdale, FL
| | - Lan Chen
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Michael T Falta
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Yang Wang
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Angela M Mitchell
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Shaikh M Atif
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Douglas G Mack
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Alex N Tinega
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Lisa A Maier
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Medicine, National Jewish Health, Denver, CO
| | - Shaodong Dai
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Clemencia Pinilla
- Center for Translational Science, Florida International University, Port St. Lucie, FL
| | - Johan Grunewald
- Department of Medicine, Solna, Karolinska University Hospital, Stockholm, Sweden
| | - Andrew P Fontenot
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
18
|
Reed B, Crawford F, Hill RC, Jin N, White J, Krovi SH, Marrack P, Hansen K, Kappler JW. Lysosomal cathepsin creates chimeric epitopes for diabetogenic CD4 T cells via transpeptidation. J Exp Med 2021; 218:211485. [PMID: 33095259 PMCID: PMC7590512 DOI: 10.1084/jem.20192135] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 08/06/2020] [Accepted: 09/10/2020] [Indexed: 01/15/2023] Open
Abstract
The identification of the peptide epitopes presented by major histocompatibility complex class II (MHCII) molecules that drive the CD4 T cell component of autoimmune diseases has presented a formidable challenge over several decades. In type 1 diabetes (T1D), recent insight into this problem has come from the realization that several of the important epitopes are not directly processed from a protein source, but rather pieced together by fusion of different peptide fragments of secretory granule proteins to create new chimeric epitopes. We have proposed that this fusion is performed by a reverse proteolysis reaction called transpeptidation, occurring during the catabolic turnover of pancreatic proteins when secretory granules fuse with lysosomes (crinophagy). Here, we demonstrate several highly antigenic chimeric epitopes for diabetogenic CD4 T cells that are produced by digestion of the appropriate inactive fragments of the granule proteins with the lysosomal protease cathepsin L (Cat-L). This pathway has implications for how self-tolerance can be broken peripherally in T1D and other autoimmune diseases.
Collapse
Affiliation(s)
- Brendan Reed
- Department of Biomedical Research, National Jewish Health, Denver, CO.,Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO.,Research Division, Barbara Davis Center for Diabetes, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Frances Crawford
- Department of Biomedical Research, National Jewish Health, Denver, CO
| | - Ryan C Hill
- Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Niyun Jin
- Department of Biomedical Research, National Jewish Health, Denver, CO.,Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO.,Research Division, Barbara Davis Center for Diabetes, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Janice White
- Department of Biomedical Research, National Jewish Health, Denver, CO
| | - S Harsha Krovi
- Department of Biomedical Research, National Jewish Health, Denver, CO.,Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Philippa Marrack
- Department of Biomedical Research, National Jewish Health, Denver, CO.,Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO.,Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Kirk Hansen
- Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - John W Kappler
- Department of Biomedical Research, National Jewish Health, Denver, CO.,Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO.,Research Division, Barbara Davis Center for Diabetes, Anschutz Medical Campus, University of Colorado, Aurora, CO.,Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado, Aurora, CO
| |
Collapse
|
19
|
Dileepan T, Malhotra D, Kotov DI, Kolawole EM, Krueger PD, Evavold BD, Jenkins MK. MHC class II tetramers engineered for enhanced binding to CD4 improve detection of antigen-specific T cells. Nat Biotechnol 2021; 39:943-948. [PMID: 33941928 PMCID: PMC10666075 DOI: 10.1038/s41587-021-00893-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/12/2021] [Indexed: 12/15/2022]
Abstract
The ability to identify T cells that recognize specific peptide antigens bound to major histocompatibility complex (MHC) molecules has enabled enumeration and molecular characterization of the lymphocytes responsible for cell-mediated immunity. Fluorophore-labeled peptide:MHC class I (p:MHCI) tetramers are well-established reagents for identifying antigen-specific CD8+ T cells by flow cytometry, but efforts to extend the approach to CD4+ T cells have been less successful, perhaps owing to lower binding strength between CD4 and MHC class II (MHCII) molecules. Here we show that p:MHCII tetramers engineered by directed evolution for enhanced CD4 binding outperform conventional tetramers for the detection of cognate T cells. Using the engineered tetramers, we identified about twice as many antigen-specific CD4+ T cells in mice immunized against multiple peptides than when using traditional tetramers. CD4 affinity-enhanced p:MHCII tetramers, therefore, allow direct sampling of antigen-specific CD4+ T cells that cannot be accessed with conventional p:MHCII tetramer technology. These new reagents could provide a deeper understanding of the T cell repertoire.
Collapse
Affiliation(s)
- Thamotharampillai Dileepan
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Deepali Malhotra
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- AstraZeneca, Gaithersburg, MD, USA
| | - Dmitri I Kotov
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- University of California, Berkeley, Berkeley, CA, USA
| | - Elizabeth M Kolawole
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Peter D Krueger
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Brian D Evavold
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Marc K Jenkins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
20
|
Rockinger GA, Guillaume P, Cachot A, Saillard M, Speiser DE, Coukos G, Harari A, Romero PJ, Schmidt J, Jandus C. Optimized combinatorial pMHC class II multimer labeling for precision immune monitoring of tumor-specific CD4 T cells in patients. J Immunother Cancer 2021; 8:jitc-2019-000435. [PMID: 32448802 PMCID: PMC7253008 DOI: 10.1136/jitc-2019-000435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND With immunotherapy gaining increasing approval for treatment of different tumor types, scientists rely on cutting edge methods for the monitoring of immune responses and biomarker development in patients. Due to the lack of tools to efficiently detect rare circulating human tumor-specific CD4 T cells, their characterization in patients still remains very limited. METHODS We have used combinatorial staining strategies with peptide major histocompatibility complex class II (pMHCII) multimer constructs of different alleles to establish an optimized staining procedure for in vitro and direct ex-vivo visualization of tumor-specific CD4 T cells, in patient samples. Furthermore, we have generated reversible multimers to achieve optimal cell staining and yet disassemble prior to in vitro cell expansion, thus preventing activation induced cell death. RESULTS We observed a vastly improved detection of tumor-specific, viral-specific and bacterial-specific cells with our optimization methods compared with the non-optimized staining procedure. By increasing the variety of fluorochromes used to label the pMHCII multimers, we were also able to increase the parallel detection of different specificities within one sample, including antigen-specific CD8 T cells. A decrease in cell viability was observed when using the full optimization method, but this was mitigated by the removal of neuraminidase and the use of reversible multimers. CONCLUSION This new optimized staining procedure represents an advance toward better detection and analysis of antigen-specific CD4 T cells. It should facilitate state-of-the art precision monitoring of tumor-specific CD4 T cells and contribute to accelerate the use and the targeting of these cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Georg Alexander Rockinger
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch of Immunology, Epalinges, Switzerland
| | - Philippe Guillaume
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch of Immunology, Epalinges, Switzerland
| | - Amélie Cachot
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch of Immunology, Epalinges, Switzerland
| | - Margaux Saillard
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch of Immunology, Epalinges, Switzerland
| | - Daniel E Speiser
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Georges Coukos
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch of Immunology, Epalinges, Switzerland
| | - Alexandre Harari
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch of Immunology, Epalinges, Switzerland
| | - Pedro J Romero
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Julien Schmidt
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch of Immunology, Epalinges, Switzerland
| | - Camilla Jandus
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland .,Ludwig Institute for Cancer Research, Lausanne Branch of Immunology, Epalinges, Switzerland
| |
Collapse
|
21
|
Falta MT, Crawford JC, Tinega AN, Landry LG, Crawford F, Mack DG, Martin AK, Atif SM, Li L, Santos RG, Nakayama M, Kappler JW, Maier LA, Thomas PG, Pinilla C, Fontenot AP. Beryllium-specific CD4+ T cells induced by chemokine neoantigens perpetuate inflammation. J Clin Invest 2021; 131:144864. [PMID: 33630763 PMCID: PMC8087207 DOI: 10.1172/jci144864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
Discovering dominant epitopes for T cells, particularly CD4+ T cells, in human immune-mediated diseases remains a significant challenge. Here, we used bronchoalveolar lavage (BAL) cells from HLA-DP2-expressing patients with chronic beryllium disease (CBD), a debilitating granulomatous lung disorder characterized by accumulations of beryllium-specific (Be-specific) CD4+ T cells in the lung. We discovered lung-resident CD4+ T cells that expressed a disease-specific public CDR3β T cell receptor motif and were specific to Be-modified self-peptides derived from C-C motif ligand 4 (CCL4) and CCL3. HLA-DP2-CCL/Be tetramer staining confirmed that these chemokine-derived peptides represented major antigenic targets in CBD. Furthermore, Be induced CCL3 and CCL4 secretion in the lungs of mice and humans. In a murine model of CBD, the addition of LPS to Be oxide exposure enhanced CCL4 and CCL3 secretion in the lung and significantly increased the number and percentage of CD4+ T cells specific for the HLA-DP2-CCL/Be epitope. Thus, we demonstrate a direct link between Be-induced innate production of chemokines and the development of a robust adaptive immune response to those same chemokines presented as Be-modified self-peptides, creating a cycle of innate and adaptive immune activation.
Collapse
Affiliation(s)
- Michael T. Falta
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jeremy C. Crawford
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Alex N. Tinega
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laurie G. Landry
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Douglas G. Mack
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Allison K. Martin
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shaikh M. Atif
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Li Li
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Radleigh G. Santos
- Department of Mathematics, Nova Southeastern University, Ft. Lauderdale, Florida, USA
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - John W. Kappler
- Department of Biomedical Research and
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lisa A. Maier
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | - Andrew P. Fontenot
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
22
|
Klawon DEJ, Gilmore DC, Leonard JD, Miller CH, Chao JL, Walker MT, Duncombe RK, Tung KS, Adams EJ, Savage PA. Altered selection on a single self-ligand promotes susceptibility to organ-specific T cell infiltration. J Exp Med 2021; 218:212038. [PMID: 33914024 PMCID: PMC8091134 DOI: 10.1084/jem.20200701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 12/17/2020] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
For the large array of self-peptide/MHC class II (pMHC-II) complexes displayed in the body, it is unclear whether CD4+ T cell tolerance must be imparted for each individual complex or whether pMHC-II–nonspecific bystander mechanisms are sufficient to confer tolerance by acting broadly on T cells reactive to multiple self-pMHC-II ligands. Here, via reconstitution of T cell–deficient mice, we demonstrate that altered T cell selection on a single prostate-specific self-pMHC-II ligand renders recipient mice susceptible to prostate-specific T cell infiltration. Mechanistically, this self-pMHC-II complex is required for directing antigen-specific cells into the Foxp3+ regulatory T cell lineage but does not induce clonal deletion to a measurable extent. Thus, our data demonstrate that polyclonal T reg cells are unable to functionally compensate for a breach in tolerance to a single self-pMHC-II complex in this setting, revealing vulnerabilities in antigen-nonspecific bystander mechanisms of immune tolerance.
Collapse
Affiliation(s)
| | - Dana C Gilmore
- Department of Pathology, University of Chicago, Chicago, IL
| | - John D Leonard
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL
| | | | - Jaime L Chao
- Department of Pathology, University of Chicago, Chicago, IL
| | | | - Ryan K Duncombe
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL
| | - Kenneth S Tung
- Department of Pathology, University of Virginia, Charlottesville, VA
| | - Erin J Adams
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL
| | - Peter A Savage
- Department of Pathology, University of Chicago, Chicago, IL
| |
Collapse
|
23
|
Sood A, Lebel MÈ, Dong M, Fournier M, Vobecky SJ, Haddad É, Delisle JS, Mandl JN, Vrisekoop N, Melichar HJ. CD5 levels define functionally heterogeneous populations of naïve human CD4 + T cells. Eur J Immunol 2021; 51:1365-1376. [PMID: 33682083 PMCID: PMC8251777 DOI: 10.1002/eji.202048788] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 12/27/2020] [Accepted: 03/04/2021] [Indexed: 11/30/2022]
Abstract
Studies in murine models show that subthreshold TCR interactions with self‐peptide are required for thymic development and peripheral survival of naïve T cells. Recently, differences in the strength of tonic TCR interactions with self‐peptide, as read‐out by cell surface levels of CD5, were associated with distinct effector potentials among sorted populations of T cells in mice. However, whether CD5 can also be used to parse functional heterogeneity among human T cells is less clear. Our study demonstrates that CD5 levels correlate with TCR signal strength in human naïve CD4+ T cells. Further, we describe a relationship between CD5 levels on naïve human CD4+ T cells and binding affinity to foreign peptide, in addition to a predominance of CD5hi T cells in the memory compartment. Differences in gene expression and biases in cytokine production potential between CD5lo and CD5hi naïve human CD4+ T cells are consistent with observations in mice. Together, these data validate the use of CD5 surface levels as a marker of heterogeneity among human naïve CD4+ T cells with important implications for the identification of functionally biased T‐ cell populations that can be exploited to improve the efficacy of adoptive cell therapies.
Collapse
Affiliation(s)
- Aditi Sood
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Ève Lebel
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada
| | - Mengqi Dong
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada
| | - Marilaine Fournier
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada
| | - Suzanne J Vobecky
- Service de Chirurgie Cardiaque, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Élie Haddad
- Département de Pédiatrie, Université de Montréal, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Jean-Sébastien Delisle
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada.,Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
| | - Judith N Mandl
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Nienke Vrisekoop
- Department of Respiratory Medicine, Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada.,Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
24
|
Willis RA, Ramachandiran V, Shires JC, Bai G, Jeter K, Bell DL, Han L, Kazarian T, Ugwu KC, Laur O, Contreras-Alcantara S, Long DL, Altman JD. Production of Class II MHC Proteins in Lentiviral Vector-Transduced HEK-293T Cells for Tetramer Staining Reagents. Curr Protoc 2021; 1:e36. [PMID: 33539685 PMCID: PMC7880703 DOI: 10.1002/cpz1.36] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Class II major histocompatibility complex peptide (MHC-IIp) multimers are precisely engineered reagents used to detect T cells specific for antigens from pathogens, tumors, and self-proteins. While the related Class I MHC/peptide (MHC-Ip) multimers are usually produced from subunits expressed in E. coli, most Class II MHC alleles cannot be produced in bacteria, and this has contributed to the perception that MHC-IIp reagents are harder to produce. Herein, we present a robust constitutive expression system for soluble biotinylated MHC-IIp proteins that uses stable lentiviral vector-transduced derivatives of HEK-293T cells. The expression design includes allele-specific peptide ligands tethered to the amino-terminus of the MHC-II β chain via a protease-cleavable linker. Following cleavage of the linker, HLA-DM is used to catalyze efficient peptide exchange, enabling high-throughput production of many distinct MHC-IIp complexes from a single production cell line. Peptide exchange is monitored using either of two label-free methods, native isoelectric focusing gel electrophoresis or matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry of eluted peptides. Together, these methods produce MHC-IIp complexes that are highly homogeneous and that form the basis for excellent MHC-IIp multimer reagents. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Lentivirus production and expression line creation Support Protocol 1: Six-well assay for estimation of production cell line yield Support Protocol 2: Universal ELISA for quantifying proteins with fused leucine zippers and His-tags Basic Protocol 2: Cultures for production of Class II MHC proteins Basic Protocol 3: Purification of Class II MHC proteins by anti-leucine zipper affinity chromatography Alternate Protocol 1: IMAC purification of His-tagged Class II MHC Support Protocol 3: Protein concentration measurements and adjustments Support Protocol 4: Polishing purification by anion-exchange chromatography Support Protocol 5: Estimating biotinylation percentage by streptavidin precipitation Basic Protocol 4: Peptide exchange Basic Protocol 5: Analysis of peptide exchange by matrix-assisted laser desorption/ionization (MALDI) mass spectrometry Alternate Protocol 2: Native isoelectric focusing to validate MHC-II peptide loading Basic Protocol 6: Multimerization Basic Protocol 7: Staining cells with Class II MHC tetramers.
Collapse
Affiliation(s)
- Richard A Willis
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Vasanthi Ramachandiran
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - John C Shires
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Ge Bai
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Kelly Jeter
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Donielle L Bell
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Lixia Han
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Tamara Kazarian
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Kyla C Ugwu
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Oskar Laur
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Custom Cloning Core Facility, Emory University School of Medicine, Atlanta, Georgia
| | - Susana Contreras-Alcantara
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - Dale L Long
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
| | - John D Altman
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Yerkes National Primate Research Center, Atlanta, Georgia
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Center for AIDS Research, Emory University, Atlanta, Georgia
| |
Collapse
|
25
|
Kolawole EM, Lamb TJ, Evavold BD. Relationship of 2D Affinity to T Cell Functional Outcomes. Int J Mol Sci 2020; 21:E7969. [PMID: 33120989 PMCID: PMC7662510 DOI: 10.3390/ijms21217969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022] Open
Abstract
T cells are critical for a functioning adaptive immune response and a strong correlation exists between T cell responses and T cell receptor (TCR): peptide-loaded MHC (pMHC) binding. Studies that utilize pMHC tetramer, multimers, and assays of three-dimensional (3D) affinity have provided advancements in our understanding of T cell responses across different diseases. However, these technologies focus on higher affinity and avidity T cells while missing the lower affinity responders. Lower affinity TCRs in expanded polyclonal populations almost always constitute a significant proportion of the response with cells mediating different effector functions associated with variation in the proportion of high and low affinity T cells. Since lower affinity T cells expand and are functional, a fully inclusive view of T cell responses is required to accurately interpret the role of affinity for adaptive T cell immunity. For example, low affinity T cells are capable of inducing autoimmune disease and T cells with an intermediate affinity have been shown to exhibit an optimal anti-tumor response. Here, we focus on how affinity of the TCR may relate to T cell phenotype and provide examples where 2D affinity influences functional outcomes.
Collapse
Affiliation(s)
| | | | - Brian D. Evavold
- Department of Pathology, University of Utah, 15 N Medical Drive, Salt Lake City, UT 84112, USA; (E.M.K.); (T.J.L.)
| |
Collapse
|
26
|
Hoffmann MM, Slansky JE. T-cell receptor affinity in the age of cancer immunotherapy. Mol Carcinog 2020; 59:862-870. [PMID: 32386086 DOI: 10.1002/mc.23212] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
The strength of the interaction between T-cell receptors (TCRs) and their ligands, peptide/major histocompatibility complex complexes (pMHCs), is one of the most frequently discussed and investigated features of T cells in immuno-oncology today. Although there are many molecules on the surface of T cells that interact with ligands on other cells, the TCR/pMHC is the only receptor-ligand pair that offers antigen specificity and dictates the functional response of the T cell. The strength of the TCR/pMHC interaction, along with the environment in which this interaction takes place, is key to how the T cell will respond. The TCR repertoire of T cells that interact with tumor-associated antigens is vast, although typically of low affinity. Here, we focus on the low-affinity interactions between TCRs from CD8+ T cells and different models used in immuno-oncology.
Collapse
Affiliation(s)
- Michele M Hoffmann
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jill E Slansky
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
27
|
Li KP, Ladle BH, Kurtulus S, Sholl A, Shanmuganad S, Hildeman DA. T-cell receptor signal strength and epigenetic control of Bim predict memory CD8 + T-cell fate. Cell Death Differ 2020; 27:1214-1224. [PMID: 31558776 PMCID: PMC7206134 DOI: 10.1038/s41418-019-0410-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/31/2019] [Accepted: 08/21/2019] [Indexed: 12/24/2022] Open
Abstract
Most effector CD8+ T cells die, while some persist and become either "effector" (TEM) or "central" (TCM) memory T cells. Paradoxically, effector CD8+ T cells with greater memory potential have higher levels of the pro-apoptotic molecule Bim. Here, we report, using a novel Bim-mCherry knock-in mouse, that cells with high levels of Bim preferentially develop into TCM cells. Bim levels remained stable and were regulated by DNA methylation at the Bim promoter. Notably, high levels of Bcl-2 were required for Bimhi cells to survive. Using Nur77-GFP mice as an indicator of TCR signal strength, Nur77 levels correlated with Bim expression and Nur77hi cells also selectively developed into TCM cells. Altogether, these data show that Bim levels and TCR signal strength are predictive of TEM- vs. TCM-cell fate. Further, given the many other biologic functions of Bim, these mice will have broad utility beyond CD8+ T-cell fate.
Collapse
Affiliation(s)
- Kun-Po Li
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Brian H Ladle
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Johns Hopkins Hospital, 1800 Orleans Street, The Charlotte R. Bloomberg Children's Center Building, 11th Floor, Baltimore, MD, 21287, USA
| | - Sema Kurtulus
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Allyson Sholl
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Sharmila Shanmuganad
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - David A Hildeman
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
28
|
Raynor J, Lin A, Hummel SA, Lampe K, Jordan M, Hoebe K, Hildeman DA. The Variable Genomic NK Cell Receptor Locus Is a Key Determinant of CD4+ T Cell Responses During Viral Infection. Front Immunol 2020; 11:197. [PMID: 32153566 PMCID: PMC7044186 DOI: 10.3389/fimmu.2020.00197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/27/2020] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence points to a key role for NK cells in controlling adaptive immune responses. In studies examining the role of CD1d on CD4+ T cell responses, we found that a line of CD1d-deficient mice on the C57BL/6J background had a homozygous 129 locus on chromosome 6 containing the entire NK cell gene cluster. Mice possessing this locus (C57BL/6.NKC129) displayed a >10-fold reduction in antigen-specific CD4+ T cell responses after intracranial infection with lymphocytic choriomeningitis virus (LCMV). Neither parental strain displayed defects in viral-specific CD4+ T cell responses. Interestingly, following infection, increased numbers of NK cells accumulated in the lymph nodes of C57BL/6.NKC129 mice and displayed enhanced in vivo functionality. Moreover, depletion of NK cells with anti-asialo-GM-1 antibody in C57BL/6.NKC129 mice resulted in a >20-fold increase in viral-specific CD4+ T cell responses. Mechanistically, we found that dendritic cell antigen presentation and early type I IFN production were significantly decreased in C57BL/6.NKC129 mice, but were restored in perforin-deficient C57BL/6.NKC129 mice or following NK depletion. Together, these data reveal that the variable genomic regions containing the activating/inhibitory NK cell receptors are key determinants of antigen-specific CD4+ T cell responses, controlling type I IFN production and the antigen-presenting capacity of dendritic cells.
Collapse
Affiliation(s)
- Jana Raynor
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Adora Lin
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Sarah A Hummel
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Kristin Lampe
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michael Jordan
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Kasper Hoebe
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David A Hildeman
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
29
|
Zhang Z, Legoux FP, Vaughan SW, Moon JJ. Opposing peripheral fates of tissue-restricted self antigen-specific conventional and regulatory CD4 + T cells. Eur J Immunol 2019; 50:63-72. [PMID: 31580477 DOI: 10.1002/eji.201948180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/29/2019] [Accepted: 09/27/2019] [Indexed: 11/08/2022]
Abstract
The development of self antigen-specific T cells is influenced by how the self antigen is expressed. Here, we created a mouse in which a model self antigen is conditionally expressed in different tissue environments. Using peptide:MHCII tetramer-based cell enrichment methods, we examined the development of corresponding endogenous self antigen-specific CD4+ T cell populations. While ubiquitous self antigen expression resulted in efficient deletion of self antigen-specific T cells in the thymus, some tissue-restricted expression patterns resulted in partial deletion of the population in peripheral lymphoid organs. Deletion specifically affected Foxp3- conventional T cells (Tconv) with a bias towards high avidity TCR expressing cells in the case of thymic, but not peripheral deletion. In contrast, Foxp3+ Treg exhibited elevated frequencies with increased TCR avidity. T cells surviving deletion were functionally impaired, with Tconv cells exhibiting more impairment than Tregs. Collectively, our results illustrate how postthymic recognition of tissue-restricted self antigens results in opposing developmental fates for Tconv and Treg cell subsets.
Collapse
Affiliation(s)
- Zimeng Zhang
- Center for Immunology and Inflammatory Diseases, and Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, and Harvard Medical School, Charlestown, MA, USA.,Program in Immunology, Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Francois P Legoux
- Center for Immunology and Inflammatory Diseases, and Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, and Harvard Medical School, Charlestown, MA, USA
| | - Spencer W Vaughan
- Center for Immunology and Inflammatory Diseases, and Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, and Harvard Medical School, Charlestown, MA, USA
| | - James J Moon
- Center for Immunology and Inflammatory Diseases, and Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
30
|
Ciabattini A, Olivieri R, Lazzeri E, Medaglini D. Role of the Microbiota in the Modulation of Vaccine Immune Responses. Front Microbiol 2019; 10:1305. [PMID: 31333592 PMCID: PMC6616116 DOI: 10.3389/fmicb.2019.01305] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/27/2019] [Indexed: 12/12/2022] Open
Abstract
The human immune system and the microbiota co-evolve, and their balanced relationship is based on crosstalk between the two systems through the course of life. This tight association and the overall composition and richness of the microbiota play an important role in the modulation of host immunity and may impact the immune response to vaccination. The availability of innovative technologies, such as next-generation sequencing (NGS) and correlated bioinformatics tools, allows a deeper investigation of the crosstalk between the microbiota and human immune responses. This review discusses the current knowledge on the influence of the microbiota on the immune response to vaccination and novel tools to deeply analyze the impact of the microbiome on vaccine responses.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Raffaela Olivieri
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elisa Lazzeri
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
31
|
Garcia-Garijo A, Fajardo CA, Gros A. Determinants for Neoantigen Identification. Front Immunol 2019; 10:1392. [PMID: 31293573 PMCID: PMC6601353 DOI: 10.3389/fimmu.2019.01392] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/03/2019] [Indexed: 12/22/2022] Open
Abstract
All tumors accumulate genetic alterations, some of which can give rise to mutated, non-self peptides presented by human leukocyte antigen (HLA) molecules and elicit T-cell responses. These immunogenic mutated peptides, or neoantigens, are foreign in nature and display exquisite tumor specificity. The correlative evidence suggesting they play an important role in the effectiveness of various cancer immunotherapies has triggered the development of vaccines and adoptive T-cell therapies targeting them. However, the systematic identification of personalized neoantigens in cancer patients, a critical requisite for the success of these therapies, remains challenging. A growing amount of evidence supports that only a small fraction of all tumor somatic non-synonymous mutations (NSM) identified represent bona fide neoantigens; mutated peptides that are processed, presented on the cell surface HLA molecules of cancer cells and are capable of triggering immune responses in patients. Here, we provide an overview of the existing strategies to identify candidate neoantigens and to evaluate their immunogenicity, two factors that impact on neoantigen identification. We will focus on their strengths and limitations to allow readers to rationally select and apply the most suitable method for their specific laboratory setting.
Collapse
Affiliation(s)
| | | | - Alena Gros
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
32
|
Kong YY, Kwok WW. Identification of Human Antigen-Specific CD4 + T-Cells with Peptide-MHC Multimer Technologies. Methods Mol Biol 2019; 1988:375-386. [PMID: 31147953 DOI: 10.1007/978-1-4939-9450-2_26] [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] [Indexed: 06/09/2023]
Abstract
The development of peptide-MHC class II multimers has provided a novel approach in studying antigen-specific CD4+ T-cells and extended the knowledge of these T cells in various disease settings, including infectious diseases, autoimmune diseases, cancer, and allergies. This chapter discusses the various applications of the peptide-MHC class II multimer technologies, specifically their uses in the evaluation of antigen-specific CD4+ T-cells ex vivo, and their uses in epitope identification.
Collapse
Affiliation(s)
- Ying Ying Kong
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
33
|
Williams T, Krovi HS, Landry LG, Crawford F, Jin N, Hohenstein A, DeNicola ME, Michels AW, Davidson HW, Kent SC, Gapin L, Kappler JW, Nakayama M. Development of T cell lines sensitive to antigen stimulation. J Immunol Methods 2018; 462:65-73. [PMID: 30165064 DOI: 10.1016/j.jim.2018.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/15/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022]
Abstract
Immortalized T cells such as T cell hybridomas, transfectomas, and transductants are useful tools to study tri-molecular complexes consisting of peptide, MHC, and T cell receptor (TCR) molecules. These cells have been utilized for antigen discovery studies for decades due to simplicity and rapidness of growing cells. However, responsiveness to antigen stimulation is typically less sensitive compared to primary T cells, resulting in occasional false negative outcomes especially for TCRs having low affinity to a peptide-MHC complex (pMHC). To overcome this obstacle, we genetically engineered T cell hybridomas to express additional CD3 molecules as well as CD4 with two amino acid substitutions that increase affinity to MHC class II molecules. The manipulated T cell hybridomas that were further transduced with retroviral vectors encoding TCRs of interest responded to cognate antigens more robustly than non-manipulated cells without evoking non-antigen specific reactivity. Of importance, the manipulation with CD3 and mutated human CD4 expression was effective in increasing responsiveness of T cell hybridomas to a wide variety of TCR, peptide, and MHC combinations across class II genetic loci (i.e. HLA-DR, HLA-DQ, HLA-DP, and murine H2-IA) and species (i.e. both humans and mice), and thus will be useful to identify antigen specificity of T cells.
Collapse
Affiliation(s)
- Theodore Williams
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA
| | - Harsha S Krovi
- Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19(th) Avenue, Aurora, CO 80045, USA
| | - Laurie G Landry
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA
| | - Frances Crawford
- Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Niyun Jin
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA
| | - Anita Hohenstein
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA
| | - Megan E DeNicola
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts School of Medicine, 368 Plantation Street, ASC7-2012, Worcester, MA 01605, USA
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA
| | - Howard W Davidson
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19(th) Avenue, Aurora, CO 80045, USA
| | - Sally C Kent
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts School of Medicine, 368 Plantation Street, ASC7-2012, Worcester, MA 01605, USA
| | - Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19(th) Avenue, Aurora, CO 80045, USA
| | - John W Kappler
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19(th) Avenue, Aurora, CO 80045, USA; Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA; Program in Structural Biology and Biochemistry, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, 1775 Aurora Court, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19(th) Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
34
|
Miller ML, McIntosh CM, Williams JB, Wang Y, Hollinger MK, Isaad NJ, Moon JJ, Gajewski TF, Chong AS, Alegre ML. Distinct Graft-Specific TCR Avidity Profiles during Acute Rejection and Tolerance. Cell Rep 2018; 24:2112-2126. [PMID: 30134172 PMCID: PMC6142813 DOI: 10.1016/j.celrep.2018.07.067] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/19/2018] [Accepted: 07/18/2018] [Indexed: 11/17/2022] Open
Abstract
Mechanisms implicated in robust transplantation tolerance at the cellular level can be broadly categorized into those that inhibit alloreactive T cells intrinsically (clonal deletion and dysfunction) or extrinsically through regulation. Here, we investigated whether additional population-level mechanisms control T cells by examining whether therapeutically induced peripheral transplantation tolerance could influence T cell populations' avidity for alloantigens. Whereas T cells with high avidity preferentially accumulated during acute rejection of allografts, the alloreactive T cells in tolerant recipients retained a low-avidity profile, comparable to naive mice despite evidence of activation. These contrasting avidity profiles upon productive versus tolerogenic stimulation were durable and persisted upon alloantigen re-encounter in the absence of any immunosuppression. Thus, peripheral transplantation tolerance involves control of alloreactive T cells at the population level, in addition to the individual cell level. Controlling expansion or eliminating high-affinity, donor-specific T cells long term may be desirable to achieve robust transplantation tolerance in the clinic.
Collapse
Affiliation(s)
- Michelle L Miller
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA
| | - Christine M McIntosh
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA
| | - Jason B Williams
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Ying Wang
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA
| | - Maile K Hollinger
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA
| | - Noel J Isaad
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA
| | - James J Moon
- Center for Immunology and Inflammatory Diseases and Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Charlestown, MA 02129, USA
| | - Thomas F Gajewski
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Anita S Chong
- Department of Surgery, Section of Transplantation, The University of Chicago, Chicago, IL 60637, USA
| | - Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
35
|
Martínez-Usatorre A, Donda A, Zehn D, Romero P. PD-1 Blockade Unleashes Effector Potential of Both High- and Low-Affinity Tumor-Infiltrating T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:792-803. [PMID: 29875150 DOI: 10.4049/jimmunol.1701644] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/09/2018] [Indexed: 12/21/2022]
Abstract
Antitumor T cell responses involve CD8+ T cells with high affinity for mutated self-antigen and low affinity for nonmutated tumor-associated Ag. Because of the highly individual nature of nonsynonymous somatic mutations in tumors, however, immunotherapy relies often on an effective engagement of low-affinity T cells. In this study, we studied the role of T cell affinity during peripheral priming with single-peptide vaccines and during the effector phase in the tumor. To that end, we compared the antitumor responses after OVA257-264 (N4) peptide vaccination of CD8+ T cells carrying TCRs with high (OT-1) and low (OT-3) avidity for the N4 peptide in B16.N4 tumor-bearing C57BL/6 mice. Additionally, we assessed the response of OT-1 cells to either high-affinity (B16.N4) or low-affinity (B16.T4) Ag-expressing tumors after high-affinity (N4) or low-affinity (T4) peptide vaccination. We noticed that although low-affinity tumor-specific T cells expand less than high-affinity T cells, they express lower levels of inhibitory receptors and produce more cytokines. Interestingly, tumor-infiltrating CD8+ T cells show similar in vivo re-expansion capacity to their counterparts in secondary lymphoid organs when transferred to tumor-free hosts, suggesting that T cells in tumors may be rekindled upon relief of tumor immunosuppression. Moreover, our results show that αPD-1 treatment enhances tumor control of high- and low-affinity ligand-expressing tumors, suggesting that combination of high-affinity peripheral priming by altered peptide ligands and checkpoint blockade may enable tumor control upon low-affinity Ag recognition in the tumor.
Collapse
Affiliation(s)
- Amaia Martínez-Usatorre
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, 1066 Epalinges, Switzerland; and
| | - Alena Donda
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, 1066 Epalinges, Switzerland; and
| | - Dietmar Zehn
- School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Pedro Romero
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, 1066 Epalinges, Switzerland; and
| |
Collapse
|
36
|
Andargachew R, Martinez RJ, Kolawole EM, Evavold BD. CD4 T Cell Affinity Diversity Is Equally Maintained during Acute and Chronic Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:19-30. [PMID: 29777029 DOI: 10.4049/jimmunol.1800295] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
TCR affinity for peptide MHC dictates the functional efficiency of T cells and their propensity to differentiate into effectors and form memory. However, in the context of chronic infections, it is unclear what the overall profile of TCR affinity for Ag is and if it differs from acute infections. Using the comprehensive affinity analysis provided by the two-dimensional micropipette adhesion frequency assay and the common indirect affinity evaluation methods of MHC class II tetramer and functional avidity, we tracked IAb GP61-80-specific cells in the mouse model of acute (Armstrong) and chronic (clone 13) lymphocytic choriomeningitis virus infection. In each response, we show CD4 T cell population affinity peaks at the effector phase and declines with memory. Of interest, the range and average relative two-dimensional affinity was equivalent between acute and chronic infection, indicating chronic Ag exposure did not skew TCR affinity. In contrast, functional and tetramer avidity measurements revealed divergent results and lacked a consistent correlation with TCR affinity. Our findings highlight that the immune system maintains a diverse range in TCR affinity even under the pressures of chronic Ag stimulation.
Collapse
Affiliation(s)
- Rakieb Andargachew
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Ryan J Martinez
- School of Medicine, Emory University, Atlanta, GA 30322; and
| | - Elizabeth M Kolawole
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Brian D Evavold
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
37
|
Rius C, Attaf M, Tungatt K, Bianchi V, Legut M, Bovay A, Donia M, Thor Straten P, Peakman M, Svane IM, Ott S, Connor T, Szomolay B, Dolton G, Sewell AK. Peptide-MHC Class I Tetramers Can Fail To Detect Relevant Functional T Cell Clonotypes and Underestimate Antigen-Reactive T Cell Populations. THE JOURNAL OF IMMUNOLOGY 2018; 200:2263-2279. [PMID: 29483360 PMCID: PMC5857646 DOI: 10.4049/jimmunol.1700242] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 01/29/2018] [Indexed: 12/01/2022]
Abstract
Peptide-MHC (pMHC) multimers, usually used as streptavidin-based tetramers, have transformed the study of Ag-specific T cells by allowing direct detection, phenotyping, and enumeration within polyclonal T cell populations. These reagents are now a standard part of the immunology toolkit and have been used in many thousands of published studies. Unfortunately, the TCR-affinity threshold required for staining with standard pMHC multimer protocols is higher than that required for efficient T cell activation. This discrepancy makes it possible for pMHC multimer staining to miss fully functional T cells, especially where low-affinity TCRs predominate, such as in MHC class II–restricted responses or those directed against self-antigens. Several recent, somewhat alarming, reports indicate that pMHC staining might fail to detect the majority of functional T cells and have prompted suggestions that T cell immunology has become biased toward the type of cells amenable to detection with multimeric pMHC. We use several viral- and tumor-specific pMHC reagents to compare populations of human T cells stained by standard pMHC protocols and optimized protocols that we have developed. Our results confirm that optimized protocols recover greater populations of T cells that include fully functional T cell clonotypes that cannot be stained by regular pMHC-staining protocols. These results highlight the importance of using optimized procedures that include the use of protein kinase inhibitor and Ab cross-linking during staining to maximize the recovery of Ag-specific T cells and serve to further highlight that many previous quantifications of T cell responses with pMHC reagents are likely to have considerably underestimated the size of the relevant populations.
Collapse
Affiliation(s)
- Cristina Rius
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Meriem Attaf
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Katie Tungatt
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Valentina Bianchi
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Mateusz Legut
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Amandine Bovay
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom.,Department of Oncology and Ludwig Cancer Research, Lausanne University Hospital, Epalinges VD 1066, Switzerland
| | - Marco Donia
- Centre for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Per Thor Straten
- Centre for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Mark Peakman
- Department of Immunobiology, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
| | - Inge Marie Svane
- Centre for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Sascha Ott
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Tom Connor
- Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom; and.,Cardiff University School of Biosciences, Cardiff CF10 3AX, United Kingdom
| | - Barbara Szomolay
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Andrew K Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom; .,Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom; and
| |
Collapse
|
38
|
Abstract
Methods that enable the identification of virus-specific CD4 and CD8 T cells are key to our understanding of how the adaptive immune response controls viral infection. Here we describe two distinct methods to evaluate the T cell response to influenza A virus (IAV). The number and phenotype of T cells that respond to natural IAV epitopes can be assessed by flow cytometry using MHC class I and class II tetramers. Using this system, IAV-specific T cells can be tracked in various organs within the same animal, or, in different cohorts, the response can be evaluated at several time points following infection. While providing clear quantitative data, flow cytometry cannot provide any information about T cell location within the lung or interactions between responding T cells and other cell types. Here we also describe a method to examine activated CD4 T cells in the lungs of living animals using multiphoton intravital microscopy, thus providing real-time analysis of T cell behavior during an infection.
Collapse
Affiliation(s)
- Robert A Benson
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, The University of Glasgow, Glasgow, G12 8TA, UK
| | - Jennifer C Lawton
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, The University of Glasgow, Glasgow, G12 8TA, UK
| | - Megan K L MacLeod
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, The University of Glasgow, B516, Sir Graeme Davies Building, 120 University Place, Glasgow, G12 8TA, UK.
| |
Collapse
|
39
|
Glassman CR, Parrish HL, Lee MS, Kuhns MS. Reciprocal TCR-CD3 and CD4 Engagement of a Nucleating pMHCII Stabilizes a Functional Receptor Macrocomplex. Cell Rep 2018; 22:1263-1275. [PMID: 29386113 PMCID: PMC5813697 DOI: 10.1016/j.celrep.2017.12.104] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/07/2017] [Accepted: 12/27/2017] [Indexed: 12/24/2022] Open
Abstract
CD4+ T cells convert the time that T cell receptors (TCRs) interact with peptides embedded within class II major histocompatibility complex molecules (pMHCII) into signals that direct cell-fate decisions. In principle, TCRs relay information to intracellular signaling motifs of the associated CD3 subunits, while CD4 recruits the kinase Lck to those motifs upon coincident detection of pMHCII. But the mechanics by which this occurs remain enigmatic. In one model, the TCR and CD4 bind pMHCII independently, while in another, CD4 interacts with a composite surface formed by the TCR-CD3 complex bound to pMHCII. Here, we report that the duration of TCR-pMHCII interactions impact CD4 binding to MHCII. In turn, CD4 increases TCR confinement to pMHCII via reciprocal interactions involving membrane distal and proximal CD4 ectodomains. The data suggest that a precisely assembled macrocomplex functions to reliably convert TCR-pMHCII confinement into reproducible signals that orchestrate adaptive immunity.
Collapse
Affiliation(s)
- Caleb R Glassman
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Heather L Parrish
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Mark S Lee
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Michael S Kuhns
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA; The BIO-5 Institute, The University of Arizona College of Medicine, Tucson, AZ 85724, USA; The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, AZ 85724, USA; The University of Arizona Cancer Center, The University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| |
Collapse
|
40
|
Jandus C, Usatorre AM, Viganò S, Zhang L, Romero P. The Vast Universe of T Cell Diversity: Subsets of Memory Cells and Their Differentiation. Methods Mol Biol 2018; 1514:1-17. [PMID: 27787788 DOI: 10.1007/978-1-4939-6548-9_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The T cell receptor confers specificity for antigen recognition to T cells. By the first encounter with the cognate antigen, reactive T cells initiate a program of expansion and differentiation that will define not only the ultimate quantity of specific cells that will be generated, but more importantly their quality and functional heterogeneity. Recent achievements using mouse model infection systems have helped to shed light into the complex network of factors that dictate and sustain memory T cell differentiation, ranging from antigen load, TCR signal strength, metabolic fitness, transcriptional programs, and proliferative potential. The different models of memory T cell differentiation are discussed in this chapter, and key phenotypic and functional attributes of memory T cell subsets are presented, both for mouse and human cells. Therapeutic manipulation of memory T cell generation is expected to provide novel unique ways to optimize current immunotherapies, both in infection and cancer.
Collapse
Affiliation(s)
- Camilla Jandus
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Amaia Martínez Usatorre
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Selena Viganò
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Lianjun Zhang
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Pedro Romero
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| |
Collapse
|
41
|
C-terminal modification of the insulin B:11-23 peptide creates superagonists in mouse and human type 1 diabetes. Proc Natl Acad Sci U S A 2017; 115:162-167. [PMID: 29255035 PMCID: PMC5776820 DOI: 10.1073/pnas.1716527115] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Insulin is a target of CD4 T cells in type 1 diabetes in mice and humans. Why the major epitope in the insulin B chain is presented poorly to the diabetogenic CD4 T cells by the disease-associated major histocompatibility class II (MHCII) alleles has been highly debated. Here we present high-resolution mouse and human MHCII structures and T-cell functional data to show that C-terminal modifications of this epitope are required for binding and presentation in the appropriate position in the MHCII binding groove. These results suggest that pancreas-specific posttranslational modifications of this peptide may play a role in the induction of diabetes and explain how the pathogenic T cells escape deletion in the thymus. A polymorphism at β57 in some major histocompatibility complex class II (MHCII) alleles of rodents and humans is associated with a high risk for developing type 1 diabetes (T1D). However, a highly diabetogenic insulin B chain epitope within the B:9–23 peptide is presented poorly by these alleles to a variety of mouse and human CD4 T cells isolated from either nonobese diabetic (NOD) mice or humans with T1D. We have shown for both species that mutations at the C-terminal end of this epitope dramatically improve presentation to these T cells. Here we present the crystal structures of these mutated peptides bound to mouse IAg7 and human HLA-DQ8 that show how the mutations function to improve T-cell activation. In both peptide binding grooves, the mutation of B:22R to E in the peptide changes a highly unfavorable side chain for the p9 pocket to an optimal one that is dependent on the β57 polymorphism, accounting for why these peptides bind much better to these MHCIIs. Furthermore, a second mutation of the adjacent B:21 (E to G) removes a side chain from the surface of the complex that is highly unfavorable for a subset of NOD mouse CD4 cells, thereby greatly enhancing their response to the complex. These results point out the similarities between the mouse and human responses to this B chain epitope in T1D and suggest there may be common posttranslational modifications at the C terminus of the peptide in vivo to create the pathogenic epitopes in both species.
Collapse
|
42
|
Determining T-cell specificity to understand and treat disease. Nat Biomed Eng 2017; 1:784-795. [DOI: 10.1038/s41551-017-0143-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023]
|
43
|
Using DR52c/Ni 2+ mimotope tetramers to detect Ni 2+ reactive CD4 + T cells in patients with joint replacement failure. Toxicol Appl Pharmacol 2017; 331:69-75. [PMID: 28554661 DOI: 10.1016/j.taap.2017.05.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 11/20/2022]
Abstract
T cell mediated hypersensitivity to nickel (Ni2+) is one of the most common causes of allergic contact dermatitis. Ni2+ sensitization may also contribute to the failure of Ni2+ containing joint implants, and revision to non-Ni2+ containing hardware can be costly and debilitating. Previously, we identified Ni2+ mimotope peptides, which are reactive to a CD4+ T cell clone, ANi2.3 (Vα1, Vβ17), isolated from a Ni2+ hypersensitive patient with contact dermatitis. This T cell is restricted to the major histocompatibility complex class II (MHCII) molecule, Human Leukocyte Antigen (HLA)-DR52c (DRA, DRB3*0301). However, it is not known if Ni2+ induced T cell responses in sensitized joint replacement failure patients are similar to subjects with Ni2+ induced contact dermatitis. Here, we generated DR52c/Ni2+ mimotope tetramers, and used them to test if the same Ni2+ T cell activation mechanism could be generalized to Ni2+ sensitized patients with associated joint implant failure. We confirmed the specificity of these tetramers by staining of ANi2.3T cell transfectomas. The DR52c/Ni2+ mimotope tetramer detected Ni2+ reactive CD4+ T cells in the peripheral blood mononuclear cells (PBMC) of patients identified as Ni2+ sensitized by patch testing and a positive Ni2+ LPT. When HLA-typed by a DR52 specific antibody, three out of four patients were DR52 positive. In one patient, Ni2+ stimulation induced the expansion of Vβ17 positive CD4+ T cells from 0.8% to 13.3%. We found that the percentage of DR52 positivity and Vβ17 usage in Ni2+ sensitized joint failure patients are similar to Ni sensitized skin allergy patients. Ni2+ independent mimotope tetramers may be a useful tool to identify the Ni2+ reactive CD4+ T cells.
Collapse
|
44
|
Bentzen AK, Hadrup SR. Evolution of MHC-based technologies used for detection of antigen-responsive T cells. Cancer Immunol Immunother 2017; 66:657-666. [PMID: 28314956 PMCID: PMC5406421 DOI: 10.1007/s00262-017-1971-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/03/2017] [Indexed: 01/07/2023]
Abstract
T cell-mediated recognition of peptide-major histocompatibility complex (pMHC) class I and II molecules is crucial for the control of intracellular pathogens and cancer, as well as for stimulation and maintenance of efficient cytotoxic responses. Such interactions may also play a role in the development of autoimmune diseases. Novel insights into this mechanism are crucial to understanding disease development and establishing new treatment strategies. MHC multimers have been used for detection of antigen-responsive T cells since the first report by Altman et al. showed that tetramerization of pMHC class I molecules provided sufficient stability to T cell receptor (TCR)-pMHC interactions, allowing detection of MHC multimer-binding T cells using flow cytometry. Since this breakthrough the scientific community has aimed for expanding the capacity of MHC multimer-based detection technologies to facilitate large-scale epitope discovery and immune monitoring in limited biological material. Screening of T cell specificity using large libraries of pMHC molecules is suitable for analyses of T cell recognition potentially at genome-wide levels rather than analyses restricted to a selection of model antigens. Such strategies provide novel insights into the immune specificities involved in disease development and response to immunotherapy, and extend fundamental knowledge related to T cell recognition patterns and cross-recognition by TCRs. MHC multimer-based technologies have now evolved from detection of 1-2 different T cell specificities per cell sample, to include more than 1000 evaluable pMHC molecules using novel technologies. Here, we provide an overview of MHC multimer-based detection technologies developed over two decades, focusing primarily on MHC class I interactions.
Collapse
Affiliation(s)
- Amalie Kai Bentzen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Sine Reker Hadrup
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark.
| |
Collapse
|
45
|
Martinez RJ, Andargachew R, Martinez HA, Evavold BD. Low-affinity CD4+ T cells are major responders in the primary immune response. Nat Commun 2016; 7:13848. [PMID: 27976744 PMCID: PMC5234832 DOI: 10.1038/ncomms13848] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 11/04/2016] [Indexed: 12/21/2022] Open
Abstract
A robust primary immune response has been correlated with the precursor number of antigen-specific T cells, as identified using peptide MHCII tetramers. However, these tetramers identify only the highest-affinity T cells. Here we show the entire CD4+ T-cell repertoire, inclusive of low-affinity T cells missed by tetramers, using a T-cell receptor (TCR) signalling reporter and micropipette assay to quantify naive precursors and expanded populations. In vivo limiting dilution assays reveal hundreds more precursor T cells than previously thought, with higher-affinity tetramer-positive T cells, comprising only 5-30% of the total antigen-specific naive repertoire. Lower-affinity T cells maintain their predominance as the primary immune response progresses, with no enhancement of survival of T cells with high-affinity TCRs. These findings demonstrate that affinity for antigen does not control CD4+ T-cell entry into the primary immune response, as a diverse range in affinity is maintained from precursor through peak of T-cell expansion.
Collapse
Affiliation(s)
- Ryan J. Martinez
- Department of Microbiology and Immunology, Emory University, 1510 Clifton Rd NE, Atlanta Georgia, 30322, USA
| | - Rakieb Andargachew
- Department of Microbiology and Immunology, Emory University, 1510 Clifton Rd NE, Atlanta Georgia, 30322, USA
| | - Hunter A. Martinez
- Department of Microbiology and Immunology, Emory University, 1510 Clifton Rd NE, Atlanta Georgia, 30322, USA
| | - Brian D. Evavold
- Department of Microbiology and Immunology, Emory University, 1510 Clifton Rd NE, Atlanta Georgia, 30322, USA
| |
Collapse
|
46
|
Altman JD, Davis MM. MHC‐Peptide Tetramers to Visualize Antigen‐Specific T Cells. ACTA ACUST UNITED AC 2016; 115:17.3.1-17.3.44. [DOI: 10.1002/cpim.14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Mark M. Davis
- Stanford University School of Medicine and The Howard Hughes Medical Institute Palo Alto California
| |
Collapse
|
47
|
Noges LE, White J, Cambier JC, Kappler JW, Marrack P. Contamination of DNase Preparations Confounds Analysis of the Role of DNA in Alum-Adjuvanted Vaccines. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:1221-30. [PMID: 27357147 PMCID: PMC4974487 DOI: 10.4049/jimmunol.1501565] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 06/02/2016] [Indexed: 11/19/2022]
Abstract
Aluminum salt (alum) adjuvants have been used for many years as adjuvants for human vaccines because they are safe and effective. Despite its widespread use, the means by which alum acts as an adjuvant remains poorly understood. Recently, it was shown that injected alum is rapidly coated with host chromatin within mice. Experiments suggested that the host DNA in the coating chromatin contributed to alum's adjuvant activity. Some of the experiments used commercially purchased DNase and showed that coinjection of these DNase preparations with alum and Ag reduced the host's immune response to the vaccine. In this study, we report that some commercial DNase preparations are contaminated with proteases. These proteases are responsible for most of the ability of DNase preparations to inhibit alum's adjuvant activity. Nevertheless, DNase somewhat reduces responses to some Ags with alum. The effect of DNase is independent of its ability to cleave DNA, suggesting that alum improves CD4 responses to Ag via a pathway other than host DNA sensing.
Collapse
Affiliation(s)
- Laura E Noges
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and
| | - Janice White
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - John C Cambier
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and
| | - John W Kappler
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and Howard Hughes Medical Institute, National Jewish Health, Denver, CO 80206
| | - Philippa Marrack
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and Howard Hughes Medical Institute, National Jewish Health, Denver, CO 80206
| |
Collapse
|
48
|
T cells from hemophilia A subjects recognize the same HLA-restricted FVIII epitope with a narrow TCR repertoire. Blood 2016; 128:2043-2054. [PMID: 27471234 DOI: 10.1182/blood-2015-11-682468] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 07/18/2016] [Indexed: 12/15/2022] Open
Abstract
Factor VIII (FVIII)-neutralizing antibodies ("inhibitors") are a serious problem in hemophilia A (HA). The aim of this study was to characterize HLA-restricted T-cell responses from a severe HA subject with a persistent inhibitor and from 2 previously studied mild HA inhibitor subjects. Major histocompatibility complex II tetramers corresponding to both of the severe HA subject's HLA-DRA-DRB1 alleles were loaded with peptides spanning FVIII-A2, C1, and C2 domains. Interestingly, only 1 epitope was identified, in peptide FVIII2194-2213, and it was identical to the HLA-DRA*01-DRB1*01:01-restricted epitope recognized by the mild HA subjects. Multiple T-cell clones and polyclonal lines having different avidities for the peptide-loaded tetramer were isolated from all subjects. Only high- and medium-avidity T cells proliferated and secreted cytokines when stimulated with FVIII2194-2213 T-cell receptor β (TCRB) gene sequencing of 15 T-cell clones from the severe HA subject revealed that all high-avidity clones expressed the same TCRB gene. High-throughput immunosequencing of high-, medium-, and low-avidity cells sorted from a severe HA polyclonal line revealed that 94% of the high-avidity cells expressed the same TCRB gene as the high-avidity clones. TCRB sequencing of clones and lines from the mild HA subjects also identified a limited TCRB gene repertoire. These results suggest a limited number of epitopes in FVIII drive inhibitor responses and that the T-cell repertoires of FVIII-responsive T cells can be quite narrow. The limited diversity of both epitopes and TCRB gene usage suggests that targeting of specific epitopes and/or T-cell clones may be a promising approach to achieve tolerance to FVIII.
Collapse
|
49
|
Liebick M, Schläger C, Oppermann M. Analysis of Chemokine Receptor Trafficking by Site-Specific Biotinylation. PLoS One 2016; 11:e0157502. [PMID: 27310579 PMCID: PMC4911081 DOI: 10.1371/journal.pone.0157502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/31/2016] [Indexed: 12/17/2022] Open
Abstract
Chemokine receptors undergo internalization and desensitization in response to ligand activation. Internalized receptors are either preferentially directed towards recycling pathways (e.g. CCR5) or sorted for proteasomal degradation (e.g. CXCR4). Here we describe a method for the analysis of receptor internalization and recycling based on specific Bir A-mediated biotinylation of an acceptor peptide coupled to the receptor, which allows a more detailed analysis of receptor trafficking compared to classical antibody-based detection methods. Studies on constitutive internalization of the chemokine receptors CXCR4 (12.1% ± 0.99% receptor internalization/h) and CCR5 (13.7% ± 0.68%/h) reveals modulation of these processes by inverse (TAK779; 10.9% ± 0.95%/h) or partial agonists (Met-CCL5; 15.6% ± 0.5%/h). These results suggest an actively driven internalization process. We also demonstrate the advantages of specific biotinylation compared to classical antibody detection during agonist-induced receptor internalization, which may be used for immunofluorescence analysis as well. Site-specific biotinylation may be applicable to studies on trafficking of transmembrane proteins, in general.
Collapse
MESH Headings
- Amides/pharmacology
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/isolation & purification
- Basophils/cytology
- Basophils/drug effects
- Basophils/metabolism
- Biotin/chemistry
- Biotin/metabolism
- Biotinylation
- CCR5 Receptor Antagonists/pharmacology
- Carbon-Nitrogen Ligases/genetics
- Carbon-Nitrogen Ligases/metabolism
- Cell Line, Tumor
- Chemokine CCL5/pharmacology
- Escherichia coli Proteins/genetics
- Escherichia coli Proteins/metabolism
- Gene Expression
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Mice
- Protein Transport/drug effects
- Quaternary Ammonium Compounds/pharmacology
- Rats
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Receptors, CXCR5/antagonists & inhibitors
- Receptors, CXCR5/genetics
- Receptors, CXCR5/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Transfection
Collapse
Affiliation(s)
- Marcel Liebick
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| | - Christian Schläger
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| | - Martin Oppermann
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| |
Collapse
|
50
|
Control of T cell antigen reactivity via programmed TCR downregulation. Nat Immunol 2016; 17:379-86. [PMID: 26901151 PMCID: PMC4803589 DOI: 10.1038/ni.3386] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/22/2015] [Indexed: 12/17/2022]
Abstract
The T cell receptor (TCR) is unique in that its affinity for ligand is unknown prior to encounter and can vary by orders of magnitude. How the immune system regulates individual T cells that display highly different reactivity to antigen remains unclear. Here we identified that activated CD4+ T cells, at the peak of clonal expansion, persistently downregulate TCR expression in proportion to the strength of initial antigen recognition. This programmed response increases the threshold for cytokine production and recall proliferation in a clone-specific manner, ultimately excluding clones with the highest antigen reactivities. Thus, programmed TCR downregulation represents a negative feedback mechanism to constrain T cell effector function with a suitable time delay, thereby allowing pathogen control while avoiding excess inflammatory damage.
Collapse
|