1
|
Billiet L, Jansen H, Pille M, Boehme L, Sanchez Sanchez G, De Cock L, Goetgeluk G, Pascal E, De Munter S, Deseins L, Ingels J, Michiels T, De Vos R, Zolfaghari A, Vandamme N, Roels J, Kerre T, Dmitriev RI, Taghon T, Vermijlen D, Vandekerckhove B. ThymoSpheres culture: A model to study human polyclonal unconventional T cells. Eur J Immunol 2024; 54:e2451265. [PMID: 39246170 PMCID: PMC11628907 DOI: 10.1002/eji.202451265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
In vitro cultures remain crucial for studying the fundamental mechanisms of human T-cell development. Here, we introduce a novel in vitro cultivation system based on ThymoSpheres (TS): dense spheroids consisting of DLL4-expressing stromal cells and human hematopoietic precursor cells, in the absence of thymic epithelial cells. These spheroids are subsequently cultured at the air-liquid interphase. TS generate large numbers of mature T cells, are easy to manipulate, scalable, and can be repeatably sampled to monitor T-cell differentiation. The mature T cells generated from primary human hematopoietic precursor cells were extensively characterized using single-cell RNA and combined T-cell receptor (TCR) sequencing. These predominantly CD8α T cells exhibit transcriptional and TCR CDR3 characteristics similar to the recently described human polyclonal αβ unconventional T cell (UTC) lineage. This includes the expression of hallmark genes associated with agonist selection, such as IKZF2 (Helios), and the expression of various natural killer receptors. The TCR repertoire of these UTCs is polyclonal and enriched for CDR3-associated autoreactive features and early rearrangements of the TCR-α chain. In conclusion, TS cultures offer an intriguing platform to study the development of this human polyclonal UTC lineage and its inducing selection mechanisms.
Collapse
MESH Headings
- Humans
- Cell Differentiation/immunology
- Cells, Cultured
- Cell Culture Techniques/methods
- Thymus Gland/cytology
- Thymus Gland/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Spheroids, Cellular/immunology
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
Collapse
|
2
|
Praveena T, Le Nours J. State of play in the molecular presentation and recognition of anti-tumor lipid-based analogues. Front Immunol 2024; 15:1479382. [PMID: 39669569 PMCID: PMC11635198 DOI: 10.3389/fimmu.2024.1479382] [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: 08/12/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
The Natural Killer T cells (NKT) are a unique subset of T lymphocytes that recognize lipid-based antigens that are presented by the monomorphic MHC-I-like molecule, CD1d. Over 30 years ago, the discovery of the glycolipid α-Galactosylceramide (α-GalCer) from the marine sponge Agelas mauritianus, as a potent activator of the invariant Natural Killer T (iNKT) cells, has attracted great attention for its use in cancer immunotherapy. However, α-GalCer can initiate both pro-inflammatory T helper cell 1 (Th1) and anti-inflammatory Th2 type immune responses that can result in either enhanced or suppressed immunity in a somewhat unpredictable manner. Th1 polarized immune response is often correlated with an optimal anti-tumor immunity, and therefore α-GalCer did not fully offer the desired potential as an anti-tumor therapeutic. Over the past decades, considerable efforts have then been invested into the design and development of novel synthetic α-GalCer analogues that will direct a more efficient immune response towards the production of Th1 biased cytokines. In this minireview, we will discuss how subtle modifications in the chemical nature of a number of α-GalCer derivatives varied immune responses. Whilst some of these analogues showed potential in enhancing stability within CD1d and directing favourable immune responses for tumor immunotherapy, their responses in mice also highlighted the need for further research in humanized models to overcome translational challenges and optimize therapeutic efficacy.
Collapse
Affiliation(s)
| | - Jérôme Le Nours
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
3
|
Rodriguez-Sevilla JJ, Colla S. T-cell dysfunctions in myelodysplastic syndromes. Blood 2024; 143:1329-1343. [PMID: 38237139 DOI: 10.1182/blood.2023023166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 03/25/2024] Open
Abstract
ABSTRACT Escape from immune surveillance is a hallmark of cancer. Immune deregulation caused by intrinsic and extrinsic cellular factors, such as altered T-cell functions, leads to immune exhaustion, loss of immune surveillance, and clonal proliferation of tumoral cells. The T-cell immune system contributes to the pathogenesis, maintenance, and progression of myelodysplastic syndrome (MDS). Here, we comprehensively reviewed our current biological knowledge of the T-cell compartment in MDS and recent advances in the development of immunotherapeutic strategies, such as immune checkpoint inhibitors and T-cell- and antibody-based adoptive therapies that hold promise to improve the outcome of patients with MDS.
Collapse
Affiliation(s)
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
4
|
Billiet L, De Cock L, Sanchez Sanchez G, Mayer RL, Goetgeluk G, De Munter S, Pille M, Ingels J, Jansen H, Weening K, Pascal E, Raes K, Bonte S, Kerre T, Vandamme N, Seurinck R, Roels J, Lavaert M, Van Nieuwerburgh F, Leclercq G, Taghon T, Impens F, Menten B, Vermijlen D, Vandekerckhove B. Single-cell profiling identifies a novel human polyclonal unconventional T cell lineage. J Exp Med 2023; 220:e20220942. [PMID: 36939517 PMCID: PMC10037106 DOI: 10.1084/jem.20220942] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/22/2022] [Accepted: 02/27/2023] [Indexed: 03/21/2023] Open
Abstract
In the human thymus, a CD10+ PD-1+ TCRαβ+ differentiation pathway diverges from the conventional single positive T cell lineages at the early double-positive stage. Here, we identify the progeny of this unconventional lineage in antigen-inexperienced blood. These unconventional T cells (UTCs) in thymus and blood share a transcriptomic profile, characterized by hallmark transcription factors (i.e., ZNF683 and IKZF2), and a polyclonal TCR repertoire with autoreactive features, exhibiting a bias toward early TCRα chain rearrangements. Single-cell RNA sequencing confirms a common developmental trajectory between the thymic and blood UTCs and clearly delineates this unconventional lineage in blood. Besides MME+ recent thymic emigrants, effector-like clusters are identified in this heterogeneous lineage. Expression of Helios and KIR and a decreased CD8β expression are characteristics of this lineage. This UTC lineage could be identified in adult blood and intestinal tissues. In summary, our data provide a comprehensive characterization of the polyclonal unconventional lineage in antigen-inexperienced blood and identify the adult progeny.
Collapse
Affiliation(s)
- Lore Billiet
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Laurenz De Cock
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
- Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Rupert L. Mayer
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- VIB Proteomics Core, VIB, Ghent, Belgium
| | - Glenn Goetgeluk
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Stijn De Munter
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Melissa Pille
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Joline Ingels
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Hanne Jansen
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Karin Weening
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Eva Pascal
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Killian Raes
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Sarah Bonte
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Tessa Kerre
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Niels Vandamme
- VIB Single Cell Core, VIB, Ghent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Ruth Seurinck
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Jana Roels
- VIB Single Cell Core, VIB, Ghent, Belgium
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Marieke Lavaert
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Filip Van Nieuwerburgh
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Georges Leclercq
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Francis Impens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- VIB Proteomics Core, VIB, Ghent, Belgium
| | - Björn Menten
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
- Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
5
|
Song W, Wang G, Wang C, Liu L, Zhang L, Zhang R, Zhang H, Shi K. Case Report: An unclassified T cell lymphoma subtype with co-expression of TCR αβ and γ chains revealed by single cell sequencing. Front Immunol 2023; 14:1184383. [PMID: 37325644 PMCID: PMC10266344 DOI: 10.3389/fimmu.2023.1184383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
Background T cell lymphomas (TCL) are a group of heterogeneous diseases with over 40 subtypes. In this study, we identified a novel TCL subtype which was featured by a unique T cell receptor (TCR) presentation, α, β and γ chains were co-existing in a single malignant T cell. Case presentation A 45-year-old male patient was diagnosed T cell lymphoma after 2-month of abdominal distension and liver enlargement. Combining histology review, PET-CT scanning and immunophenotyes, the patient was not classified to any existing TCL subtypes. To better understand this unclassified TCL case, we performed single cell RNA sequencing paired with TCR sequencing on the patient's PBMC and bone marrow samples. To our surprise, we identified that the malignant T cells had a very rare TCR combination, by expressing two α chains, one β chain and one γ chain simultaneously. We further studied the molecular pathogenesis and tumor cell heterogeneity of this rare TCL subtype. A set of potential therapeutic targets were identified from the transcriptome data, such as CCL5, KLRG1 and CD38. Conclusions We identified the first TCL case co-expressing α, β and γ chains and dissected its molecular pathogenesis, providing valuable information for precision medicine options for this novel TCL subtype.
Collapse
Affiliation(s)
- Wei Song
- Department of Radiology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Gang Wang
- Department of Radiology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Cheng Wang
- Innovec Biotherapeutics, Inc., Beijing, China
| | - Lulu Liu
- Innovec Biotherapeutics, Inc., Beijing, China
| | | | - Ruoyu Zhang
- Innovec Biotherapeutics, Inc., Beijing, China
| | - Haixi Zhang
- Department of Hematology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Province Clinical Center for Hematologic Disease, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Hematologic Disease, Kunming, Yunnan, China
| | - Keqian Shi
- Department of Hematology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Province Clinical Center for Hematologic Disease, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Hematologic Disease, Kunming, Yunnan, China
| |
Collapse
|
6
|
Tang Y, Ma S, Lin S, Wu Y, Chen S, Liu G, Ma L, Wang Z, Jiang L, Wang Y. Cell-free protein synthesis of CD1E and B2M protein and in vitro interaction. Protein Expr Purif 2023; 203:106209. [PMID: 36460227 DOI: 10.1016/j.pep.2022.106209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022]
Abstract
CD1E, one of the most important glycolipid antigens on T cell membranes, is required for glycolipid antigen presentation on the cell surface. Cell-based recombinant expression systems have many limitations for synthesizing transmembrane proteins such as CD1E, including low protein yields and miss folding. To overcome these challenges, here we successfully synthesized high-quality soluble CD1E using an E.coli cell-free protein synthesis system (CFPS) with the aid of detergent. Following purification by Ni2+ affinity chromatography, we were able to obtain CD1E with ≥90% purity. Furthermore, we used the string website to predict the protein interaction network of CD1E and identified a potential binding partner━B2M. Similarly, we synthesized soluble B2M in the E.coli CFPS. Finally, we verified the interaction between CD1E and B2M by using Surface Plasmon Resonance (SPR). Taken together, the methods described here provide an alternative way to obtain active transmembrane protein and may facilitate future structural and functional studies on CD1E.
Collapse
Affiliation(s)
- Yajie Tang
- College of Biology and Food Engineering, Anyang Institute of Technology, Anyang, 455000, China
| | - Shengming Ma
- College of Biology and Food Engineering, Anyang Institute of Technology, Anyang, 455000, China
| | - Sen Lin
- Anyang Kindstar Global Medical Laboratory LTD, Anyang, Henan province, 455000, China
| | - Yinrong Wu
- College of Biology and Food Engineering, Anyang Institute of Technology, Anyang, 455000, China
| | - Siyang Chen
- College of Biology and Food Engineering, Anyang Institute of Technology, Anyang, 455000, China
| | - Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Lisong Ma
- State Key Laboratory of North China Crop Improvement and RegμLation, College of Horticulture, Hebei Agricultural University, Baoding, 071001, China
| | - Zaihua Wang
- Guangdong Provincial Key Lab of Ornamental Plant Germplasm Innovation and Utilization, Environmental Horticulture Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Lele Jiang
- Surgical Diagnostics Pty Ltd, Roseville, Sydney, 2069, Australia.
| | - Yao Wang
- College of Biology and Food Engineering, Anyang Institute of Technology, Anyang, 455000, China.
| |
Collapse
|
7
|
del Rio Oliva M, Mellett M, Basler M. Immunoproteasome inhibition attenuates experimental psoriasis. Front Immunol 2022; 13:1075615. [PMID: 36591277 PMCID: PMC9798438 DOI: 10.3389/fimmu.2022.1075615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Psoriasis is an autoimmune skin disease associated with multiple comorbidities. The immunoproteasome is a special form of the proteasome expressed in cells of hematopoietic origin. Methods The therapeutic use of ONX 0914, a selective inhibitor of the immunoproteasome, was investigated in Card14ΔE138+/- mice, which spontaneously develop psoriasis-like symptoms, and in the imiquimod murine model. Results In both models, treatment with ONX 0914 significantly reduced skin thickness, inflammation scores, and pathological lesions in the analyzed skin tissue. Furthermore, immunoproteasome inhibition normalized the expression of several pro-inflammatory genes in the ear and significantly reduced the inflammatory infiltrate, accompanied by a significant alteration in the αβ+ and γδ+ T cell subsets. Discussion ONX 0914 ameliorated psoriasis-like symptoms in two different murine psoriasis models, which supports the use of immunoproteasome inhibitors as a therapeutic treatment in psoriasis.
Collapse
Affiliation(s)
- Marta del Rio Oliva
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Mark Mellett
- Department of Dermatology, University Hospital Zürich (USZ), Zürich, Switzerland
- Faculty of Medicine, University of Zürich (UZH), Zürich, Switzerland
| | - Michael Basler
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| |
Collapse
|
8
|
Song Y, Liu Y, Teo HY, Liu H. Targeting Cytokine Signals to Enhance γδT Cell-Based Cancer Immunotherapy. Front Immunol 2022; 13:914839. [PMID: 35747139 PMCID: PMC9210953 DOI: 10.3389/fimmu.2022.914839] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/09/2022] [Indexed: 12/28/2022] Open
Abstract
γδT cells represent a small percentage of T cells in circulation but are found in large numbers in certain organs. They are considered to be innate immune cells that can exert cytotoxic functions on target cells without MHC restriction. Moreover, γδT cells contribute to adaptive immune response via regulating other immune cells. Under the influence of cytokines, γδT cells can be polarized to different subsets in the tumor microenvironment. In this review, we aimed to summarize the current understanding of antigen recognition by γδT cells, and the immune regulation mediated by γδT cells in the tumor microenvironment. More importantly, we depicted the polarization and plasticity of γδT cells in the presence of different cytokines and their combinations, which provided the basis for γδT cell-based cancer immunotherapy targeting cytokine signals.
Collapse
Affiliation(s)
- Yuan Song
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yonghao Liu
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Huey Yee Teo
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- *Correspondence: Haiyan Liu,
| |
Collapse
|
9
|
Walsh ES, Tollison TS, Brochu HN, Shaw BI, Diveley KR, Chou H, Law L, Kirk AD, Gale M, Peng X. Single-Cell-Based High-Throughput Ig and TCR Repertoire Sequencing Analysis in Rhesus Macaques. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:762-771. [PMID: 34987112 PMCID: PMC8820446 DOI: 10.4049/jimmunol.2100824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/17/2021] [Indexed: 02/03/2023]
Abstract
Recent advancements in microfluidics and high-throughput sequencing technologies have enabled recovery of paired H and L chains of Igs and VDJ and VJ chains of TCRs from thousands of single cells simultaneously in humans and mice. Despite rhesus macaques being one of the most well-studied model organisms for the human adaptive immune response, high-throughput single-cell immune repertoire sequencing assays are not yet available due to the complexity of these polyclonal receptors. We used custom primers that capture all known rhesus macaque Ig and TCR isotypes and chains that are fully compatible with a commercial solution for single-cell immune repertoire profiling. Using these rhesus-specific assays, we sequenced Ig and TCR repertoires in >60,000 cells from cryopreserved rhesus PBMCs, splenocytes, and FACS-sorted B and T cells. We were able to recover every Ig isotype and TCR chain, measure clonal expansion in proliferating T cells, and pair Ig and TCR repertoires with gene expression profiles of the same single cells. Our results establish the ability to perform high-throughput immune repertoire analysis in rhesus macaques at the single-cell level.
Collapse
Affiliation(s)
- Evan S. Walsh
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27607,Bioinformatics Graduate Program, North Carolina State University, Raleigh, NC 27695
| | - Tammy S. Tollison
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27607
| | - Hayden N. Brochu
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27607,Bioinformatics Graduate Program, North Carolina State University, Raleigh, NC 27695
| | - Brian I. Shaw
- Department of Surgery, Duke University, Durham, NC 27710
| | - Kayleigh R. Diveley
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27607,Genetics Graduate Program, North Carolina State University, Raleigh, NC 27695
| | - Hsuan Chou
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27607
| | - Lynn Law
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Allan D. Kirk
- Department of Surgery, Duke University, Durham, NC 27710
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA 98109,Center for Innate Immunity and Immune Diseases, University of Washington, Seattle, WA 98109,Washington National Primate Research Center, University of Washington, Seattle, WA 98121
| | - Xinxia Peng
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27607,Bioinformatics Graduate Program, North Carolina State University, Raleigh, NC 27695,Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695
| |
Collapse
|
10
|
Pothast CR, Dijkland RC, Thaler M, Hagedoorn RS, Kester MGD, Wouters AK, Hiemstra PS, van Hemert MJ, Gras S, Falkenburg JHF, Heemskerk MHM. SARS-CoV-2-specific CD4 + and CD8 + T cell responses can originate from cross-reactive CMV-specific T cells. eLife 2022; 11:82050. [PMID: 36408799 PMCID: PMC9822249 DOI: 10.7554/elife.82050] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/13/2022] [Indexed: 11/22/2022] Open
Abstract
Detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) specific CD4+ and CD8+ T cells in SARS-CoV-2-unexposed donors has been explained by the presence of T cells primed by other coronaviruses. However, based on the relatively high frequency and prevalence of cross-reactive T cells, we hypothesized cytomegalovirus (CMV) may induce these cross-reactive T cells. Stimulation of pre-pandemic cryo-preserved peripheral blood mononuclear cells (PBMCs) with SARS-CoV-2 peptides revealed that frequencies of SARS-CoV-2-specific T cells were higher in CMV-seropositive donors. Characterization of these T cells demonstrated that membrane-specific CD4+ and spike-specific CD8+ T cells originate from cross-reactive CMV-specific T cells. Spike-specific CD8+ T cells recognize SARS-CoV-2 spike peptide FVSNGTHWF (FVS) and dissimilar CMV pp65 peptide IPSINVHHY (IPS) presented by HLA-B*35:01. These dual IPS/FVS-reactive CD8+ T cells were found in multiple donors as well as severe COVID-19 patients and shared a common T cell receptor (TCR), illustrating that IPS/FVS-cross-reactivity is caused by a public TCR. In conclusion, CMV-specific T cells cross-react with SARS-CoV-2, despite low sequence homology between the two viruses, and may contribute to the pre-existing immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Cilia R Pothast
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Romy C Dijkland
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Melissa Thaler
- Department of Medical Microbiology, Leiden University Medical CenterLeidenNetherlands
| | - Renate S Hagedoorn
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Michel GD Kester
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Anne K Wouters
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical CenterLeidenNetherlands
| | - Martijn J van Hemert
- Department of Medical Microbiology, Leiden University Medical CenterLeidenNetherlands
| | - Stephanie Gras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe UniversityVictoriaAustralia,Department of Biochemistry and Molecular Biology, Monash UniversityClaytonAustralia
| | | | | |
Collapse
|
11
|
Almeida CF, Smith DGM, Cheng TY, Harpur CM, Batleska E, Nguyen-Robertson CV, Nguyen T, Thelemann T, Reddiex SJJ, Li S, Eckle SBG, Van Rhijn I, Rossjohn J, Uldrich AP, Moody DB, Williams SJ, Pellicci DG, Godfrey DI. Benzofuran sulfonates and small self-lipid antigens activate type II NKT cells via CD1d. Proc Natl Acad Sci U S A 2021; 118:e2104420118. [PMID: 34417291 PMCID: PMC8403964 DOI: 10.1073/pnas.2104420118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Natural killer T (NKT) cells detect lipids presented by CD1d. Most studies focus on type I NKT cells that express semi-invariant αβ T cell receptors (TCR) and recognize α-galactosylceramides. However, CD1d also presents structurally distinct lipids to NKT cells expressing diverse TCRs (type II NKT cells), but our knowledge of the antigens for type II NKT cells is limited. An early study identified a nonlipidic NKT cell agonist, phenyl pentamethyldihydrobenzofuransulfonate (PPBF), which is notable for its similarity to common sulfa drugs, but its mechanism of NKT cell activation remained unknown. Here, we demonstrate that a range of pentamethylbenzofuransulfonates (PBFs), including PPBF, activate polyclonal type II NKT cells from human donors. Whereas these sulfa drug-like molecules might have acted pharmacologically on cells, here we demonstrate direct contact between TCRs and PBF-treated CD1d complexes. Further, PBF-treated CD1d tetramers identified type II NKT cell populations expressing αβTCRs and γδTCRs, including those with variable and joining region gene usage (TRAV12-1-TRAJ6) that was conserved across donors. By trapping a CD1d-type II NKT TCR complex for direct mass-spectrometric analysis, we detected molecules that allow the binding of CD1d to TCRs, finding that both selected PBF family members and short-chain sphingomyelin lipids are present in these complexes. Furthermore, the combination of PPBF and short-chain sphingomyelin enhances CD1d tetramer staining of PPBF-reactive T cell lines over either molecule alone. This study demonstrates that nonlipidic small molecules, which resemble sulfa drugs implicated in systemic hypersensitivity and drug allergy reactions, are targeted by a polyclonal population of type II NKT cells in a CD1d-restricted manner.
Collapse
Affiliation(s)
- Catarina F Almeida
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Dylan G M Smith
- School of Chemistry, The University of Melbourne, Melbourne, VIC 3052, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Tan-Yun Cheng
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Boston, MA 02115
| | - Chris M Harpur
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Elena Batleska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Catriona V Nguyen-Robertson
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Tram Nguyen
- School of Chemistry, The University of Melbourne, Melbourne, VIC 3052, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Tamara Thelemann
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Scott J J Reddiex
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Shihan Li
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Sidonia B G Eckle
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Ildiko Van Rhijn
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Boston, MA 02115
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, University Utrecht, 3584CL Utrecht, Netherlands
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Adam P Uldrich
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - D Branch Moody
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Boston, MA 02115;
| | - Spencer J Williams
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia;
- School of Chemistry, The University of Melbourne, Melbourne, VIC 3052, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Daniel G Pellicci
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
12
|
Gherardin NA, Redmond SJ, McWilliam HEG, Almeida CF, Gourley KHA, Seneviratna R, Li S, De Rose R, Ross FJ, Nguyen-Robertson CV, Su S, Ritchie ME, Villadangos JA, Moody DB, Pellicci DG, Uldrich AP, Godfrey DI. CD36 family members are TCR-independent ligands for CD1 antigen-presenting molecules. Sci Immunol 2021; 6:6/60/eabg4176. [PMID: 34172588 DOI: 10.1126/sciimmunol.abg4176] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/01/2021] [Accepted: 05/18/2021] [Indexed: 12/22/2022]
Abstract
CD1c presents lipid-based antigens to CD1c-restricted T cells, which are thought to be a major component of the human T cell pool. However, the study of CD1c-restricted T cells is hampered by the presence of an abundantly expressed, non-T cell receptor (TCR) ligand for CD1c on blood cells, confounding analysis of TCR-mediated CD1c tetramer staining. Here, we identified the CD36 family (CD36, SR-B1, and LIMP-2) as ligands for CD1c, CD1b, and CD1d proteins and showed that CD36 is the receptor responsible for non-TCR-mediated CD1c tetramer staining of blood cells. Moreover, CD36 blockade clarified tetramer-based identification of CD1c-restricted T cells and improved identification of CD1b- and CD1d-restricted T cells. We used this technique to characterize CD1c-restricted T cells ex vivo and showed diverse phenotypic features, TCR repertoire, and antigen-specific subsets. Accordingly, this work will enable further studies into the biology of CD1 and human CD1-restricted T cells.
Collapse
Affiliation(s)
- Nicholas A Gherardin
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Samuel J Redmond
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Hamish E G McWilliam
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Catarina F Almeida
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Katherine H A Gourley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Rebecca Seneviratna
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Shihan Li
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Robert De Rose
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Fiona J Ross
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Catriona V Nguyen-Robertson
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Shian Su
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3053, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Matthew E Ritchie
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3053, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - D Branch Moody
- Division of Rheumatology, Immunity, and Inflammation, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel G Pellicci
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia.,Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
13
|
Abstract
In all human cells, human leukocyte antigen (HLA) class I glycoproteins assemble with a peptide and take it to the cell surface for surveillance by lymphocytes. These include natural killer (NK) cells and γδ T cells of innate immunity and αβ T cells of adaptive immunity. In healthy cells, the presented peptides derive from human proteins, to which lymphocytes are tolerant. In pathogen-infected cells, HLA class I expression is perturbed. Reduced HLA class I expression is detected by KIR and CD94:NKG2A receptors of NK cells. Almost any change in peptide presentation can be detected by αβ CD8+ T cells. In responding to extracellular pathogens, HLA class II glycoproteins, expressed by specialized antigen-presenting cells, present peptides to αβ CD4+ T cells. In comparison to the families of major histocompatibility complex (MHC) class I, MHC class II and αβ T cell receptors, the antigenic specificity of the γδ T cell receptors is incompletely understood.
Collapse
Affiliation(s)
- Zakia Djaoud
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA; ,
| | - Peter Parham
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA; ,
| |
Collapse
|
14
|
Paul S, Pearlman AH, Douglass J, Mog BJ, Hsiue EHC, Hwang MS, DiNapoli SR, Konig MF, Brown PA, Wright KM, Sur S, Gabelli SB, Li Y, Ghiaur G, Pardoll DM, Papadopoulos N, Bettegowda C, Kinzler KW, Zhou S, Vogelstein B. TCR β chain-directed bispecific antibodies for the treatment of T cell cancers. Sci Transl Med 2021; 13:eabd3595. [PMID: 33649188 PMCID: PMC8236299 DOI: 10.1126/scitranslmed.abd3595] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/30/2020] [Accepted: 02/03/2021] [Indexed: 12/27/2022]
Abstract
Immunotherapies such as chimeric antigen receptor (CAR) T cells and bispecific antibodies redirect healthy T cells to kill cancer cells expressing the target antigen. The pan-B cell antigen-targeting immunotherapies have been remarkably successful in treating B cell malignancies. Such therapies also result in the near-complete loss of healthy B cells, but this depletion is well tolerated by patients. Although analogous targeting of pan-T cell markers could, in theory, help control T cell cancers, the concomitant healthy T cell depletion would result in severe and unacceptable immunosuppression. Thus, therapies directed against T cell cancers require more selective targeting. Here, we describe an approach to target T cell cancers through T cell receptor (TCR) antigens. Each T cell, normal or malignant, expresses a unique TCR β chain generated from 1 of 30 TCR β chain variable gene families (TRBV1 to TRBV30). We hypothesized that bispecific antibodies targeting a single TRBV family member expressed in malignant T cells could promote killing of these cancer cells, while preserving healthy T cells that express any of the other 29 possible TRBV family members. We addressed this hypothesis by demonstrating that bispecific antibodies targeting TRBV5-5 (α-V5) or TRBV12 (α-V12) specifically lyse relevant malignant T cell lines and patient-derived T cell leukemias in vitro. Treatment with these antibodies also resulted in major tumor regressions in mouse models of human T cell cancers. This approach provides an off-the-shelf, T cell cancer selective targeting approach that preserves enough healthy T cells to maintain cellular immunity.
Collapse
Affiliation(s)
- Suman Paul
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA.
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Alexander H Pearlman
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Jacqueline Douglass
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Brian J Mog
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Emily Han-Chung Hsiue
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Michael S Hwang
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Sarah R DiNapoli
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Maximilian F Konig
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Division of Rheumatology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Patrick A Brown
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Katharine M Wright
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Surojit Sur
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Sandra B Gabelli
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yana Li
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Gabriel Ghiaur
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Nickolas Papadopoulos
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Chetan Bettegowda
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Kenneth W Kinzler
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Shibin Zhou
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Bert Vogelstein
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA.
- Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Ludwig Center and Lustgarten Laboratory, at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
15
|
Novel Molecular Insights into Human Lipid-Mediated T Cell Immunity. Int J Mol Sci 2021; 22:ijms22052617. [PMID: 33807663 PMCID: PMC7961386 DOI: 10.3390/ijms22052617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
T cells represent a critical arm of our immune defense against pathogens. Over the past two decades, considerable inroads have been made in understanding the fundamental principles underpinning the molecular presentation of peptide-based antigens by the Major Histocompatibility Complex molecules (MHC-I and II), and their molecular recognition by specialized subsets of T cells. However, some T cells can recognize lipid-based antigens presented by MHC-I-like molecules that belong to the Cluster of Differentiation 1 (CD1) family. Here, we will review the advances that have been made in the last five years to understand the molecular mechanisms orchestrating the presentation of novel endogenous and exogenous lipid-based antigens by the CD1 glycoproteins and their recognition by specific populations of CD1-reactive T cells.
Collapse
|
16
|
Edwards SC, Sutton CE, Ladell K, Grant EJ, McLaren JE, Roche F, Dash P, Apiwattanakul N, Awad W, Miners KL, Lalor SJ, Ribot JC, Baik S, Moran B, McGinley A, Pivorunas V, Dowding L, Macoritto M, Paez-Cortez J, Slavin A, Anderson G, Silva-Santos B, Hokamp K, Price DA, Thomas PG, McLoughlin RM, Mills KHG. A population of proinflammatory T cells coexpresses αβ and γδ T cell receptors in mice and humans. J Exp Med 2020; 217:133848. [PMID: 32106283 PMCID: PMC7201916 DOI: 10.1084/jem.20190834] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 11/29/2019] [Accepted: 01/17/2020] [Indexed: 12/18/2022] Open
Abstract
T cells are classically recognized as distinct subsets that express αβ or γδ TCRs. We identify a novel population of T cells that coexpress αβ and γδ TCRs in mice and humans. These hybrid αβ-γδ T cells arose in the murine fetal thymus by day 16 of ontogeny, underwent αβ TCR-mediated positive selection into CD4+ or CD8+ thymocytes, and constituted up to 10% of TCRδ+ cells in lymphoid organs. They expressed high levels of IL-1R1 and IL-23R and secreted IFN-γ, IL-17, and GM-CSF in response to canonically restricted peptide antigens or stimulation with IL-1β and IL-23. Hybrid αβ-γδ T cells were transcriptomically distinct from conventional γδ T cells and displayed a hyperinflammatory phenotype enriched for chemokine receptors and homing molecules that facilitate migration to sites of inflammation. These proinflammatory T cells promoted bacterial clearance after infection with Staphylococcus aureus and, by licensing encephalitogenic Th17 cells, played a key role in the development of autoimmune disease in the central nervous system.
Collapse
Affiliation(s)
- Sarah C Edwards
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Caroline E Sutton
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Emma J Grant
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK.,Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - James E McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Fiona Roche
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Pradyot Dash
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Nopporn Apiwattanakul
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN.,Division of Infectious Diseases, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Walid Awad
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Kelly L Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Stephen J Lalor
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Julie C Ribot
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Song Baik
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Edgbaston, Birmingham, UK
| | - Barry Moran
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Aoife McGinley
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | | | - Graham Anderson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Edgbaston, Birmingham, UK
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Karsten Hokamp
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Rachel M McLoughlin
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kingston H G Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
17
|
TCR Recognition of Peptide-MHC-I: Rule Makers and Breakers. Int J Mol Sci 2020; 22:ijms22010068. [PMID: 33374673 PMCID: PMC7793522 DOI: 10.3390/ijms22010068] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
T cells are a critical part of the adaptive immune system that are able to distinguish between healthy and unhealthy cells. Upon recognition of protein fragments (peptides), activated T cells will contribute to the immune response and help clear infection. The major histocompatibility complex (MHC) molecules, or human leukocyte antigens (HLA) in humans, bind these peptides to present them to T cells that recognise them with their surface T cell receptors (TCR). This recognition event is the first step that leads to T cell activation, and in turn can dictate disease outcomes. The visualisation of TCR interaction with pMHC using structural biology has been crucial in understanding this key event, unravelling the parameters that drive this interaction and their impact on the immune response. The last five years has been the most productive within the field, wherein half of current unique TCR-pMHC-I structures to date were determined within this time. Here, we review the new insights learned from these recent TCR-pMHC-I structures and their impact on T cell activation.
Collapse
|
18
|
Perez C, Gruber I, Arber C. Off-the-Shelf Allogeneic T Cell Therapies for Cancer: Opportunities and Challenges Using Naturally Occurring "Universal" Donor T Cells. Front Immunol 2020; 11:583716. [PMID: 33262761 PMCID: PMC7685996 DOI: 10.3389/fimmu.2020.583716] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022] Open
Abstract
Chimeric antigen receptor (CAR) engineered T cell therapies individually prepared for each patient with autologous T cells have recently changed clinical practice in the management of B cell malignancies. Even though CARs used to redirect polyclonal T cells to the tumor are not HLA restricted, CAR T cells are also characterized by their endogenous T cell receptor (TCR) repertoire. Tumor-antigen targeted TCR-based T cell therapies in clinical trials are thus far using “conventional” αβ-TCRs that recognize antigens presented as peptides in the context of the major histocompatibility complex. Thus, both CAR- and TCR-based adoptive T cell therapies (ACTs) are dictated by compatibility of the highly polymorphic HLA molecules between donors and recipients in order to avoid graft-versus-host disease and rejection. The development of third-party healthy donor derived well-characterized off-the-shelf cell therapy products that are readily available and broadly applicable is an intensive area of research. While genome engineering provides the tools to generate “universal” donor cells that can be redirected to cancers, we will focus our attention on third-party off-the-shelf strategies with T cells that are characterized by unique natural features and do not require genome editing for safe administration. Specifically, we will discuss the use of virus-specific T cells, lipid-restricted (CD1) T cells, MR1-restricted T cells, and γδ-TCR T cells. CD1- and MR1-restricted T cells are not HLA-restricted and have the potential to serve as a unique source of universal TCR sequences to be broadly applicable in TCR-based ACT as their targets are presented by the monomorphic CD1 or MR1 molecules on a wide variety of tumor types. For each cell type, we will summarize the stage of preclinical and clinical development and discuss opportunities and challenges to deliver off-the-shelf targeted cellular therapies against cancer.
Collapse
Affiliation(s)
- Cynthia Perez
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Isabelle Gruber
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Caroline Arber
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Castro CD, Boughter CT, Broughton AE, Ramesh A, Adams EJ. Diversity in recognition and function of human γδ T cells. Immunol Rev 2020; 298:134-152. [PMID: 33136294 DOI: 10.1111/imr.12930] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/17/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022]
Abstract
As interest increases in harnessing the potential power of tissue-resident cells for human health and disease, γδ T cells have been thrust into the limelight due to their prevalence in peripheral tissues, their sentinel-like phenotypes, and their unique antigen recognition capabilities. This review focuses primarily on human γδ T cells, highlighting their distinctive characteristics including antigen recognition, function, and development, with an emphasis on where they differ from their αβ T cell comparators, as well as from γδ T cell populations in the mouse. We review the antigens that have been identified thus far to regulate members of the human Vδ1 population and discuss what players are involved in transducing phosphoantigen-mediated signals to human Vγ9Vδ2 T cells. We also briefly review distinguishing features of these cells in terms of TCR signaling, use of coreceptor and costimulatory molecules and their development. These cells have great potential to be harnessed in a clinical setting, but caution must be taken to understand their unique capabilities and how they differ from the populations to which they are commonly compared.
Collapse
Affiliation(s)
- Caitlin D Castro
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Christopher T Boughter
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL, USA
| | - Augusta E Broughton
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Amrita Ramesh
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Erin J Adams
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, USA
| |
Collapse
|
20
|
Shepherd FR, McLaren JE. T Cell Immunity to Bacterial Pathogens: Mechanisms of Immune Control and Bacterial Evasion. Int J Mol Sci 2020; 21:E6144. [PMID: 32858901 PMCID: PMC7504484 DOI: 10.3390/ijms21176144] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
The human body frequently encounters harmful bacterial pathogens and employs immune defense mechanisms designed to counteract such pathogenic assault. In the adaptive immune system, major histocompatibility complex (MHC)-restricted αβ T cells, along with unconventional αβ or γδ T cells, respond to bacterial antigens to orchestrate persisting protective immune responses and generate immunological memory. Research in the past ten years accelerated our knowledge of how T cells recognize bacterial antigens and how many bacterial species have evolved mechanisms to evade host antimicrobial immune responses. Such escape mechanisms act to corrupt the crosstalk between innate and adaptive immunity, potentially tipping the balance of host immune responses toward pathological rather than protective. This review examines the latest developments in our knowledge of how T cell immunity responds to bacterial pathogens and evaluates some of the mechanisms that pathogenic bacteria use to evade such T cell immunosurveillance, to promote virulence and survival in the host.
Collapse
Affiliation(s)
| | - James E. McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
| |
Collapse
|
21
|
Thymic development of unconventional T cells: how NKT cells, MAIT cells and γδ T cells emerge. Nat Rev Immunol 2020; 20:756-770. [DOI: 10.1038/s41577-020-0345-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2020] [Indexed: 12/11/2022]
|
22
|
Yazdanifar M, Barbarito G, Bertaina A, Airoldi I. γδ T Cells: The Ideal Tool for Cancer Immunotherapy. Cells 2020; 9:E1305. [PMID: 32456316 PMCID: PMC7290982 DOI: 10.3390/cells9051305] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/27/2022] Open
Abstract
γδ T cells have recently gained considerable attention as an attractive tool for cancer adoptive immunotherapy due to their potent anti-tumor activity and unique role in immunosurveillance. The remarkable success of engineered T cells for the treatment of hematological malignancies has revolutionized the field of adoptive cell immunotherapy. Accordingly, major efforts are underway to translate this exciting technology to the treatment of solid tumors and the development of allogeneic therapies. The unique features of γδ T cells, including their major histocompatibility complex (MHC)-independent anti-cancer activity, tissue tropism, and multivalent response against a broad spectrum of the tumors, render them ideal for designing universal 'third-party' cell products, with the potential to overcome the challenges of allogeneic cell therapy. In this review, we describe the crucial role of γδ T cells in anti-tumor immunosurveillance and we summarize the different approaches used for the ex vivo and in vivo expansion of γδ T cells suitable for the development of novel strategies for cancer therapy. We further discuss the different transduction strategies aiming at redirecting or improving the function of γδ T cells, as well as, the considerations for the clinical applications.
Collapse
Affiliation(s)
- Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94305, USA; (M.Y.); (G.B.)
| | - Giulia Barbarito
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94305, USA; (M.Y.); (G.B.)
| | - Alice Bertaina
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94305, USA; (M.Y.); (G.B.)
| | - Irma Airoldi
- Laboratorio Cellule Staminali post-natali e Terapie Cellulari, IRCCS G. Gaslini, Via G. Gaslini 5, 16147 Genova, Italy
| |
Collapse
|
23
|
Ramírez-Salinas GL, García-Machorro J, Rojas-Hernández S, Campos-Rodríguez R, de Oca ACM, Gomez MM, Luciano R, Zimic M, Correa-Basurto J. Bioinformatics design and experimental validation of influenza A virus multi-epitopes that induce neutralizing antibodies. Arch Virol 2020; 165:891-911. [PMID: 32060794 PMCID: PMC7222995 DOI: 10.1007/s00705-020-04537-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/11/2019] [Indexed: 02/01/2023]
Abstract
Pandemics caused by influenza A virus (IAV) are responsible for the deaths of millions of humans around the world. One of these pandemics occurred in Mexico in 2009. Despite the impact of IAV on human health, there is no effective vaccine. Gene mutations and translocation of genome segments of different IAV subtypes infecting a single host cell make the development of a universal vaccine difficult. The design of immunogenic peptides using bioinformatics tools could be an interesting strategy to increase the success of vaccines. In this work, we used the predicted amino acid sequences of the neuraminidase (NA) and hemagglutinin (HA) proteins of different IAV subtypes to perform multiple alignments, epitope predictions, molecular dynamics simulations, and experimental validation. Peptide selection was based on the following criteria: promiscuity, protein surface exposure, and the degree of conservation among different medically relevant IAV strains. These peptides were tested using immunological assays to test their ability to induce production of antibodies against IAV. We immunized rabbits and mice and measured the levels of IgG and IgA antibodies in serum samples and nasal washes. Rabbit antibodies against the peptides P11 and P14 (both of which are hybrids of NA and HA) recognized HA from both group 1 (H1, H2, and H5) and group 2 (H3 and H7) IAV and also recognized the purified NA protein from the viral stock (influenza A Puerto Rico/916/34). IgG antibodies from rabbits immunized with P11 and P14 were capable of recognizing viral particles and inhibited virus hemagglutination. Additionally, intranasal immunization of mice with P11 and P14 induced specific IgG and IgA antibodies in serum and nasal mucosa, respectively. Interestingly, the IgG antibodies were found to have neutralizing capability. In conclusion, the peptides designed through in silico studies were validated in experimental assays.
Collapse
Affiliation(s)
- G Lizbeth Ramírez-Salinas
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotécnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11340, México
| | - Jazmín García-Machorro
- Laboratorio de medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11340, México.
| | - Saúl Rojas-Hernández
- Laboratorio de Inmunología celular, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11340, México
| | - Rafael Campos-Rodríguez
- Laboratorio de Bioquímica. Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11340, México
| | - Arturo Contis-Montes de Oca
- Laboratorio de Inmunología celular, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11340, México
| | - Miguel Medina Gomez
- Laboratorio de medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11340, México
| | - Rocío Luciano
- Laboratorio de medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11340, México
| | - Mirko Zimic
- Laboratorio de Bioinformática y Biología Molecular, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotécnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11340, México.
| |
Collapse
|
24
|
Gutierrez-Arcelus M, Teslovich N, Mola AR, Polidoro RB, Nathan A, Kim H, Hannes S, Slowikowski K, Watts GFM, Korsunsky I, Brenner MB, Raychaudhuri S, Brennan PJ. Lymphocyte innateness defined by transcriptional states reflects a balance between proliferation and effector functions. Nat Commun 2019; 10:687. [PMID: 30737409 PMCID: PMC6368609 DOI: 10.1038/s41467-019-08604-4] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 01/21/2019] [Indexed: 02/06/2023] Open
Abstract
How innate T cells (ITC), including invariant natural killer T (iNKT) cells, mucosal-associated invariant T (MAIT) cells, and γδ T cells, maintain a poised effector state has been unclear. Here we address this question using low-input and single-cell RNA-seq of human lymphocyte populations. Unbiased transcriptomic analyses uncover a continuous ‘innateness gradient’, with adaptive T cells at one end, followed by MAIT, iNKT, γδ T and natural killer cells at the other end. Single-cell RNA-seq reveals four broad states of innateness, and heterogeneity within canonical innate and adaptive populations. Transcriptional and functional data show that innateness is characterized by pre-formed mRNA encoding effector functions, but impaired proliferation marked by decreased baseline expression of ribosomal genes. Together, our data shed new light on the poised state of ITC, in which innateness is defined by a transcriptionally-orchestrated trade-off between rapid cell growth and rapid effector function. Innate T cells (ITC) contain many subsets and are poised to promptly respond to antigens and pathogens, but how this poised state is maintained is still unclear. Here the authors perform single-cell RNA-seq to align the various ITC subsets along an ‘innateness gradient’ that is associated with changes in proliferation and effector functions.
Collapse
Affiliation(s)
- Maria Gutierrez-Arcelus
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA, 02115.,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA.,Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nikola Teslovich
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA, 02115.,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA.,Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Alex R Mola
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Rafael B Polidoro
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Aparna Nathan
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA, 02115.,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA.,Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hyun Kim
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA, 02115.,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA.,Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Susan Hannes
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA, 02115.,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA.,Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kamil Slowikowski
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA, 02115.,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA.,Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gerald F M Watts
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ilya Korsunsky
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA, 02115.,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA.,Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael B Brenner
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Soumya Raychaudhuri
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA, 02115. .,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA. .,Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA. .,Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA. .,Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PL, UK.
| | - Patrick J Brennan
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
25
|
Unappreciated diversity within the pool of CD1d-restricted T cells. Semin Cell Dev Biol 2018; 84:42-47. [DOI: 10.1016/j.semcdb.2017.11.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/11/2022]
|
26
|
Godfrey DI, Le Nours J, Andrews DM, Uldrich AP, Rossjohn J. Unconventional T Cell Targets for Cancer Immunotherapy. Immunity 2018; 48:453-473. [PMID: 29562195 DOI: 10.1016/j.immuni.2018.03.009] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 02/07/2023]
Abstract
Most studies on the immunotherapeutic potential of T cells have focused on CD8 and CD4 T cells that recognize peptide antigens (Ag) presented by polymorphic major histocompatibility complex (MHC) class I and MHC class II molecules, respectively. However, unconventional T cells, which interact with MHC class Ib and MHC-I like molecules, are also implicated in tumor immunity, although their role therein is unclear. These include unconventional T cells targeting MHC class Ib molecules such as HLA-E and its murine ortholog Qa-1b, natural killer T (NKT) cells, mucosal associated invariant T (MAIT) cells, and γδ T cells. Here, we review the current understanding of the roles of these unconventional T cells in tumor immunity and discuss why further studies into the immunotherapeutic potential of these cells is warranted.
Collapse
Affiliation(s)
- Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Jérôme Le Nours
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Daniel M Andrews
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia; Institute of Infection and Immunity, Cardiff University, School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
27
|
Trottein F, Paget C. Natural Killer T Cells and Mucosal-Associated Invariant T Cells in Lung Infections. Front Immunol 2018; 9:1750. [PMID: 30116242 PMCID: PMC6082944 DOI: 10.3389/fimmu.2018.01750] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
The immune system has been traditionally divided into two arms called innate and adaptive immunity. Typically, innate immunity refers to rapid defense mechanisms that set in motion within minutes to hours following an insult. Conversely, the adaptive immune response emerges after several days and relies on the innate immune response for its initiation and subsequent outcome. However, the recent discovery of immune cells displaying merged properties indicates that this distinction is not mutually exclusive. These populations that span the innate-adaptive border of immunity comprise, among others, CD1d-restricted natural killer T cells and MR1-restricted mucosal-associated invariant T cells. These cells have the unique ability to swiftly activate in response to non-peptidic antigens through their T cell receptor and/or to activating cytokines in order to modulate many aspects of the immune response. Despite they recirculate all through the body via the bloodstream, these cells mainly establish residency at barrier sites including lungs. Here, we discuss the current knowledge into the biology of these cells during lung (viral and bacterial) infections including activation mechanisms and functions. We also discuss future strategies targeting these cell types to optimize immune responses against respiratory pathogens.
Collapse
Affiliation(s)
- François Trottein
- Univ. Lille, U1019 – UMR 8204 – CIIL – Centre d’Infection et d’Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Christophe Paget
- Institut National de la Santé et de la Recherche Médicale U1100, Centre d’Etude des Pathologies Respiratoires (CEPR), Tours, France
- Université de Tours, Tours, France
| |
Collapse
|
28
|
Cotton RN, Shahine A, Rossjohn J, Moody DB. Lipids hide or step aside for CD1-autoreactive T cell receptors. Curr Opin Immunol 2018; 52:93-99. [PMID: 29738961 DOI: 10.1016/j.coi.2018.04.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/17/2018] [Indexed: 12/31/2022]
Abstract
Peptide and lipid antigens are presented to T cells when bound to MHC or CD1 proteins, respectively. The general paradigm of T cell antigen recognition is that T cell receptors (TCRs) co-recognize an epitope comprised of the antigen and antigen presenting molecule. Here we review the latest studies in which T cells operate outside the co-recognition paradigm: TCRs can broadly contact CD1 itself, but not the carried lipid. The essential structural feature in these new mechanisms is a large 'antigen free' zone on the outer surface of certain antigen presenting molecules. Whereas peptides dominate the exposed surface of MHC-peptide complexes, all human CD1 proteins have a closed, antigen-free surface, which is known as the A' roof. These new structural models help to interpret recent biological studies of CD1 autoreactive T cells in vivo, which have now been broadly observed in studies on TCR-transgenic mice, healthy humans and patients with autoimmune disease.
Collapse
Affiliation(s)
- Rachel N Cotton
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Adam Shahine
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia; Institute of Infection and Immunity, Cardiff University, School of Medicine, Heath Park, Cardiff CF14 4XN, United Kingdom
| | - D Branch Moody
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Gras S, Van Rhijn I, Shahine A, Le Nours J. Molecular recognition of microbial lipid-based antigens by T cells. Cell Mol Life Sci 2018; 75:1623-1639. [PMID: 29340708 PMCID: PMC6328055 DOI: 10.1007/s00018-018-2749-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/17/2017] [Accepted: 01/08/2018] [Indexed: 02/06/2023]
Abstract
The immune system has evolved to protect hosts from pathogens. T cells represent a critical component of the immune system by their engagement in host defence mechanisms against microbial infections. Our knowledge of the molecular recognition by T cells of pathogen-derived peptidic antigens that are presented by the major histocompatibility complex glycoproteins is now well established. However, lipids represent an additional, distinct chemical class of molecules that when presented by the family of CD1 antigen-presenting molecules can serve as antigens, and be recognized by specialized subsets of T cells leading to antigen-specific activation. Over the past decades, numerous CD1-presented self- and bacterial lipid-based antigens have been isolated and characterized. However, our understanding at the molecular level of T cell immunity to CD1 molecules presenting microbial lipid-based antigens is still largely unexplored. Here, we review the insights and the molecular basis underpinning the recognition of microbial lipid-based antigens by T cells.
Collapse
Affiliation(s)
- Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia
| | - Ildiko Van Rhijn
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital/Harvard Medical School, Boston, USA
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, University Utrecht, Utrecht, The Netherlands
| | - Adam Shahine
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia
| | - Jérôme Le Nours
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
30
|
Simões AE, Di Lorenzo B, Silva-Santos B. Molecular Determinants of Target Cell Recognition by Human γδ T Cells. Front Immunol 2018; 9:929. [PMID: 29755480 PMCID: PMC5934422 DOI: 10.3389/fimmu.2018.00929] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/16/2018] [Indexed: 12/22/2022] Open
Abstract
The unique capabilities of gamma-delta (γδ) T cells to recognize cells under stressed conditions, particularly infected or transformed cells, and killing them or regulating the immune response against them, paved the way to the development of promising therapeutic strategies for cancer and infectious diseases. From a mechanistic standpoint, numerous studies have unveiled a remarkable flexibility of γδ T cells in employing their T cell receptor and/or NK cell receptors for target cell recognition, even if the relevant ligands often remain uncertain. Here, we review the accumulated knowledge on the diverse mechanisms of target cell recognition by γδ T cells, focusing on human γδ T cells, to provide an integrated perspective of their therapeutic potential in cancer and infectious diseases.
Collapse
Affiliation(s)
- André E Simões
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Biagio Di Lorenzo
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal.,Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Bruno Silva-Santos
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
31
|
Rosskopf S, Leitner J, Paster W, Morton LT, Hagedoorn RS, Steinberger P, Heemskerk MHM. A Jurkat 76 based triple parameter reporter system to evaluate TCR functions and adoptive T cell strategies. Oncotarget 2018; 9:17608-17619. [PMID: 29707134 PMCID: PMC5915142 DOI: 10.18632/oncotarget.24807] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
Adoptive T cell therapy using TCR transgenic autologous T cells has shown great potential for the treatment of tumor patients. Thorough characterization of genetically reprogrammed T cells is necessary to optimize treatment success. Here, we describe the generation of triple parameter reporter T cells based on the Jurkat 76 T cell line for the evaluation of TCR and chimeric antigen receptor functions as well as adoptive T cell strategies. This Jurkat subline is devoid of endogenous TCR alpha and TCR beta chains, thereby circumventing the problem of TCR miss-pairing and unexpected specificities. The resultant reporter cells allow simultaneous determination of the activity of the transcription factors NF-κB, NFAT and AP-1 that play key roles in T cell activation. Human TCRs directed against tumor and virus antigens were introduced and reporter responses were determined using tumor cell lines endogenously expressing the antigens of interest or via addition of antigenic peptides. Finally, we demonstrate that coexpression of adhesion molecules like CD2 and CD226 as well as CD28 chimeric receptors represents an effective strategy to augment the response of TCR-transgenic reporters to cells presenting cognate antigens.
Collapse
Affiliation(s)
- Sandra Rosskopf
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Paster
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Laura T Morton
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Renate S Hagedoorn
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
32
|
Vermijlen D, Gatti D, Kouzeli A, Rus T, Eberl M. γδ T cell responses: How many ligands will it take till we know? Semin Cell Dev Biol 2018; 84:75-86. [PMID: 29402644 DOI: 10.1016/j.semcdb.2017.10.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 09/06/2017] [Accepted: 10/09/2017] [Indexed: 12/20/2022]
Abstract
γδ T cells constitute a sizeable and non-redundant fraction of the total T cell pool in all jawed vertebrates, but in contrast to conventional αβ T cells they are not restricted by classical MHC molecules. Progress in our understanding of the role of γδ T cells in the immune system has been hampered, and is being hampered, by the considerable lack of knowledge regarding the antigens γδ T cells respond to. The past few years have seen a wealth of data regarding the TCR repertoires of distinct γδ T cell populations and a growing list of confirmed and proposed molecules that are recognised by γδ T cells in different species. Yet, the physiological contexts underlying the often restricted TCR usage and the chemical diversity of γδ T cell ligands remain largely unclear, and only few structural studies have confirmed direct ligand recognition by the TCR. We here review the latest progress in the identification and validation of putative γδ T cell ligands and discuss the implications of such findings for γδ T cell responses in health and disease.
Collapse
Affiliation(s)
- David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics and Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Belgium.
| | - Deborah Gatti
- Department of Pharmacotherapy and Pharmaceutics and Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Belgium
| | - Ariadni Kouzeli
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Teja Rus
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom; Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
33
|
Conserved Vδ1 Binding Geometry in a Setting of Locus-Disparate pHLA Recognition by δ/αβ T Cell Receptors (TCRs): Insight into Recognition of HIV Peptides by TCRs. J Virol 2017; 91:JVI.00725-17. [PMID: 28615212 DOI: 10.1128/jvi.00725-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 06/02/2017] [Indexed: 11/20/2022] Open
Abstract
Given the limited set of T cell receptor (TCR) V genes that are used to create TCRs that are reactive to different ligands, such as major histocompatibility complex (MHC) class I, MHC class II, and MHC-like proteins (for example, MIC molecules and CD1 molecules), the Vδ1 segment can be rearranged with Dδ-Jδ-Cδ or Jα-Cα segments to form classical γδTCRs or uncommon αβTCRs using a Vδ1 segment (δ/αβTCR). Here we have determined two complex structures of the δ/αβTCRs (S19-2 and TU55) bound to different locus-disparate MHC class I molecules with HIV peptides (HLA-A*2402-Nef138-10 and HLA-B*3501-Pol448-9). The overall binding modes resemble those of classical αβTCRs but display a strong tilt binding geometry of the Vδ1 domain toward the HLA α1 helix, due to a conserved extensive interaction between the CDR1δ loop and the N-terminal region of the α1 helix (mainly in position 62). The aromatic amino acids of the CDR1δ loop exploit different conformations ("aromatic ladder" or "aromatic hairpin") to accommodate distinct MHC helical scaffolds. This tolerance helps to explain how a particular TCR V region can similarly dock onto multiple MHC molecules and thus may potentially explain the nature of TCR cross-reactivity. In addition, the length of the CDR3δ loop could affect the extent of tilt binding of the Vδ1 domain, and adaptively, the pairing Vβ domains adjust their mass centers to generate differential MHC contacts, hence probably ensuring TCR specificity for a certain peptide-MHC class I (pMHC-I). Our data have provided further structural insights into the TCR recognition of classical pMHC-I molecules, unifying cross-reactivity and specificity.IMPORTANCE The specificity of αβ T cell recognition is determined by the CDR loops of the αβTCR, and the general mode of binding of αβTCRs to pMHC has been established over the last decade. Due to the intrinsic genomic structure of the TCR α/δ chain locus, some Vδ segments can rearrange with the Cα segment, forming a hybrid VδCαVβCβ TCR, the δ/αβTCR. However, the basis for the molecular recognition of such TCRs of their ligands is elusive. Here an αβTCR using the Vδ1 segment, S19-2, was isolated from an HIV-infected patient in an HLA-A*24:02-restricted manner. We then solved the crystal structures of the S19-2 TCR and another δ/αβTCR, TU55, bound to their respective ligands, revealing a conserved Vδ1 binding feature. Further binding kinetics analysis revealed that the S19-2 and TU55 TCRs bind pHLA very tightly and in a long-lasting manner. Our results illustrate the mode of binding of a TCR using the Vδ1 segment to its ligand, virus-derived pHLA.
Collapse
|
34
|
Zajonc DM. The CD1 family: serving lipid antigens to T cells since the Mesozoic era. Immunogenetics 2016; 68:561-76. [PMID: 27368414 DOI: 10.1007/s00251-016-0931-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/22/2016] [Indexed: 12/20/2022]
Abstract
Class I-like CD1 molecules are in a family of antigen-presenting molecules that bind lipids and lipopeptides, rather than peptides for immune surveillance by T cells. Since CD1 lacks the high degree of polymorphism found in their major histocompatibility complex (MHC) class I molecules, different species express different numbers of CD1 isotypes, likely to be able to present structurally diverse classes of lipid antigens. In this review, we will present a historical overview of the structures of the different human CD1 isotypes and also discuss species-specific adaptations of the lipid-binding groove. We will discuss how single amino acid changes alter the shape and volume of the CD1 binding groove, how these minor changes can give rise to different numbers of binding pockets, and how these pockets affect the lipid repertoire that can be presented by any given CD1 protein. We will compare the structures of various lipid antigens and finally, we will discuss recognition of CD1-presented lipid antigens by antigen receptors on T cells (TCRs).
Collapse
Affiliation(s)
- Dirk M Zajonc
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, CA, 92037, USA. .,Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000, Ghent, Belgium.
| |
Collapse
|
35
|
Deciphering the clinical relevance of allo-human leukocyte antigen cross-reactivity in mediating alloimmunity following transplantation. Curr Opin Organ Transplant 2016; 21:29-39. [PMID: 26575852 DOI: 10.1097/mot.0000000000000264] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW Despite a growing awareness regarding the potential of cross-reactive virus-specific memory T cells to mediate alloimmunity, there has been limited clinical evaluation on allograft immunopathology. This review will explore published models of human T-cell cross-reactivity and discuss criteria required to drive this mechanism as a contributing cause of allograft dysfunction in transplantation. RECENT FINDINGS Published models of human allogeneic (allo)-human leukocyte antigen (HLA) cross-reactivity have enabled dissection of the cross-reactive T cell receptor/peptide/major histocompatibility complex (TCR/peptide/MHC) interaction. In many of the models, the cross-reactive T cells express a unique TCR, although the relevance of a public cross-reactive TCR repertoire has yet to be determined. Equally, allopeptide identity, a vital component driving cross-recognition, remains unknown in the majority of models thereby prompting further characterization utilizing novel technologies. Although clinical studies examining the presence and impact of specific cross-reactive virus-specific T cells have been minimally explored, the existing data suggest that there may be a marginal set of requirements that need to be satisfied before the potentially damaging effects of allo-HLA cross-reactivity can be realized. SUMMARY Our understanding of allo-HLA cross-reactivity continues to evolve as improved technology and novel strategies allow us to better question the contribution of allo-HLA cross-reactivity in clinically relevant allograft dysfunction.
Collapse
|
36
|
Abstract
CD1- and MHC-related molecule-1 (MR1)-restricted T lymphocytes recognize nonpeptidic antigens, such as lipids and small metabolites, and account for a major fraction of circulating and tissue-resident T cells. They represent a readily activated, long-lasting population of effector cells and contribute to the early phases of immune response, orchestrating the function of other cells. This review addresses the main aspects of their immunological functions, including antigen and T cell receptor repertoires, mechanisms of nonpeptidic antigen presentation, and the current evidence for their participation in human and experimental diseases.
Collapse
Affiliation(s)
- Lucia Mori
- Department of Biomedicine, Basel University Hospital and Basel University, CH-4031 Basel, Switzerland; , , .,Singapore Immunology Network, A*STAR, 138648 Singapore
| | - Marco Lepore
- Department of Biomedicine, Basel University Hospital and Basel University, CH-4031 Basel, Switzerland; , ,
| | - Gennaro De Libero
- Department of Biomedicine, Basel University Hospital and Basel University, CH-4031 Basel, Switzerland; , , .,Singapore Immunology Network, A*STAR, 138648 Singapore
| |
Collapse
|
37
|
Atypical natural killer T-cell receptor recognition of CD1d-lipid antigens. Nat Commun 2016; 7:10570. [PMID: 26875526 PMCID: PMC4756352 DOI: 10.1038/ncomms10570] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/29/2015] [Indexed: 11/25/2022] Open
Abstract
Crucial to Natural Killer T (NKT) cell function is the interaction between their T-cell receptor (TCR) and CD1d-antigen complex. However, the diversity of the NKT cell repertoire and the ensuing interactions with CD1d-antigen remain unclear. We describe an atypical population of CD1d–α-galactosylceramide (α-GalCer)-reactive human NKT cells that differ markedly from the prototypical TRAV10-TRAJ18-TRBV25-1+ type I NKT cell repertoire. These cells express a range of TCR α- and β-chains that show differential recognition of glycolipid antigens. Two atypical NKT TCRs (TRAV21-TRAJ8-TRBV7–8 and TRAV12-3-TRAJ27-TRBV6-5) bind orthogonally over the A′-pocket of CD1d, adopting distinct docking modes that contrast with the docking mode of all type I NKT TCR-CD1d-antigen complexes. Moreover, the interactions with α-GalCer differ between the type I and these atypical NKT TCRs. Accordingly, diverse NKT TCR repertoire usage manifests in varied docking strategies and specificities towards CD1d–α-GalCer and related antigens, thus providing far greater scope for diverse glycolipid antigen recognition. The invariant αβTCR of type I NKT cells recognizes a lipid α-GalCer presented by CD1d. Here the authors describe atypical α-GalCer-reactive NKT cells with diverse TCRs, which bind to CD1d-α-GalCer in a manner distinct from type I NKT cells, thus unveiling greater diversity in lipid antigen recognition.
Collapse
|
38
|
Roy S, Ly D, Castro CD, Li NS, Hawk AJ, Altman JD, Meredith SC, Piccirilli JA, Moody DB, Adams EJ. Molecular Analysis of Lipid-Reactive Vδ1 γδ T Cells Identified by CD1c Tetramers. THE JOURNAL OF IMMUNOLOGY 2016; 196:1933-42. [PMID: 26755823 DOI: 10.4049/jimmunol.1502202] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/05/2015] [Indexed: 02/06/2023]
Abstract
CD1c is abundantly expressed on human dendritic cells (DC) and B cells, where it binds and displays lipid Ags to T cells. In this study, we report that CD1c tetramers carrying Mycobacterium tuberculosis phosphomycoketide bind γδ TCRs. An unbiased method of ligand-based TCR selection detects interactions only with Vδ1(+) TCRs, and mutational analyses demonstrate a role of the Vδ1 domain during recognition. These results strengthen evidence for a role of CD1c in the γδ T cell response, providing biophysical evidence for CD1c-γδ TCR interactions and a named foreign Ag. Surprisingly, TCRs also bind CD1c complexes formed with diverse lipids such as lysophosphatidylcholine, sulfatide, or mannosyl-phosophomycoketide, but not lipopeptide ligands. Dissection of TCR interactions with CD1c carrying foreign Ags, permissive ligands, and nonpermissive lipid ligands clarifies the molecular basis of the frequently observed but poorly understood phenomenon of mixed self- and foreign Ag reactivity in the CD1 system.
Collapse
Affiliation(s)
- Sobhan Roy
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Dalam Ly
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115; Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Caitlin D Castro
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637; Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Nan-Sheng Li
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Andrew J Hawk
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637; Department of Pathology, University of Chicago, Chicago, IL 60637
| | - John D Altman
- Department of Microbiology and Immunology, Emory Vaccine Center at Yerkes, Emory University School of Medicine, Atlanta, GA 30329; and
| | - Stephen C Meredith
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637; Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Joseph A Piccirilli
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637; Department of Chemistry, University of Chicago, Chicago, IL 60637
| | - D Branch Moody
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115;
| | - Erin J Adams
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637; Committee on Immunology, University of Chicago, Chicago, IL 60637;
| |
Collapse
|
39
|
Van Rhijn I, Moody DB. Donor Unrestricted T Cells: A Shared Human T Cell Response. THE JOURNAL OF IMMUNOLOGY 2015; 195:1927-32. [PMID: 26297792 DOI: 10.4049/jimmunol.1500943] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The now-famous term "restriction" derived from experiments in which T cells from Donor A failed to recognize Ags presented by cells from Donor B. Restriction results from interdonor variation in MHC genes. Donor restriction dominates immunologists' thinking about the T cell response because it governs organ transplantation and hinders the discovery of disease-associated Ags. However, other T cells can be considered "donor unrestricted" because their targets, CD1a, CD1b, CD1c, CD1d, or MR1, are expressed in a similar form among all humans. A striking feature of donor unrestricted T cells is the expression of invariant TCRs with nearly species-wide distribution. In this article, we review new evidence that donor unrestricted T cells are common in humans. NKT cells, mucosa-associated invariant T cells, and germline-encoded mycolyl-reactive T cells operate outside of the familiar principles of the MHC system, providing a broader picture of T cell function and new opportunities for therapy.
Collapse
Affiliation(s)
- Ildiko Van Rhijn
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584CL Utrecht, the Netherlands
| | - D Branch Moody
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| |
Collapse
|
40
|
Legut M, Cole DK, Sewell AK. The promise of γδ T cells and the γδ T cell receptor for cancer immunotherapy. Cell Mol Immunol 2015; 12:656-68. [PMID: 25864915 PMCID: PMC4716630 DOI: 10.1038/cmi.2015.28] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 03/01/2015] [Indexed: 12/13/2022] Open
Abstract
γδ T cells form an important part of adaptive immune responses against infections and malignant transformation. The molecular targets of human γδ T cell receptors (TCRs) remain largely unknown, but recent studies have confirmed the recognition of phosphorylated prenyl metabolites, lipids in complex with CD1 molecules and markers of cellular stress. All of these molecules are upregulated on various cancer types, highlighting the potential importance of the γδ T cell compartment in cancer immunosurveillance and paving the way for the use of γδ TCRs in cancer therapy. Ligand recognition by the γδ TCR often requires accessory/co-stimulatory stress molecules on both T cells and target cells; this cellular stress context therefore provides a failsafe against harmful self-reactivity. Unlike αβ T cells, γδ T cells recognise their targets irrespective of HLA haplotype and therefore offer exciting possibilities for off-the-shelf, pan-population cancer immunotherapies. Here, we present a review of known ligands of human γδ T cells and discuss the promise of harnessing these cells for cancer treatment.
Collapse
MESH Headings
- Antigen Presentation
- Antigens, CD1/genetics
- Antigens, CD1/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Clinical Trials as Topic
- Gene Expression Regulation, Neoplastic/immunology
- Hemiterpenes/immunology
- Humans
- Immunotherapy/methods
- Ligands
- Models, Molecular
- Monitoring, Immunologic
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Organophosphorus Compounds/immunology
- Phosphorylation
- Protein Structure, Tertiary
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- Mateusz Legut
- Division of Infection and Immunity and Systems Immunity University Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - David K Cole
- Division of Infection and Immunity and Systems Immunity University Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - Andrew K Sewell
- Division of Infection and Immunity and Systems Immunity University Research Institute, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
41
|
Van Rhijn I, Godfrey DI, Rossjohn J, Moody DB. Lipid and small-molecule display by CD1 and MR1. Nat Rev Immunol 2015; 15:643-54. [PMID: 26388332 PMCID: PMC6944187 DOI: 10.1038/nri3889] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The antigen-presenting molecules CD1 and MHC class I-related protein (MR1) display lipids and small molecules to T cells. The antigen display platforms in the four CD1 proteins are laterally asymmetrical, so that the T cell receptor (TCR)-binding surfaces are comprised of roofs and portals, rather than the long grooves seen in the MHC antigen-presenting molecules. TCRs can bind CD1 proteins with left-sided or right-sided footprints, creating unexpected modes of antigen recognition. The use of tetramers of human CD1a, CD1b, CD1c or MR1 proteins now allows detailed analysis of the human T cell repertoire, which has revealed new invariant TCRs that bind CD1b molecules and are different from those that define natural killer T cells and mucosal-associated invariant T cells.
Collapse
MESH Headings
- Antigen Presentation/immunology
- Antigens, CD1/chemistry
- Antigens, CD1/immunology
- Antigens, CD1/metabolism
- Histocompatibility Antigens Class I/chemistry
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Humans
- Lipids/chemistry
- Lipids/immunology
- Minor Histocompatibility Antigens
- Models, Molecular
- Protein Binding/immunology
- Protein Structure, Tertiary
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Ildiko Van Rhijn
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - D Branch Moody
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
42
|
Abstract
Over two decades ago, it was discovered that the human T-cell repertoire contains T cells that do not recognize peptide antigens in the context of MHC molecules but instead respond to lipid antigens presented by CD1 antigen-presenting molecules. The ability of T cells to 'see' lipid antigens bound to CD1 enables these lymphocytes to sense changes in the lipid composition of cells and tissues as a result of infections, inflammation, or malignancies. Although foreign lipid antigens have been shown to function as antigens for CD1-restricted T cells, many CD1-restricted T cells do not require foreign antigens for activation but instead can be activated by self-lipids presented by CD1. This review highlights recent developments in the field, including the identification of common mammalian lipids that function as autoantigens for αβ and γδ T cells, a novel mode of T-cell activation whereby CD1a itself rather than lipids serves as the autoantigen, and various mechanisms by which the activation of CD1-autoreactive T cells is regulated. As CD1 can induce T-cell effector functions in the absence of foreign antigens, multiple mechanisms are in place to regulate this self-reactivity, and stimulatory CD1-lipid complexes appear to be tightly controlled in space and time.
Collapse
|
43
|
Wolf BJ, Tatituri RVV, Almeida CF, Le Nours J, Bhowruth V, Johnson D, Uldrich AP, Hsu FF, Brigl M, Besra GS, Rossjohn J, Godfrey DI, Brenner MB. Identification of a Potent Microbial Lipid Antigen for Diverse NKT Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:2540-51. [PMID: 26254340 DOI: 10.4049/jimmunol.1501019] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/10/2015] [Indexed: 01/17/2023]
Abstract
Semi-invariant/type I NKT cells are a well-characterized CD1d-restricted T cell subset. The availability of potent Ags and tetramers for semi-invariant/type I NKT cells allowed this population to be extensively studied and revealed their central roles in infection, autoimmunity, and tumor immunity. In contrast, diverse/type II NKT (dNKT) cells are poorly understood because the lipid Ags that they recognize are largely unknown. We sought to identify dNKT cell lipid Ag(s) by interrogating a panel of dNKT mouse cell hybridomas with lipid extracts from the pathogen Listeria monocytogenes. We identified Listeria phosphatidylglycerol as a microbial Ag that was significantly more potent than a previously characterized dNKT cell Ag, mammalian phosphatidylglycerol. Further, although mammalian phosphatidylglycerol-loaded CD1d tetramers did not stain dNKT cells, the Listeria-derived phosphatidylglycerol-loaded tetramers did. The structure of Listeria phosphatidylglycerol was distinct from mammalian phosphatidylglycerol because it contained shorter, fully-saturated anteiso fatty acid lipid tails. CD1d-binding lipid-displacement studies revealed that the microbial phosphatidylglycerol Ag binds significantly better to CD1d than do counterparts with the same headgroup. These data reveal a highly potent microbial lipid Ag for a subset of dNKT cells and provide an explanation for its increased Ag potency compared with the mammalian counterpart.
Collapse
Affiliation(s)
- Benjamin J Wolf
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Raju V V Tatituri
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Catarina F Almeida
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging at University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jérôme Le Nours
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Veemal Bhowruth
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Darryl Johnson
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging at University of Melbourne, Parkville, Victoria 3010, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging at University of Melbourne, Parkville, Victoria 3010, Australia
| | - Fong-Fu Hsu
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University, St. Louis, MO 63110
| | - Manfred Brigl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jamie Rossjohn
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, United Kingdom
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging at University of Melbourne, Parkville, Victoria 3010, Australia
| | - Michael B Brenner
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115;
| |
Collapse
|
44
|
Macho-Fernandez E, Brigl M. The Extended Family of CD1d-Restricted NKT Cells: Sifting through a Mixed Bag of TCRs, Antigens, and Functions. Front Immunol 2015; 6:362. [PMID: 26284062 PMCID: PMC4517383 DOI: 10.3389/fimmu.2015.00362] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/04/2015] [Indexed: 01/21/2023] Open
Abstract
Natural killer T (NKT) cells comprise a family of specialized T cells that recognize lipid antigens presented by CD1d. Based on their T cell receptor (TCR) usage and antigen specificities, CD1d-restricted NKT cells have been divided into two main subsets: type I NKT cells that use a canonical invariant TCR α-chain and recognize α-galactosylceramide (α-GalCer), and type II NKT cells that use a more diverse αβ TCR repertoire and do not recognize α-GalCer. In addition, α-GalCer-reactive NKT cells that use non-canonical αβ TCRs and CD1d-restricted T cells that use γδ or δ/αβ TCRs have recently been identified, revealing further diversity among CD1d-restricted T cells. Importantly, in addition to their distinct antigen specificities, functional differences are beginning to emerge between the different members of the CD1d-restricted T cell family. In this review, while using type I NKT cells as comparison, we will focus on type II NKT cells and the other non-invariant CD1d-restricted T cell subsets, and discuss our current understanding of the antigens they recognize, the formation of stimulatory CD1d/antigen complexes, the modes of TCR-mediated antigen recognition, and the mechanisms and consequences of their activation that underlie their function in antimicrobial responses, anti-tumor immunity, and autoimmunity.
Collapse
Affiliation(s)
- Elodie Macho-Fernandez
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Manfred Brigl
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Attaf M, Legut M, Cole DK, Sewell AK. The T cell antigen receptor: the Swiss army knife of the immune system. Clin Exp Immunol 2015; 181:1-18. [PMID: 25753381 PMCID: PMC4469151 DOI: 10.1111/cei.12622] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2015] [Indexed: 01/01/2023] Open
Abstract
The mammalian T cell receptor (TCR) orchestrates immunity by responding to many billions of different ligands that it has never encountered before and cannot adapt to at the protein sequence level. This remarkable receptor exists in two main heterodimeric isoforms: αβ TCR and γδ TCR. The αβ TCR is expressed on the majority of peripheral T cells. Most αβ T cells recognize peptides, derived from degraded proteins, presented at the cell surface in molecular cradles called major histocompatibility complex (MHC) molecules. Recent reports have described other αβ T cell subsets. These 'unconventional' T cells bear TCRs that are capable of recognizing lipid ligands presented in the context of the MHC-like CD1 protein family or bacterial metabolites bound to the MHC-related protein 1 (MR1). γδ T cells constitute a minority of the T cell pool in human blood, but can represent up to half of total T cells in tissues such as the gut and skin. The identity of the preferred ligands for γδ T cells remains obscure, but it is now known that this receptor can also functionally engage CD1-lipid, or immunoglobulin (Ig) superfamily proteins called butyrophilins in the presence of pyrophosphate intermediates of bacterial lipid biosynthesis. Interactions between TCRs and these ligands allow the host to discriminate between self and non-self and co-ordinate an attack on the latter. Here, we describe how cells of the T lymphocyte lineage and their antigen receptors are generated and discuss the various modes of antigen recognition by these extraordinarily versatile receptors.
Collapse
Affiliation(s)
- M Attaf
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - M Legut
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - D K Cole
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - A K Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
46
|
Adams EJ, Gu S, Luoma AM. Human gamma delta T cells: Evolution and ligand recognition. Cell Immunol 2015; 296:31-40. [PMID: 25991474 DOI: 10.1016/j.cellimm.2015.04.008] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 04/19/2015] [Accepted: 04/24/2015] [Indexed: 01/26/2023]
Abstract
The γδ T cell lineage in humans remains much of an enigma due to the low number of defined antigens, the non-canonical ways in which these cells respond to their environment and difficulty in tracking this population in vivo. In this review, we survey a comparative evolutionary analysis of the primate V, D and J gene segments and contrast these findings with recent progress in defining antigen recognition by different populations of γδ T cells in humans. Signatures of both purifying and diversifying selection at the Vδ and Vγ gene loci are placed into context of Vδ1+ γδ T cell recognition of CD1d presenting different lipids, and Vγ 9Vδ2 T cell modulation by pyrophosphate-based phosphoantigens through the butyrophilins BTN3A. From this comparison, it is clear that co-evolution between γδ TCRs and these ligands is likely occurring, but the diversity inherent in these recombined receptors is an important feature in ligand surveillance.
Collapse
Affiliation(s)
- Erin J Adams
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, University of Chicago, Chicago, IL 60637, USA; Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA.
| | - Siyi Gu
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Adrienne M Luoma
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
47
|
Hansen VL, Miller RD. The Evolution and Structure of Atypical T Cell Receptors. Results Probl Cell Differ 2015; 57:265-78. [PMID: 26537385 DOI: 10.1007/978-3-319-20819-0_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The T cell receptor structure and genetic organization have been thought to have been stable in vertebrate evolution relative to the immunoglobulins. For the most part, this has been true and the content and organization of T cell receptor genes has been fairly conserved over the past 400 million years of gnathostome evolution. Analyses of TCRδ chains in a broad range of vertebrate lineages over the past decade have revealed a remarkable and previously unrealized degree of plasticity. This plasticity can generally be described in two forms. The first is broad use of antibody heavy chain variable genes in place of the conventional Vδ. The second form containing an unusual three extracellular domain structures has evolved independently in both cartilaginous fishes and mammals. Two well-studied vertebrate lineages, the eutherian mammals such as mice and humans and teleost fishes, lack any of these alternative TCR forms, contributing to why they went undiscovered for so long after the initial description of the conventional TCR chains three decades ago. This chapter describes the state of knowledge of these unusual TCR forms, both their structure and genetics, and current ideas on their function.
Collapse
Affiliation(s)
- Victoria L Hansen
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Robert D Miller
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|