1
|
Fan T, Tai C, Sleiman KC, Cutcliffe MP, Kim H, Liu Y, Li J, Xin G, Grashel M, Baert L, Ekeocha C, Vergenes P, Lima S, Lo WL, Lin J, Hanaoka B, Tankersley TN, Wang M, Zhang X, Tsokos GC, Jarjour W, Longman R, Wu HJJ. Aberrant T follicular helper cells generated by T H17 cell plasticity in the gut promote extraintestinal autoimmunity. Nat Immunol 2025; 26:790-804. [PMID: 40307450 DOI: 10.1038/s41590-025-02125-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/12/2025] [Indexed: 05/02/2025]
Abstract
Much remains unknown regarding T follicular helper 17 (TFH17) cells commonly found in autoimmune patients. We previously showed that (and here ask why) egress of gut segmented filamentous bacteria (SFB)-induced TFH cells from Peyer's patches (PP) to systemic sites promotes arthritis. We found splenic TFH17 cells are gut derived. Functional analyses using fate-mapping mice revealed a c-Maf-dependent and SFB-induced TH17-to-TFH cell reprogramming that dominantly occurs in PPs. Unlike conventional TFH cells, TH17-derived TFH cells are highly migratory and atypically concentrated in the dark zone of germinal centers (GCs). Compared to conventional TFH cells, TH17-derived TFH cells express higher levels of TFH-associated functional molecules and more robustly conjugate with B cells. Gain- and loss-of-function studies demonstrated their dominance in promoting GC B cells and arthritis. Notably, murine gut TH17-derived TFH signatures exist in rheumatoid arthritis patients. Thus, gut T cell plasticity generates atypical, potent TFH cells promoting systemic autoimmunity.
Collapse
Affiliation(s)
- Tingting Fan
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Chi Tai
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kiah C Sleiman
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Madeline P Cutcliffe
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Haram Kim
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ye Liu
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jianying Li
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Gang Xin
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology; The Ohio State Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Mollyanna Grashel
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Laurie Baert
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Chinwe Ekeocha
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Paige Vergenes
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Svetlana Lima
- Jill Roberts Center and Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Wan-Lin Lo
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Judith Lin
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Beatriz Hanaoka
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Trevor N Tankersley
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Beijing, China
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Wael Jarjour
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Randy Longman
- Jill Roberts Center and Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Hsin-Jung Joyce Wu
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA.
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA.
- Pelotonia Institute for Immuno-Oncology; The Ohio State Comprehensive Cancer Center, Columbus, Ohio, USA.
- Arizona Arthritis Center, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
2
|
Hu Y, Huang J, Wang S, Sun X, Wang X, Yu H. Deciphering Autoimmune Diseases: Unveiling the Diagnostic, Therapeutic, and Prognostic Potential of Immune Repertoire Sequencing. Inflammation 2025; 48:676-695. [PMID: 38914737 DOI: 10.1007/s10753-024-02079-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
Autoimmune diseases (AIDs) are immune system disorders where the body exhibits an immune response to its own antigens, causing damage to its own tissues and organs. The pathogenesis of AIDs is incompletely understood. However, recent advances in immune repertoire sequencing (IR-seq) technology have opened-up a new avenue to study the IR. These studies have revealed the prevalence in IR alterations, potentially inducing AIDs by disrupting immune tolerance and thereby contributing to our comprehension of AIDs. IR-seq harbors significant potential for the clinical diagnosis, personalized treatment, and prognosis of AIDs. This article reviews the application and progress of IR-seq in diseases, such as multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and type 1 diabetes, to enhance our understanding of the pathogenesis of AIDs and offer valuable references for the diagnosis and treatment of AIDs.
Collapse
Affiliation(s)
- Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China.
| |
Collapse
|
3
|
Sui S, Wei X, Zhu Y, Feng Q, Zha X, Mao L, Huang B, Lei W, Chen G, Zhan H, Chen H, Feng R, Zeng C, Li Y, Luo OJ. Single-Cell Multiomics Reveals TCR Clonotype-Specific Phenotype and Stemness Heterogeneity of T-ALL Cells. Cell Prolif 2025; 58:e13786. [PMID: 39675761 PMCID: PMC11969251 DOI: 10.1111/cpr.13786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
T-cell acute lymphoblastic leukaemia (T-ALL) is a heterogeneous malignant disease with high relapse and mortality rates. To characterise the multiomics features of T-ALL, we conducted integrative analyses using single-cell RNA, TCR and chromatin accessibility sequencing on pre- and post-treatment peripheral blood and bone marrow samples of the same patients. We found that there is transcriptional rewiring of gene regulatory networks in T-ALL cells. Some transcription factors, such as TCF3 and KLF3, showed differences in activity and expression levels between T-ALL and normal T cells and were associated with the prognosis of T-ALL patients. Furthermore, we identified multiple malignant TCR clonotypes among the T-ALL cells, where the clonotypes consisted of distinct combinations of the same TCR α and β chain per patient. The T-ALL cells displayed clonotype-specific immature thymocyte cellular characteristics and response to chemotherapy. Remarkably, T-ALL cells with an orphan TCRβ chain displayed the strongest stemness and resistance to chemotherapy. Our study provided transcriptome and epigenome characterisation of T-ALL cells categorised by TCR clonotypes, which may be helpful for the development of novel predictive markers to evaluate treatment effectiveness for T-ALL.
Collapse
MESH Headings
- Humans
- Single-Cell Analysis/methods
- Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Phenotype
- Gene Regulatory Networks
- Transcriptome
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Male
- Female
- Multiomics
Collapse
Affiliation(s)
- Songnan Sui
- Department of Systems Biomedical Sciences, School of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of MedicineJinan UniversityGuangzhouChina
- Department of Hematology, First Affiliated HospitalJinan UniversityGuangzhouChina
- Central People's Hospital of ZhanjiangZhanjiangChina
- Zhanjiang Key Laboratory of Leukemia Pathogenesis and Targeted Therapy ResearchZhanjiangChina
| | - Xiaolei Wei
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yue Zhu
- Department of Systems Biomedical Sciences, School of MedicineJinan UniversityGuangzhouChina
| | - Qiuyue Feng
- Department of Systems Biomedical Sciences, School of MedicineJinan UniversityGuangzhouChina
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Lipeng Mao
- Department of Systems Biomedical Sciences, School of MedicineJinan UniversityGuangzhouChina
| | - Boya Huang
- Department of Systems Biomedical Sciences, School of MedicineJinan UniversityGuangzhouChina
| | - Wen Lei
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of MedicineJinan UniversityGuangzhouChina
- Guangdong Second Provincial General Hospital, Integrated Chinese and Western Medicine Postdoctoral Research Station, School of MedicineJinan UniversityGuangzhouChina
| | - Guobing Chen
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of MedicineJinan UniversityGuangzhouChina
| | - Huien Zhan
- Department of Hematology, First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Huan Chen
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ru Feng
- Department of Hematology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Chengwu Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of MedicineJinan UniversityGuangzhouChina
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of MedicineJinan UniversityGuangzhouChina
- Department of Hematology, First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of MedicineJinan UniversityGuangzhouChina
| |
Collapse
|
4
|
Enache A, Carty SA, Babushok DV. Origins of T-cell-mediated autoimmunity in acquired aplastic anaemia. Br J Haematol 2025; 206:1035-1053. [PMID: 39836983 PMCID: PMC11985373 DOI: 10.1111/bjh.19993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/28/2024] [Indexed: 01/23/2025]
Abstract
Acquired aplastic anaemia (AA) is an autoimmune bone marrow failure disease resulting from a cytotoxic T-cell-mediated attack on haematopoietic stem and progenitor cells (HSPCs). Despite significant progress in understanding the T-cell repertoire alterations in AA, identifying specific pathogenic T cells in AA patients has remained elusive, primarily due to the unknown antigenic targets of the autoimmune attack. In this review, we will synthesize findings from several decades of research to critically evaluate the current knowledge on T-cell repertoires in AA. We will highlight new insights gained from recent in vitro studies of candidate autoreactive T cells isolated from AA patients and will discuss efforts to identify shared T-cell clonotypes in AA. Finally, we will discuss emerging evidence on the potential T-cell cross-reactivity between HSPC and common viral epitopes that may contribute to the development of AA in some patients. We conclude by highlighting the areas of consensus and limitations, as well as the ongoing uncertainties, and we identify promising directions for future research in the field.
Collapse
MESH Headings
- Anemia, Aplastic/genetics
- Anemia, Aplastic/immunology
- Autoimmunity/genetics
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Hematopoietic Stem Cells/immunology
- Epitopes, T-Lymphocyte/immunology
- Antigens, Viral/immunology
- Immune Tolerance
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Lymphocyte Activation
- Humans
- Gene Rearrangement, T-Lymphocyte
- T-Lymphocytes, Cytotoxic/immunology
- Viruses/immunology
Collapse
Affiliation(s)
- Aura Enache
- Drexel University College of MedicineDrexel UniversityPhiladelphiaPennsylvaniaUSA
- Division of Hematology‐Oncology, Department of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Shannon A. Carty
- Division of Hematology and Oncology, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Daria V. Babushok
- Division of Hematology‐Oncology, Department of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Comprehensive Bone Marrow Failure Center, Department of PediatricsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
5
|
Kazansky DB, Kalinina AA, Khromykh LM. Memory T Cells: Investigation of Original Models with Transgenic T Cell Receptors. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:161-172. [PMID: 40254396 DOI: 10.1134/s0006297924603940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 04/22/2025]
Abstract
This review summarizes the research data on original mouse models developed in the laboratory of regulatory mechanisms in immunity of the Research Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation. Transfer of the genes of individual α- and β-chains of T cell receptors (TCRs) of memory cells has resulted in production of transgenic animal lines valuable for studying T lymphocyte homeostasis and patterns of formation of their activation profile markers. Investigation of the transgenic models revealed new features of immune selection and tumor progression. In particular, the fundamental property of some TCRs, termed "chain-centricity", has been confirmed; it involves dominance of one of the TCR chains during recognition of the MHC (major histocompatibility complex)/peptide complex. This property makes it possible to artificially generate a significant pool of immunocompetent T cells so it could be used in adoptive immunotherapy for oncological and infectious diseases. Transfer of the dominant active TCR α-chains provides the possibility for constructing organisms with innate specific immunological resistance to certain pathogens. The results of recent studies indicate that TCR, determining the T lymphocyte relationship with its MHC microenvironment, has an instructive role in formation of its functions and phenotype. One of these functions may be production of cyclophilin A by the cortisone-resistant memory cells localized in thymus. The evidence has been accumulated that expression of TCR with a certain structure and specificity is a sufficient condition for formation of the functional potential of memory cells in a T cell, regardless of its former interaction with antigenic MHC/peptide complexes.
Collapse
Affiliation(s)
- Dmitry B Kazansky
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia.
| | - Anastasiia A Kalinina
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| | - Ludmila M Khromykh
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| |
Collapse
|
6
|
Sserwadda H, Lee JH, Lee BH, Jung S, Bang YJ, Cho BK, Nam HJ, Choi SJ, Gong JR, Choi HS, Jung CW, Chung H, Nam H, Kim ER, Kim HJ, Park CG, Kim YH. Superloaded Multiplexed scRNA-seq Data Preserves Primary Immune Cell Heterogeneity but Necessitates Stringent Doublet Removal. Immunol Invest 2025:1-17. [PMID: 39882751 DOI: 10.1080/08820139.2025.2457039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
BACKGROUND Single-cell RNA sequencing (scRNA-seq) has improved our ability to characterize rare cell populations. In practice, cells from different tissues or donors are simultaneously loaded onto the instrument (multiplexed) at the recommended (standard loading) or higher (superloading) numbers to save time and money. Although cost-effective, superloading can stymie computational analyses owing to high multiplet rates and sample complexity. METHODS We compared the effects of superloading on multiplexed single-cell gene expression and T cell receptor (TCR) data generated from human thymus and blood samples from different donors. RESULTS Minimal transcriptomic differences were observed between the data generated by either standard or superloading. Irrespective of the loading cell number, we found that over 50% of the T cells expressing multiple TCR chains were doublets. CONCLUSION Multiple samples can be run simultaneously without compromising data quality and subsequent analyses. However, an additional doublet removal step based on TCR configuration may improve the accuracy of T cell analysis.
Collapse
Affiliation(s)
- Henry Sserwadda
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jung Ho Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Brian H Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Sunyoung Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Yoon Ji Bang
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Beom Keun Cho
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Hyo Jeong Nam
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - So-Jung Choi
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jeong-Ryeol Gong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Hyun Seung Choi
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Chong Wook Jung
- Department of Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Hyeyeon Chung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Hyunsung Nam
- Genomic Medicine Institute, Seoul National University, Seoul, South Korea
| | - Eung Re Kim
- Department of Thoracic and Cardiovascular Surgery, Sejong General Hospital, Bucheon, South Korea
| | - Hyun Je Kim
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
- Genomic Medicine Institute, Seoul National University, Seoul, South Korea
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Dermatology, Seoul National University Hospital, Seoul, South Korea
- Transplantation Research Institute, Seoul National University Medical Research Center, Seoul, South Korea
- Department of Basic Research, PB Immune Therapeutics Inc, Seoul, South Korea
| | - Chung-Gyu Park
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea
- Transplantation Research Institute, Seoul National University Medical Research Center, Seoul, South Korea
- Department of Basic Research, PB Immune Therapeutics Inc, Seoul, South Korea
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Seoul National University Hospital, Seoul, South Korea
| | - Yong-Hee Kim
- Transplantation Research Institute, Seoul National University Medical Research Center, Seoul, South Korea
- Department of Basic Research, PB Immune Therapeutics Inc, Seoul, South Korea
| |
Collapse
|
7
|
Dolton G, Thomas H, Tan LR, Rius Rafael C, Doetsch S, Ionescu GA, Cardo LF, Crowther MD, Behiry E, Morin T, Caillaud ME, Srai D, Parolini L, Hasan MS, Fuller A, Topley K, Wall A, Hopkins JR, Omidvar N, Alvares C, Zabkiewicz J, Frater J, Szomolay B, Sewell AK. MHC-related protein 1-restricted recognition of cancer via a semi-invariant TCR-α chain. J Clin Invest 2025; 135:e181895. [PMID: 39744940 PMCID: PMC11684821 DOI: 10.1172/jci181895] [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: 04/11/2024] [Accepted: 10/29/2024] [Indexed: 01/16/2025] Open
Abstract
The T cell antigen presentation platform MR1 consists of 6 allomorphs in humans that differ by no more than 5 amino acids. The principal function of this highly conserved molecule involves presenting microbial metabolites to the abundant mucosal-associated invariant T (MAIT) cell subset. Recent developments suggest that the role of MR1 extends to presenting antigens from cancer cells, a function dependent on the K43 residue in the MR1 antigen binding cleft. Here, we successfully cultured cancer-activated, MR1-restricted T cells from multiple donors and confirmed that they recognized a wide range of cancer types expressing the most common MR1*01 and/or MR1*02 allomorphs (over 95% of the population), while remaining inert to healthy cells including healthy B cells and monocytes. Curiously, in all but one donor these T cells were found to incorporate a conserved TCR-α chain motif, CAXYGGSQGNLIF (where X represents 3-5 amino acids), because of pairing between 10 different TRAV genes and the TRAJ42 gene segment. This semi-invariance in the TCR-α chain is reminiscent of MAIT cells and suggests recognition of a conserved antigen bound to K43.
Collapse
MESH Headings
- Humans
- Minor Histocompatibility Antigens/genetics
- Minor Histocompatibility Antigens/immunology
- Minor Histocompatibility Antigens/metabolism
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/metabolism
- Neoplasms/immunology
- Neoplasms/genetics
- Neoplasms/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Mucosal-Associated Invariant T Cells/immunology
- Mucosal-Associated Invariant T Cells/metabolism
- Antigen Presentation
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Amino Acid Motifs
- Cell Line, Tumor
Collapse
Affiliation(s)
- Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Hannah Thomas
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Li Rong Tan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Cristina Rius Rafael
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Stephanie Doetsch
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Giulia-Andreea Ionescu
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Lucia F. Cardo
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Michael D. Crowther
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Enas Behiry
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Théo Morin
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Marine E. Caillaud
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Devinder Srai
- Nuffield Department of Medicine and Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Lucia Parolini
- Nuffield Department of Medicine and Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Md Samiul Hasan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Anna Fuller
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Katie Topley
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Aaron Wall
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jade R. Hopkins
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Nader Omidvar
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Caroline Alvares
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Joanna Zabkiewicz
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - John Frater
- Nuffield Department of Medicine and NIHR Biomedical Research Centre University of Oxford, Oxford, United Kingdom
| | - Barbara Szomolay
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunology Research Institute, Cardiff University Cardiff, United Kingdom
| | - Andrew K. Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunology Research Institute, Cardiff University Cardiff, United Kingdom
- Division of Infection and Immunity, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
8
|
Pogorelyy MV, Kirk AM, Adhikari S, Minervina AA, Sundararaman B, Vegesana K, Brice DC, Scott ZB, Thomas PG. TIRTL-seq: Deep, quantitative, and affordable paired TCR repertoire sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613345. [PMID: 39345544 PMCID: PMC11430070 DOI: 10.1101/2024.09.16.613345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
ɑ/β T cells are key players in adaptive immunity. The specificity of T cells is determined by the sequences of the hypervariable T cell receptor (TCR) ɑ and β chains. Although bulk TCR sequencing offers a cost-effective approach for in-depth TCR repertoire profiling, it does not provide chain pairings, which are essential for determining T cell specificity. In contrast, single-cell TCR sequencing technologies produce paired chain data, but are limited in throughput to thousands of cells and are cost-prohibitive for cohort-scale studies. Here, we present TIRTL-seq (Throughput-Intensive Rapid TCR Library sequencing), a novel approach that generates ready-to-sequence TCR libraries from live cells in less than 7 hours. The protocol is optimized for use with non-contact liquid handlers in an automation-friendly 384-well plate format. Reaction volume miniaturization reduces library preparation costs to <$0.50 per well. The core principle of TIRTL-seq is the parallel generation of hundreds of libraries providing multiple biological replicates from a single sample that allows precise inference of both frequencies of individual clones and TCR chain pairings from well-occurrence patterns. We demonstrate scalability of our approach up to 1 million unique paired αβTCR clonotypes corresponding to over 30 million T cells per sample at a cost of less than $2000. For a sample of 10 million cells the cost is ~$200. We benchmarked TIRTL-seq against state-of-the-art 5'RACE bulk TCR-seq and 10x Genomics Chromium technologies on longitudinal samples. We show that TIRTL-seq is able to quantitatively identify expanding and contracting clonotypes between timepoints while providing accurate TCR chain pairings, including distinct temporal dynamics of SARS-CoV-2-specific and EBV-specific CD8+ T cell responses after infection. While clonal expansion was followed by sharp contraction for SARS-CoV-2 specific TCRs, EBV-specific TCRs remained stable once established. The sequences of both ɑ and β TCR chains are essential for determining T cell specificity. As the field moves towards greater applications in diagnostics and immunotherapy that rely on TCR specificity, we anticipate that our scalable paired TCR sequencing methodology will be instrumental for collecting large paired-chain datasets and ultimately extracting therapeutically relevant information from the TCR repertoire.
Collapse
Affiliation(s)
| | | | | | | | | | - Kasi Vegesana
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David C Brice
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Paul G Thomas
- St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
9
|
Xu Y, Yuan Y, Mou L, Hui L, Zhang X, Yao X, Li J. scRNA+TCR-seq reveals the pivotal role of dual receptor T lymphocytes in the pathogenesis of Kawasaki disease and during IVIG treatment. Front Immunol 2024; 15:1457687. [PMID: 39421738 PMCID: PMC11484261 DOI: 10.3389/fimmu.2024.1457687] [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: 07/01/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Kawasaki disease (KD), a common cause of acquired heart disease in children in developed countries, is primarily treated with intravenous immunoglobulin (IVIG), but some children demonstrate IVIG resistance with increased coronary artery injury risk. T cells have been demonstrated to be involved in the pathogenesis of KD and its treatment with IVIG. However, the role and mechanism of dual TCR T lymphocytes in the occurrence of KD and IVIG therapy remain unclear. Methods This study, based on scRNA-seq combined with TCR-seq technology, clustered the peripheral blood mononuclear cells of 3 healthy controls and 6 KD patients before and after IVIG treatment. Comparative analysis was conducted to investigate the differences in the proportion of single/dual receptor T cells, the characteristics of CDR3 repertoires, cell types, and the expression of transcription factors among the three groups. The study aimed to explore the correlation between dual TCR T cells and KD as well as IVIG treatment. Results In our experimental results, we observed the presence of dual TCR T cells in all three groups. However, compared to the healthy control group and the IVIG-treated group, the KD patients before IVIG treatment exhibited a lower proportion of dual TCR T cells, with variability between samples, ranging from 4% to 15%. Notably, after IVIG treatment, the proportion of dual TCR T cells significantly increased, stabilizing above 12%, and these T cells also exhibited clonal expansion and a preference for V gene usage. In addition we found differences in dual TCR T cell subsets among the three groups, for example, IVIG treatment increases the proportion of dual TCR Treg cells, but it still remains below that of healthy control groups, significantly higher proportions of both dual TCR CD8 central and effector memory T cells in IVIG-treated KD patients, and differences in the expression of transcription factors between single and dual TCR T cells. These results suggest dual TCR T cells correlate with KD and IVIG treatment. Conclusion Dual TCR T lymphocytes, especially dual TCR CD8 T cells and Treg cells, play crucial roles in the pathogenesis of KD and during IVIG treatment, providing strong support for further elucidating KD pathogenesis and optimizing treatment strategies.
Collapse
Affiliation(s)
- Yuanyuan Xu
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Yi Yuan
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Lanlan Mou
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Linhu Hui
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Xing Zhang
- Department of Cardiology, Kunming Children’s Hospital, Kunming, China
| | - Xinsheng Yao
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Jun Li
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
10
|
Martins YC, Rosa-Gonçalves P, Daniel-Ribeiro CT. Theories of immune recognition: Is anybody right? Immunology 2024; 173:274-285. [PMID: 39034280 DOI: 10.1111/imm.13839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/04/2024] [Indexed: 07/23/2024] Open
Abstract
The clonal selection theory (CST) is the centrepiece of the current paradigm used to explain immune recognition and memory. Throughout the past decades, the original CST had been expanded and modified to explain new experimental evidences since its original publication by Burnet. This gave origin to new paradigms that govern experimental immunology nowadays, such as the associative recognition of antigen model and the stranger/danger signal model. However, these new theories also do not fully explain experimental findings such as natural autoimmune immunoglobulins, idiotypic networks, low and high dose tolerance, and dual-receptor T and B cells. To make sense of these empirical data, some authors have been trying to change the paradigm of immune cognition using a systemic approach, analogies with brain processing and concepts from second-order cybernetics. In the present paper, we review the CST and some of the theories/hypotheses derived from it, focusing on immune recognition. We point out their main weaknesses and highlight arguments made by their opponents and believers. We conclude that, until now, none of the proposed theories can fully explain the totality of immune phenomena and that a theory of everything is needed in immunology.
Collapse
Affiliation(s)
- Yuri Chaves Martins
- Department of Anesthesiology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Pamela Rosa-Gonçalves
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz and Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz and Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Peng Q, Xu Y, Yao X. scRNA+ TCR-seq revealed dual TCR T cells antitumor response in the TME of NSCLC. J Immunother Cancer 2024; 12:e009376. [PMID: 39237261 PMCID: PMC11381643 DOI: 10.1136/jitc-2024-009376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 09/07/2024] Open
Abstract
The intricate origins, subsets, and characteristics of TCR (T Cell Receptor) s, along with the mechanisms underpinning the antitumor response of tumor-infiltrating T lymphocytes within the tumor microenvironment (TME) remain enigmatic. Recently, the advent of single-cell RNA+TCR-sequencing (scRNA+TCR seq) has revolutionized TME analysis, providing unprecedented insight into the origins, cell subsets, TCR CDR3 compositions, and the expression patterns of response/depletion factors within individual tumor-infiltrating T lymphocytes. Our analysis of the shared scRNA+TCR seq dataset revealed a substantial presence of dual TCR T cells, characterized by clonal hyperplasia and remarkable migratory prowess across various tissues, including blood, normal, peritumoral, and tumor tissues in non-small cell lung cancer patients. Notably, dual TCR CD8+T cells predominantly fell within the CXCL13+subset, displaying potent antitumor activity and a strong preference for tumor tissue residency. Conversely, dual TCR CD4+T cells were predominantly classified as CD5+ or LMNA+subsets, exhibiting a more even distribution across diverse tissue types. By harnessing scRNA+TCR seq and other cutting-edge technologies, we can delve deeper into the effects and mechanisms that regulate the antitumor response or tolerance of dual TCR T cells. This innovative approach holds immense promise in offering fresh perspectives and avenues for advancing research on TIL (Tumor infiltrating lymphocyte)s within the TME.
Collapse
Affiliation(s)
- Qi Peng
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Yuanyuan Xu
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Xinsheng Yao
- Department of Immunology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
12
|
Manjili MH, Manjili SH. The quantum model of T-cell activation: Revisiting immune response theories. Scand J Immunol 2024; 100:e13375. [PMID: 38750629 PMCID: PMC11250909 DOI: 10.1111/sji.13375] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/15/2024] [Accepted: 04/29/2024] [Indexed: 07/16/2024]
Abstract
Our understanding of the immune response is far from complete, missing out on more detailed explanations that could be provided by molecular insights. To bridge this gap, we introduce the quantum model of T-cell activation. This model suggests that the transfer of energy during protein phosphorylation within T cells is not a continuous flow but occurs in discrete bursts, or 'quanta', of phosphates. This quantized energy transfer is mediated by oscillating cycles of receptor phosphorylation and dephosphorylation, initiated by dynamic 'catch-slip' pulses in the peptide-major histocompatibility complex-T-cell receptor (pMHC-TcR) interactions. T-cell activation is predicated upon achieving a critical threshold of catch-slip pulses at the pMHC-TcR interface. Costimulation is relegated to a secondary role, becoming crucial only when the frequency of pMHC-TcR catch-slip pulses does not meet the necessary threshold for this quanta-based energy transfer. Therefore, our model posits that it is the quantum nature of energy transfer-not the traditional signal I or signal II-that plays the decisive role in T-cell activation. This paradigm shift highlights the importance of understanding T-cell activation through a quantum lens, offering a potentially transformative perspective on immune response regulation.
Collapse
Affiliation(s)
- Masoud H. Manjili
- Department of Microbiology & Immunology, VCU School of Medicine
- Massey Comprehensive Cancer Center, 401 College Street, Richmond, VA, 23298, USA
| | - Saeed H. Manjili
- AMF Automation Technologies LLC, 2115 W. Laburnum Ave., Richmond, VA 23227
| |
Collapse
|
13
|
de Greef PC, Njeru SN, Benz C, Fillatreau S, Malissen B, Agenès F, de Boer RJ, Kirberg J. The TCR assigns naive T cells to a preferred lymph node. SCIENCE ADVANCES 2024; 10:eadl0796. [PMID: 39047099 PMCID: PMC11268406 DOI: 10.1126/sciadv.adl0796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 06/21/2024] [Indexed: 07/27/2024]
Abstract
Naive T cells recirculate between the spleen and lymph nodes where they mount immune responses when meeting dendritic cells presenting foreign antigen. As this may happen anywhere, naive T cells ought to visit all lymph nodes. Here, deep sequencing almost-complete TCR repertoires led to a comparison of different lymph nodes within and between individual mice. We find strong evidence for a deterministic CD4/CD8 lineage choice and a consistent spatial structure. Specifically, some T cells show a preference for one or multiple lymph nodes, suggesting that their TCR interacts with locally presented (self-)peptides. These findings are mirrored in TCR-transgenic mice showing localized CD69 expression, retention, and cell division. Thus, naive T cells intermittently sense antigenically dissimilar niches, which is expected to affect their homeostatic competition.
Collapse
MESH Headings
- Animals
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Mice
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Mice, Transgenic
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Lectins, C-Type/metabolism
- Lectins, C-Type/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Peter C. de Greef
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, Netherlands
| | | | - Claudia Benz
- Division of Immunology, Paul-Ehrlich-Institut, IMG53, Langen, Germany
| | - Simon Fillatreau
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Université Paris Cité, Faculté de Médecine, Paris, France
- AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Bernard Malissen
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Fabien Agenès
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
- Inserm, Délégation Régionale Auvergne Rhône Alpes, 69500 Bron, France
| | - Rob J. de Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, Netherlands
| | - Jörg Kirberg
- Division of Immunology, Paul-Ehrlich-Institut, IMG53, Langen, Germany
| |
Collapse
|
14
|
Yuanyuanxu, Qipeng, Qingqingma, Yao X. scRNA + TCR-seq revealed the dual TCR pTh17 and Treg T cells involvement in autoimmune response in ankylosing spondylitis. Int Immunopharmacol 2024; 135:112279. [PMID: 38796963 DOI: 10.1016/j.intimp.2024.112279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE Th17 and Treg play important roles in AS, but their single and dual TCR pairing types, ratios, and CDR3 characteristics remain unknown. METHODS Single-cell RNA + TCR-seq results from six AS patients were used to cluster T-cell subpopulations and analyze the single and dual TCR T cell ratio, diversity/clonality/overlap of CDR3, and expression of transcription factors. RESULTS 1. AS patients have about 10% of dual TCR T cells, and SFMC have decreased diversity CDR3 libraries and significant clonal proliferation compared to PBMC. 2. Dual TCR ratio: memory T > naive T; pTh 17 > Th17; Treg /Th17/Th1/EM significantly higher than naive CD4 + T/CM, Pathogenic Th17 cells contain clonally proliferating single TCR and dual TCR cells. 3. The expression of single TCR and dual TCR transcription factors of each T cell subpopulation was basically the same, but there was differential expression of characteristic transcription factors, e.g. Foxp3, CTLA4, STAT5B, IL10RB, LAG3 in dual TCR Treg was higher than that of single TCR Treg; TNFSF10/12, TNFRSF4/14, CCL5, KLRB1 in dual TCR pTh17 were significantly higher than those in single TCR pTh17. 4. Between naive CD4 + T, pTh17, Th1 and Treg, there are partially identity identical tcr paired cells. CONCLUSIONS The high proportion of dual TCR T cells such as pTh17 and Treg in AS and the high expression of some transcription factors suggested a close association with self-response in AS; The overlap of CDR3 between Th1, Th17,pTh17, and Treg in AS suggested that the subpopulations may be differentiated from each other to regulate the inflammatory homeostasis and progression.
Collapse
Affiliation(s)
- Yuanyuanxu
- Department of Immunology, Center of Immunomolecular Engineering, Zunyi Medical University, Zunyi, China
| | - Qipeng
- Department of Immunology, Center of Immunomolecular Engineering, Zunyi Medical University, Zunyi, China
| | - Qingqingma
- Aerospace Hospital of Zunyi Medical University (Hospital 417), China
| | - Xinsheng Yao
- Department of Immunology, Center of Immunomolecular Engineering, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
15
|
Uhlemann H, Epp K, Klesse C, Link-Rachner CS, Surendranath V, Günther UP, Schetelig J, Heidenreich F. Shape of the art: TCR-repertoire after allogeneic hematopoietic cell transplantation. Best Pract Res Clin Haematol 2024; 37:101558. [PMID: 39098804 DOI: 10.1016/j.beha.2024.101558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/03/2024] [Accepted: 06/27/2024] [Indexed: 08/06/2024]
Abstract
The human adaptive immune repertoire is characterized by specificity and diversity to provide immunity against past and future tasks. Such tasks are mainly infections but also malignant transformations of cells. With its multiple lines of defense, the human immune system contains both, rapid reaction forces and the potential to capture, disassemble and analyze strange structures in order to teach the adaptive immune system and mount a specific immune response. Prevention and mitigation of autoimmunity is of equal importance. In the context of allogeneic hematopoietic cell transplantation (HCT) specific challenges exist with the transfer of cells from the adapted donor immune system to the immunosuppressed recipient. Those challenges are immunogenetic disparity between donor and host, reconstitution of immunity early after HCT by expansion of mature immune effector cells, and impaired thymic function, if the recipient is an adult (as it is the case in most HCTs). The possibility to characterize the adaptive immune repertoire by massively parallel sequencing of T-cell receptor gene rearrangements allows for a much more detailed characterization of the T-cell repertoire. In addition, high-dimensional characterization of immune effector cells based on their immunophenotype and single cell RNA sequencing allow for much deeper insights in adaptive immune responses. We here review, existing - still incomplete - information on immune reconstitution after allogeneic HCT. Building on the technological advances much deeper insights into immune recovery after HCT and adaptive immune responses and can be expected in the coming years.
Collapse
Affiliation(s)
- Heike Uhlemann
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany.
| | - Katharina Epp
- University Hospital Carl Gustav Carus, Dresden, Germany
| | | | | | | | | | - Johannes Schetelig
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany
| | - Falk Heidenreich
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany
| |
Collapse
|
16
|
Croce G, Bobisse S, Moreno DL, Schmidt J, Guillame P, Harari A, Gfeller D. Deep learning predictions of TCR-epitope interactions reveal epitope-specific chains in dual alpha T cells. Nat Commun 2024; 15:3211. [PMID: 38615042 PMCID: PMC11016097 DOI: 10.1038/s41467-024-47461-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/03/2024] [Indexed: 04/15/2024] Open
Abstract
T cells have the ability to eliminate infected and cancer cells and play an essential role in cancer immunotherapy. T cell activation is elicited by the binding of the T cell receptor (TCR) to epitopes displayed on MHC molecules, and the TCR specificity is determined by the sequence of its α and β chains. Here, we collect and curate a dataset of 17,715 αβTCRs interacting with dozens of class I and class II epitopes. We use this curated data to develop MixTCRpred, an epitope-specific TCR-epitope interaction predictor. MixTCRpred accurately predicts TCRs recognizing several viral and cancer epitopes. MixTCRpred further provides a useful quality control tool for multiplexed single-cell TCR sequencing assays of epitope-specific T cells and pinpoints a substantial fraction of putative contaminants in public databases. Analysis of epitope-specific dual α T cells demonstrates that MixTCRpred can identify α chains mediating epitope recognition. Applying MixTCRpred to TCR repertoires from COVID-19 patients reveals enrichment of clonotypes predicted to bind an immunodominant SARS-CoV-2 epitope. Overall, MixTCRpred provides a robust tool to predict TCRs interacting with specific epitopes and interpret TCR-sequencing data from both bulk and epitope-specific T cells.
Collapse
Affiliation(s)
- Giancarlo Croce
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
- Agora Cancer Research Centre, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Sara Bobisse
- Agora Cancer Research Centre, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Dana Léa Moreno
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
- Agora Cancer Research Centre, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Julien Schmidt
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Philippe Guillame
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Centre, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - David Gfeller
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.
- Agora Cancer Research Centre, Lausanne, Switzerland.
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland.
| |
Collapse
|
17
|
Kalinina AA, Khromykh LM, Kazansky DB. T Cell Receptor Chain Centricity: The Phenomenon and Potential Applications in Cancer Immunotherapy. Int J Mol Sci 2023; 24:15211. [PMID: 37894892 PMCID: PMC10607890 DOI: 10.3390/ijms242015211] [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: 08/31/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
T cells are crucial players in adaptive anti-cancer immunity. The gene modification of T cells with tumor antigen-specific T cell receptors (TCRs) was a milestone in personalized cancer immunotherapy. TCR is a heterodimer (either α/β or γ/δ) able to recognize a peptide antigen in a complex with self-MHC molecules. Although traditional concepts assume that an α- and β-chain contribute equally to antigen recognition, mounting data reveal that certain receptors possess chain centricity, i.e., one hemi-chain TCR dominates antigen recognition and dictates its specificity. Chain-centric TCRs are currently poorly understood in terms of their origin and the functional T cell subsets that express them. In addition, the ratio of α- and β-chain-centric TCRs, as well as the exact proportion of chain-centric TCRs in the native repertoire, is generally still unknown today. In this review, we provide a retrospective analysis of studies that evidence chain-centric TCRs, propose patterns of their generation, and discuss the potential applications of such receptors in T cell gene modification for adoptive cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Dmitry B. Kazansky
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia
| |
Collapse
|
18
|
Kusuda M, Nakasone H, Yoshimura K, Okada Y, Tamaki M, Matsuoka A, Ishikawa T, Meno T, Nakamura Y, Kawamura M, Takeshita J, Kawamura S, Yoshino N, Misaki Y, Gomyo A, Tanihara A, Kimura SI, Kako S, Kanda Y. Gene expression and TCR amino acid sequences selected by HLA-A02:01-restricted CTLs specific to HTLV-1 in ATL patients. Br J Haematol 2023; 202:578-588. [PMID: 37317804 DOI: 10.1111/bjh.18918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023]
Abstract
Adult T-cell leukaemia/lymphoma (ATL) is an aggressive malignancy of peripheral T cells caused by human T-cell lymphotropic virus type-1 (HTLV-1). Tax is the most important regulatory protein for HTLV-1. We aimed to reveal a unique amino acid sequence (AA) of complementarity-determining region 3 (CDR3) of the T-cell receptor (TCR)β and TCRα chains of HLA-A*02:01-restricted Tax11-19 -specific cytotoxic T cells (Tax-CTLs). The gene expression profiles (GEP) of Tax-CTLs were assessed by the next-generation sequence (NGS) method with SMARTer technology. Tax-CTLs seemed to be oligoclonal, and their gene compositions were skewed. The unique motifs of 'DSWGK' in TCRα and 'LAG' in TCRβ at CDR3 were observed in almost all patients. Tax-CTL clones harbouring the 'LAG' motif with BV28 had a higher binding score than those without either of them, besides a higher binding score associated with longer survival. Tax-CTLs established from a single cell showed killing activities against Tax-peptide-pulsed HLA-A2+ T2 cell lines. GEP of Tax-CTLs revealed that genes associated with immune response activity were well preserved in long-term survivors with stable status. These methods and results can help us better understand immunity against ATL, and should contribute to future studies on the clinical application of adoptive T-cell therapies.
Collapse
Affiliation(s)
- Machiko Kusuda
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Hideki Nakasone
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
- Division of Stem Cell Regulation, Center for Molecular Medicine, Jichi Medicial University, Shimotsuke, Japan
| | - Kazuki Yoshimura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Yosuke Okada
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Masaharu Tamaki
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Akari Matsuoka
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Takuto Ishikawa
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Tomohiro Meno
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Yuhei Nakamura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Masakatsu Kawamura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Junko Takeshita
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Shunto Kawamura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Nozomu Yoshino
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Yukiko Misaki
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Ayumi Gomyo
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Aki Tanihara
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Shun-Ichi Kimura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Shinichi Kako
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| |
Collapse
|
19
|
Zhu L, Peng Q, Li J, Wu Y, Wang J, Zhou D, Ma L, Yao X. scRNA-seq revealed the special TCR β & α V(D)J allelic inclusion rearrangement and the high proportion dual (or more) TCR-expressing cells. Cell Death Dis 2023; 14:487. [PMID: 37524693 PMCID: PMC10390570 DOI: 10.1038/s41419-023-06004-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 08/02/2023]
Abstract
Allelic exclusion, one lymphocyte expresses one antigen receptor, is a fundamental mechanism of immunological self-tolerance and highly specific immune responses to pathogens. However, the phenomenon of V(D)J allelic inclusion (incomplete allelic exclusion or allelic escape) rearrangement and dual TCR T cells have been discovered by multiple laboratories. Despite continuous new discoveries, the proportion and underlying mechanism of dual TCR has been puzzling immunologists. In this study, we observed the presence of single T cells expressing multiple TCR chains in all samples, with the proportion of 15%, 10%, and 20% in the human thymus, human peripheral blood, and mouse lymphoid organs, respectively. The proportion of T cells possessing multiple T-cell receptors (TCR) varied significantly in different physiological states and developmental stages. By analyzing RSS category, RSS direction, and V(D)J gene position at TR locus of T cells which contain multiple TCR chains, we creatively found that one of TCR β (or TCR α) should originate from the transcription of V(D)J combination in T-cell receptor excision circle (TREC) formed after the twice successful rearrangement in the same chromosome. Moreover, human V30 (or mouse V31) gene may participate in reverse recombination and transcription to prevent allelic exclusion. In general, high proportion of T cells with multiple TCR at the transcriptome level was first made public, and we proposed a novel mechanism of secondary (or more) TCR rearrangement on a single chromosome. Our findings also indicated that the single-cell sequencing data should be classified according to the single, multiple, and abnormal TCR when analyzing the T-cell repertoire.
Collapse
Affiliation(s)
- Lanwei Zhu
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Qi Peng
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Jun Li
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Yingjie Wu
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Jiayi Wang
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Dewei Zhou
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Long Ma
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Xinsheng Yao
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
20
|
Shen Y, Voigt A, Leng X, Rodriguez AA, Nguyen CQ. A current and future perspective on T cell receptor repertoire profiling. Front Genet 2023; 14:1159109. [PMID: 37408774 PMCID: PMC10319011 DOI: 10.3389/fgene.2023.1159109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023] Open
Abstract
T cell receptors (TCR) play a vital role in the immune system's ability to recognize and respond to foreign antigens, relying on the highly polymorphic rearrangement of TCR genes. The recognition of autologous peptides by adaptive immunity may lead to the development and progression of autoimmune diseases. Understanding the specific TCR involved in this process can provide insights into the autoimmune process. RNA-seq (RNA sequencing) is a valuable tool for studying TCR repertoires by providing a comprehensive and quantitative analysis of the RNA transcripts. With the development of RNA technology, transcriptomic data must provide valuable information to model and predict TCR and antigen interaction and, more importantly, identify or predict neoantigens. This review provides an overview of the application and development of bulk RNA-seq and single-cell (SC) RNA-seq to examine the TCR repertoires. Furthermore, discussed here are bioinformatic tools that can be applied to study the structural biology of peptide/TCR/MHC (major histocompatibility complex) and predict antigenic epitopes using advanced artificial intelligence tools.
Collapse
Affiliation(s)
- Yiran Shen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Alexandria Voigt
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Xuebing Leng
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Amy A. Rodriguez
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Cuong Q. Nguyen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, United States
- Center of Orphaned Autoimmune Diseases, University of Florida, Gainesville, FL, United States
| |
Collapse
|
21
|
Jang HJ, Caron C, Lee CK, Wang L, Jama B, Bui JD, Morris GP. Dual receptor T cells mediate effective antitumor immune responses via increased recognition of tumor antigens. J Immunother Cancer 2023; 11:e006472. [PMID: 37188395 PMCID: PMC10186424 DOI: 10.1136/jitc-2022-006472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Discovery that ~16% of T cells naturally co-express two T-cell receptor (TCR) clonotypes prompts examining the role of dual TCR cells in immune functions. METHODS Using TCRα-reporter transgenic mice, enabling unambiguous identification of single-TCR and dual-TCR cells, we tested the role of dual TCR cells in antitumor immune responses against immune-responsive syngeneic 6727 sarcoma and immune-resistant B16F10 melanoma. RESULTS Dual TCR cells were specifically increased among tumor-infiltrating lymphocytes (TILs) in both models, indicating selective advantage in antitumor responses. Phenotype and single-cell gene expression analyses identified dual TCR are predominant during the effective antitumor response, demonstrating selectively increased activation in the TIL compartment and skewing toward an effector memory phenotype. Absence of dual TCR cells impaired immune response to B16F10 but not 6727, suggesting that dual TCR cells may be more influential in responses against poorly immunogenic tumors. Dual TCR cells demonstrated an advantage in recognition of B16F10-derived neoantigens in vitro, providing a mechanistic basis for their antitumor reactivity. CONCLUSIONS These results discover an unrecognized role for dual TCR cells in protective immune function and identify these cells and their TCRs as a potential resource for antitumor immunotherapy.
Collapse
Affiliation(s)
- Hyun J Jang
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Christine Caron
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Calvin K Lee
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Lu Wang
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Burhan Jama
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Jack D Bui
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Gerald P Morris
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
22
|
Muhowski EM, Rogers LM. Dual TCR-Expressing T Cells in Cancer: How Single-Cell Technologies Enable New Investigation. Immunohorizons 2023; 7:299-306. [PMID: 37129560 PMCID: PMC10354877 DOI: 10.4049/immunohorizons.2200062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/11/2023] [Indexed: 05/03/2023] Open
Abstract
TCR diversity measures are often used to understand the immune response in cancer. Traditional measures of diversity rely on bulk RNA sequencing (RNAseq) of the β-chain variable regions. However, the full αβ TCR repertoire is a combination of both the α- and β-chains, which are encoded by separate genes. In contrast with bulk RNAseq, single-cell RNAseq (scRNAseq) allows paired chain analyses, yielding a more accurate measure of the repertoire. Interestingly, ∼30% of mature peripheral T cells express multiple TCR alleles (e.g., two α-chains) and may exhibit dual Ag specificity. scRNAseq has become increasingly common, and data from both human and animal studies are publicly available. However, routine workflows discard secondary TCR alleles and focus on a single TCR clone per cell. This perspectives piece emphasizes why this may not be good practice and highlights unanswered questions in the field of T cell dual specificity.
Collapse
|
23
|
Kalinina AA, Ziganshin RK, Silaeva YY, Sharova NI, Nikonova MF, Persiyantseva NA, Gorkova TG, Antoshina EE, Trukhanova LS, Donetskova AD, Komogorova VV, Litvina MM, Mitin AN, Zamkova MA, Bruter AV, Khromykh LM, Kazansky DB. Physiological and Functional Effects of Dominant Active TCRα Expression in Transgenic Mice. Int J Mol Sci 2023; 24:ijms24076527. [PMID: 37047500 PMCID: PMC10094918 DOI: 10.3390/ijms24076527] [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: 03/02/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
A T cell receptor (TCR) consists of α- and β-chains. Accumulating evidence suggests that some TCRs possess chain centricity, i.e., either of the hemi-chains can dominate in antigen recognition and dictate the TCR’s specificity. The introduction of TCRα/β into naive lymphocytes generates antigen-specific T cells that are ready to perform their functions. Transgenesis of the dominant active TCRα creates transgenic animals with improved anti-tumor immune control, and adoptive immunotherapy with TCRα-transduced T cells provides resistance to infections. However, the potential detrimental effects of the dominant hemi-chain TCR’s expression in transgenic animals have not been well investigated. Here, we analyzed, in detail, the functional status of the immune system of recently generated 1D1a transgenic mice expressing the dominant active TCRα specific to the H2-Kb molecule. In their age dynamics, neither autoimmunity due to the random pairing of transgenic TCRα with endogenous TCRβ variants nor significant disturbances in systemic homeostasis were detected in these mice. Although the specific immune response was considerably enhanced in 1D1a mice, responses to third-party alloantigens were not compromised, indicating that the expression of dominant active TCRα did not limit immune reactivity in transgenic mice. Our data suggest that TCRα transgene expression could delay thymic involution and maintain TCRβ repertoire diversity in old transgenic mice. The detected changes in the systemic homeostasis in 1D1a transgenic mice, which are minor and primarily transient, may indicate variations in the ontogeny of wild-type and transgenic mouse lines.
Collapse
Affiliation(s)
- Anastasiia A. Kalinina
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
| | - Rustam Kh. Ziganshin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya st. 16/10, 117997 Moscow, Russia
| | - Yulia Yu. Silaeva
- Institute of Gene Biology, Russian Academy of Sciences, Vavilova st. 34/5, 119334 Moscow, Russia
| | - Nina I. Sharova
- National Research Center, Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe sh., 24, 115522 Moscow, Russia
| | - Margarita F. Nikonova
- National Research Center, Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe sh., 24, 115522 Moscow, Russia
| | - Nadezda A. Persiyantseva
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
| | - Tatiana G. Gorkova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
| | - Elena E. Antoshina
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
| | - Lubov S. Trukhanova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
| | - Almira D. Donetskova
- National Research Center, Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe sh., 24, 115522 Moscow, Russia
| | - Victoria V. Komogorova
- National Research Center, Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe sh., 24, 115522 Moscow, Russia
| | - Marina M. Litvina
- National Research Center, Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe sh., 24, 115522 Moscow, Russia
| | - Alexander N. Mitin
- National Research Center, Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe sh., 24, 115522 Moscow, Russia
| | - Maria A. Zamkova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
- Institute of Gene Biology, Russian Academy of Sciences, Vavilova st. 34/5, 119334 Moscow, Russia
| | - Alexandra V. Bruter
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Ludmila M. Khromykh
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
| | - Dmitry B. Kazansky
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Kashirskoe sh., 24, 115478 Moscow, Russia
- Correspondence:
| |
Collapse
|
24
|
Sanromán ÁF, Joshi K, Au L, Chain B, Turajlic S. TCR sequencing: applications in immuno-oncology research. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 17:100373. [PMID: 36908996 PMCID: PMC9996383 DOI: 10.1016/j.iotech.2023.100373] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
•T-cell receptor (TCR) interaction with major histocompatibility complex-antigen complexes leads to antitumour responses.•TCR sequencing analysis allows characterisation of T cells that recognise tumour neoantigens.•T-cell clonal revival and clonal replacement potentially underpin immunotherapy responses.
Collapse
Affiliation(s)
- Á F Sanromán
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
| | - K Joshi
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, UK.,Renal and Skin Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - L Au
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Australia
| | - B Chain
- Division of Infection and Immunity, University College London, London, UK.,Department of Computer Science, University College London, London, UK
| | - S Turajlic
- Renal and Skin Unit, The Royal Marsden NHS Foundation Trust, London, UK.,Melanoma and Kidney Cancer Team, The Institute of Cancer Research, London, UK
| |
Collapse
|
25
|
Identification of T Cell Receptors Targeting a Neoantigen Derived from Recurrently Mutated FGFR3. Cancers (Basel) 2023; 15:cancers15041031. [PMID: 36831375 PMCID: PMC9953830 DOI: 10.3390/cancers15041031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Immunotherapies, including immune checkpoint blockades, play a critically important role in cancer treatments. For immunotherapies, neoantigens, which are generated by somatic mutations in cancer cells, are thought to be good targets due to their tumor specificity. Because neoantigens are unique in individual cancers, it is challenging to develop personalized immunotherapy targeting neoantigens. In this study, we screened "shared neoantigens", which are specific types of neoantigens derived from mutations observed commonly in a subset of cancer patients. Using exome sequencing data in the Cancer Genome Atlas (TCGA), we predicted shared neoantigen peptides and performed in vitro screening of shared neoantigen-reactive CD8+ T cells using peripheral blood from healthy donors. We examined the functional activity of neoantigen-specific T cell receptors (TCRs) by generating TCR-engineered T cells. Among the predicted shared neoantigens from TCGA data, we found that the mutated FGFR3Y373C peptide induced antigen-specific CD8+ T cells from the donor with HLA-A*02:06 via an ELISPOT assay. Subsequently, we obtained FGFR3Y373C-specific CD8+ T cell clones and identified two different sets of TCRs specifically reactive to FGFR3Y373C. We found that the TCR-engineered T cells expressing FGFR3Y373C-specific TCRs recognized the mutated FGFR3Y373C peptide but not the corresponding wild-type peptide. These two FGFR3Y373C-specific TCR-engineered T cells showed cytotoxic activity against mutated FGFR3Y373C-loaded cells. These results imply the possibility of strategies of immunotherapies targeting shared neoantigens, including cancer vaccines and TCR-engineered T cell therapies.
Collapse
|
26
|
Dascalu S, Preston SG, Dixon RJ, Flammer PG, Fiddaman S, Boyd A, Sealy JE, Sadeyen JR, Kaspers B, Velge P, Iqbal M, Bonsall MB, Smith AL. The influences of microbial colonisation and germ-free status on the chicken TCRβ repertoire. Front Immunol 2023; 13:1052297. [PMID: 36685492 PMCID: PMC9847582 DOI: 10.3389/fimmu.2022.1052297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
Microbial colonisation is paramount to the normal development of the immune system, particularly at mucosal sites. However, the relationships between the microbiome and the adaptive immune repertoire have mostly been explored in rodents and humans. Here, we report a high-throughput sequencing analysis of the chicken TCRβ repertoire and the influences of microbial colonisation on tissue-resident TCRβ+ cells. The results reveal that the microbiome is an important driver of TCRβ diversity in both intestinal tissues and the bursa of Fabricius, but not in the spleen. Of note, public TCRβ sequences (shared across individuals) make a substantial contribution to the repertoire. Additionally, different tissues exhibit biases in terms of their V family and J gene usage, and these effects were influenced by the gut-associated microbiome. TCRβ clonal expansions were identified in both colonised and germ-free birds, but differences between the groups were indicative of an influence of the microbiota. Together, these findings provide an insight into the avian adaptive immune system and the influence of the microbiota on the TCRβ repertoire.
Collapse
Affiliation(s)
- Stefan Dascalu
- Department of Biology, University of Oxford, Oxford, United Kingdom
- Avian Influenza Research Group, The Pirbright Institute, Pirbright, United Kingdom
| | - Stephen G. Preston
- Department of Biology, University of Oxford, Oxford, United Kingdom
- UCL School of Pharmacy, University College London, London, United Kingdom
| | - Robert J. Dixon
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | | | - Steven Fiddaman
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Amy Boyd
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Joshua E. Sealy
- Avian Influenza Research Group, The Pirbright Institute, Pirbright, United Kingdom
| | - Jean-Remy Sadeyen
- Avian Influenza Research Group, The Pirbright Institute, Pirbright, United Kingdom
| | - Bernd Kaspers
- Veterinary Faculty, Ludwig Maximillians University of Munich, Planegg, Germany
| | - Philippe Velge
- Institut National de la Recherche Agronomique (INRAE), Université François Rabelais de Tours, Unités Mixtes de Recherche, Infectiologie et Santé Publique (ISP), Nouzilly, France
| | - Munir Iqbal
- Avian Influenza Research Group, The Pirbright Institute, Pirbright, United Kingdom
| | | | - Adrian L. Smith
- Department of Biology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Swan SL, Mehta N, Ilich E, Shen SH, Wilkinson DS, Anderson AR, Segura T, Sanchez-Perez L, Sampson JH, Bellamkonda RV. IL7 and IL7 Flt3L co-expressing CAR T cells improve therapeutic efficacy in mouse EGFRvIII heterogeneous glioblastoma. Front Immunol 2023; 14:1085547. [PMID: 36817432 PMCID: PMC9936235 DOI: 10.3389/fimmu.2023.1085547] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy in glioblastoma faces many challenges including insufficient CAR T cell abundance and antigen-negative tumor cells evading targeting. Unfortunately, preclinical studies evaluating CAR T cells in glioblastoma focus on tumor models that express a single antigen, use immunocompromised animals, and/or pre-treat with lymphodepleting agents. While lymphodepletion enhances CAR T cell efficacy, it diminishes the endogenous immune system that has the potential for tumor eradication. Here, we engineered CAR T cells to express IL7 and/or Flt3L in 50% EGFRvIII-positive and -negative orthotopic tumors pre-conditioned with non-lymphodepleting irradiation. IL7 and IL7 Flt3L CAR T cells increased intratumoral CAR T cell abundance seven days after treatment. IL7 co-expression with Flt3L modestly increased conventional dendritic cells as well as the CD103+XCR1+ population known to have migratory and antigen cross-presenting capabilities. Treatment with IL7 or IL7 Flt3L CAR T cells improved overall survival to 67% and 50%, respectively, compared to 9% survival with conventional or Flt3L CAR T cells. We concluded that CAR T cells modified to express IL7 enhanced CAR T cell abundance and improved overall survival in EGFRvIII heterogeneous tumors pre-conditioned with non-lymphodepleting irradiation. Potentially IL7 or IL7 Flt3L CAR T cells can provide new opportunities to combine CAR T cells with other immunotherapies for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Sheridan L Swan
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Nalini Mehta
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Ekaterina Ilich
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Steven H Shen
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Daniel S Wilkinson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - Alexa R Anderson
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Tatiana Segura
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States.,Clinical Science Departments of Neurology and Dermatology, Duke University, Durham, NC, United States
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Pathology, Duke University Medical Center, Durham, NC, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - Ravi V Bellamkonda
- Department of Biology, Emory University, Atlanta, GA, United States.,Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA, United States
| |
Collapse
|
28
|
Wong P, Cina DP, Sherwood KR, Fenninger F, Sapir-Pichhadze R, Polychronakos C, Lan J, Keown PA. Clinical application of immune repertoire sequencing in solid organ transplant. Front Immunol 2023; 14:1100479. [PMID: 36865546 PMCID: PMC9971933 DOI: 10.3389/fimmu.2023.1100479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/25/2023] [Indexed: 02/16/2023] Open
Abstract
Background Measurement of T cell receptor (TCR) or B cell receptor (BCR) gene utilization may be valuable in monitoring the dynamic changes in donor-reactive clonal populations following transplantation and enabling adjustment in therapy to avoid the consequences of excess immune suppression or to prevent rejection with contingent graft damage and to indicate the development of tolerance. Objective We performed a review of current literature to examine research in immune repertoire sequencing in organ transplantation and to assess the feasibility of this technology for clinical application in immune monitoring. Methods We searched MEDLINE and PubMed Central for English-language studies published between 2010 and 2021 that examined T cell/B cell repertoire dynamics upon immune activation. Manual filtering of the search results was performed based on relevancy and predefined inclusion criteria. Data were extracted based on study and methodology characteristics. Results Our initial search yielded 1933 articles of which 37 met the inclusion criteria; 16 of these were kidney transplant studies (43%) and 21 were other or general transplantation studies (57%). The predominant method for repertoire characterization was sequencing the CDR3 region of the TCR β chain. Repertoires of transplant recipients were found to have decreased diversity in both rejectors and non-rejectors when compared to healthy controls. Rejectors and those with opportunistic infections were more likely to have clonal expansion in T or B cell populations. Mixed lymphocyte culture followed by TCR sequencing was used in 6 studies to define an alloreactive repertoire and in specialized transplant settings to track tolerance. Conclusion Methodological approaches to immune repertoire sequencing are becoming established and offer considerable potential as a novel clinical tool for pre- and post-transplant immune monitoring.
Collapse
Affiliation(s)
- Paaksum Wong
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Davide P Cina
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Karen R Sherwood
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Franz Fenninger
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ruth Sapir-Pichhadze
- Department of Medicine, Division of Nephrology, McGill University, Montreal, QC, Canada.,Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Constantin Polychronakos
- Department of Pediatrics, The Research Institute of the McGill University Health Centre and the Montreal Children's Hospital, Montreal, QC, Canada
| | - James Lan
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul A Keown
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
29
|
Radtke D, Thuma N, Schülein C, Kirchner P, Ekici AB, Schober K, Voehringer D. Th2 single-cell heterogeneity and clonal distribution at distant sites in helminth-infected mice. eLife 2022; 11:74183. [PMID: 35950748 PMCID: PMC9391044 DOI: 10.7554/elife.74183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Th2 cells provide effector functions in type 2 immune responses to helminths and allergens. Despite knowledge about molecular mechanisms of Th2 cell differentiation, there is little information on Th2 cell heterogeneity and clonal distribution between organs. To address this, we performed combined single-cell transcriptome and T-cell receptor (TCR) clonotype analysis on murine Th2 cells in mesenteric lymph nodes (MLNs) and lung after infection with Nippostrongylus brasiliensis (Nb) as a human hookworm infection model. We find organ-specific expression profiles, but also populations with conserved migration or effector/resident memory signatures that unexpectedly cluster with potentially regulatory Il10posFoxp3neg cells. A substantial MLN subpopulation with an interferon response signature suggests a role for interferon signaling in Th2 differentiation or diversification. Further RNA-inferred developmental directions indicate proliferation as a hub for differentiation decisions. Although the TCR repertoire is highly heterogeneous, we identified expanded clones and CDR3 motifs. Clonal relatedness between distant organs confirmed effective exchange of Th2 effector cells, although locally expanded clones dominated the response. We further cloned an Nb-specific TCR from an expanded clone in the lung effector cluster and describe surface markers that distinguish transcriptionally defined clusters. These results provide insights in Th2 cell subset diversity and clonal relatedness in distant organs.
Collapse
Affiliation(s)
- Daniel Radtke
- Department of Infection Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Natalie Thuma
- Department of Infection Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christine Schülein
- Institute of Clinical Microbiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Kilian Schober
- Institute of Clinical Microbiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
30
|
Cui L, Zhang C, Ding H, Feng D, Huang H, Lu Z, Liu B. Clonal Distribution and Intratumor Heterogeneity of the TCR Repertoire in Papillary Thyroid Cancer With or Without Coexistent Hashimoto's Thyroiditis. Front Immunol 2022; 13:821601. [PMID: 35720279 PMCID: PMC9203861 DOI: 10.3389/fimmu.2022.821601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/11/2022] [Indexed: 12/24/2022] Open
Abstract
The intratumor heterogeneity (ITH) of the amount and TCR repertoires of tumor infiltrating lymphocytes (TILs) in PTC with and without coexistent Hashimoto's thyroiditis (HT) are unclear. Here, we investigated the amount of T cells in tumor and corresponding normal tissues by immunohistochemical staining on 80 tumor samples and 40 normal samples from 40 patients. The immune repertoire of T cells was identified on 24 tumor samples and 12 normal samples from 12 patients using TCR high-throughput sequencing. The results demonstrated that the numbers of CD3+, CD4+ and CD8+ T cells in PTC without coexistent HT (PTC-WO) were significantly lower than those in PTC with existing HT (PTC-W). In PTC-W, the density of CD4+ TILs were generally higher when compared with CD8+ TILs. Furthermore, we found that the numbers of CD3+ T cells and their CD4+, CD8+ subtypes in tumor samples were generally higher than those in normal tissue in PTC-WO and moreover, the number of CD3+ T cells was negatively associated with TCR clonality in PTC-WO. In addition, although ITH of the TCR repertoire truly existed in PTC-W and PTC-WO, the TCR repertoires between distinct regions of the non-adjacent tumor foci were presented with a higher degree of similarity than those between tumor and matched normal tissue in PTC-WO, yet the similarity of intratumor repertoires was not significantly higher than those between tumor and corresponding normal samples in PTC-W. This research comprehensively delineated the quantity and TCR repertoire ITH of T cells in PTC-W and PTC-WO, suggesting that TILs might be reactive to tumor antigens in PTC-WO. Moreover, multiregion biopsies should be performed to precisely identify the immune background in PTC-W and PTC-WO.
Collapse
Affiliation(s)
- Likun Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Head & Neck Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chaoting Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Huirong Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Core Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dongdong Feng
- Department of Head and Neck Surgery, Center of Otolaryngology Head and Neck Surgery, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Hongying Huang
- Department of Pathology, New York University Langone Medical Center, New York, NY, United States
| | - Zheming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Baoguo Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Head & Neck Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
31
|
Sun X, Nguyen T, Achour A, Ko A, Cifello J, Ling C, Sharma J, Hiroi T, Zhang Y, Chia CW, Wood Iii W, Wu WW, Zukley L, Phue JN, Becker KG, Shen RF, Ferrucci L, Weng NP. Longitudinal analysis reveals age-related changes in the T cell receptor repertoire of human T cell subsets. J Clin Invest 2022; 132:158122. [PMID: 35708913 PMCID: PMC9433102 DOI: 10.1172/jci158122] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
A diverse T cell receptor (TCR) repertoire is essential for protection against a variety of pathogens, and TCR repertoire size is believed to decline with age. However, the precise size of human TCR repertoires, in both total and subsets of T cells, as well as their changes with age, are not fully characterized. We conducted a longitudinal analysis of the human blood TCRα and TCRβ repertoire of CD4+ and CD8+ T cell subsets using a unique molecular identifier–based (UMI-based) RNA-seq method. Thorough analysis of 1.9 × 108 T cells yielded the lower estimate of TCR repertoire richness in an adult at 3.8 × 108. Alterations of the TCR repertoire with age were observed in all 4 subsets of T cells. The greatest reduction was observed in naive CD8+ T cells, while the greatest clonal expansion was in memory CD8+ T cells, and the highest increased retention of TCR sequences was in memory CD8+ T cells. Our results demonstrated that age-related TCR repertoire attrition is subset specific and more profound for CD8+ than CD4+ T cells, suggesting that aging has a more profound effect on cytotoxic as opposed to helper T cell functions. This may explain the increased susceptibility of older adults to novel infections.
Collapse
Affiliation(s)
- Xiaoping Sun
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Thomas Nguyen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Achouak Achour
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Annette Ko
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Jeffrey Cifello
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Chen Ling
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Jay Sharma
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Toyoko Hiroi
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Yongqing Zhang
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Chee W Chia
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, United States of America
| | - William Wood Iii
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Wells W Wu
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Linda Zukley
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States of America
| | - Je-Nie Phue
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Kevin G Becker
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Rong-Fong Shen
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States of America
| | - Nan-Ping Weng
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| |
Collapse
|
32
|
Huisman W, Hageman L, Leboux DAT, Khmelevskaya A, Efimov GA, Roex MCJ, Amsen D, Falkenburg JHF, Jedema I. Public T-Cell Receptors (TCRs) Revisited by Analysis of the Magnitude of Identical and Highly-Similar TCRs in Virus-Specific T-Cell Repertoires of Healthy Individuals. Front Immunol 2022; 13:851868. [PMID: 35401538 PMCID: PMC8987591 DOI: 10.3389/fimmu.2022.851868] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/04/2022] [Indexed: 01/11/2023] Open
Abstract
Since multiple different T-cell receptor (TCR) sequences can bind to the same peptide-MHC combination and the number of TCR-sequences that can theoretically be generated even exceeds the number of T cells in a human body, the likelihood that many public identical (PUB-I) TCR-sequences frequently contribute to immune responses has been estimated to be low. Here, we quantitatively analyzed the TCR-repertoires of 190 purified virus-specific memory T-cell populations, directed against 21 epitopes of Cytomegalovirus, Epstein-Barr virus and Adenovirus isolated from 29 healthy individuals, and determined the magnitude, defined as prevalence within the population and frequencies within individuals, of PUB-I TCR and of TCR-sequences that are highly-similar (PUB-HS) to these PUB-I TCR-sequences. We found that almost one third of all TCR nucleotide-sequences represented PUB-I TCR amino-acid (AA) sequences and found an additional 12% of PUB-HS TCRs differing by maximally 3 AAs. We illustrate that these PUB-I and PUB-HS TCRs were structurally related and contained shared core-sequences in their TCR-sequences. We found a prevalence of PUB-I and PUB-HS TCRs of up to 50% among individuals and showed frequencies of virus-specific PUB-I and PUB-HS TCRs making up more than 10% of each virus-specific T-cell population. These findings were confirmed by using an independent TCR-database of virus-specific TCRs. We therefore conclude that the magnitude of the contribution of PUB-I and PUB-HS TCRs to these virus-specific T-cell responses is high. Because the T cells from these virus-specific memory TCR-repertoires were the result of successful control of the virus in these healthy individuals, these PUB-HS TCRs and PUB-I TCRs may be attractive candidates for immunotherapy in immunocompromised patients that lack virus-specific T cells to control viral reactivation.
Collapse
Affiliation(s)
- Wesley Huisman
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands.,Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory for Blood Cell Research, Amsterdam, Netherlands
| | - Lois Hageman
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Didier A T Leboux
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Alexandra Khmelevskaya
- Laboratory of Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| | - Grigory A Efimov
- Laboratory of Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| | - Marthe C J Roex
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Derk Amsen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory for Blood Cell Research, Amsterdam, Netherlands
| | | | - Inge Jedema
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
33
|
Rapid identification of tumor-reactive T-cell receptors by RNA preamplification-based single-cell sequencing. J Immunol Methods 2022; 504:113260. [PMID: 35331733 DOI: 10.1016/j.jim.2022.113260] [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: 02/21/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 11/23/2022]
Abstract
T-cell receptor (TCR)-transduced T (TCR-T) cell therapy has shown promising efficacy in the clinical treatment of malignant cancers. However, the populations covered by reported TCRs are still limited. Tumor infiltrating lymphocytes (TILs) are natural reservoirs of tumor-reactive T cells and TCRs. Approaches are required for the fast and cost-effective identification of tumor-reactive TCRs from TILs. The widely employed TCR identification approaches by the clonal expansion of TILs involve a TCR singularization process for the direct pairing of TCR Vα and the Vβ chain. However, the clonal expansion of T cells is well known to require extensive time and effort due to the involvement of T cell cultures. Several single-cell multiplexing PCR methods followed by Sanger sequencing have been developed, representing a cost-effective and fast approach for single-cell TCR identification. In this study, an RNA-based preamplification step was included in the single-cell TCR sequencing, which can reduce the multiplexing PCR amplification to one round. Moreover, the cDNA product of RNA preamplification is derived from the whole genome mRNA, instead of TCR mRNA only by multiplexing primers-based DNA preamplification, which is valuable for many other analyses (e.g., phenotypic analysis) of the tumor-reactive T cells that can be correlated with the identified TCRs. The feasibility for both single α chain and dual α chain TILs of this approach highlights its potential value as a rapid and cost-effective sequencing strategy for the development of TCR-T therapies for solid cancers.
Collapse
|
34
|
Suliman S, Kjer-Nielsen L, Iwany SK, Lopez Tamara K, Loh L, Grzelak L, Kedzierska K, Ocampo TA, Corbett AJ, McCluskey J, Rossjohn J, León SR, Calderon R, Lecca-Garcia L, Murray MB, Moody DB, Van Rhijn I. Dual TCR-α Expression on Mucosal-Associated Invariant T Cells as a Potential Confounder of TCR Interpretation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1389-1395. [PMID: 35246495 PMCID: PMC9359468 DOI: 10.4049/jimmunol.2100275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 01/12/2022] [Indexed: 05/20/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells are innate-like T cells that are highly abundant in human blood and tissues. Most MAIT cells have an invariant TCRα-chain that uses T cell receptor α-variable 1-2 (TRAV1-2) joined to TRAJ33/20/12 and recognizes metabolites from bacterial riboflavin synthesis bound to the Ag-presenting molecule MHC class I related (MR1). Our attempts to identify alternative MR1-presented Ags led to the discovery of rare MR1-restricted T cells with non-TRAV1-2 TCRs. Because altered Ag specificity likely alters affinity for the most potent known Ag, 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil (5-OP-RU), we performed bulk TCRα- and TCRβ-chain sequencing and single-cell-based paired TCR sequencing on T cells that bound the MR1-5-OP-RU tetramer with differing intensities. Bulk sequencing showed that use of V genes other than TRAV1-2 was enriched among MR1-5-OP-RU tetramerlow cells. Although we initially interpreted these as diverse MR1-restricted TCRs, single-cell TCR sequencing revealed that cells expressing atypical TCRα-chains also coexpressed an invariant MAIT TCRα-chain. Transfection of each non-TRAV1-2 TCRα-chain with the TCRβ-chain from the same cell demonstrated that the non-TRAV1-2 TCR did not bind the MR1-5-OP-RU tetramer. Thus, dual TCRα-chain expression in human T cells and competition for the endogenous β-chain explains the existence of some MR1-5-OP-RU tetramerlow T cells. The discovery of simultaneous expression of canonical and noncanonical TCRs on the same T cell means that claims of roles for non-TRAV1-2 TCR in MR1 response must be validated by TCR transfer-based confirmation of Ag specificity.
Collapse
Affiliation(s)
- Sara Suliman
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;
- Division of Experimental Medicine, Department of Medicine, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA
| | - Lars Kjer-Nielsen
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sarah K Iwany
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kattya Lopez Tamara
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Socios en Salud Sucursal Perú, Lima, Peru
| | - Liyen Loh
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ludivine Grzelak
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Tonatiuh A Ocampo
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Alexandra J Corbett
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | | | | | | | - Megan B Murray
- Department of Global Health and Social Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Division of Global Health Equity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; and
| | - D Branch Moody
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ildiko Van Rhijn
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
35
|
Rodríguez-Caparrós A, Álvarez-Santiago J, López-Castellanos L, Ruiz-Rodríguez C, Valle-Pastor MJ, López-Ros J, Angulo Ú, Andrés-León E, Suñé C, Hernández-Munain C. Differently Regulated Gene-Specific Activity of Enhancers Located at the Boundary of Subtopologically Associated Domains: TCRα Enhancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:910-928. [PMID: 35082160 DOI: 10.4049/jimmunol.2000864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/05/2021] [Indexed: 11/19/2022]
Abstract
Enhancers activate transcription through long-distance interactions with their cognate promoters within a particular subtopologically associated domain (sub-TAD). The TCRα enhancer (Eα) is located at the sub-TAD boundary between the TCRα and DAD1 genes and regulates transcription toward both sides in an ∼1-Mb region. Analysis of Eα activity in transcribing the unrearranged TCRα gene at the 5'-sub-TAD has defined Eα as inactive in CD4-CD8- thymocytes, active in CD4+CD8+ thymocytes, and strongly downregulated in CD4+ and CD8+ thymocytes and αβ T lymphocytes. Despite its strongly reduced activity, Eα is still required for high TCRα transcription and expression of TCRαβ in mouse and human T lymphocytes, requiring collaboration with distant sequences for such functions. Because VαJα rearrangements in T lymphocytes do not induce novel long-range interactions between Eα and other genomic regions that remain in cis after recombination, strong Eα connectivity with the 3'-sub-TAD might prevent reduced transcription of the rearranged TCRα gene. Our analyses of transcriptional enhancer dependence during T cell development and non-T lineage tissues at the 3'-sub-TAD revealed that Eα can activate the transcription of specific genes, even when it is inactive to transcribe the TCRα gene at the 5'-sub-TAD. Hence distinct requirements for Eα function are necessary at specific genes at both sub-TADs, implying that enhancers do not merely function as chromatin loop anchors that nucleate the formation of factor condensates to increase gene transcription initiated at their cognate promoters. The observed different regulated Eα activity for activating specific genes at its flanking sub-TADs may be a general feature for enhancers located at sub-TAD boundaries.
Collapse
Affiliation(s)
- Alonso Rodríguez-Caparrós
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - Jesús Álvarez-Santiago
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - Laura López-Castellanos
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - Candela Ruiz-Rodríguez
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - María Jesús Valle-Pastor
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - Jennifer López-Ros
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - Úrsula Angulo
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - Eduardo Andrés-León
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - Carlos Suñé
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| | - Cristina Hernández-Munain
- Institute of Parasitology and Biomedicine López-Neyra-Spanish National Research Council and Health Science Technology Park, Granada, Spain
| |
Collapse
|
36
|
Bystander T cells in cancer immunology and therapy. NATURE CANCER 2022; 3:143-155. [PMID: 35228747 DOI: 10.1038/s43018-022-00335-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 01/11/2022] [Indexed: 01/10/2023]
Abstract
Cancer-specific T cells are required for effective anti-cancer immunity and have a central role in cancer immunotherapy. However, emerging evidence suggests that only a small fraction of tumor-infiltrating T cells are cancer specific, and T cells that recognize cancer-unrelated antigens (so-called 'bystanders') are abundant. Although the role of cancer-specific T cells in anti-cancer immunity has been well established, the implications of bystander T cells in tumors are only beginning to be understood. It is becoming increasingly clear that bystander T cells are not a homogeneous group of cells but, instead, they differ in their specificities, their activation states and effector functions. In this Perspective, we discuss recent studies of bystander T cells in tumors, including experimental and computational approaches that enable their identification and functional analysis and viewpoints on how these insights could be used to develop new therapeutic approaches for cancer immunotherapy.
Collapse
|
37
|
Liu H, Pan W, Tang C, Tang Y, Wu H, Yoshimura A, Deng Y, He N, Li S. The methods and advances of adaptive immune receptors repertoire sequencing. Theranostics 2021; 11:8945-8963. [PMID: 34522220 PMCID: PMC8419057 DOI: 10.7150/thno.61390] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
The adaptive immune response is a powerful tool, capable of recognizing, binding to, and neutralizing a vast number of internal and external threats via T or B lymphatic receptors with widespread sets of antigen specificities. The emergence of high-throughput sequencing technology and bioinformatics provides opportunities for research in the fields of life sciences and medicine. The analysis and annotation for immune repertoire data can reveal biologically meaningful information, including immune prediction, target antigens, and effective evaluation. Continuous improvements of the immunological repertoire sequencing methods and analysis tools will help to minimize the experimental and calculation errors and realize the immunological information to meet the clinical requirements. That said, the clinical application of adaptive immune repertoire sequencing requires appropriate experimental methods and standard analytical tools. At the population cell level, we can acquire the overview of cell groups, but the information about a single cell is not obtained accurately. The information that is ignored may be crucial for understanding the heterogeneity of each cell, gene expression and drug response. The combination of high-throughput sequencing and single-cell technology allows us to obtain single-cell information with low-cost and high-throughput. In this review, we summarized the current methods and progress in this area.
Collapse
Affiliation(s)
- Hongmei Liu
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China
| | - Wenjing Pan
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China
| | - Congli Tang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Yujie Tang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hu-nan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Yan Deng
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China
| | - Nongyue He
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Song Li
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China
| |
Collapse
|
38
|
Lu YC, Zheng Z, Lowery FJ, Gartner JJ, Prickett TD, Robbins PF, Rosenberg SA. Direct identification of neoantigen-specific TCRs from tumor specimens by high-throughput single-cell sequencing. J Immunother Cancer 2021; 9:jitc-2021-002595. [PMID: 34321276 PMCID: PMC8320258 DOI: 10.1136/jitc-2021-002595] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 01/22/2023] Open
Abstract
Background Recognition of neoantigens by T cells plays a major role in cancer immunotherapy. Identification of neoantigen-specific T-cell receptors (TCRs) has become a critical research tool for studying T cell-mediated responses after immunotherapy. In addition, neoantigen-specific TCRs can be used to modify the specificity of T cells for T cell-based therapies targeting tumor-specific mutations. Although several techniques have been developed to identify TCR sequences, these techniques still require a significant amount of labor, making them impractical in the clinical setting. Methods Thanks to the availability of high-throughput single-cell sequencing, we developed a new process to isolate neoantigen-specific TCR sequences. This process included the isolation of tumor-infiltrating T cells from a tumor specimen and the stimulation of T cells by neoantigen-loaded dendritic cells, followed by single-cell sequencing for TCR and T-cell activation markers, interferon-γ and interleukin-2. Results In this study, potential neoantigen-specific TCRs were isolated from three melanoma and three colorectal tumor specimens. These TCRs were then synthesized and transduced into autologous T cells, followed by testing the recognition of neoantigens. A total of 28 neoantigen-specific TCRs were identified by this process. If identical TCR sequences were detected from two or more single cells, this approach was highly reliable (100%, 19 out of 19 TCRs). Conclusion This single-cell approach provides an efficient process to isolate antigen-specific TCRs for research and clinical applications.
Collapse
Affiliation(s)
- Yong-Chen Lu
- Surgery Branch, National Institutes of Health, Bethesda, Maryland, USA
- Department of Pathology and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Zhili Zheng
- Surgery Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Frank J Lowery
- Surgery Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Jared J Gartner
- Surgery Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Todd D Prickett
- Surgery Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Paul F Robbins
- Surgery Branch, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
39
|
Poorebrahim M, Mohammadkhani N, Mahmoudi R, Gholizadeh M, Fakhr E, Cid-Arregui A. TCR-like CARs and TCR-CARs targeting neoepitopes: an emerging potential. Cancer Gene Ther 2021; 28:581-589. [PMID: 33654227 PMCID: PMC8203496 DOI: 10.1038/s41417-021-00307-7] [Citation(s) in RCA: 35] [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: 11/20/2020] [Revised: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 01/31/2023]
Abstract
Neoepitopes or neoantigens are a spectrum of unique mutations presented in a particular patient's tumor. Neoepitope-based adoptive therapies have the potential of tumor eradication without undue damaging effect on normal tissues. In this context, methods based on the T cell receptor (TCR) engineering or chimeric antigen receptors (CARs) have shown great promise. This review focuses on the TCR-like CARs and TCR-CARs directed against tumor-derived epitopes, with a concerted view on neoepitopes. We also address the current limitations of the field to know how to harness the full benefits of this approach and thereby design a sustained and specific antitumor therapy.
Collapse
MESH Headings
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/therapeutic use
- Humans
- Immunotherapy, Adoptive
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Chimeric Antigen/genetics
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Niloufar Mohammadkhani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Reza Mahmoudi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Monireh Gholizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Fakhr
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Angel Cid-Arregui
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
40
|
Ma Y, Ou J, Lin T, Chen L, Chen J, Wang M. Next Generation Sequencing-Based Identification of T-Cell Receptors for Immunotherapy Against Hepatocellular Carcinoma. Hepatol Commun 2021; 5:1106-1119. [PMID: 34141993 PMCID: PMC8183181 DOI: 10.1002/hep4.1697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/19/2021] [Accepted: 02/04/2021] [Indexed: 01/02/2023] Open
Abstract
Hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) remains a global health concern, and HBV proteins may be ideal targets for T cell-based immunotherapy for HCC. There is a need for fast and efficient identification of HBV-specific T cell receptors (TCRs) for the development of TCR-transduced T (TCR-T) cell-based immunotherapy. Two widely employed TCR identification approaches, T cell clonal expansion and single-cell sequencing, involve a TCR singularization process for the direct identification of Vα and Vβ pairs of TCR chains. Clonal expansion of T cells is well known to have tedious time and effort requirements due to the use of T cell cultures, whereas single-cell sequencing is limited by the requirements of cell sorting and the preparation of a single-cell immune-transcriptome library as well as the massive cost of the whole procedure. Here, we present a next-generation sequencing (NGS)-based HBV-specific TCR identification that does not require the TCR singularization process. Conclusion: Two pairing strategies, ranking-based strategy and α-β chain mixture-based strategy, have proved to be useful for NGS-based TCR identification, particularly for polyclonal T cells purified by a peptide-major histocompatibility complex (pMHC) multimer-based approach. Functional evaluation confirmed the specificity and avidity of two identified HBV-specific TCRs, which may potentially be used to produce TCR-T cells to treat patients with HBV-related HCC.
Collapse
Affiliation(s)
- Yipeng Ma
- Department of Research and DevelopmentShenzhen Institute for Innovation and Translational MedicineShenzhen International Biological Valley-Life Science Industrial ParkShenzhenChina
| | - Jiayu Ou
- Department of Research and DevelopmentShenzhen Institute for Innovation and Translational MedicineShenzhen International Biological Valley-Life Science Industrial ParkShenzhenChina
| | - Tong Lin
- Department of Research and DevelopmentShenzhen Institute for Innovation and Translational MedicineShenzhen International Biological Valley-Life Science Industrial ParkShenzhenChina
| | - Lei Chen
- Department of Research and DevelopmentShenzhen Institute for Innovation and Translational MedicineShenzhen International Biological Valley-Life Science Industrial ParkShenzhenChina
| | - Junhui Chen
- Intervention and Cell Therapy CenterPeking University Shenzhen HospitalShenzhenChina
| | - Mingjun Wang
- Department of Research and DevelopmentShenzhen Institute for Innovation and Translational MedicineShenzhen International Biological Valley-Life Science Industrial ParkShenzhenChina
| |
Collapse
|
41
|
Sun B, Bugarin-Estrada E, Overend LE, Walker CE, Tucci FA, Bashford-Rogers RJM. Double-jeopardy: scRNA-seq doublet/multiplet detection using multi-omic profiling. CELL REPORTS METHODS 2021; 1:None. [PMID: 34278374 PMCID: PMC8262260 DOI: 10.1016/j.crmeth.2021.100008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/09/2021] [Accepted: 03/26/2021] [Indexed: 11/25/2022]
Abstract
The computational detection and exclusion of cellular doublets and/or multiplets is a cornerstone for the identification the true biological signals from single-cell RNA sequencing (scRNA-seq) data. Current methods do not sensitively identify both heterotypic and homotypic doublets and/or multiplets. Here, we describe a machine learning approach for doublet/multiplet detection utilizing VDJ-seq and/or CITE-seq data to predict their presence based on transcriptional features associated with identified hybrid droplets. This approach highlights the utility of leveraging multi-omic single-cell information for the generation of high-quality datasets. Our method has high sensitivity and specificity in inflammatory-cell-dominant scRNA-seq samples, thus presenting a powerful approach to ensuring high-quality scRNA-seq data.
Collapse
Affiliation(s)
- Bo Sun
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | | | | | | | | |
Collapse
|
42
|
Sponaas AM, Waage A, Vandsemb EN, Misund K, Børset M, Sundan A, Slørdahl TS, Standal T. Bystander Memory T Cells and IMiD/Checkpoint Therapy in Multiple Myeloma: A Dangerous Tango? Front Immunol 2021; 12:636375. [PMID: 33679794 PMCID: PMC7928324 DOI: 10.3389/fimmu.2021.636375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/26/2021] [Indexed: 12/19/2022] Open
Abstract
In this review article we discuss the role of the memory T cells in multiple myeloma (MM) and how they may influence immune responses in patients that received immunomodulating drugs and check point therapy.
Collapse
Affiliation(s)
- Anne Marit Sponaas
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Anders Waage
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Hematology, St.Olavs Hospital, Trondheim, Norway
| | - Esten N Vandsemb
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristine Misund
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Magne Børset
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Immunology and Transfusion Medicine, St.Olavs Hospital, Trondheim, Norway
| | - Anders Sundan
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tobias Schmidt Slørdahl
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Hematology, St.Olavs Hospital, Trondheim, Norway
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Clinical and Molecular Medicine, Center of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
43
|
Paria BC, Levin N, Lowery F, Pasetto A, Deniger DC, Parkhurst MR, Yossef R, Kim SP, Florentin M, Ngo L, Ray S, Krishna S, Robbins PF, Rosenberg SA. Rapid Identification and Evaluation of Neoantigen-reactive T-Cell Receptors From Single Cells. J Immunother 2021; 44:1-8. [PMID: 33086340 PMCID: PMC7725897 DOI: 10.1097/cji.0000000000000342] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Engineered T cells expressing tumor-specific T-cell receptors (TCRs) are emerging as a mode of personalized cancer immunotherapy that requires identification of TCRs against the products of known driver mutations and novel mutations in a timely fashion. We present a nonviral and non-next-generation sequencing platform for rapid, and efficient neoantigen-specific TCR identification and evaluation that does not require the use of recombinant cloning techniques. The platform includes an innovative method of TCRα detection using Sanger sequencing, TCR pairings and the use of TCRα/β gene fragments for putative TCR evaluation. Using patients' samples, we validated and compared our new methods head-to-head with conventional approaches used for TCR discovery. Development of a unique demultiplexing method for identification of TCRα, adaptation of synthetic TCRs for gene transfer, and a reliable reporter system significantly shortens TCR discovery time over conventional methods and increases throughput to facilitate testing prospective personalized TCRs for adoptive cell therapy.
Collapse
Affiliation(s)
- Biman C. Paria
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Noam Levin
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Frank Lowery
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Anna Pasetto
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Drew C. Deniger
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Maria R. Parkhurst
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Rami Yossef
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Sanghyun P. Kim
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Maria Florentin
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Lien Ngo
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Satyajit Ray
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Sri Krishna
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Paul F. Robbins
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | | |
Collapse
|
44
|
TCRα reporter mice reveal contribution of dual TCRα expression to T cell repertoire and function. Proc Natl Acad Sci U S A 2020; 117:32574-32583. [PMID: 33288689 DOI: 10.1073/pnas.2013188117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
It is known that a subpopulation of T cells expresses two T cell receptor (TCR) clonotypes, though the extent and functional significance of this is not established. To definitively evaluate dual TCRα cells, we generated mice with green fluorescent protein and red fluorescent protein reporters linked to TCRα, revealing that ∼16% of T cells express dual TCRs, notably higher than prior estimates. Importantly, dual TCR expression has functional consequences, as dual TCR cells predominated response to lymphocytic choriomeningitis virus infection, comprising up to 60% of virus-specific CD4+ and CD8+ T cells during acute responses. Dual receptor expression selectively influenced immune memory, as postinfection memory CD4+ populations contained significantly increased frequencies of dual TCR cells. These data reveal a previously unappreciated contribution of dual TCR cells to the immune repertoire and highlight their potential effects on immune responses.
Collapse
|
45
|
Abstract
T cells are an integral component of the adaptive immune response via the recognition of peptides by the cell surface-expressed T cell receptor (TCR). Rearrangement of the TCR genes results in a highly polymorphic repertoire on the T cells within a given individual. Although the diverse repertoire is beneficial for immune responses to foreign pathogens, recognition of self-peptides by T cells can contribute to the development of autoimmune disorders. Increasing evidence supports a pathogenic role for T cells in autoimmune pathology, and it is of interest to determine the TCR repertoires involved in autoimmune disease development. In this review, we summarize methodologies and advancements in the TCR sequencing field and discuss recent studies focused on TCR sequencing in a variety of autoimmune conditions. The rapidly evolving methodology of TCR sequencing has the potential to allow for a better understanding of autoimmune disease pathogenesis, identify disease-specific biomarkers, and aid in developing therapies to prevent and treat a number of these disorders.
Collapse
Affiliation(s)
- Angela M Mitchell
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO, USA, 80045
| | - Aaron W Michels
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO, USA, 80045
| |
Collapse
|
46
|
Kalinina AA, Nesterenko LN, Bruter AV, Balunets DV, Chudakov DM, Izraelson M, Britanova OV, Khromykh LM, Kazansky DB. Adoptive Immunotherapy Based on Chain-Centric TCRs in Treatment of Infectious Diseases. iScience 2020; 23:101854. [PMID: 33313494 PMCID: PMC7721641 DOI: 10.1016/j.isci.2020.101854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/03/2020] [Accepted: 11/19/2020] [Indexed: 12/29/2022] Open
Abstract
Complications after vaccination, lack of vaccines against certain infections, and the emergence of antibiotic-resistant microorganisms point to the need for alternative ways of protection and treatment of infectious diseases. Here, we proposed a therapeutic approach to control salmonellosis based on adoptive cell therapy. We showed that the T cell receptor (TCR) repertoire of salmonella-specific memory cells contains 20% of TCR variants with the dominant-active α-chain. Transduction of intact T lymphocytes with the dominant salmonella-specific TCRα led to their enhanced in vitro proliferation in response to salmonella. Adoptive transfer of transduced T cells resulted in a significant decrease in bacterial loads in mice infected with salmonella before or after the adoptive transfer. We demonstrated that adoptive immunotherapy based on T cells, transduced with dominant-specific TCRα could be successfully applied for treatment and prevention of infectious diseases and represent a useful addition to vaccination and existing therapeutic strategies. A regular TCR repertoire of memory T cells contains alpha-chain-centric TCRs Dominant-active TCRα, paired with random TCRβ, recognizes specific microbial antigens Adoptive immunotherapy could be applied for treatment of infections
Collapse
Affiliation(s)
- Anastasiia A Kalinina
- Federal State Budgetary Institution "N. N. Blokhin National Medical Research Center of Oncology" оf the Ministry of Health of the Russian Federation, 115478 Moscow, Russia
| | - Ludmila N Nesterenko
- "N. F. Gamaleya National Research Center of Epidemiology and Microbiology", the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Alexandra V Bruter
- Federal State Budgetary Institution "N. N. Blokhin National Medical Research Center of Oncology" оf the Ministry of Health of the Russian Federation, 115478 Moscow, Russia.,Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Denis V Balunets
- "N. F. Gamaleya National Research Center of Epidemiology and Microbiology", the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Dmitriy M Chudakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Mark Izraelson
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Olga V Britanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ludmila M Khromykh
- Federal State Budgetary Institution "N. N. Blokhin National Medical Research Center of Oncology" оf the Ministry of Health of the Russian Federation, 115478 Moscow, Russia
| | - Dmitry B Kazansky
- Federal State Budgetary Institution "N. N. Blokhin National Medical Research Center of Oncology" оf the Ministry of Health of the Russian Federation, 115478 Moscow, Russia
| |
Collapse
|
47
|
Gill RG, Burrack AL. Diverse Routes of Allograft Tolerance Disruption by Memory T Cells. Front Immunol 2020; 11:580483. [PMID: 33117387 PMCID: PMC7578217 DOI: 10.3389/fimmu.2020.580483] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Memory T lymphocytes constitute a significant problem in tissue and organ transplantation due their contribution to early rejection and their relative resistance to tolerance-promoting therapies. Memory cells generated by environmental antigen exposure, as with T cells in general, harbor a high frequency of T cell receptors (TCR) spontaneously cross-reacting with allogeneic major histocompatibility complex (MHC) molecules. This phenomenon, known as ‘heterologous’ immunity, is thought to be a key barrier to transplant tolerance induction since such memory cells can potentially react directly with essentially any prospective allograft. In this review, we describe two additional concepts that expand this commonly held view of how memory cells contribute to transplant immunity and tolerance disruption. Firstly, autoimmunity is an additional response that can comprise an endogenously generated form of heterologous alloimmunity. However, unlike heterologous immunity generated as a byproduct of indiscriminate antigen sensitization, autoimmunity can generate T cells that have the unusual potential to interact with the graft either through the recognition of graft-bearing autoantigens or by their cross-reactive (heterologous) alloimmune specificity to MHC molecules. Moreover, we describe an additional pathway, independent of significant heterologous immunity, whereby immune memory to vaccine- or pathogen-induced antigens also may impair tolerance induction. This latter form of immune recognition indirectly disrupts tolerance by the licensing of naïve alloreactive T cells by vaccine/pathogen directed memory cells recognizing the same antigen-presenting cell in vivo. Thus, there appear to be recognition pathways beyond typical heterologous immunity through which memory T cells can directly or indirectly impact allograft immunity and tolerance.
Collapse
Affiliation(s)
- Ronald G Gill
- Departments of Surgery and Immunology and Microbiology, University of Colorado Denver, Aurora, CO, United States
| | - Adam L Burrack
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
48
|
Biernacki MA, Foster KA, Woodward KB, Coon ME, Cummings C, Cunningham TM, Dossa RG, Brault M, Stokke J, Olsen TM, Gardner K, Estey E, Meshinchi S, Rongvaux A, Bleakley M. CBFB-MYH11 fusion neoantigen enables T cell recognition and killing of acute myeloid leukemia. J Clin Invest 2020; 130:5127-5141. [PMID: 32831296 PMCID: PMC7524498 DOI: 10.1172/jci137723] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Proteins created from recurrent fusion genes like CBFB-MYH11 are prevalent in acute myeloid leukemia (AML), often necessary for leukemogenesis, persistent throughout the disease course, and highly leukemia specific, making them attractive neoantigen targets for immunotherapy. A nonameric peptide derived from a prevalent CBFB-MYH11 fusion protein was found to be immunogenic in HLA-B*40:01+ donors. High-avidity CD8+ T cell clones isolated from healthy donors killed CBFB-MYH11+ HLA-B*40:01+ AML cell lines and primary human AML samples in vitro. CBFB-MYH11-specific T cells also controlled CBFB-MYH11+ HLA-B*40:01+ AML in vivo in a patient-derived murine xenograft model. High-avidity CBFB-MYH11 epitope-specific T cell receptors (TCRs) transduced into CD8+ T cells conferred antileukemic activity in vitro. Our data indicate that the CBFB-MYH11 fusion neoantigen is naturally presented on AML blasts and enables T cell recognition and killing of AML. We provide proof of principle for immunologically targeting AML-initiating fusions and demonstrate that targeting neoantigens has clinical relevance even in low-mutational frequency cancers like fusion-driven AML. This work also represents a first critical step toward the development of TCR T cell immunotherapy targeting fusion gene-driven AML.
Collapse
Affiliation(s)
- Melinda A. Biernacki
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine
| | - Kimberly A. Foster
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kyle B. Woodward
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Michael E. Coon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Carrie Cummings
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Tanya M. Cunningham
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Robson G. Dossa
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Michelle Brault
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jamie Stokke
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Pediatrics, and
| | - Tayla M. Olsen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Elihu Estey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Pediatrics, and
| | - Anthony Rongvaux
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Pediatrics, and
| |
Collapse
|
49
|
Park IA, Rajaei H, Kim YA, Lee H, Lee H, Seo JH, Heo SH, Song IH, Gong G, Lee HJ. T cell receptor repertoires of ex vivo-expanded tumor-infiltrating lymphocytes from breast cancer patients. Immunol Res 2020; 68:233-245. [PMID: 32886262 DOI: 10.1007/s12026-020-09150-8] [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: 01/25/2020] [Accepted: 08/14/2020] [Indexed: 10/23/2022]
Abstract
A higher level of tumor-infiltrating lymphocytes (TILs) is associated with better prognosis in breast cancer patients. Adoptive transfer of lymphocytes coupled with conventional therapies has appealed to many clinicians and investigators as an effective treatment strategy for cancer patients, which necessitates efficient activation and expansion of cytotoxic T lymphocytes precisely targeting cancer cells. To comprehensively understand composition of TILs and to provide a grounding in adoptive T cell therapy, we analyzed the T cell receptor (TCR) repertoires in ex vivo-expanded TILs from nine breast cancer patients via next-generation sequencing. For the three of them, TCR repertoires of TILs gathered after the initial culture during 2 weeks were additionally analyzed and compared to those of TILs that underwent ex vivo rapid expansion procedure (REP). Diversity of TCR repertoire was variable among the patients. V/J segment usage in the clonotypes was similar among patients, with variable distribution of read counts for each V/J segment. The top 50% of most frequently observed VJ combinations was present in > 80% of the total clonotypes. Compared with TCGA data, the samples contained a similar amount of recurrent CDR3 sequences, but clonotype expansion was variable among the samples. In terms of clinicopathologic factor, presence of in vitro reactivity among triple-negative breast cancer cases seemed to be related to lower Shannon's index, but p value was not statistically significant. In addition, the proportion of CD45RO+ cells out of CD8+ T cells were negatively correlated with Shannon's diversity index for both TCRα and TCRβ chains (p = 0.010) via Spearman test. In this study, we identified a heterogeneous pattern of expanded T cell clones and stable usage of V/J segments in ex vivo-expanded TILs from breast cancer patients. Further large-scale studies are requisite to elucidate the clinical significance of TCR repertoires.
Collapse
Affiliation(s)
- In Ah Park
- Department of Pathology and Tranlational Genomics, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Hajar Rajaei
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Young-Ae Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Hyeonjin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Heejae Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Jeong-Han Seo
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Sun-Hee Heo
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - In Hye Song
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
50
|
Hu F, Huang D, Luo Y, Zhou P, Lv C, Wang K, Weng Q, Liu X, Guan Y, Geng Y, Du J, Chen J, Wang J, Wu H. Hematopoietic lineage-converted T cells carrying tumor-associated antigen-recognizing TCRs effectively kill tumor cells. J Immunother Cancer 2020; 8:jitc-2019-000498. [PMID: 32669292 PMCID: PMC7368548 DOI: 10.1136/jitc-2019-000498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2020] [Indexed: 11/04/2022] Open
Abstract
Tumor-associated antigen (TAA) T-cell receptor (TCR) gene-engineered T cells exhibit great potential in antitumor immunotherapy. Considering the high costs and low availability of patient-derived peripheral blood T cells, substantial efforts have been made to explore alternatives to natural T cells. We previously reported that enforced expression of Hoxb5 converted B cells into induced T (iT) cells in vivo Here, we successfully regenerated naive OT1 (major histocompatibility complex I restricted ovalbumin antigen) iT cells (OT1-iT) in vivo by expressing Hoxb5 in pro-pre-B cells in the OT1 transgenic mouse. The OT1-iT cells can be activated and expanded in vitro in the presence of tumor cells. Particularly, these regenerated OT1-iT cells effectively eradicated tumor cells expressing the TAA (ovalbumin) both in vitro and in vivo This study provides insights into the translational applications of blood lineage-transdifferentiated T cells in immunotherapy.
Collapse
Affiliation(s)
- Fangxiao Hu
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Dehao Huang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuxuan Luo
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Peiqing Zhou
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Cui Lv
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Kaitao Wang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qitong Weng
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaofei Liu
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangzhou, Guangdong, China
| | - Yuxian Guan
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Yang Geng
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangzhou, Guangdong, China
| | - Juan Du
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangzhou, Guangdong, China
| | - Jiekai Chen
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangzhou, Guangdong, China
| | - Jinyong Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China .,CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,University of Chinese Academy of Sciences, Beijing, China.,Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangzhou, Guangdong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Hongling Wu
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China .,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangzhou, Guangdong, China
| |
Collapse
|