151
|
Induction of neoantigen-reactive T cells from healthy donors. Nat Protoc 2019; 14:1926-1943. [DOI: 10.1038/s41596-019-0170-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 03/21/2019] [Indexed: 12/21/2022]
|
152
|
Thakkar N, Bailey-Kellogg C. Balancing sensitivity and specificity in distinguishing TCR groups by CDR sequence similarity. BMC Bioinformatics 2019; 20:241. [PMID: 31092185 PMCID: PMC6521430 DOI: 10.1186/s12859-019-2864-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/29/2019] [Indexed: 12/18/2022] Open
Abstract
Background Repertoire sequencing is enabling deep explorations into the cellular immune response, including the characterization of commonalities and differences among T cell receptor (TCR) repertoires from different individuals, pathologies, and antigen specificities. In seeking to understand the generality of patterns observed in different groups of TCRs, it is necessary to balance how well each pattern represents the diversity among TCRs from one group (sensitivity) vs. how many TCRs from other groups it also represents (specificity). The variable complementarity determining regions (CDRs), particularly the third CDRs (CDR3s) interact with major histocompatibility complex (MHC)-presented epitopes from putative antigens, and thus encode the determinants of recognition. Results We here systematically characterize the predictive power that can be obtained from CDR3 sequences, using representative, readily interpretable methods for evaluating CDR sequence similarity and then clustering and classifying sequences based on similarity. An initial analysis of CDR3s of known structure, clustered by structural similarity, helps calibrate the limits of sequence diversity among CDRs that might have a common mode of interaction with presented epitopes. Subsequent analyses demonstrate that this same range of sequence similarity strikes a favorable specificity/sensitivity balance in distinguishing twins from non-twins based on overall CDR3 repertoires, classifying CDR3 repertoires by antigen specificity, and distinguishing general pathologies. Conclusion We conclude that within a fairly broad range of sequence similarity, matching CDR3 sequences are likely to share specificities. Electronic supplementary material The online version of this article (10.1186/s12859-019-2864-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Neerja Thakkar
- Department of Computer Science, Dartmouth, Hanover, NH, USA
| | | |
Collapse
|
153
|
Profiling the TRB and IGH repertoire of patients with H5N6 Avian Influenza Virus Infection by high-throughput sequencing. Sci Rep 2019; 9:7429. [PMID: 31092835 PMCID: PMC6520366 DOI: 10.1038/s41598-019-43648-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/29/2019] [Indexed: 11/16/2022] Open
Abstract
Avian Influenza A (H5N6) Virus causes severe influenza disease in humans and is manifested by acute respiratory distress syndrome, multi-organ failure, and high mortality rates. T cells recognize antigens specifically through a membrane protein T cell receptor (TCR). To ward off a wide variety of pathogens, the human adaptive immune system harbors a vast array of TCRs, which are collectively referred to as the TCR repertoire. The B cell receptor (BCR) is involved in inducing the humoral immune response. The generation of a diverse T cell and B cell repertoire is essential for protection against infection. In this study, multiplex PCR based on genomic DNA amplicons and Illumina high-throughput sequencing (HTS) were applied to study the characteristics and polymorphisms of the TRB and IGH repertoire in the peripheral blood mononuclear cells (PBMCs) from two H5N6 AIV patients and six healthy donors (NC). The CDR3 average length in the AIV group was different from the NC group. The TRBV12-3, TRBV12-4, and TRBV15 gene segments and TRBV30/TRBJ1-2, TRBV12-3/TRBJ1-1 and IGHV3-11/IGHJ6 gene segment pairings also exhibited a higher usage in the PBMCs of AIV donors and may provide more information for generating more effective T/B cell targeted diagnosis/protection strategies.
Collapse
|
154
|
Hassert M, Brien JD, Pinto AK. Mouse Models of Heterologous Flavivirus Immunity: A Role for Cross-Reactive T Cells. Front Immunol 2019; 10:1045. [PMID: 31143185 PMCID: PMC6520664 DOI: 10.3389/fimmu.2019.01045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
Most of the world is at risk of being infected with a flavivirus such as dengue virus, West Nile virus, yellow fever virus, Japanese encephalitis virus, tick-borne encephalitis virus, and Zika virus, significantly impacting millions of lives. Importantly, many of these genetically similar viruses co-circulate within the same geographic regions, making it likely for individuals living in areas of high flavivirus endemicity to be infected with multiple flaviviruses during their lifetime. Following a flavivirus infection, a robust virus-specific T cell response is generated and the memory recall of this response has been demonstrated to provide long-lasting immunity, protecting against reinfection with the same pathogen. However, multiple studies have shown that this flavivirus specific T cell response can be cross-reactive and active during heterologous flavivirus infection, leading to the question: How does immunity to one flavivirus shape immunity to the next, and how does this impact disease? It has been proposed that in some cases unfavorable disease outcomes may be caused by lower avidity cross-reactive memory T cells generated during a primary flavivirus infection that preferentially expand during a secondary heterologous infection and function sub optimally against the new pathogen. While in other cases, these cross-reactive cells still have the potential to facilitate cross-protection. In this review, we focus on cross-reactive T cell responses to flaviviruses and the concepts and consequences of T cell cross-reactivity, with particular emphasis linking data generated using murine models to our new understanding of disease outcomes following heterologous flavivirus infection.
Collapse
Affiliation(s)
- Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
155
|
Bonfiglio MF, Weinstein DM. Allergic reactions to small-molecule drugs: Will we move from reaction to prediction? Am J Health Syst Pharm 2019; 76:574-580. [DOI: 10.1093/ajhp/zxz035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Mark F Bonfiglio
- Lloyd L Gregory School of Pharmacy, Palm Beach Atlantic University, Palm Beach Gardens, FL
| | | |
Collapse
|
156
|
Wehr P, Purvis H, Law S, Thomas R. Dendritic cells, T cells and their interaction in rheumatoid arthritis. Clin Exp Immunol 2019; 196:12-27. [PMID: 30589082 PMCID: PMC6422662 DOI: 10.1111/cei.13256] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2018] [Indexed: 12/27/2022] Open
Abstract
Dendritic cells (DCs) are the key professional antigen-presenting cells which bridge innate and adaptive immune responses, inducing the priming and differentiation of naive to effector CD4+ T cells, the cross-priming of CD8+ T cells and the promotion of B cell antibody responses. DCs also play a critical role in the maintenance of immune homeostasis and tolerance. DC-T cell interactions underpin the generation of an autoimmune response in rheumatoid arthritis (RA). Here we describe the function of DCs and review evidence for DC and T cell involvement in RA pathogenesis, in particular through the presentation of self-peptide by DCs that triggers differentiation and activation of autoreactive T cells. Finally, we discuss the emerging field of targeting the DC-T cell interaction for antigen-specific immunotherapy of RA.
Collapse
Affiliation(s)
- P. Wehr
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra HospitalBrisbaneAustralia
| | - H. Purvis
- King's College London, Academic Department of Rheumatology, Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and MedicineLondonUK
| | - S.‐C. Law
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra HospitalBrisbaneAustralia
| | - R. Thomas
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra HospitalBrisbaneAustralia
| |
Collapse
|
157
|
Khosravi-Maharlooei M, Obradovic A, Misra A, Motwani K, Holzl M, Seay HR, DeWolf S, Nauman G, Danzl N, Li H, Ho SH, Winchester R, Shen Y, Brusko TM, Sykes M. Crossreactive public TCR sequences undergo positive selection in the human thymic repertoire. J Clin Invest 2019; 129:2446-2462. [PMID: 30920391 DOI: 10.1172/jci124358] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We investigated human T-cell repertoire formation using high throughput TCRβ CDR3 sequencing in immunodeficient mice receiving human hematopoietic stem cells (HSCs) and human thymus grafts. Replicate humanized mice generated diverse and highly divergent repertoires. Repertoire narrowing and increased CDR3β sharing was observed during thymocyte selection. While hydrophobicity analysis implicated self-peptides in positive selection of the overall repertoire, positive selection favored shorter shared sequences that had reduced hydrophobicity at positions 6 and 7 of CDR3βs, suggesting weaker interactions with self-peptides than unshared sequences, possibly allowing escape from negative selection. Sharing was similar between autologous and allogeneic thymi and occurred between different cell subsets. Shared sequences were enriched for allo-crossreactive CDR3βs and for Type 1 diabetes-associated autoreactive CDR3βs. Single-cell TCR-sequencing showed increased sharing of CDR3αs compared to CDR3βs between mice. Our data collectively implicate preferential positive selection for shared human CDR3βs that are highly cross-reactive. While previous studies suggested a role for recombination bias in producing "public" sequences in mice, our study is the first to demonstrate a role for thymic selection. Our results implicate positive selection for promiscuous TCRβ sequences that likely evade negative selection, due to their low affinity for self-ligands, in the abundance of "public" human TCRβ sequences.
Collapse
Affiliation(s)
- Mohsen Khosravi-Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA
| | - Aleksandar Obradovic
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA
| | - Aditya Misra
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA
| | - Keshav Motwani
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Markus Holzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA
| | - Howard R Seay
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Susan DeWolf
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA
| | - Grace Nauman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA.,Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, New York, USA
| | - Nichole Danzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA
| | - Haowei Li
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA
| | - Siu-Hong Ho
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA
| | | | - Yufeng Shen
- Center for Computational Biology and Bioinformatics, and
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, New York, USA.,Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, New York, USA.,Department of Surgery, Columbia University Medical Center, Columbia University, New York, New York, USA
| |
Collapse
|
158
|
Schober K, Buchholz VR, Busch DH. TCR repertoire evolution during maintenance of CMV-specific T-cell populations. Immunol Rev 2019; 283:113-128. [PMID: 29664573 DOI: 10.1111/imr.12654] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During infections and cancer, the composition of the T-cell receptor (TCR) repertoire of antigen-specific CD8+ T cells changes over time. TCR avidity is thought to be a major driver of this process, thereby interacting with several additional regulators of T-cell responses to form a composite immune response architecture. Infections with latent viruses, such as cytomegalovirus (CMV), can lead to large T-cell responses characterized by an oligoclonal TCR repertoire. Here, we review the current status of experimental studies and theoretical models of TCR repertoire evolution during CMV infection. We will particularly discuss the degree to which this process may be determined through structural TCR avidity. As engineered TCR-redirected T cells have moved into the spotlight for providing more effective immunotherapies, it is essential to understand how the key features of a given TCR influence T-cell expansion and maintenance in settings of infection or malignancy. Deeper insights into these mechanisms will improve our basic understanding of T-cell immunology and help to identify optimal TCRs for immunotherapy.
Collapse
Affiliation(s)
- Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany.,Focus Group 'Clinical Cell Processing and Purification', Institute for Advanced Study, TUM, Munich, Germany.,National Centre for Infection Research (DZIF), Munich, Germany
| |
Collapse
|
159
|
Ma J, Sun G, Zhu P, Liu S, Ou M, Chen Z, Zou C, Chan FL, Dai Y, Sui W. Determination of the complexity and diversity of the TCR β-chain CDR3 repertoire in bladder cancer using high-throughput sequencing. Oncol Lett 2019; 17:3808-3816. [PMID: 30881501 PMCID: PMC6403507 DOI: 10.3892/ol.2019.10015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 12/06/2018] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to investigate the complexity and diversity of the T lymphocyte immune repertoire in patients with bladder cancer. To do so, the immune state of patients was assessed. The study also aimed to elucidate the aetiology and pathogenesis of bladder cancer to provide a novel theoretical basis for disease prevention, diagnosis, treatment and prognosis monitoring. Cancerous and paracancerous (control) tissue samples were collected from five patients diagnosed with muscle-invasive bladder cancer. Multiplex polymerase chain reaction and Illumina high-throughput sequencing were used to determine the characteristics and clonal diversity of the T-cell receptor (TCR) β-chain complementarity-determining region 3 (CDR3) gene in the cancerous and paracancerous tissues of patients with bladder cancer. The degree of clonal expansion in malignant samples was significantly higher than in adjacent samples. Furthermore, ΤCRβ variable (TRBV), ΤCRβ diversity (TRBD) and ΤCRβ joining (TRBJ) repertoires were significantly different in cancerous samples compared with adjacent samples. In addition, 13 identified V-J pairs were highly expressed in cancerous samples whereas they had low expression in control samples. In conclusion, the degree of T-cell clonal expansion in bladder cancerous tissue was higher than in paracancerous tissue, whereas the immune diversity of the tissues of patients with bladder cancer was significantly lower. The DNA sequence and amino acid sequences, and V-J combination level may be used to comprehensively understand the diversity and characteristics of TCR CDR3 in bladder cancer and paracancerous tissues, and to evaluate the immune status of bladder cancer to develop therapeutic targets and biomarkers for prognosis monitoring.
Collapse
Affiliation(s)
- Jingsheng Ma
- Nephrology Department, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin No. 181 Hospital (The Affiliated Guilim Hospital of Southern Medical University), Guilin, Guangxi 541002, P.R. China
| | - Guoping Sun
- Central Laboratory of Shenzhen Pingshan People's Hospital, Shenzhen, Guangdong 518118, P.R. China
| | - Peng Zhu
- Central Laboratory of Shenzhen Pingshan People's Hospital, Shenzhen, Guangdong 518118, P.R. China
| | - Song Liu
- Clinical Medical Research Center of The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Minglin Ou
- Clinical Medical Research Center of The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Zhiqiang Chen
- Clinical Medical Research Center of The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Chang Zou
- Clinical Medical Research Center of The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Frank Leing Chan
- Cancer Biology and Experimental Therapeutics, School of Biomedical Sciences, The Chinese University of Hong Kong, SAR, P.R. China
| | - Yong Dai
- Clinical Medical Research Center of The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Weiguo Sui
- Nephrology Department, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin No. 181 Hospital (The Affiliated Guilim Hospital of Southern Medical University), Guilin, Guangxi 541002, P.R. China
| |
Collapse
|
160
|
Jiang N, Schonnesen AA, Ma KY. Ushering in Integrated T Cell Repertoire Profiling in Cancer. Trends Cancer 2019; 5:85-94. [PMID: 30755308 PMCID: PMC6544389 DOI: 10.1016/j.trecan.2018.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/28/2022]
Abstract
Advances in immune profiling techniques have dramatically changed the cancer immunotherapy and monitoring landscape. High-throughput protein and gene expression technologies have paved the way for the discovery of therapeutic targets and biomarkers, and have made monitoring therapeutic response possible through the ability to independently assay the phenotype, specificity, exhaustion status, and lineage of single T cells. Although valuable insights into response profiling have been gained with current technologies, it has become evident that single-method profiling is insufficient to accurately capture an antitumor T cell response. We discuss and propose new methods that combine multiple axes of analysis to provide a comprehensive analysis of T cell repertoire in the fight against cancer.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA; LIVESTRONG Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA.
| | - Alexandra A Schonnesen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Ke-Yue Ma
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
161
|
Sprouse ML, Bates NA, Felix KM, Wu HJJ. Impact of gut microbiota on gut-distal autoimmunity: a focus on T cells. Immunology 2019; 156:305-318. [PMID: 30560993 DOI: 10.1111/imm.13037] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/07/2018] [Accepted: 11/25/2018] [Indexed: 12/14/2022] Open
Abstract
The immune system is essential for maintaining a delicate balance between eliminating pathogens and maintaining tolerance to self-tissues to avoid autoimmunity. An enormous and complex community of gut microbiota provides essential health benefits to the host, particularly by regulating immune homeostasis. Many of the metabolites derived from commensals can impact host health by directly regulating the immune system. Many autoimmune diseases arise from an imbalance between pathogenic effector T cells and regulatory T (Treg) cells. Recent interest has emerged in understanding how cross-talk between gut microbiota and the host immune system promotes autoimmune development by controlling the differentiation and plasticity of T helper and Treg cells. At the molecular level, our recent study, along with others, demonstrates that asymptomatic colonization by commensal bacteria in the gut is capable of triggering autoimmune disease by molecular mimicking self-antigen and skewing the expression of dual T-cell receptors on T cells. Dysbiosis, an imbalance of the gut microbiota, is involved in autoimmune development in both mice and humans. Although it is well known that dysbiosis can impact diseases occurring within the gut, growing literature suggests that dysbiosis also causes the development of gut-distal/non-gut autoimmunity. In this review, we discuss recent advances in understanding the potential molecular mechanisms whereby gut microbiota induces autoimmunity, and the evidence that the gut microbiota triggers gut-distal autoimmune diseases.
Collapse
Affiliation(s)
- Maran L Sprouse
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Nicholas A Bates
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Krysta M Felix
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Hsin-Jung Joyce Wu
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA.,Arizona Arthritis Center, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
162
|
Huth A, Liang X, Krebs S, Blum H, Moosmann A. Antigen-Specific TCR Signatures of Cytomegalovirus Infection. THE JOURNAL OF IMMUNOLOGY 2018; 202:979-990. [PMID: 30587531 DOI: 10.4049/jimmunol.1801401] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/19/2018] [Indexed: 11/19/2022]
Abstract
CMV is a prevalent human pathogen. The virus cannot be eliminated from the body, but is kept in check by CMV-specific T cells. Patients with an insufficient T cell response, such as transplant recipients, are at high risk of developing CMV disease. However, the CMV-specific T cell repertoire is complex, and it is not yet clear which T cells protect best against virus reactivation and disease. In this study, we present a highly resolved characterization of CMV-specific human CD8+ T cells based on enrichment by specific peptide stimulation and mRNA sequencing of their TCR β-chains (TCRβ). Our analysis included recently identified T cell epitopes restricted through HLA-C, whose presentation is resistant to viral immunomodulation, and well-studied HLA-B-restricted epitopes. In eight healthy virus carriers, we identified a total of 1052 CMV-specific TCRβ sequences. HLA-C-restricted, CMV-specific TCRβ clonotypes dominated the ex vivo T cell response and contributed the highest-frequency clonotype of the entire repertoire in two of eight donors. We analyzed sharing and similarity of CMV-specific TCRβ sequences and identified 63 public or related sequences belonging to 17 public TCRβ families. In our cohort, and in an independent cohort of 352 donors, the cumulative frequency of these public TCRβ family members was a highly discriminatory indicator of carrying both CMV infection and the relevant HLA type. Based on these findings, we propose CMV-specific TCRβ signatures as a biomarker for an antiviral T cell response to identify patients in need of treatment and to guide future development of immunotherapy.
Collapse
Affiliation(s)
- Alina Huth
- German Center for Infection Research Group Host Control of Viral Latency and Reactivation, Research Unit Gene Vectors, Helmholtz Center Munich, 81377 Munich, Germany.,Deutsches Zentrum für Infektionsforschung, 81377 Munich, Germany; and
| | - Xiaoling Liang
- German Center for Infection Research Group Host Control of Viral Latency and Reactivation, Research Unit Gene Vectors, Helmholtz Center Munich, 81377 Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Andreas Moosmann
- German Center for Infection Research Group Host Control of Viral Latency and Reactivation, Research Unit Gene Vectors, Helmholtz Center Munich, 81377 Munich, Germany; .,Deutsches Zentrum für Infektionsforschung, 81377 Munich, Germany; and
| |
Collapse
|
163
|
Vojtek I, Buchy P, Doherty TM, Hoet B. Would immunization be the same without cross-reactivity? Vaccine 2018; 37:539-549. [PMID: 30591255 DOI: 10.1016/j.vaccine.2018.12.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/07/2018] [Accepted: 12/04/2018] [Indexed: 01/08/2023]
Abstract
"Cross-reactivity" (the observed immune response against pathogen types not specifically targeted by the vaccine antigen composition) and "cross-protection" (clinical protection against related non-vaccine microorganism types) are vaccinology concepts that are attracting renewed interest in the context of disease prevention. National health authorities are collecting mounting evidence of the importance of cross-reactivity. For some vaccines, this has been substantiated by cross-protection data from clinical studies and/or post-licensure data, where their introduction into immunization programmes has shown beneficial impacts on disease caused by related non-vaccine microorganisms. This knowledge has influenced the way new vaccines are designed, developed, and evaluated in real-life settings. Some of the new vaccines are now designed with the specific aim of having a greater breadth of protection. Ideal vaccine antigens therefore include epitopes with conserved homology across related pathogen types, because it is not always possible to include the antigens of all the individual types of a given pathogen species. The use of novel adjuvants with greater immunostimulatory properties can also contribute to improved overall vaccine cross-reactivity, as could the use of antigen delivery platforms. The growing body of evidence allows us to better understand the full impact of vaccines - beyond vaccine-type disease - which should be taken into consideration when assessing the full value of vaccination programmes.
Collapse
Affiliation(s)
- Ivo Vojtek
- GSK, Avenue Fleming 20, 1300 Wavre, Belgium.
| | | | | | | |
Collapse
|
164
|
Riley TP, Baker BM. The intersection of affinity and specificity in the development and optimization of T cell receptor based therapeutics. Semin Cell Dev Biol 2018; 84:30-41. [DOI: 10.1016/j.semcdb.2017.10.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 10/07/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022]
|
165
|
Rojas M, Restrepo-Jiménez P, Monsalve DM, Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, Leung PS, Ansari AA, Gershwin ME, Anaya JM. Molecular mimicry and autoimmunity. J Autoimmun 2018; 95:100-123. [DOI: 10.1016/j.jaut.2018.10.012] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 12/15/2022]
|
166
|
Kavazović I, Polić B, Wensveen FM. Cheating the Hunger Games; Mechanisms Controlling Clonal Diversity of CD8 Effector and Memory Populations. Front Immunol 2018; 9:2831. [PMID: 30555492 PMCID: PMC6281969 DOI: 10.3389/fimmu.2018.02831] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/16/2018] [Indexed: 11/23/2022] Open
Abstract
Effector and memory CD8 T cells have an intrinsic difference in the way they must approach antigen; effector cells need to address the pathogen at hand and therefore favor outgrowth of only high-affinity clones. In contrast, the memory pool benefits from greater clonal diversity to recognize and eliminate pathogens with mutations in their immunogenic epitopes. Effector and memory fates are ultimately the result of the same three signals that control T cell activation; T cell receptor (TCR) engagement together with co-stimulation and cytokines. Great progress has been made in our understanding of the transcriptional programs that drive effector or memory differentiation. However, how these two different programs result from the same initial cues is still a matter of debate. An emerging image is that not only the classical three signals determine T cell differentiation, but also the ability of cells to access these signals relative to that of other activated clones. Inter-clonal competition is therefore not only a selective force, but also a mediator of CD8 T cell fate. How this is regulated on a transcriptional level, especially in the context of a selective “hunger game” based on antigen-affinity in which only cells of high-affinity are supposed to survive, is still poorly defined. In this review, we discuss recent literature that illustrates how antigen-affinity dependent inter-clonal competition shapes effector and memory populations in an environment of antigen affinity-driven selection. We argue that fine-tuning of TCR signal intensity presents an attractive target for regulating the scope of CD8 T cell vaccines.
Collapse
Affiliation(s)
- Inga Kavazović
- Department of Histology & Embryology University of Rijeka, Rijeka, Croatia
| | - Bojan Polić
- Department of Histology & Embryology University of Rijeka, Rijeka, Croatia
| | - Felix M Wensveen
- Department of Histology & Embryology University of Rijeka, Rijeka, Croatia.,Department of Experimental Immunology, Amsterdam University Medical Center University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
167
|
powerTCR: A model-based approach to comparative analysis of the clone size distribution of the T cell receptor repertoire. PLoS Comput Biol 2018; 14:e1006571. [PMID: 30485278 PMCID: PMC6287877 DOI: 10.1371/journal.pcbi.1006571] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 12/10/2018] [Accepted: 10/19/2018] [Indexed: 11/19/2022] Open
Abstract
Sequencing of the T cell receptor (TCR) repertoire is a powerful tool for deeper study of immune response, but the unique structure of this type of data makes its meaningful quantification challenging. We introduce a new method, the Gamma-GPD spliced threshold model, to address this difficulty. This biologically interpretable model captures the distribution of the TCR repertoire, demonstrates stability across varying sequencing depths, and permits comparative analysis across any number of sampled individuals. We apply our method to several datasets and obtain insights regarding the differentiating features in the T cell receptor repertoire among sampled individuals across conditions. We have implemented our method in the open-source R package powerTCR. A more detailed understanding of the immune response can unlock critical information concerning diagnosis and treatment of disease. Here, in particular, we study T cells through T cell receptor sequencing, as T cells play a vital role in immune response. One important feature of T cell receptor sequencing data is the frequencies of each receptor in a given sample. These frequencies harbor global information about the landscape of the immune response. We introduce a flexible method that extracts this information by modeling the distribution of these frequencies, and show that it can be used to quantify differences in samples from individuals of different biological conditions.
Collapse
|
168
|
Bentzen AK, Such L, Jensen KK, Marquard AM, Jessen LE, Miller NJ, Church CD, Lyngaa R, Koelle DM, Becker JC, Linnemann C, Schumacher TNM, Marcatili P, Nghiem P, Nielsen M, Hadrup SR. T cell receptor fingerprinting enables in-depth characterization of the interactions governing recognition of peptide-MHC complexes. Nat Biotechnol 2018; 36:nbt.4303. [PMID: 30451992 PMCID: PMC9452375 DOI: 10.1038/nbt.4303] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/24/2018] [Indexed: 02/08/2023]
Abstract
The promiscuous nature of T-cell receptors (TCRs) allows T cells to recognize a large variety of pathogens, but makes it challenging to understand and control T-cell recognition. Existing technologies provide limited information about the key requirements for T-cell recognition and the ability of TCRs to cross-recognize structurally related elements. Here we present a 'one-pot' strategy for determining the interactions that govern TCR recognition of peptide-major histocompatibility complex (pMHC). We measured the relative affinities of TCRs to libraries of barcoded peptide-MHC variants and applied this knowledge to understand the recognition motif, here termed the TCR fingerprint. The TCR fingerprints of 16 different TCRs were identified and used to predict and validate cross-recognized peptides from the human proteome. The identified fingerprints differed among TCRs recognizing the same epitope, demonstrating the value of this strategy for understanding T-cell interactions and assessing potential cross-recognition before selection of TCRs for clinical development.
Collapse
Affiliation(s)
- Amalie K Bentzen
- Department of Micro and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Lina Such
- Department of Micro and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Kamilla K Jensen
- Department of Bio and Health Informatics, Technical University of Denmark, Lyngby, Denmark
| | - Andrea M Marquard
- Department of Micro and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Leon E Jessen
- Department of Micro and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
- Department of Bio and Health Informatics, Technical University of Denmark, Lyngby, Denmark
| | - Natalie J Miller
- Department of Medicine, Divisions of Dermatology, University of Washington, Seattle, Washington, USA
| | - Candice D Church
- Department of Medicine, Divisions of Dermatology, University of Washington, Seattle, Washington, USA
| | - Rikke Lyngaa
- Department of Micro and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Division of Vaccine and Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jürgen C Becker
- Translational Skin Cancer Research, University Hospital Essen and University of Duisburg-Essen, Essen, Germany
| | - Carsten Linnemann
- Department of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ton N M Schumacher
- Department of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paolo Marcatili
- Department of Bio and Health Informatics, Technical University of Denmark, Lyngby, Denmark
| | - Paul Nghiem
- Department of Medicine, Divisions of Dermatology, University of Washington, Seattle, Washington, USA
| | - Morten Nielsen
- Department of Bio and Health Informatics, Technical University of Denmark, Lyngby, Denmark
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Sine R Hadrup
- Department of Micro and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
169
|
Bai Y, Wang D, Li W, Huang Y, Ye X, Waite J, Barry T, Edelmann KH, Levenkova N, Guo C, Skokos D, Wei Y, Macdonald LE, Fury W. Evaluation of the capacities of mouse TCR profiling from short read RNA-seq data. PLoS One 2018; 13:e0207020. [PMID: 30439982 PMCID: PMC6237323 DOI: 10.1371/journal.pone.0207020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/22/2018] [Indexed: 11/18/2022] Open
Abstract
Profiling T cell receptor (TCR) repertoire via short read transcriptome sequencing (RNA-Seq) has a unique advantage of probing simultaneously TCRs and the genome-wide RNA expression of other genes. However, compared to targeted amplicon approaches, the shorter read length is more prone to mapping error. In addition, only a small percentage of the genome-wide reads may cover the TCR loci and thus the repertoire could be significantly under-sampled. Although this approach has been applied in a few studies, the utility of transcriptome sequencing in probing TCR repertoires has not been evaluated extensively. Here we present a systematic assessment of RNA-Seq in TCR profiling. We evaluate the power of both Fluidigm C1 full-length single cell RNA-Seq and bulk RNA-Seq in characterizing the repertoires of different diversities under either naïve conditions or after immunogenic challenges. Standard read length and sequencing coverage were employed so that the evaluation was conducted in accord with the current RNA-Seq practices. Despite high sequencing depth in bulk RNA-Seq, we encountered difficulty quantifying TCRs with low transcript abundance (<1%). Nevertheless, top enriched TCRs with an abundance of 1–3% or higher can be faithfully detected and quantified. When top TCR sequences are of interest and transcriptome sequencing is available, it is worthwhile to conduct a TCR profiling using the RNA-Seq data.
Collapse
Affiliation(s)
- Yu Bai
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
- * E-mail: (YB); (WF)
| | - David Wang
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Wentian Li
- Robert S. Boas Center for Genomics & Human Genetics, Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York, United States of America
| | - Ying Huang
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Xuan Ye
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Janelle Waite
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Thomas Barry
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Kurt H. Edelmann
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Natasha Levenkova
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Chunguang Guo
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Dimitris Skokos
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Yi Wei
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Lynn E. Macdonald
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
| | - Wen Fury
- Regeneron Pharmaceuticals, Tarrytown, New York, United States of America
- * E-mail: (YB); (WF)
| |
Collapse
|
170
|
Jin YB, Luo W, Zhang GY, Lin KR, Cui JH, Chen XP, Pan YM, Mao XF, Tang J, Wang YJ. TCR repertoire profiling of tumors, adjacent normal tissues, and peripheral blood predicts survival in nasopharyngeal carcinoma. Cancer Immunol Immunother 2018; 67:1719-1730. [PMID: 30155576 PMCID: PMC11028245 DOI: 10.1007/s00262-018-2237-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/23/2018] [Indexed: 12/29/2022]
Abstract
The T-cell immune responses in nasopharyngeal carcinoma patients have been extensively investigated recently for designing adoptive immunotherapy or immune checkpoint blockade therapy. However, the distribution characteristics of T cells associated with NPC pathogenesis are largely unknown. We performed deep sequencing for TCR repertoire profiling on matched tumor/adjacent normal tissue from 15 NPC patients and peripheral blood from 39 NPC patients, 39 patients with other nasopharyngeal diseases, and 33 healthy controls. We found that a lower diversity of TCR repertoire in tumors than paired tissues or a low similarity between the paired tissues was associated with a poor prognosis in NPC. A more diverse TCR repertoire was identified in the peripheral blood of NPC patients relative to the controls; this was related to a significant decrease in the proportion of high-frequency TCR clones in NPC. Higher diversity in peripheral blood of NPC patients was associated with a worse prognosis. Due to the peculiarity of the Vβ gene usage patterns in the peripheral blood of NPC patients, 15 Vβ genes were selected to distinguish NPC patients from controls by the least absolute shrinkage and selection operator analysis. We identified 11 clonotypes shared by tumors and peripheral blood samples from different NPC patients, defined as "NPC-associated" that might have value in adoptive immunotherapy. In conclusion, we here report the systematic and overall characteristics of the TCR repertoire in tumors, adjacent normal tissues, and peripheral blood of NPC patients. The data obtained may be relevant to future clinical studies in the setting of immunotherapy for NPC patients.
Collapse
Affiliation(s)
- Ya-Bin Jin
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Wei Luo
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China.
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China.
| | - Guo-Yi Zhang
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Cancer Center, Foshan Hospital, Sun Yat-sen University, Foshan, 528000, Guangdong, China
| | - Kai-Rong Lin
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Jin-Huan Cui
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Xiang-Ping Chen
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Ying-Ming Pan
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Xiao-Fan Mao
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Jun Tang
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Otolaryngology Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, Foshan, 528000, Guangdong, China
| | - Yue-Jian Wang
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China.
- Otolaryngology Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, Foshan, 528000, Guangdong, China.
| |
Collapse
|
171
|
Zhang J, Hu M, Wang B, Gao J, Wang L, Li L, Chen S, Cui B, Gu W, Wang W, Ning G. Comprehensive assessment of T-cell repertoire following autologous hematopoietic stem cell transplantation for treatment of type 1 diabetes using high-throughput sequencing. Pediatr Diabetes 2018; 19:1229-1237. [PMID: 30022578 DOI: 10.1111/pedi.12728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/17/2018] [Accepted: 07/09/2018] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE We aimed to investigate T-cell receptor (TCR) repertoires in type 1 diabetes (T1D) patients receiving autologous hematopoietic stem cell transplantation (AHSCT) treatment. METHODS High-throughput deep TCR beta (TCRB) chain sequencing was performed to assess millions of individual TCRs in five T1D patients receiving AHSCT treatment and another five patients receiving insulin treatment during 12 months of follow-up. RESULTS No significant changes in TCRB sequence reads, complementarity-determining region 3 (CDR3) sequences, or the usage of TCRB VJ gene-segments were observed at 12 months after AHSCT. Compared with the baseline, the usage of TCRB VJ gene-segments at 12 months decreased in the insulin treatment group (1836.4 ± 437.7 vs 2763.6 ± 390.6, P = 0.015), and the change rates were larger than those undergoing AHSCT (-0.62 ± 0.16 vs 0.06 ± 0.45, P = 0.002). Changes in the TCR repertoire were smaller after AHSCT than those with insulin treatment (P = 2.2*10-32 ). TCRBV 7-7/TCRBJ 2-5 was depleted after AHSCT while expanded with insulin treatment. TCRBV 12-4, TCRBV 10-3, TCRBV 12-3/TCRBJ 1-2 were expanded after AHSCT while ablated with insulin treatment. CONCLUSIONS We found that AHSCT is safe without reduction in the diversity of TCR repertoires and TCR repertoires tend to be more stable after AHSCT. Furthermore, these four candidate TCRBV/TCRBJ gene usages on CDR3 regions may act as therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Juanjuan Zhang
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China
| | - Min Hu
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Bokai Wang
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China
| | - Jie Gao
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China
| | - Li Wang
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China
| | - Li Li
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China
| | - Sisi Chen
- Beijing Genomics Institute, Shenzhen, Guangdong, China
| | - Bin Cui
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic diseases, Ruijin Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases and Shanghai E-institute for Endocrinology, Shanghai, China.,Laboratory for Endocrine & Metabolic Diseases, Institute of Health Science, Shanghai JiaoTong University, School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
172
|
Yan Q, Wang L, Lai L, Liu S, Chen H, Zhang J, Dai Y, Sui W. Next generation sequencing reveals novel alterations in B-cell heavy chain receptor repertoires associated with acute-on-chronic liver failure. Int J Mol Med 2018; 43:243-255. [PMID: 30365073 PMCID: PMC6257861 DOI: 10.3892/ijmm.2018.3946] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 08/30/2018] [Indexed: 12/30/2022] Open
Abstract
Acute-on-chronic liver failure (ACLF) is a newly-defined serious syndrome with major features of acute decompensation (AD) of hepatic cirrhosis, liver failure and failure of multiple other organs. To date, the mechanism underlying the development and progression of ACLF remains to be fully elucidated. It has been noted that ACLF is associated with immune dysregulation. However, studies have mainly focused on T-cell responses. The present study aimed to determine the composition and alterations of B-cell receptor (BCR) heavy chain repertoires associated with ACLF using next generation sequencing (NGS). A total of six patients with hepatitis B virus (HBV)-related ACLF and six healthy control subjects were prospectively enrolled in the present study. The B-cell immunoglobulin heavy chain (IGH) repertoires in peripheral blood mononuclear cells (PBMCs) obtained from the patients with HBV-related ACLF and the control subjects were analyzed using NGS, coupled with multiplex polymerase chain reaction, were Illumina sequenced, and were further characterized using the international ImMunoGeneTics database. The distribution of the BCR complementarity-determining region 3 (CDR3) variable (V), diversity (D) and joining (J) and V-J gene segments were found to be comparable between the ACLF and control groups. Of note, the degree of clonal expansion in the ACLF group was significantly higher than that in the control group (P<0.05). Furthermore, a t-test of the distribution ratio of the V, D, J and V-J combinations in patients with ACLF and control subjects revealed differentially expressed genes. In total, six genes were upregulated and 19 genes were downregulated in response to ACLF. The difference between these two groups was statistically significant (P<0.05). The approach used in the present study was feasible and effective for analyzing peripheral B-cell repertoires in HBV-related ACLF. These results provide direct evidence that the BCR repertoire is important in immune responses, autoimmunity and alloreactivity, and that there is a link between the BCR repertoire and HBV-ACLF. Therefore, ACLF-specific BCR CDR3 sequences hold promise for therapeutic benefit to HBV-ACLF in the future.
Collapse
Affiliation(s)
- Qiang Yan
- Department of Nephrology, No. 181 Hospital of Guilin, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Diseases Research, Guilin, Guangxi 541002, P.R. China
| | - Lei Wang
- Department of Nephrology, No. 181 Hospital of Guilin, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Diseases Research, Guilin, Guangxi 541002, P.R. China
| | - Liusheng Lai
- Department of Nephrology, No. 181 Hospital of Guilin, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Diseases Research, Guilin, Guangxi 541002, P.R. China
| | - Song Liu
- Clinical Medical Research Center, Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Huaizhou Chen
- Department of Nephrology, No. 181 Hospital of Guilin, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Diseases Research, Guilin, Guangxi 541002, P.R. China
| | - Jiaxing Zhang
- Department of Nephrology, No. 181 Hospital of Guilin, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Diseases Research, Guilin, Guangxi 541002, P.R. China
| | - Yong Dai
- Clinical Medical Research Center, Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Weiguo Sui
- Department of Nephrology, No. 181 Hospital of Guilin, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin Key Laboratory of Kidney Diseases Research, Guilin, Guangxi 541002, P.R. China
| |
Collapse
|
173
|
Lanzer KG, Cookenham T, Reiley WW, Blackman MA. Virtual memory cells make a major contribution to the response of aged influenza-naïve mice to influenza virus infection. IMMUNITY & AGEING 2018; 15:17. [PMID: 30093911 PMCID: PMC6081820 DOI: 10.1186/s12979-018-0122-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
Background A diverse repertoire of naïve T cells is thought to be essential for a robust response to new infections. However, a key aspect of aging of the T cell compartment is a decline in numbers and diversity of peripheral naïve T cells. We have hypothesized that the age-related decline in naïve T cells forces the immune system to respond to new infections using cross-reactive memory T cells generated to previous infections that dominate the aged peripheral T cell repertoire. Results Here we confirm that the CD8 T cell response of aged, influenza-naïve mice to primary infection with influenza virus is dominated by T cells that derive from the memory T cell pool. These cells exhibit the phenotypic characteristics of virtual memory cells rather than true memory cells. Furthermore, we find that the repertoire of responding CD8 T cells is constrained compared with that of young mice, and differs significantly between individual aged mice. After infection, these virtual memory CD8 T cells effectively develop into granzyme-producing effector cells, and clear virus with kinetics comparable to naïve CD8 T cells from young mice. Conclusions The response of aged, influenza-naive mice to a new influenza infection is mediated largely by memory CD8 T cells. However, unexpectedly, they have the phenotype of VM cells. In response to de novo influenza virus infection, the VM cells develop into granzyme-producing effector cells and clear virus with comparable kinetics to young CD8 T cells.
Collapse
Affiliation(s)
| | - Tres Cookenham
- Trudeau Institute, 154 Algonquin Avenue, Saranac Lake, NY 12983 USA
| | - William W Reiley
- Trudeau Institute, 154 Algonquin Avenue, Saranac Lake, NY 12983 USA
| | | |
Collapse
|
174
|
Migalska M, Sebastian A, Radwan J. Profiling of the TCRβ repertoire in non-model species using high-throughput sequencing. Sci Rep 2018; 8:11613. [PMID: 30072736 PMCID: PMC6072738 DOI: 10.1038/s41598-018-30037-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/19/2018] [Indexed: 02/06/2023] Open
Abstract
In recent years, immune repertoire profiling with high-throughput sequencing (HTS) has advanced our understanding of adaptive immunity. However, fast progress in the field applied mostly to human and mouse research, with only few studies devoted to other model vertebrates. We present the first in-depth characterization of the T-cell receptor (TCR) repertoire in a non-model mammal (bank vole, Myodes glareolus), widely used in ecological and evolutionary research. We used RNA from spleens, 5′RACE and HTS to describe V and J segments of TCRβ, qualitatively characterize preferential V–J segment usage and CDR3 length distribution. Overall orthology to murine genes was preserved, with 11 J and 37 V genes found in voles (although 3 V genes lacked a close orthologue). Further, we implemented unique molecular identifiers for quantitative analysis of CDR3 repertoire with stringent error correction. A conservative, lower bound estimation of the TCRβ repertoire was similar to that found for mice (1.7–2.3 × 105 clonotypes). We hope that by providing an easy-to-follow molecular protocol and on-line bioinformatics tools that do not require reference sequences (AmpliTCR and AmpliCDR3), we will encourage HTS immune repertoire profiling in other non-model vertebrates, thus opening new research avenues in e.g. comparative immunology, ecology and evolutionary biology.
Collapse
Affiliation(s)
- Magdalena Migalska
- Evolutionary Biology Group, Faculty of Biology, Adam Mickiewicz University, ul. Umultowska 89, 61-614, Poznan, Poland.
| | - Alvaro Sebastian
- Evolutionary Biology Group, Faculty of Biology, Adam Mickiewicz University, ul. Umultowska 89, 61-614, Poznan, Poland.,Instituto Aragonés de Empleo (INAEM), c/Royo Villanova 1, 50007, Zaragoza, Spain
| | - Jacek Radwan
- Evolutionary Biology Group, Faculty of Biology, Adam Mickiewicz University, ul. Umultowska 89, 61-614, Poznan, Poland
| |
Collapse
|
175
|
Unexpected High Incidence of Human Herpesvirus-6 Encephalitis after Naive T Cell-Depleted Graft of Haploidentical Stem Cell Transplantation in Pediatric Patients. Biol Blood Marrow Transplant 2018; 24:2316-2323. [PMID: 30031939 DOI: 10.1016/j.bbmt.2018.07.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/10/2018] [Indexed: 11/21/2022]
Abstract
The CD45RA T cell depletion (TCD) method has been used to deplete naive T cells, preventing graft-versus-host disease (GVHD) but preserving memory cells, providing immediate functional T cells with anti-infection, antileukemia, and antirejection effects. We describe a series of 25 consecutive high-risk patients with leukemia who received haploidentical hematopoietic stem cell transplantation (haplo-HSCT) with CD45RA TCD. Each patient received 2 cell products: 1 created by CD34 positive selection and the other through CD45RA depletion from the CD34 negative fraction by a CliniMACS device. CD45RA-depleted haplo-HSCT was well tolerated, with rapid engraftment and low risk of severe acute GVHD and chronic GVHD. Although this treatment achieved a good control of viral reactivations, such as cytomegalovirus and adenovirus, we observed an unexpectedly high rate of limbic encephalitis due to human herpesvirus-6 (HHV-6; 8 cases). Characteristically, the infection appeared early in almost all patients, just after the engraftment. Although no patient died from encephalitis, 1 patient showed neuropsychological sequelae, and another experienced secondary graft failure just after the HHV-6 reactivation.
Collapse
|
176
|
De Simone M, Rossetti G, Pagani M. Single Cell T Cell Receptor Sequencing: Techniques and Future Challenges. Front Immunol 2018; 9:1638. [PMID: 30072991 PMCID: PMC6058020 DOI: 10.3389/fimmu.2018.01638] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/03/2018] [Indexed: 12/19/2022] Open
Abstract
The peculiarity of T cell is their ability to recognize an infinite range of self and foreign antigens. This ability is achieved during thymic development through a complex molecular mechanism based on somatic recombination that leads to the expression of a very heterogeneous population of surface antigen receptors, the T Cell Receptors (TCRs). TCRs are cell specific and represent a sort of “molecular tag” of T cells and have been widely studied to monitor the dynamics of T cells in terms of clonality and diversity in several contexts including lymphoid malignancies, infectious diseases, autoimmune diseases, and tumor immunology. In this review, we provide an overview of the strategies used to investigate the TCR repertoire from the pioneering techniques based on the V segments identification to the revolution introduced by Next-Generation Sequencing that allows for high-throughput sequencing of alpha and beta chains. Single cell based approaches brought the analysis to a higher level of complexity and now provide the opportunity to sequence paired alpha and beta chains. We also discuss novel approaches that through the integration of TCR tracking and mRNA single cell sequencing offer a valuable tool to associate antigen specificity to transcriptional dynamics and to understand the molecular mechanisms of T cell plasticity.
Collapse
Affiliation(s)
- Marco De Simone
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Grazisa Rossetti
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Massimiliano Pagani
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
177
|
Brucella Peptide Cross-Reactive Major Histocompatibility Complex Class I Presentation Activates SIINFEKL-Specific T Cell Receptor-Expressing T Cells. Infect Immun 2018; 86:IAI.00281-18. [PMID: 29735518 PMCID: PMC6013681 DOI: 10.1128/iai.00281-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 04/25/2018] [Indexed: 12/23/2022] Open
Abstract
Brucella spp. are intracellular pathogenic bacteria remarkable in their ability to escape immune surveillance and therefore inflict a state of chronic disease within the host. To enable further immune response studies, Brucella was engineered to express the well-characterized chicken ovalbumin (OVA). Surprisingly, we found that CD8 T cells bearing T cell receptors (TCR) nominally specific for the OVA peptide SIINFEKL (OT-1) reacted to parental Brucella-infected targets as well as OVA-expressing Brucella variants in cytotoxicity assays. Furthermore, splenocytes from Brucella-immunized mice produced gamma interferon (IFN-γ) and exhibited cytotoxicity in response to SIINFEKL-pulsed target cells.To determine if the SIINFEKL-reactive OT-1 TCR could be cross-reacting to Brucella peptides, we searched the Brucella proteome using an algorithm to generate a list of near-neighbor nonamer peptides that would bind to H2Kb. Selecting five Brucella peptide candidates, along with controls, we verified that several of these peptides mimicked SIINFEKL, resulting in T cell activation through the “SIINFEKL-specific” TCR. Activation was dependent on peptide concentration as well as sequence. Our results underscore the complexity and ubiquity of cross-reactivity in T cell recognition. This cross-reactivity may enable microbes such as Brucella to escape immune surveillance by presenting peptides similar to those of the host and may also lead to the activation of autoreactive T cells.
Collapse
|
178
|
Sun G, Qiu L, Cheng Z, Pan W, Qiu J, Zou C, Xie N, Liu S, Zhu P, Zeng J, Dai Y. Association of the characteristics of B- and T-cell repertoires with papillary thyroid carcinoma. Oncol Lett 2018; 16:1584-1592. [PMID: 30008841 PMCID: PMC6036450 DOI: 10.3892/ol.2018.8800] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 11/16/2017] [Indexed: 11/16/2022] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer. Complementarity-determining region 3 (CDR3) of B-cell receptors (BCRs) and T-cell receptors are the major site of antigen recognition, which determines a unique clone type, and are considered to be the representative of the disease. In the present study, high-throughput sequencing was used to analyze the association of characteristics of the BCR immunoglobulin heavy chain (IGH) and the T-cell receptor β chain (TRB) CDR3 genes in PTC and corresponding pericarcinous tissues from patients. A difference of CDR3 length distributions of total IGH CDR3 sequences between the two groups was revealed. IGHV3-11/IGHJ6, TRBV2/TRBJ1-2 and TRBV2/TRBJ1-1 may be biomarkers for the development of PTC. Furthermore, it was revealed that the extent of the common clonotype expressions at the amino acid level was slightly higher compared with the nucleotide level. The Shannon entropy demonstrated a diversity reduction in PTC compared with the pericarcinous group, and the highly expended clone (HEC) expression of PTC was higher compared with that of the corresponding pericarcinous group. Additionally, the highest clone frequency percentage of IGH and TRB was at 0.1–1.0% degree of expansion, as HEC expression was higher in PTC compared with the matched group. There was no shared clone of HECs in the two groups either at the amino acid level or at the nucleotide expression level. The differential expression of CDR3 sequences of PTC have been identified in the present study. Further research is required for assessing the immune repertoire size, diversity, cloning tracking and finding public clones of T-cell and B-cell populations in the development of PTC.
Collapse
Affiliation(s)
- Guoping Sun
- Core Laboratory, Pingshan New District People's Hospital, Shenzhen, Guangdong 518118, P.R. China
| | - Lumei Qiu
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Zhiqiang Cheng
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Weibing Pan
- Core Laboratory, Pingshan New District People's Hospital, Shenzhen, Guangdong 518118, P.R. China
| | - Jingjun Qiu
- Core Laboratory, Pingshan New District People's Hospital, Shenzhen, Guangdong 518118, P.R. China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Ni Xie
- Core Laboratory, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Song Liu
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Peng Zhu
- Core Laboratory, Pingshan New District People's Hospital, Shenzhen, Guangdong 518118, P.R. China
| | - Jun Zeng
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
179
|
Souquette A, Thomas PG. Past Life and Future Effects-How Heterologous Infections Alter Immunity to Influenza Viruses. Front Immunol 2018; 9:1071. [PMID: 29872429 PMCID: PMC5972221 DOI: 10.3389/fimmu.2018.01071] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/30/2018] [Indexed: 12/21/2022] Open
Abstract
Influenza virus frequently mutates due to its error-prone polymerase. This feature contributes to influenza virus’s ability to evade pre-existing immunity, leading to annual epidemics and periodic pandemics. T cell memory plays a key protective role in the face of an antigenically distinct influenza virus strain because T cell targets are often derived from conserved internal proteins, whereas humoral immunity targets are often sites of increased mutation rates that are tolerated by the virus. Most studies of influenza T cell memory are conducted in naive, specific pathogen free mice and do not account for repetitive influenza infection throughout a lifetime, sequential acute heterologous infections between influenza infections, or heterologous chronic co-infections. By contrast to these mouse models, humans often experience numerous influenza infections, encounter heterologous acute infections between influenza infections, and are infected with at least one chronic virus. In this review, we discuss recent advances in understanding the effects of heterologous infections on the establishment and maintenance of CD8+ T cell immunological memory. Understanding the various factors that affect immune memory can provide insights into the development of more effective vaccines and increase reproducibility of translational studies between animal models and clinical results.
Collapse
Affiliation(s)
- Aisha Souquette
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
180
|
Intrinsic and extrinsic contributors to defective CD8+ T cell responses with aging. Exp Gerontol 2018; 105:140-145. [DOI: 10.1016/j.exger.2018.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 12/20/2022]
|
181
|
Pusch E, Renz H, Skevaki C. Respiratory virus-induced heterologous immunity: Part of the problem or part of the solution? ALLERGO JOURNAL 2018; 27:28-45. [PMID: 32300267 PMCID: PMC7149200 DOI: 10.1007/s15007-018-1580-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/15/2018] [Indexed: 12/31/2022]
Abstract
Purpose To provide current knowledge on respiratory virus-induced heterologous immunity (HI) with a focus on humoral and cellular cross-reactivity. Adaptive heterologous immune responses have broad implications on infection, autoimmunity, allergy and transplant immunology. A better understanding of the mechanisms involved might ultimately open up possibilities for disease prevention, for example by vaccination. Methods A structured literature search was performed using Medline and PubMed to provide an overview of the current knowledge on respiratory-virus induced adaptive HI. Results In HI the immune response towards one antigen results in an alteration of the immune response towards a second antigen. We provide an overview of respiratory virus-induced HI, including viruses such as respiratory syncytial virus (RSV), rhinovirus (RV), coronavirus (CoV) and influenza virus (IV). We discuss T cell receptor (TCR) and humoral cross-reactivity as mechanisms of HI involving those respiratory viruses. Topics covered include HI between respiratory viruses as well as between respiratory viruses and other pathogens. Newly developed vaccines, which have the potential to provide protection against multiple virus strains are also discussed. Furthermore, respiratory viruses have been implicated in the development of autoimmune diseases, such as narcolepsy, Guillain-Barré syndrome, type 1 diabetes or myocarditis. Finally, we discuss the role of respiratory viruses in asthma and the hygiene hypothesis, and review our recent findings on HI between IV and allergens, which leads to protection from experimental asthma. Conclusion Respiratory-virus induced HI may have protective but also detrimental effects on the host. Respiratory viral infections contribute to asthma or autoimmune disease development, but on the other hand, a lack of microbial encounter is associated with an increasing number of allergic as well as autoimmune diseases. Future research might help identify the elements which determine a protective or detrimental outcome in HI-based mechanisms.
Collapse
Affiliation(s)
- Emanuel Pusch
- Institute of Laboratory Medicine, Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Harald Renz
- Institute of Laboratory Medicine, Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine, Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| |
Collapse
|
182
|
Holland CJ, MacLachlan BJ, Bianchi V, Hesketh SJ, Morgan R, Vickery O, Bulek AM, Fuller A, Godkin A, Sewell AK, Rizkallah PJ, Wells S, Cole DK. In Silico and Structural Analyses Demonstrate That Intrinsic Protein Motions Guide T Cell Receptor Complementarity Determining Region Loop Flexibility. Front Immunol 2018; 9:674. [PMID: 29696015 PMCID: PMC5904202 DOI: 10.3389/fimmu.2018.00674] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/19/2018] [Indexed: 11/13/2022] Open
Abstract
T-cell immunity is controlled by T cell receptor (TCR) binding to peptide major histocompatibility complexes (pMHCs). The nature of the interaction between these two proteins has been the subject of many investigations because of its central role in immunity against pathogens, cancer, in autoimmunity, and during organ transplant rejection. Crystal structures comparing unbound and pMHC-bound TCRs have revealed flexibility at the interaction interface, particularly from the perspective of the TCR. However, crystal structures represent only a snapshot of protein conformation that could be influenced through biologically irrelevant crystal lattice contacts and other factors. Here, we solved the structures of three unbound TCRs from multiple crystals. Superposition of identical TCR structures from different crystals revealed some conformation differences of up to 5 Å in individual complementarity determining region (CDR) loops that are similar to those that have previously been attributed to antigen engagement. We then used a combination of rigidity analysis and simulations of protein motion to reveal the theoretical potential of TCR CDR loop flexibility in unbound state. These simulations of protein motion support the notion that crystal structures may only offer an artifactual indication of TCR flexibility, influenced by crystallization conditions and crystal packing that is inconsistent with the theoretical potential of intrinsic TCR motions.
Collapse
Affiliation(s)
- Christopher J Holland
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom.,Immunocore, Abingdon, United Kingdom
| | - Bruce J MacLachlan
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Valentina Bianchi
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom.,Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Sophie J Hesketh
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom.,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Richard Morgan
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Owen Vickery
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Anna M Bulek
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Anna Fuller
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Andrew Godkin
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Andrew K Sewell
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Pierre J Rizkallah
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Stephen Wells
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - David K Cole
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom.,Immunocore, Abingdon, United Kingdom
| |
Collapse
|
183
|
Ritter J, Zimmermann K, Jöhrens K, Mende S, Seegebarth A, Siegmund B, Hennig S, Todorova K, Rosenwald A, Daum S, Hummel M, Schumann M. T-cell repertoires in refractory coeliac disease. Gut 2018; 67:644-653. [PMID: 28188172 PMCID: PMC5868243 DOI: 10.1136/gutjnl-2016-311816] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 12/01/2016] [Accepted: 12/08/2016] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Refractory coeliac disease (RCD) is a potentially hazardous complication of coeliac disease (CD). In contrast to RCD type I, RCD type II is a precursor entity of enteropathy-associated T-cell lymphoma (EATL), which is associated with clonally expanding T-cells that are also found in the sequentially developing EATL. Using high-throughput sequencing (HTS), we aimed to establish the small-intestinal T-cell repertoire (TCR) in CD and RCD to unravel the role of distinct T-cell clonotypes in RCD pathogenesis. DESIGN DNA extracted from duodenal mucosa specimens of controls (n=9), active coeliacs (n=10), coeliacs on a gluten-free diet (n=9), RCD type I (n=8), RCD type II (n=8) and unclassified Marsh I cases (n=3) collected from 2002 to 2013 was examined by TCRβ-complementarity-determining regions 3 (CDR3) multiplex PCR followed by HTS of the amplicons. RESULTS On average, 106 sequence reads per sample were generated consisting of up to 900 individual TCRβ rearrangements. In RCD type II, the most frequent clonotypes (ie, sequence reads with identical CDR3) represent in average 42.6% of all TCRβ rearrangements, which was significantly higher than in controls (6.8%; p<0.01) or RCD type I (6.7%; p<0.01). Repeat endoscopies in individual patients revealed stability of clonotypes for up to several years without clinical symptoms of EATL. Dominant clonotypes identified in individual patients with RCD type II were unique and not related between patients. CD-associated, gliadin-dependent CDR3 motifs were only detectable at low frequencies. CONCLUSIONS TCRβ-HTS analysis unravels the TCR in CD and allows detailed analysis of individual TCRβ rearrangements. Dominant TCRβ sequences identified in patients with RCD type II are unique and not homologous to known gliadin-specific TCR sequences, supporting the assumption that these clonal T-cells expand independent of gluten stimulation.
Collapse
Affiliation(s)
- Julia Ritter
- Institute of Pathology, Charité—University Medicine, Berlin, Germany
| | - Karin Zimmermann
- Institute of Pathology, Charité—University Medicine, Berlin, Germany
| | - Korinna Jöhrens
- Institute of Pathology, Charité—University Medicine, Berlin, Germany
| | - Stefanie Mende
- Institute of Pathology, Charité—University Medicine, Berlin, Germany
| | - Anke Seegebarth
- Institute of Pathology, Charité—University Medicine, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—University Medicine, Berlin, Germany
| | | | - Kremena Todorova
- Center for Tumor Medicine, Charité—University Medicine, Berlin, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, and Comprehensive Cancer Center Mainfranken (CCCMF), Würzburg, Germany
| | - Severin Daum
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—University Medicine, Berlin, Germany
| | - Michael Hummel
- Institute of Pathology, Charité—University Medicine, Berlin, Germany
| | - Michael Schumann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—University Medicine, Berlin, Germany,Berlin Institute of Health, Berlin, Germany,Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
184
|
Clemens EB, van de Sandt C, Wong SS, Wakim LM, Valkenburg SA. Harnessing the Power of T Cells: The Promising Hope for a Universal Influenza Vaccine. Vaccines (Basel) 2018; 6:vaccines6020018. [PMID: 29587436 PMCID: PMC6027237 DOI: 10.3390/vaccines6020018] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Next-generation vaccines that utilize T cells could potentially overcome the limitations of current influenza vaccines that rely on antibodies to provide narrow subtype-specific protection and are prone to antigenic mismatch with circulating strains. Evidence from animal models shows that T cells can provide heterosubtypic protection and are crucial for immune control of influenza virus infections. This has provided hope for the design of a universal vaccine able to prime against diverse influenza virus strains and subtypes. However, multiple hurdles exist for the realisation of a universal T cell vaccine. Overall primary concerns are: extrapolating human clinical studies, seeding durable effective T cell resident memory (Trm), population human leucocyte antigen (HLA) coverage, and the potential for T cell-mediated immune escape. Further comprehensive human clinical data is needed during natural infection to validate the protective role T cells play during infection in the absence of antibodies. Furthermore, fundamental questions still exist regarding the site, longevity and duration, quantity, and phenotype of T cells needed for optimal protection. Standardised experimental methods, and eventually simplified commercial assays, to assess peripheral influenza-specific T cell responses are needed for larger-scale clinical studies of T cells as a correlate of protection against influenza infection. The design and implementation of a T cell-inducing vaccine will require a consensus on the level of protection acceptable in the community, which may not provide sterilizing immunity but could protect the individual from severe disease, reduce the length of infection, and potentially reduce transmission in the community. Therefore, increasing the standard of care potentially offered by T cell vaccines should be considered in the context of pandemic preparedness and zoonotic infections, and in combination with improved antibody vaccine targeting methods. Current pandemic vaccine preparedness measures and ongoing clinical trials under-utilise T cell-inducing vaccines, reflecting the myriad questions that remain about how, when, where, and which T cells are needed to fight influenza virus infection. This review aims to bring together basic fundamentals of T cell biology with human clinical data, which need to be considered for the implementation of a universal vaccine against influenza that harnesses the power of T cells.
Collapse
Affiliation(s)
- E Bridie Clemens
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Carolien van de Sandt
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Sook San Wong
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Sophie A Valkenburg
- HKU Pasteur Research Pole, School of Public Health, University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
185
|
Pusch E, Renz H, Skevaki C. Respiratory virus-induced heterologous immunity: Part of the problem or part of the solution? ACTA ACUST UNITED AC 2018; 27:79-96. [PMID: 32226720 PMCID: PMC7100437 DOI: 10.1007/s40629-018-0056-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/15/2018] [Indexed: 12/13/2022]
Abstract
Purpose To provide current knowledge on respiratory virus-induced heterologous immunity (HI) with a focus on humoral and cellular cross-reactivity. Adaptive heterologous immune responses have broad implications on infection, autoimmunity, allergy and transplant immunology. A better understanding of the mechanisms involved might ultimately open up possibilities for disease prevention, for example by vaccination. Methods A structured literature search was performed using Medline and PubMed to provide an overview of the current knowledge on respiratory-virus induced adaptive HI. Results In HI the immune response towards one antigen results in an alteration of the immune response towards a second antigen. We provide an overview of respiratory virus-induced HI, including viruses such as respiratory syncytial virus (RSV), rhinovirus (RV), coronavirus (CoV) and influenza virus (IV). We discuss T cell receptor (TCR) and humoral cross-reactivity as mechanisms of HI involving those respiratory viruses. Topics covered include HI between respiratory viruses as well as between respiratory viruses and other pathogens. Newly developed vaccines which have the potential to provide protection against multiple virus strains are also discussed. Furthermore, respiratory viruses have been implicated in the development of autoimmune diseases, such as narcolepsy, Guillain–Barré syndrome, type 1 diabetes or myocarditis. Finally, we discuss the role of respiratory viruses in asthma and the hygiene hypothesis, and review our recent findings on HI between IV and allergens, which leads to protection from experimental asthma. Conclusion Respiratory-virus induced HI may have protective but also detrimental effects on the host. Respiratory viral infections contribute to asthma or autoimmune disease development, but on the other hand, a lack of microbial encounter is associated with an increasing number of allergic as well as autoimmune diseases. Future research might help identify the elements which determine a protective or detrimental outcome in HI-based mechanisms.
Collapse
Affiliation(s)
- Emanuel Pusch
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| |
Collapse
|
186
|
Lyons JJ, Milner JD. Primary atopic disorders. J Exp Med 2018; 215:1009-1022. [PMID: 29549114 PMCID: PMC5881472 DOI: 10.1084/jem.20172306] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/21/2018] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
Important insights from monogenic disorders into the immunopathogenesis of allergic diseases and reactions are discussed. Monogenic disorders have provided fundamental insights into human immunity and the pathogenesis of allergic diseases. The pathways identified as critical in the development of atopy range from focal defects in immune cells and epithelial barrier function to global changes in metabolism. A major goal of studying heritable single-gene disorders that lead to severe clinical allergic diseases is to identify fundamental pathways leading to hypersensitivity that can be targeted to provide novel therapeutic strategies for patients with allergic diseases, syndromic and nonsyndromic alike. Here, we review known single-gene disorders leading to severe allergic phenotypes in humans, discuss how the revealed pathways fit within our current understanding of the atopic diathesis, and propose how some pathways might be targeted for therapeutic benefit.
Collapse
Affiliation(s)
- Jonathan J Lyons
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
187
|
Muraille E. Diversity Generator Mechanisms Are Essential Components of Biological Systems: The Two Queen Hypothesis. Front Microbiol 2018; 9:223. [PMID: 29487592 PMCID: PMC5816788 DOI: 10.3389/fmicb.2018.00223] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 01/30/2018] [Indexed: 01/02/2023] Open
Abstract
Diversity is widely known to fuel adaptation and evolutionary processes and increase robustness at the population, species and ecosystem levels. The Neo-Darwinian paradigm proposes that the diversity of biological entities is the consequence of genetic changes arising spontaneously and randomly, without regard for their usefulness. However, a growing body of evidence demonstrates that the evolutionary process has shaped mechanisms, such as horizontal gene transfer mechanisms, meiosis and the adaptive immune system, which has resulted in the regulated generation of diversity among populations. Though their origins are unrelated, these diversity generator (DG) mechanisms share common functional properties. They (i) contribute to the great unpredictability of the composition and/or behavior of biological systems, (ii) favor robustness and collectivism among populations and (iii) operate mainly by manipulating the systems that control the interaction of living beings with their environment. The definition proposed here for DGs is based on these properties and can be used to identify them according to function. Interestingly, prokaryotic DGs appear to be mainly reactive, as they generate diversity in response to environmental stress. They are involved in the widely described Red Queen/arms race/Cairnsian dynamic. The emergence of multicellular organisms harboring K selection traits (longer reproductive life cycle and smaller population size) has led to the acquisition of a new class of DGs that act anticipatively to stress pressures and generate a distinct dynamic called the “White Queen” here. The existence of DGs leads to the view of evolution as a more “intelligent” and Lamarckian-like process. Their repeated selection during evolution could be a neglected example of convergent evolution and suggests that some parts of the evolutionary process are tightly constrained by ecological factors, such as the population size, the generation time and the intensity of selective pressure. The ubiquity of DGs also suggests that regulated auto-generation of diversity is a fundamental property of life.
Collapse
Affiliation(s)
- Eric Muraille
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
188
|
Abstract
HLA associations, T cell receptor (TCR) repertoire bias, and sex bias have independently been shown for many diseases. While some immunological differences between the sexes have been described, they do not fully explain bias in men toward many infections/cancers, and toward women in autoimmunity. Next-generation TCR variable beta chain (TCRBV) immunosequencing of 824 individuals was evaluated in a multiparametric analysis including HLA-A -B/MHC class I background, TCRBV usage, sex, age, ethnicity, and TCRBV selection/expansion dynamics. We found that HLA-associated shaping of TCRBV usage differed between the sexes. Furthermore, certain TCRBVs were selected and expanded in unison. Correlations between these TCRBV relationships and biochemical similarities in HLA-binding positions were different in CD8 T cells of patients with autoimmune diseases (multiple sclerosis and rheumatoid arthritis) compared with healthy controls. Within patients, men showed higher TCRBV relationship Spearman's rhos in relation to HLA-binding position similarities compared with women. In line with this, CD8 T cells of men with autoimmune diseases also showed higher degrees of TCRBV perturbation compared with women. Concerted selection and expansion of CD8 T cells in patients with autoimmune diseases, but especially in men, appears to be less dependent on high HLA-binding similarity than in CD4 T cells. These findings are consistent with studies attributing autoimmunity to processes of epitope spreading and expansion of low-avidity T cell clones and may have further implications for the interpretation of pathogenic mechanisms of infectious and autoimmune diseases with known HLA associations. Reanalysis of some HLA association studies, separating the data by sex, could be informative.
Collapse
|
189
|
Spear TT, Wang Y, Smith TW, Simms PE, Garrett-Mayer E, Hellman LM, Baker BM, Nishimura MI. Altered Peptide Ligands Impact the Diversity of Polyfunctional Phenotypes in T Cell Receptor Gene-Modified T Cells. Mol Ther 2018; 26:996-1007. [PMID: 29503203 DOI: 10.1016/j.ymthe.2018.01.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/23/2022] Open
Abstract
The use of T cell receptor (TCR) gene-modified T cells in adoptive cell transfer has had promising clinical success, but often, simple preclinical evaluation does not necessarily accurately predict treatment efficacy or safety. Preclinical studies generally evaluate one or a limited number of type 1 cytokines to assess antigen recognition. However, recent studies have implicated other "typed" T cells in effective anti-tumor/viral immunity, and limited functional evaluations may underestimate cross-reactivity. In this study, we use an altered peptide ligand (APL) model and multi-dimensional flow cytometry to evaluate polyfunctionality of TCR gene-modified T cells. Evaluating six cytokines and the lytic marker CD107a on a per cell basis revealed remarkably diverse polyfunctional phenotypes within a single T cell culture and among peripheral blood lymphocyte (PBL) donors. This polyfunctional assessment identified unexpected phenotypes, including cells producing both type 1 and type 2 cytokines, and highlighted interferon γneg (IFNγneg) antigen-reactive populations overlooked in our previous studies. Additionally, APLs skewed functional phenotypes to be less polyfunctional, which was not necessarily related to changes in TCR-peptide-major histocompatibility complex (pMHC) affinity. A better understanding of gene-modified T cell functional diversity may help identify optimal therapeutic phenotypes, predict clinical responses, anticipate off-target recognition, and improve the design and delivery of TCR gene-modified T cells.
Collapse
Affiliation(s)
- Timothy T Spear
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153, USA.
| | - Yuan Wang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Thomas W Smith
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153, USA
| | - Patricia E Simms
- Flow Cytometry Core Facility, Office of Research Services, Loyola University Chicago, Maywood, IL, 60153 USA
| | - Elizabeth Garrett-Mayer
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29415, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29415, USA
| | - Lance M Hellman
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Brian M Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Michael I Nishimura
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
190
|
Fontaine M, Vogel I, Van Eycke YR, Galuppo A, Ajouaou Y, Decaestecker C, Kassiotis G, Moser M, Leo O. Regulatory T cells constrain the TCR repertoire of antigen-stimulated conventional CD4 T cells. EMBO J 2018; 37:398-412. [PMID: 29263148 PMCID: PMC5793804 DOI: 10.15252/embj.201796881] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 11/17/2017] [Accepted: 11/27/2017] [Indexed: 01/22/2023] Open
Abstract
To analyze the potential role of Tregs in controlling the TCR repertoire breadth to a non-self-antigen, a TCRβ transgenic mouse model (EF4.1) expressing a limited, yet polyclonal naïve T-cell repertoire was used. The response of EF4.1 mice to an I-Ab-associated epitope of the F-MuLV envelope protein is dominated by clones expressing a Vα2 gene segment, thus allowing a comprehensive analysis of the TCRα repertoire in a relatively large cohort of mice. Control and Treg-depleted EF4.1 mice were immunized, and the extent of the Vα2-bearing, antigen-specific TCR repertoire was characterized by high-throughput sequencing and spectratyping analysis. In addition to increased clonal expansion and acquisition of effector functions, Treg depletion led to the expression of a more diverse TCR repertoire comprising several private clonotypes rarely observed in control mice or in the pre-immune repertoire. Injection of anti-CD86 antibodies in vivo led to a strong reduction in TCR diversity, suggesting that Tregs may influence TCR repertoire diversity by modulating costimulatory molecule availability. Collectively, these studies illustrate an additional mechanism whereby Tregs control the immune response to non-self-antigens.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- B7-2 Antigen/immunology
- Cells, Cultured
- Friend murine leukemia virus/immunology
- Lymphocyte Depletion
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes, Regulatory/immunology
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Martina Fontaine
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Isabel Vogel
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Yves-Rémi Van Eycke
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratories of Image, Signal processing & Acoustics Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Adrien Galuppo
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Yousra Ajouaou
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Christine Decaestecker
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratories of Image, Signal processing & Acoustics Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, London, UK
- Department of Medicine Faculty of Medicine, Imperial College London London, UK
| | - Muriel Moser
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Oberdan Leo
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| |
Collapse
|
191
|
Giudice V, Feng X, Lin Z, Hu W, Zhang F, Qiao W, Ibanez MDPF, Rios O, Young NS. Deep sequencing and flow cytometric characterization of expanded effector memory CD8 +CD57 + T cells frequently reveals T-cell receptor Vβ oligoclonality and CDR3 homology in acquired aplastic anemia. Haematologica 2018; 103:759-769. [PMID: 29419434 PMCID: PMC5927970 DOI: 10.3324/haematol.2017.176701] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/30/2017] [Indexed: 11/24/2022] Open
Abstract
Oligoclonal expansion of CD8+ CD28− lymphocytes has been considered indirect evidence for a pathogenic immune response in acquired aplastic anemia. A subset of CD8+ CD28− cells with CD57 expression, termed effector memory cells, is expanded in several immune-mediated diseases and may have a role in immune surveillance. We hypothesized that effector memory CD8+CD28−CD57+ cells may drive aberrant oligoclonal expansion in aplastic anemia. We found CD8+CD57+ cells frequently expanded in the blood of aplastic anemia patients, with oligoclonal characteristics by flow cytometric Vβ usage analysis: skewing in 1–5 Vβ families and frequencies of immunodominant clones ranging from 1.98% to 66.5%. Oligoclonal characteristics were also observed in total CD8+ cells from aplastic anemia patients with CD8+CD57+ cell expansion by T-cell receptor deep sequencing, as well as the presence of 1–3 immunodominant clones. Oligoclonality was confirmed by T-cell receptor repertoire deep sequencing of enriched CD8+CD57+ cells, which also showed decreased diversity compared to total CD4+ and CD8+ cell pools. From analysis of complementarity-determining region 3 sequences in the CD8+ cell pool, a total of 29 sequences were shared between patients and controls, but these sequences were highly expressed in aplastic anemia subjects and also present in their immunodominant clones. In summary, expansion of effector memory CD8+ T cells is frequent in aplastic anemia and mirrors Vβ oligoclonal expansion. Flow cytometric Vβ usage analysis combined with deep sequencing technologies allows high resolution characterization of the T-cell receptor repertoire, and might represent a useful tool in the diagnosis and periodic evaluation of aplastic anemia patients. (Registered at clinicaltrials.gov identifiers: 00001620, 01623167, 00001397, 00071045, 00081523, 00961064)
Collapse
Affiliation(s)
- Valentina Giudice
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Zenghua Lin
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA.,Department of Hematology, Affiliated Hospital of Nantong University, Jiangsu, China
| | - Wei Hu
- BGI Genomics, BGI-Shenzhen, China
| | | | | | | | - Olga Rios
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| |
Collapse
|
192
|
Nie J, Zhang Y, Li X, Chen M, Liu C, Han W. DNA demethylating agent decitabine broadens the peripheral T cell receptor repertoire. Oncotarget 2018; 7:37882-37892. [PMID: 27191266 PMCID: PMC5122357 DOI: 10.18632/oncotarget.9352] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/28/2016] [Indexed: 01/08/2023] Open
Abstract
Purpose Decitabine, a promising epi-immunotherapeutic agent has shown clinical responses in solid tumor patients, while the anti-tumor mechanisms were unclear. We aimed to investigate the immunomodulatory effect of decitabine in peripheral T cells. Experimental design We applied next-generation sequencing to investigate the complementarity-determining region 3 (CDR3) of the TCRβ gene, the diversity of which acts as the prerequisite for the host immune system to recognize the universal foreign antigens. We collected the peripheral blood mononuclear cells (PBMCs) from 4 patients, at baseline and after 2 cycles of low-dose decitabine therapy. Results An increase of the unique productive sequences of the CDR3 of TCRβ was observed in all of the 4 patients after decitabine treatment, which was characterized by a lower abundance of expanded clones and increased TCR diversity compared with before decitabine treatment. Further analysis showed a tendency for CD4 T cells with an increased CD4/CD8 ratio in response to decitabine therapy. In addition, the genome-wide expression alterations confirmed the effects of decitabine on immune reconstitution, and the increase of TCR excision circles (TRECs) was validated. Conclusions The low-dose DNMT inhibitor decitabine broadens the peripheral T cell repertoire, providing a novel role for the epigenetic modifying agent in anti-tumor immune enhancement.
Collapse
Affiliation(s)
- Jing Nie
- Department of Immunology, Institute of Basic Medical Science, PLA General Hospital, Beijing, 100853, China
| | - Yan Zhang
- Department of Biological Therapy, PLA General Hospital, Beijing, 100853, China
| | - Xiang Li
- Department of Immunology, Institute of Basic Medical Science, PLA General Hospital, Beijing, 100853, China
| | - Meixia Chen
- Department of Biological Therapy, PLA General Hospital, Beijing, 100853, China
| | - Chuanjie Liu
- Department of Immunology, Institute of Basic Medical Science, PLA General Hospital, Beijing, 100853, China
| | - Weidong Han
- Department of Immunology, Institute of Basic Medical Science, PLA General Hospital, Beijing, 100853, China.,Department of Biological Therapy, PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
193
|
Chew WL. Immunity to CRISPR Cas9 and Cas12a therapeutics. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10. [PMID: 29083112 DOI: 10.1002/wsbm.1408] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 12/27/2022]
Abstract
Genome-editing therapeutics are poised to treat human diseases. As we enter clinical trials with the most promising CRISPR-Cas9 and CRISPR-Cas12a (Cpf1) modalities, the risks associated with administering these foreign biomolecules into human patients become increasingly salient. Preclinical discovery with CRISPR-Cas9 and CRISPR-Cas12a systems and foundational gene therapy studies indicate that the host immune system can mount undesired responses against the administered proteins and nucleic acids, the gene-edited cells, and the host itself. These host defenses include inflammation via activation of innate immunity, antibody induction in humoral immunity, and cell death by T-cell-mediated cytotoxicity. If left unchecked, these immunological reactions can curtail therapeutic benefits and potentially lead to mortality. Ways to assay and reduce the immunogenicity of Cas9 and Cas12a proteins are therefore critical for ensuring patient safety and treatment efficacy, and for bringing us closer to realizing the vision of permanent genetic cures. WIREs Syst Biol Med 2018, 10:e1408. doi: 10.1002/wsbm.1408 This article is categorized under: Laboratory Methods and Technologies > Genetic/Genomic Methods Translational, Genomic, and Systems Medicine > Translational Medicine Translational, Genomic, and Systems Medicine > Therapeutic Methods.
Collapse
Affiliation(s)
- Wei Leong Chew
- Synthetic Biology, Genome Institute of Singapore, Singapore, Singapore
| |
Collapse
|
194
|
Artificial Methods for T Cell Activation: Critical Tools in T Cell Biology and T Cell Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:207-219. [PMID: 30471035 DOI: 10.1007/978-981-13-0445-3_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Antigen-specific immunity conferred by T lymphocytes is a result of complex molecular interactions at the immunological synapse. A variety of biomimetic approaches have been devised to artificially induce T cell activation either to study the T cell biology or to expand and prime the therapeutic T cell populations. Here we first briefly review the molecular and cellular, structural and phenotypical bases that are involved in T cell activation. The artificial methods for T cell activation are then discussed in two grand categories, the soluble (3D) and the surface-anchored (2D) platforms with their design parameters. With the growing number of successful adoptive T cell therapies, the spurring demands for effective and safe T cell expansion as well as precise control over resulting T cell functions and phenotypes warrant the extensions of engineering parameters in the development of novel methodologies for T cell activation.
Collapse
|
195
|
Park H, Kim H, Doh J. Multifunctional Microwell Arrays for Single Cell Level Functional Analysis of Lymphocytes. Bioconjug Chem 2017; 29:672-679. [DOI: 10.1021/acs.bioconjchem.7b00620] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
196
|
Yohannes DA, Freitag TL, de Kauwe A, Kaukinen K, Kurppa K, Wacklin P, Mäki M, Arstila TP, Anderson RP, Greco D, Saavalainen P. Deep sequencing of blood and gut T-cell receptor β-chains reveals gluten-induced immune signatures in celiac disease. Sci Rep 2017; 7:17977. [PMID: 29269859 PMCID: PMC5740085 DOI: 10.1038/s41598-017-18137-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/06/2017] [Indexed: 12/16/2022] Open
Abstract
Celiac disease (CD) patients mount an abnormal immune response to gluten. T-cell receptor (TCR) repertoires directed to some immunodominant gluten peptides have previously been described, but the global immune response to in vivo gluten exposure in CD has not been systematically investigated yet. Here, we characterized signatures associated with gluten directed immune activity and identified gluten-induced T-cell clonotypes from total blood and gut TCR repertoires in an unbiased manner using immunosequencing. CD patient total TCR repertoires showed increased overlap and substantially altered TRBV-gene usage in both blood and gut samples, and increased diversity in the gut during gluten exposure. Using differential abundance analysis, we identified gluten-induced clonotypes in each patient that were composed of a large private and an important public component. Hierarchical clustering of public clonotypes associated with dietary gluten exposure identified subsets of highly similar clonotypes, the most proliferative of which showing significant enrichment for the motif ASS[LF]R[SW][TD][DT][TE][QA][YF] in PBMC repertoires. These results show that CD-associated clonotypes can be identified and that common gluten associated immune response features can be characterized in vivo from total repertoires, with potential use in disease stratification and monitoring.
Collapse
Affiliation(s)
- Dawit A Yohannes
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Tobias L Freitag
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland.,Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Andrea de Kauwe
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Katri Kaukinen
- Department of Internal Medicine, Tampere University Hospital and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Kalle Kurppa
- Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Pirjo Wacklin
- Finnish Red Cross Blood Transfusion Service, Helsinki, Finland
| | - Markku Mäki
- Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - T Petteri Arstila
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland.,Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Robert P Anderson
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,ImmusanT, Inc., Cambridge, MA, USA
| | - Dario Greco
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Päivi Saavalainen
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland. .,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
197
|
Exploring viral reservoir: The combining approach of cell sorting and droplet digital PCR. Methods 2017; 134-135:98-105. [PMID: 29197654 DOI: 10.1016/j.ymeth.2017.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/24/2017] [Accepted: 11/27/2017] [Indexed: 12/12/2022] Open
Abstract
Combined antiretroviral therapy (cART) blocks different steps of HIV replication and maintains plasma viral RNA at undetectable levels. The virus can remain in long-living cells and create a reservoir where HIV can restart replicating after cART discontinuation. A persistent viral production triggers and maintains a persistent immune activation, which is a well-known feature of chronic HIV infection, and contributes either to precocious aging, or to the increased incidence of morbidity and mortality of HIV positive patients. The new frontier of the treatment of HIV infection is nowadays eradication of the virus from all host cells and tissues. For this reason, it is crucial to have a clear and precise idea of where the virus hides, and which are the cells that keep it silent. Important efforts have been made to improve the detection of viral reservoirs, and new techniques are now giving the opportunity to characterize viral reservoirs. Among these techniques, a strategic approach based upon cell sorting and droplet digital PCR (ddPCR) is opening new horizons and opportunities of research. This review provides an overview of the methods that combine cell sorting and ddPCR for the quantification of HIV DNA in different cell types, and for the detection of its maintenance.
Collapse
|
198
|
Marrack P, Krovi SH, Silberman D, White J, Kushnir E, Nakayama M, Crooks J, Danhorn T, Leach S, Anselment R, Scott-Browne J, Gapin L, Kappler J. The somatically generated portion of T cell receptor CDR3α contributes to the MHC allele specificity of the T cell receptor. eLife 2017; 6:30918. [PMID: 29148973 PMCID: PMC5701794 DOI: 10.7554/elife.30918] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/16/2017] [Indexed: 01/24/2023] Open
Abstract
Mature T cells bearing αβ T cell receptors react with foreign antigens bound to alleles of major histocompatibility complex proteins (MHC) that they were exposed to during their development in the thymus, a phenomenon known as positive selection. The structural basis for positive selection has long been debated. Here, using mice expressing one of two different T cell receptor β chains and various MHC alleles, we show that positive selection-induced MHC bias of T cell receptors is affected both by the germline encoded elements of the T cell receptor α and β chain and, surprisingly, dramatically affected by the non germ line encoded portions of CDR3 of the T cell receptor α chain. Thus, in addition to determining specificity for antigen, the non germline encoded elements of T cell receptors may help the proteins cope with the extremely polymorphic nature of major histocompatibility complex products within the species.
Collapse
Affiliation(s)
- Philippa Marrack
- Howard Hughes Medical Institute, Denver, United States.,Department of Biomedical Research, National Jewish Health, Denver, United States.,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, United States
| | - Sai Harsha Krovi
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, United States
| | - Daniel Silberman
- Department of Biomedical Research, National Jewish Health, Denver, United States.,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, United States
| | - Janice White
- Department of Biomedical Research, National Jewish Health, Denver, United States
| | - Eleanor Kushnir
- Department of Biomedical Research, National Jewish Health, Denver, United States
| | - Maki Nakayama
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, United States.,Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, United States
| | - James Crooks
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, United States
| | - Thomas Danhorn
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, United States
| | - Sonia Leach
- Department of Biomedical Research, National Jewish Health, Denver, United States.,Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, United States
| | - Randy Anselment
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, United States
| | | | - Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, United States
| | - John Kappler
- Howard Hughes Medical Institute, Denver, United States.,Department of Biomedical Research, National Jewish Health, Denver, United States.,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, United States
| |
Collapse
|
199
|
Xiao X, Cai J. Mucosal-Associated Invariant T Cells: New Insights into Antigen Recognition and Activation. Front Immunol 2017; 8:1540. [PMID: 29176983 PMCID: PMC5686390 DOI: 10.3389/fimmu.2017.01540] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 10/30/2017] [Indexed: 12/20/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells, a novel subpopulation of innate-like T cells that express an invariant T cell receptor (TCR)α chain and a diverse TCRβ chain, can recognize a distinct set of small molecules, vitamin B metabolites, derived from some bacteria, fungi but not viruses, in the context of an evolutionarily conserved major histocompatibility complex-related molecule 1 (MR1). This implies that MAIT cells may play unique and important roles in host immunity. Although viral antigens are not recognized by this limited TCR repertoire, MAIT cells are known to be activated in a TCR-independent mechanism during some viral infections, such as hepatitis C virus and influenza virus. In this article, we will review recent works in MAIT cell antigen recognition, activation and the role MAIT cells may play in the process of bacterial and viral infections and pathogenesis of non-infectious diseases.
Collapse
Affiliation(s)
- Xingxing Xiao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianping Cai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
200
|
van den Heuvel H, Heutinck KM, van der Meer-Prins EMW, Franke-van Dijk MEI, van Miert PPMC, Zhang X, Ten Berge IJM, Claas FHJ. The avidity of cross-reactive virus-specific T cells for their viral and allogeneic epitopes is variable and depends on epitope expression. Hum Immunol 2017; 79:39-50. [PMID: 29100943 DOI: 10.1016/j.humimm.2017.10.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/30/2017] [Accepted: 10/30/2017] [Indexed: 10/18/2022]
Abstract
Virus-specific T cells can recognize allogeneic HLA (allo-HLA) through cross-reactivity of their T-cell receptor (TCR). In a transplantation setting, such allo-HLA cross-reactivity may contribute to harmful immune responses towards the allograft, provided that the cross-reactive T cells get sufficiently activated upon recognition of the allo-HLA. An important determinant of T-cell activation is TCR avidity, which to date, has remained largely unexplored for allo-HLA-cross-reactive virus-specific T cells. For this purpose, cold target inhibition assays were performed using allo-HLA-cross-reactive virus-specific memory CD8+ T-cell clones as responders, and syngeneic cells loaded with viral peptide and allogeneic cells as hot (radioactively-labeled) and cold (non-radioactively-labeled) targets. CD8 dependency of the T-cell responses was assessed using interferon γ (IFNγ) enzyme-linked immunosorbent assay (ELISA) in the presence and absence of CD8-blocking antibodies. At high viral-peptide loading concentrations, T-cell clones consistently demonstrated lower avidity for allogeneic versus viral epitopes, but at suboptimal concentrations the opposite was observed. In line, anti-viral reactivity was CD8 independent at high, but not at suboptimal viral-peptide-loading concentrations. The avidity of allo-HLA-cross-reactive virus-specific memory CD8+ T cells is therefore highly dependent on epitope expression, and as a consequence, can be both higher and lower for allogeneic versus viral targets under different (patho)physiological conditions.
Collapse
Affiliation(s)
- Heleen van den Heuvel
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | - Kirstin M Heutinck
- Department of Experimental Immunology, Academic Medical Centre, Amsterdam, The Netherlands; Renal Transplant Unit, Department of Internal Medicine, Division of Internal Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| | - Ellen M W van der Meer-Prins
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Marry E I Franke-van Dijk
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Paula P M C van Miert
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Xiaoqian Zhang
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Ineke J M Ten Berge
- Renal Transplant Unit, Department of Internal Medicine, Division of Internal Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| | - Frans H J Claas
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|