1
|
Islam MZ, Zimmerman S, Lindahl A, Weidanz J, Ordovas-Montanes J, Kostic A, Luber J, Robben M. Single-cell RNA-seq reveals disease-specific CD8+ T cell clonal expansion and a high frequency of transcriptionally distinct double-negative T cells in diabetic NOD mice. PLoS One 2025; 20:e0317987. [PMID: 40106422 PMCID: PMC11922263 DOI: 10.1371/journal.pone.0317987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/08/2025] [Indexed: 03/22/2025] Open
Abstract
T cells primarily drive the autoimmune destruction of pancreatic beta cells in Type 1 diabetes (T1D). However, the profound yet uncharacterized diversity of the T cell populations in vivo has hindered obtaining a clear picture of the T cell changes that occur longitudinally during T1D onset. This study aimed to identify T cell clonal expansion and distinct transcriptomic signatures associated with T1D progression in Non-Obese Diabetic (NOD) mice. Here we profiled the transcriptome and T cell receptor (TCR) repertoire of T cells at single-cell resolution from longitudinally collected peripheral blood and pancreatic islets of NOD mice using single-cell RNA sequencing technology. We detected disease dependent development of infiltrating CD8 + T cells with altered cytotoxic and inflammatory effector states. In addition, we discovered a high frequency of transcriptionally distinct double negative (DN) T cells that fluctuate throughout T1D pathogenesis. This study identifies potential disease relevant TCR sequences and potential disease biomarkers that can be further characterized through future research.
Collapse
Affiliation(s)
- Md Zohorul Islam
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong, Victoria, Australia
| | - Sam Zimmerman
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alexis Lindahl
- Department of Animal Science, University of Illinois, Urbana-Champaign, Illinois, United States of America
| | - Jon Weidanz
- Department of Kinesiology, The University of Texas at Arlington, Texas, United States of America
- Department of Bioengineering, The University of Texas at Arlington, Texas, United States of America
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts, United States of America
| | - Aleksandar Kostic
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jacob Luber
- Department of Computer Science and Engineering, The University of Texas at Arlington, United States of America
| | - Michael Robben
- Department of Animal Science, University of Illinois, Urbana-Champaign, Illinois, United States of America
- Department of Computer Science and Engineering, The University of Texas at Arlington, United States of America
| |
Collapse
|
2
|
Wang Z, Yang Y, Chen G, Chen G, Luo J, Li Y, Shi J, Chen H. Unravelling T-cell dynamics and immune responses in initial and recurrent uveitis. Scand J Immunol 2024; 100:e13417. [PMID: 39511764 DOI: 10.1111/sji.13417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/22/2024] [Accepted: 10/01/2024] [Indexed: 11/15/2024]
Abstract
This study aimed to identify novel serological targets and investigate immune responses in patients with non-infectious uveitis, focusing on differences between initial onset and recurrent episodes. Differential gene expression analysis, immunocyte typing and T-cell receptor (TCR) gene analysis were conducted on RNA-sequenced peripheral blood samples from healthy individuals (n = 6) and non-infectious uveitis patients (n = 12), divided into 6 patients each at initial onset and recurrent stages. Peripheral blood T-cell types were analysed using flow cytometry. Bioinformatics methods included tools for RNA sequencing data processing, CIBERSORT for immune cell type prediction and specialized software for TCR repertoire analysis. Findings indicated that individuals with recurrent uveitis demonstrated a stronger adaptive immune response and a more pronounced immune imbalance compared to those with initial onset. Memory T cells were predominant in recurrent episodes, suggesting their potential role as biomarkers for disease progression. Significant differences in TCR diversity and V(D)J gene usage were observed between the various uveitis groups and healthy controls. Importantly, 38 uveitis-specific TCR sequences showed substantial expansion in the uveitis patients compared to controls. An elevated expansion of these specific TCR sequences was associated with an increased risk of uveitis development. The study highlights the critical role of adaptive immune responses and specific immune cell types in the pathogenesis of recurrent uveitis. Identification of the uveitis-specific TCR repertoire set could provide deeper insights into the disease and facilitate the development of targeted therapies for uveitis patients.
Collapse
Affiliation(s)
- Zhiruo Wang
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yuanyuan Yang
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Guochun Chen
- Clinical Immunology Research Center of Central South University, Changsha, Hunan, China
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Gong Chen
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jing Luo
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yunping Li
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jingming Shi
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Huihui Chen
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
- Clinical Immunology Research Center of Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Jia X, Zhai TY, Wang B, Zhang JA, Song RH. High-throughput T cell receptor sequencing reveals differential immune repertoires in autoimmune thyroid diseases. Mol Cell Endocrinol 2022; 550:111644. [PMID: 35429598 DOI: 10.1016/j.mce.2022.111644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 01/16/2023]
Abstract
BACKGROUND Autoimmune thyroid diseases (AITDs) are chronic autoimmune diseases specific to thyroid and mainly include Graves' disease (GD) and Hashimoto' thyroiditis (HT). The adaptive immunoreactivity of CD4+ T cells plays a crucial role in the pathogenesis of AITDs, but very little has been known about its changes in disease status. METHODS We collected peripheral CD4+ T cells from 12 GD patients, including 6 newly diagnosed GD (NGD) and 6 refractory GD (RGD) patients, 6 HT patients and 6 healthy controls, and examined the gene expression profiles and colon types of T cells receptor (TCR) β chain complementarity determining region 3 (CDR3) using high-throughput sequencing. RESULTS The TCR repertoire were significantly expanded in AITDs groups, and some TCR genes were expressed more preferentially in AITDs group than in the healthy control group, including TRBV15 (P = 0.001), TRBV4-2 (P = 0.003), TRBV9 (P = 0.007), TRBV3-2 (P = 0.012), TRBV7-8 (P = 0.015), TRBV25-1 (P = 0.019), TRBV12-4 (P = 0.019) and TRBV27 (P = 0.02) in GD patients as well as TRBV29-1 (P = 0.004), TRBV12-4 (P = 0.004), TRBV6-5 (P = 0.011), TRBV7-2 (P = 0.012), TRBV27 (P = 0.012), TRBV9 (P = 0.031) and TRBV4-2 (P = 0.032) in HT patients. Moreover, subgroup analysis showed that the difference in the TCR spectrum between the normal group and NGD was not obvious, but a large number of differential genes appeared in the RGD group. CONCLUSION TCR spectrum has changed in patients with AITDs with expanded repertoire and many upregulated TRBV genes. Moreover, this difference is not apparent in GD patients at the initial stage, but as the disease progresses, the differences in TCR profiles became more pronounced.
Collapse
Affiliation(s)
- Xi Jia
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China
| | - Tian-Yu Zhai
- Department of Endocrinology, Zhongshan Hospital of Fudan University, China
| | - Bing Wang
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China
| | - Jin-An Zhang
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China.
| | - Rong-Hua Song
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China.
| |
Collapse
|
4
|
Song R, Jia X, Zhao J, Du P, Zhang JA. T cell receptor revision and immune repertoire changes in autoimmune diseases. Int Rev Immunol 2021; 41:517-533. [PMID: 34243694 DOI: 10.1080/08830185.2021.1929954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Autoimmune disease (AID) is a condition in which the immune system breaks down and starts to attack the body. Some common AIDs include systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes mellitus and so forth. The changes in T-cell receptor (TCR) repertoire have been found in several autoimmune diseases, and may be responsible for the breakdown of peripheral immune tolerance. In this review, we discussed the processes of TCR revision in peripheral immune environment, the changes in TCR repertoire that occurred in various AIDs, and the specifically expanded T cell clones. We hope our discussion can provide insights for the future studies, helping with the discovery of disease biomarkers and expanding the strategies of immune-targeted therapy. HighlightsRestricted TCR repertoire and biased TCR-usage are found in a variety of AIDs.TCR repertoire shows tissue specificity in a variety of AID diseases.The relationship between TCR repertoire diversity and disease activity is still controversial in AIDs.Dominant TCR clonotypes may help to discover new disease biomarkers and expand the strategies of immune-targeted therapy.
Collapse
Affiliation(s)
- Ronghua Song
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xi Jia
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jing Zhao
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Peng Du
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jin-An Zhang
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
5
|
Mhanna V, Fourcade G, Barennes P, Quiniou V, Pham HP, Ritvo PG, Brimaud F, Gouritin B, Churlaud G, Six A, Mariotti-Ferrandiz E, Klatzmann D. Impaired Activated/Memory Regulatory T Cell Clonal Expansion Instigates Diabetes in NOD Mice. Diabetes 2021; 70:976-985. [PMID: 33479057 DOI: 10.2337/db20-0896] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/19/2021] [Indexed: 11/13/2022]
Abstract
Regulatory T cell (Treg) insufficiency licenses the destruction of insulin-producing pancreatic β-cells by autoreactive effector T cells (Teffs), causing spontaneous autoimmune diabetes in NOD mice. We investigated the contribution to diabetes of the T-cell receptor (TCR) repertoires of naive regulatory T cells (nTregs), activated/memory Tregs (amTregs), and CD4+ Teffs from prediabetic NOD mice and normal C57BL/6 (B6) mice. NOD mice amTreg and Teff repertoire diversity was unexpectedly higher than that of B6 mice. This was due to the presence of highly expanded clonotypes in B6 amTregs and Teffs that were largely lost in their NOD counterparts. Interleukin-2 (IL-2) administration to NOD mice restored such amTreg clonotype expansions and prevented diabetes development. In contrast, IL-2 administration only led to few or no clonotype expansions in nTregs and Teffs, respectively. Noteworthily, IL-2-expanded amTreg and nTreg clonotypes were markedly enriched in islet-antigen specific TCRs. Altogether, our results highlight the link between a reduced clonotype expansion within the activated Treg repertoire and the development of an autoimmune disease. They also indicate that the repertoire of amTregs is amenable to rejuvenation by IL-2.
Collapse
Affiliation(s)
- Vanessa Mhanna
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - Gwladys Fourcade
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - Pierre Barennes
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - Valentin Quiniou
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
- Clinical Investigation Center in Biotherapy and Inflammation-Immunopathology-Biotherapy Department, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Pitié-Salpêtrière, Paris, France
| | - Hang P Pham
- Statistics Department, ILTOO Pharma, Paris, France
| | - Paul-Gydeon Ritvo
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - Faustine Brimaud
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - Bruno Gouritin
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - Guillaume Churlaud
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
- Clinical Investigation Center in Biotherapy and Inflammation-Immunopathology-Biotherapy Department, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Pitié-Salpêtrière, Paris, France
| | - Adrien Six
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | | | - David Klatzmann
- Sorbonne Universite, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
- Clinical Investigation Center in Biotherapy and Inflammation-Immunopathology-Biotherapy Department, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Pitié-Salpêtrière, Paris, France
| |
Collapse
|
6
|
Li Y, Ma L, Dong X, Pan Y, Shi B, He X, Zhang T, Sun S, Yao X. Preliminary analysis of spatial-temporal homogeneity and heterogeneity of TCR β chain CDR3 repertoires in BALB/c mice. INTERNATIONAL JOURNAL OF MOLECULAR EPIDEMIOLOGY AND GENETICS 2019; 10:10-28. [PMID: 30911358 PMCID: PMC6420717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 01/21/2019] [Indexed: 06/09/2023]
Abstract
The T-cell response and tolerance in non-lymph tissues differs from those in lymph tissues such as the spleen and thymus. The distribution and composition of the TCR repertoires in non-lymph tissues and how they differ and associate with their counterparts in lymph tissue remain unclear. Thus, we studied the thymus, spleen, blood, liver and small intestine of BALB/c mice at the ages of one, three and five months to carry out a preliminary analysis of the spatial-temporal homogeneity and heterogeneity of the total TCR β chain CDR3 repertoire using high-throughput sequencing technology and immune bioinformatics approaches. The data show that the diversity of the CDR3 repertoires was decreased as the mouse age increased, except in the small intestine. The number of low-expanded clones in the CDR3 repertoires was greatest in the thymus, followed by the spleen, blood, liver and small intestine, and highly expanded clones had an opposite trend in the different mice ages. The thymus and the spleen showed the greatest overlap of CDR3 sequences with the other tissues across the different mice ages. The distribution of the CDR3 repertoire length was normal, with a median of 14 aa in all the mouse tissues, except the small intestine of the one-month-old mice had a median of 12 aa. In summary, the composition and characteristics of the CDR3 repertoires in the thymus were similar to those in the spleen, and repertoires in the blood were similar to those in the liver; only the small intestine showed a unique composition. These results offer a novel method to explore the source, differentiation, proliferation and response of distinct T cells in different tissues at different mice ages.
Collapse
Affiliation(s)
- Yuehong Li
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Long Ma
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Xiaoheng Dong
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Yurong Pan
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Bin Shi
- Department of Laboratory Medicine, Zunyi Medical UniversityZunyi 563003, China
| | - Xiaoyan He
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Teng Zhang
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Suhong Sun
- Department of Breast Surgery, The First Affiliated Hospital of Zunyi Medical CollegeZunyi 563003, China
| | - Xinsheng Yao
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| |
Collapse
|
7
|
Jia X, Wang B, Zhai T, Yao Q, Li Q, Zhang JA. WITHDRAWN: T cell receptor revision and immune repertoire changes in autoimmune diseases. Clin Immunol 2018:S1521-6616(18)30724-1. [PMID: 30543918 DOI: 10.1016/j.clim.2018.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Xi Jia
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Bing Wang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Tianyu Zhai
- Department of Endocrinology, Zhongshan Hospital of Fudan University, Shanghai 201508, China
| | - Qiuming Yao
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Qian Li
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Jin-An Zhang
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China.
| |
Collapse
|
8
|
de Jong A, Jabbari A, Dai Z, Xing L, Lee D, Li MM, Duvic M, Hordinsky M, Norris DA, Price V, Mackay-Wiggan J, Clynes R, Christiano AM. High-throughput T cell receptor sequencing identifies clonally expanded CD8+ T cell populations in alopecia areata. JCI Insight 2018; 3:121949. [PMID: 30282836 DOI: 10.1172/jci.insight.121949] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/29/2018] [Indexed: 01/04/2023] Open
Abstract
Alopecia areata (AA) is an autoimmune disease in which cytotoxic T cells specifically target growing hair follicles. We used high-throughput TCR sequencing in the C3H/HeJ mouse model of AA and in human AA patients to gain insight into pathogenic T cell populations and their dynamics, which revealed clonal CD8+ T cell expansions in lesional skin. In the C3H/HeJ model, we observed interindividual sharing of TCRβ chain protein sequences, which strongly supports a model of antigenic drive in AA. The overlap between the lesional TCR repertoire and a population of CD8+NKG2D+ T cells in skin-draining lymph nodes identified this subset as pathogenic effectors. In AA patients, treatment with the oral JAK inhibitor tofacitinib resulted in a decrease in clonally expanded CD8+ T cells in the scalp but also revealed that many expanded lesional T cell clones do not completely disappear from either skin or blood during treatment with tofacitinib, which may explain in part the relapse of disease after stopping treatment.
Collapse
Affiliation(s)
| | | | | | - Luzhou Xing
- Department of Pathology, Columbia University, New York, New York, USA
| | | | | | - Madeleine Duvic
- Department of Dermatology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Maria Hordinsky
- Department of Dermatology, University of Minnesota, Minneapolis, Minnesota, USA
| | - David A Norris
- Department of Dermatology, University of Colorado, Denver, Colorado, USA
| | - Vera Price
- Department of Dermatology, UCSF, San Francisco, California, USA
| | | | | | - Angela M Christiano
- Department of Dermatology and.,Department of Genetics and Development, Columbia University, New York, New York, USA
| |
Collapse
|
9
|
Chaara W, Gonzalez-Tort A, Florez LM, Klatzmann D, Mariotti-Ferrandiz E, Six A. RepSeq Data Representativeness and Robustness Assessment by Shannon Entropy. Front Immunol 2018; 9:1038. [PMID: 29868003 PMCID: PMC5962720 DOI: 10.3389/fimmu.2018.01038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/25/2018] [Indexed: 12/30/2022] Open
Abstract
High-throughput sequencing (HTS) has the potential to decipher the diversity of T cell repertoires and their dynamics during immune responses. Applied to T cell subsets such as T effector and T regulatory cells, it should help identify novel biomarkers of diseases. However, given the extreme diversity of TCR repertoires, understanding how the sequencing conditions, including cell numbers, biological and technical sampling and sequencing depth, impact the experimental outcome is critical to proper use of these data. Here, we assessed the representativeness and robustness of TCR repertoire diversity assessment according to experimental conditions. By comparative analyses of experimental datasets and computer simulations, we found that (i) for small samples, the number of clonotypes recovered is often higher than the number of cells per sample, even after removing the singletons; (ii) high-sequencing depth for small samples alters the clonotype distributions, which can be corrected by filtering the datasets using Shannon entropy as a threshold; and (iii) a single sequencing run at high depth does not ensure a good coverage of the clonotype richness in highly polyclonal populations, which can be better covered using multiple sequencing. Altogether, our results warrant better understanding and awareness of the limitation of TCR diversity analyses by HTS and justify the development of novel computational tools for improved modeling of the highly complex nature of TCR repertoires.
Collapse
Affiliation(s)
- Wahiba Chaara
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), Paris, France
| | - Ariadna Gonzalez-Tort
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | - Laura-Maria Florez
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | - David Klatzmann
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), Paris, France
| | - Encarnita Mariotti-Ferrandiz
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), Paris, France
| | - Adrien Six
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), Paris, France
| |
Collapse
|
10
|
Gieras A, Gehbauer C, Perna-Barrull D, Engler JB, Diepenbruck I, Glau L, Joosse SA, Kersten N, Klinge S, Mittrücker HW, Friese MA, Vives-Pi M, Tolosa E. Prenatal Administration of Betamethasone Causes Changes in the T Cell Receptor Repertoire Influencing Development of Autoimmunity. Front Immunol 2017; 8:1505. [PMID: 29181000 PMCID: PMC5693859 DOI: 10.3389/fimmu.2017.01505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/25/2017] [Indexed: 12/15/2022] Open
Abstract
Prenatal glucocorticoids are routinely administered to pregnant women at risk of preterm delivery in order to improve survival of the newborn. However, in half of the cases, birth occurs outside the beneficial period for lung development. Glucocorticoids are potent immune modulators and cause apoptotic death of immature T cells, and we have previously shown that prenatal betamethasone treatment at doses eliciting lung maturation induce profound thymocyte apoptosis in the offspring. Here, we asked if there are long-term consequences on the offspring’s immunity after this treatment. In the non-obese diabetic mouse model, prenatal betamethasone clearly decreased the frequency of pathogenic T cells and the incidence of type 1 diabetes (T1D). In contrast, in the lupus-prone MRL/lpr strain, prenatal glucocorticoids induced changes in the T cell repertoire that resulted in more autoreactive cells. Even though glucocorticoids transiently enhanced regulatory T cell (Treg) development, these cells did not have a protective effect in a model for multiple sclerosis which relies on a limited repertoire of pathogenic T cells for disease induction that were not affected by prenatal betamethasone. We conclude that prenatal steroid treatment, by inducing changes in the T cell receptor repertoire, has unforeseeable consequences on development of autoimmune disease. Our data should encourage further research to fully understand the consequences of this widely used treatment.
Collapse
Affiliation(s)
- Anna Gieras
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Gehbauer
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David Perna-Barrull
- Immunology Division, Germans Trias i Pujol Research Institute and Hospital, Universitat Autonoma de Barcelona, Badalona, Spain
| | - Jan Broder Engler
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Ines Diepenbruck
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Glau
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nora Kersten
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefanie Klinge
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Marta Vives-Pi
- Immunology Division, Germans Trias i Pujol Research Institute and Hospital, Universitat Autonoma de Barcelona, Badalona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Ostmeyer J, Christley S, Rounds WH, Toby I, Greenberg BM, Monson NL, Cowell LG. Statistical classifiers for diagnosing disease from immune repertoires: a case study using multiple sclerosis. BMC Bioinformatics 2017; 18:401. [PMID: 28882107 PMCID: PMC5588725 DOI: 10.1186/s12859-017-1814-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/29/2017] [Indexed: 12/29/2022] Open
Abstract
Background Deep sequencing of lymphocyte receptor repertoires has made it possible to comprehensively profile the clonal composition of lymphocyte populations. This opens the door for novel approaches to diagnose and prognosticate diseases with a driving immune component by identifying repertoire sequence patterns associated with clinical phenotypes. Indeed, recent studies support the feasibility of this, demonstrating an association between repertoire-level summary statistics (e.g., diversity) and patient outcomes for several diseases. In our own prior work, we have shown that six codons in VH4-containing genes in B cells from the cerebrospinal fluid of patients with relapsing remitting multiple sclerosis (RRMS) have higher replacement mutation frequencies than observed in healthy controls or patients with other neurological diseases. However, prior methods to date have been limited to focusing on repertoire-level summary statistics, ignoring the vast amounts of information in the millions of individual immune receptors comprising a repertoire. We have developed a novel method that addresses this limitation by using innovative approaches for accommodating the extraordinary sequence diversity of immune receptors and widely used machine learning approaches. We applied our method to RRMS, an autoimmune disease that is notoriously difficult to diagnose. Results We use the biochemical features encoded by the complementarity determining region 3 of each B cell receptor heavy chain in every patient repertoire as input to a detector function, which is fit to give the correct diagnosis for each patient using maximum likelihood optimization methods. The resulting statistical classifier assigns patients to one of two diagnosis categories, RRMS or other neurological disease, with 87% accuracy by leave-one-out cross-validation on training data (N = 23) and 72% accuracy on unused data from a separate study (N = 102). Conclusions Our method is the first to apply statistical learning to immune repertoires to aid disease diagnosis, learning repertoire-level labels from the set of individual immune repertoire sequences. This method produced a repertoire-based statistical classifier for diagnosing RRMS that provides a high degree of diagnostic capability, rivaling the accuracy of diagnosis by a clinical expert. Additionally, this method points to a diagnostic biochemical motif in the antibodies of RRMS patients, which may offer insight into the disease process. Electronic supplementary material The online version of this article (10.1186/s12859-017-1814-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jared Ostmeyer
- Department of Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9066, USA
| | - Scott Christley
- Department of Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9066, USA
| | - William H Rounds
- Department of Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9066, USA
| | - Inimary Toby
- Department of Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9066, USA
| | - Benjamin M Greenberg
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9036, USA
| | - Nancy L Monson
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9036, USA
| | - Lindsay G Cowell
- Department of Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9066, USA.
| |
Collapse
|
12
|
Thelin MA, Kissler S, Vigneault F, Watters AL, White D, Koshy ST, Vermillion SA, Mooney DJ, Serwold T, Ali OA. In Vivo Enrichment of Diabetogenic T Cells. Diabetes 2017; 66:2220-2229. [PMID: 28396510 PMCID: PMC5521861 DOI: 10.2337/db16-0946] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 03/07/2017] [Indexed: 01/13/2023]
Abstract
Dysfunctional T cells can mediate autoimmunity, but the inaccessibility of autoimmune tissues and the rarity of autoimmune T cells in the blood hinder their study. We describe a method to enrich and harvest autoimmune T cells in vivo by using a biomaterial scaffold loaded with protein antigens. In model antigen systems, we found that antigen-specific T cells become enriched within scaffolds containing their cognate antigens. When scaffolds containing lysates from an insulin-producing β-cell line were implanted subcutaneously in autoimmune diabetes-prone NOD mice, β-cell-reactive T cells homed to these scaffolds and became enriched. These T cells induced diabetes after adoptive transfer, indicating their pathogenicity. Furthermore, T-cell receptor (TCR) sequencing identified many expanded TCRs within the β-cell scaffolds that were also expanded within the pancreata of NOD mice. These data demonstrate the utility of biomaterial scaffolds loaded with disease-specific antigens to identify and study rare, therapeutically important T cells.
Collapse
Affiliation(s)
| | | | - Frederic Vigneault
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Alexander L Watters
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Des White
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA
| | - Sandeep T Koshy
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA
| | - Sarah A Vermillion
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - David J Mooney
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA
| | - Thomas Serwold
- Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Omar A Ali
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| |
Collapse
|
13
|
Oftedal BE, Ardesjö Lundgren B, Hamm D, Gan PY, Holdsworth SR, Hahn CN, Schreiber AW, Scott HS. T cell receptor assessment in autoimmune disease requires access to the most adjacent immunologically active organ. J Autoimmun 2017; 81:24-33. [DOI: 10.1016/j.jaut.2017.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/28/2017] [Accepted: 03/06/2017] [Indexed: 01/22/2023]
|
14
|
T Cell Repertoire Diversity Is Decreased in Type 1 Diabetes Patients. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:338-348. [PMID: 28024918 PMCID: PMC5200939 DOI: 10.1016/j.gpb.2016.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 10/13/2016] [Accepted: 10/25/2016] [Indexed: 02/01/2023]
Abstract
Type 1 diabetes mellitus (T1D) is an immune-mediated disease. The autoreactive T cells in T1D patients attack and destroy their own pancreatic cells. In order to systematically investigate the potential autoreactive T cell receptors (TCRs), we used a high-throughput immune repertoire sequencing technique to profile the spectrum of TCRs in individual T1D patients and controls. We sequenced the T cell repertoire of nine T1D patients, four type 2 diabetes (T2D) patients, and six nondiabetic controls. The diversity of the T cell repertoire in T1D patients was significantly decreased in comparison with T2D patients (P=7.0E-08 for CD4+ T cells, P=1.4E-04 for CD8+ T cells) and nondiabetic controls (P=2.7E-09 for CD4+ T cells, P=7.6E-06 for CD8+ T cells). Moreover, T1D patients had significantly more highly-expanded T cell clones than T2D patients (P=5.2E-06 for CD4+ T cells, P=1.9E-07 for CD8+ T cells) and nondiabetic controls (P=1.7E-07 for CD4+ T cells, P=3.3E-03 for CD8+ T cells). Furthermore, we identified a group of highly-expanded T cell receptor clones that are shared by more than two T1D patients. Although further validation in larger cohorts is needed, our data suggest that T cell receptor diversity measurements may become a valuable tool in investigating diabetes, such as using the diversity as an index to distinguish different types of diabetes.
Collapse
|
15
|
Ma L, Yang L, Bin Shi, He X, Peng A, Li Y, Zhang T, Sun S, Ma R, Yao X. Analyzing the CDR3 Repertoire with respect to TCR-Beta Chain V-D-J and V-J Rearrangements in Peripheral T Cells using HTS. Sci Rep 2016; 6:29544. [PMID: 27404392 PMCID: PMC4941575 DOI: 10.1038/srep29544] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/20/2016] [Indexed: 11/09/2022] Open
Abstract
V-D-J rearrangement of the TCR-beta chain follows the 12/23 rule and the beyond 12/23 restriction. Currently, the proportion and characteristics of TCR-beta chain V-J rearrangement is unclear. We used high-throughput sequencing to compare and analyze TCR-beta chain V-J rearrangement and V-D-J rearrangement in the CDR3 repertoires of T cells from the PBMCs of six volunteers and six BALB/c mice. The results showed that the percentage of V-J rearrangement of the volunteers was approximately 0.7%, whereas that of the mice was 2.2%. The clonality of mice V-J rearrangement was significantly reduced compared with the V-D-J rearrangement, whereas the clonality of human V-J rearrangement was slightly reduced compared with the V-D-J rearrangement. V-J rearrangement in CDR3 involved the significant usage of N, S, F and L, whereas V-D-J rearrangement in CDR3 involved the significant usage of R and G. The levels of V deletion and J deletion in V-J rearrangement were significantly reduced compared with V-D-J rearrangement. TRBD and TRBJ usage in V-J rearrangement differed from that of V-D-J rearrangement, including dominant usage of TRBV and TRBJ and their pairing. Taken together, these results provide new ideas and technology for studies of V-D-J rearrangement and V-J rearrangement in the CDR3 repertoire.
Collapse
Affiliation(s)
- Long Ma
- Department of Immunology, Research Center for Medicine &Biology, Innovation &Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563003, China
| | - Liwen Yang
- Department of Immunology, Research Center for Medicine &Biology, Innovation &Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563003, China
| | - Bin Shi
- Department of Laboratory Medicine, Zunyi Medical University, Zunyi 563003, China
| | - Xiaoyan He
- Department of Immunology, Research Center for Medicine &Biology, Innovation &Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563003, China
| | - Aihua Peng
- Department of Immunology, Research Center for Medicine &Biology, Innovation &Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563003, China
| | - Yuehong Li
- Department of Immunology, Research Center for Medicine &Biology, Innovation &Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563003, China
| | - Teng Zhang
- Department of Immunology, Research Center for Medicine &Biology, Innovation &Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563003, China
| | - Suhong Sun
- Department of Breast Surgery, The first Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Rui Ma
- Department of Immunology, Research Center for Medicine &Biology, Innovation &Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563003, China
| | - Xinsheng Yao
- Department of Immunology, Research Center for Medicine &Biology, Innovation &Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563003, China
| |
Collapse
|
16
|
Marrero I, Aguilera C, Hamm DE, Quinn A, Kumar V. High-throughput sequencing reveals restricted TCR Vβ usage and public TCRβ clonotypes among pancreatic lymph node memory CD4(+) T cells and their involvement in autoimmune diabetes. Mol Immunol 2016; 74:82-95. [PMID: 27161799 PMCID: PMC6301078 DOI: 10.1016/j.molimm.2016.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/28/2016] [Accepted: 04/28/2016] [Indexed: 01/18/2023]
Abstract
Islet-reactive memory CD4(+) T cells are an essential feature of type 1 diabetes (T1D) as they are involved in both spontaneous disease and in its recurrence after islet transplantation. Expansion and enrichment of memory T cells have also been shown in the peripheral blood of diabetic patients. Here, using high-throughput sequencing, we investigated the clonal diversity of the TCRβ repertoire of memory CD4(+) T cells in the pancreatic lymph nodes (PaLN) of non-obese diabetic (NOD) mice and examined their clonal overlap with islet-infiltrating memory CD4T cells. Both prediabetic and diabetic NOD mice exhibited a restricted TCRβ repertoire dominated by clones expressing TRBV13-2, TRBV13-1 or TRBV5 gene segments. There is a limited degree of TCRβ overlap between the memory CD4 repertoire of PaLN and pancreas as well as between the prediabetic and diabetic group. However, public TCRβ clonotypes were identified across several individual animals, some of them with sequences similar to the TCRs from the islet-reactive T cells suggesting their antigen-driven expansion. Moreover, the majority of the public clonotypes expressed TRBV13-2 (Vβ8.2) gene segment. Nasal vaccination with an immunodominat peptide derived from the TCR Vβ8.2 chain led to protection from diabetes, suggesting a critical role for Vβ8.2(+) CD4(+) memory T cells in T1D. These results suggest that memory CD4(+) T cells bearing limited dominant TRBV genes contribute to the autoimmune diabetes and can be potentially targeted for intervention in diabetes. Furthermore, our results have important implications for the identification of public T cell clonotypes as potential novel targets for immune manipulation in human T1D.
Collapse
Affiliation(s)
- Idania Marrero
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, CA 92121, USA; Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA.
| | - Carlos Aguilera
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, CA 92121, USA
| | - David E Hamm
- Adaptive Biotechnologies, 1551 Eastlake Ave E #200, Seattle, WA 98102, USA
| | - Anthony Quinn
- Department of Biological Sciences, University of Toledo, 2801 W Bancroft St., Toledo, OH 43606, USA
| | - Vipin Kumar
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, CA 92121, USA; Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
17
|
CD8(+) T-cell pathogenicity in Rasmussen encephalitis elucidated by large-scale T-cell receptor sequencing. Nat Commun 2016; 7:11153. [PMID: 27040081 PMCID: PMC4822013 DOI: 10.1038/ncomms11153] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/25/2016] [Indexed: 11/29/2022] Open
Abstract
Rasmussen encephalitis (RE) is a rare paediatric epilepsy with uni-hemispheric inflammation and progressive neurological deficits. To elucidate RE immunopathology, we applied T-cell receptor (TCR) sequencing to blood (n=23), cerebrospinal fluid (n=2) and brain biopsies (n=5) of RE patients, and paediatric controls. RE patients present with peripheral CD8+ T-cell expansion and its strength correlates with disease severity. In addition, RE is the only paediatric epilepsy with prominent T-cell expansions in the CNS. Consistently, common clones are shared between RE patients, who also share MHC-I alleles. Public RE clones share Vβ genes and length of the CDR3. Rituximab/natalizumab/basiliximab treatment does not change TCR diversity, stem cell transplantation replaces the TCR repertoire with minimal overlap between donor and recipient, as observed in individual cases. Our study supports the hypothesis of an antigen-specific attack of peripherally expanded CD8+ lymphocytes against CNS structures in RE, which might be ameliorated by restricting access to the CNS. Rasmussen Encephalitis is a rare neurological disease accompanied by inflammation and T cell infiltration in the brain. Here the authors show that the severity of this disease correlates with clonal CD8 T cell expansion.
Collapse
|
18
|
Frankl JA, Thearle MS, Desmarais C, Bogardus C, Krakoff J. T-cell receptor repertoire variation may be associated with type 2 diabetes mellitus in humans. Diabetes Metab Res Rev 2016; 32:297-307. [PMID: 26408818 PMCID: PMC6383518 DOI: 10.1002/dmrr.2720] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/07/2015] [Accepted: 08/23/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Recent work in Pima Indians, a population with high rates of obesity and type 2 diabetes mellitus (T2DM), demonstrated that human leukocyte antigen haplotype DRB1*02 carriers have an increased acute insulin response and decreased risk for the development of T2DM, implicating loss of self-tolerance in the pathogenesis of T2DM. Advances in genomic sequencing have made T-cell receptor repertoire analysis a practical mode of investigation. METHODS High-throughput sequencing of T-cell receptor complementarity-determining region 3 was carried out in male Pima Indians with normal glucose regulation (n = 11; age = 31 ± 8 years; %fat = 30.2 ± 8.7%) and the protective DRB1*02 haplotype versus those with T2DM without DRB1*02 (n = 7; age = 34 ± 8 years; %fat = 31.2 ± 4.7%). Findings were partially replicated in another cohort by assessing the predictive ability of T-cell receptor variation on risk of T2DM in Pima Indian men (n = 27; age = 28.9 ± 7.1 years; %fat = 28.8 ± 7.1%) and women (n = 20; age = 29 ± 7.0 years; %fat = 37.1 ± 6.8%) with baseline normal glucose regulation but without the protective haplotype who were invited to follow-up examinations as frequently as every 2 years where diabetes status was assessed by a 75-g oral glucose tolerance test. Of these subjects, 13 developed diabetes. RESULTS T-cell receptor complementarity-determining region 3 length was shorter in those with T2DM, and a one-nucleotide decrease in complementarity-determining region 3 length was associated with a nearly threefold increase in risk for future diabetes. The frequency of one variable gene, TRBV7-8, was higher in those with T2DM. A 1% increase in TRBV7-8 frequency was associated with a greater than threefold increase in diabetes risk. CONCLUSIONS These results indicate that T-cell autoimmunity may be an important component in progression to T2DM in Pima Indians.
Collapse
Affiliation(s)
- Joseph A. Frankl
- Obesity and Diabetes Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Marie S. Thearle
- Obesity and Diabetes Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | | | - Clifton Bogardus
- Obesity and Diabetes Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
- Correspondence to: Jonathan Krakoff, 5th Floor, Phoenix Indian Medical Center, 4212 N. 16th St, Phoenix, AZ 85016, USA.
| |
Collapse
|
19
|
Johnson JL, Jones MB, Cobb BA. Polysaccharide A from the capsule of Bacteroides fragilis induces clonal CD4+ T cell expansion. J Biol Chem 2014; 290:5007-5014. [PMID: 25540199 DOI: 10.1074/jbc.m114.621771] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
For 3 decades, the view of MHCII-dependent antigen presentation has been completely dominated by peptide antigens despite our 2004 discovery in which MHCII was shown to present processed fragments of zwitterionic capsular polysaccharides to T cells. Published findings further demonstrate that polysaccharide A (PSA) from the capsule of Bacteroides fragilis is a potent activator of CD4(+) T cells and that these T cells have important biological functions, especially in the maintenance of immunological homeostasis. However, little is known about the nature of T cell recognition of the polysaccharide-MHCII complex or the phenotype of the resulting activated cells. Here, we use next-generation sequencing of the αβT cell receptor of CD4(+) T cells from mice stimulated with PSA in comparison with protein antigen simulation and non-immunized controls and found that PSA immunization induced clonal expansion of a small subset of suppressive CD4(+)CD45RB(low) effector/memory T cells. Moreover, the sequences of the complementarity-determining region 3 (CDR3) loop from top clones indicate a lack of specific variable β and joining region use and average CDR3 loop length. There was also a preference for a zwitterionic motif within the CDR3 loop sequences, aligning well with the known requirement for a similar motif within PSA to enable T cell activation. These data support a model in which PSA, and possibly other T cell-dependent polysaccharide antigens, elicits a clonal and therefore specific CD4(+) T cell response often characterized by pairing dual-charged CDR3 loop sequences with dual-charged PSA.
Collapse
Affiliation(s)
- Jenny L Johnson
- From the Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| | - Mark B Jones
- From the Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| | - Brian A Cobb
- From the Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106.
| |
Collapse
|
20
|
Toivonen R, Arstila TP, Hänninen A. Islet-associated T-cell receptor-β CDR sequence repertoire in prediabetic NOD mice reveals antigen-driven T-cell expansion and shared usage of VβJβ TCR chains. Mol Immunol 2014; 64:127-35. [PMID: 25480393 DOI: 10.1016/j.molimm.2014.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/05/2014] [Accepted: 11/10/2014] [Indexed: 11/27/2022]
Abstract
Autoimmune destruction of pancreatic islets in the nonobese diabetic (NOD) mouse is driven by T cells recognizing various autoantigens mostly in insulin-producing beta-cells. To investigate if T-cell accumulation in islets during early insulitis is clonally predetermined, we compared the complementarity determining regions (CDR3) of T-cell receptor (TCR)β-chains present in islet-infiltrating T cells in young prediabetic NOD mice. High-throughput sequencing of TCRβ-chain DNA extracted from islets of 7-wk old NOD mice revealed a biased TCRβ-chain repertoire in all mice, as a restricted number of clones (17-36 clones) was highly overrepresented and made over 20% of total islet repertoire in each mouse. Among these clones, various Vβ and Jβ families were present but certain VβJβ combinations such as TRBV19-0-TRBJ2-7 and TRBV13-3-TRBJ2-5 were highly shared between individual mice. On TCRβ-chain CDR sequence level, many islet clones (72-146) were shared between at least two individual mice. None of them was among expanded clones in both, suggesting considerable stochasticity in the interactions between TCR and peptide-MHC, even with a limited range of autoantigens. A comparison of islet-CDR3-sequences with CRD-sequences from other tissues revealed clonal overlap with pancreatic lymph node and gut, but these repertoires did not overlap together. Our results suggest that antigen-specific T cells are expanded in pancreatic lymph node and islets, but different specificities expand in individual mice. Some islet-infiltrating T-cell specificities may have a distinct origin shared with gut-infiltrating T cells.
Collapse
Affiliation(s)
- R Toivonen
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - T P Arstila
- Haartman Institute and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| | - A Hänninen
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland.
| |
Collapse
|
21
|
Kern J, Drutel R, Leanhart S, Bogacz M, Pacholczyk R. Reduction of T cell receptor diversity in NOD mice prevents development of type 1 diabetes but not Sjögren's syndrome. PLoS One 2014; 9:e112467. [PMID: 25379761 PMCID: PMC4224485 DOI: 10.1371/journal.pone.0112467] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/09/2014] [Indexed: 12/23/2022] Open
Abstract
Non-obese diabetic (NOD) mice are well-established models of independently developing spontaneous autoimmune diseases, Sjögren’s syndrome (SS) and type 1 diabetes (T1D). The key determining factor for T1D is the strong association with particular MHCII molecule and recognition by diabetogenic T cell receptor (TCR) of an insulin peptide presented in the context of I-Ag7 molecule. For SS the association with MHCII polymorphism is weaker and TCR diversity involved in the onset of the autoimmune phase of SS remains poorly understood. To compare the impact of TCR diversity reduction on the development of both diseases we generated two lines of TCR transgenic NOD mice. One line expresses transgenic TCRβ chain originated from a pathogenically irrelevant TCR, and the second line additionally expresses transgenic TCRαmini locus. Analysis of TCR sequences on NOD background reveals lower TCR diversity on Treg cells not only in the thymus, but also in the periphery. This reduction in diversity does not affect conventional CD4+ T cells, as compared to the TCRmini repertoire on B6 background. Interestingly, neither transgenic TCRβ nor TCRmini mice develop diabetes, which we show is due to lack of insulin B:9–23 specific T cells in the periphery. Conversely SS develops in both lines, with full glandular infiltration, production of autoantibodies and hyposalivation. It shows that SS development is not as sensitive to limited availability of TCR specificities as T1D, which suggests wider range of possible TCR/peptide/MHC interactions driving autoimmunity in SS.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Autoantibodies/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Flow Cytometry
- Genetic Variation/immunology
- Insulin/genetics
- Insulin/immunology
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Salivary Glands/immunology
- Salivary Glands/metabolism
- Sjogren's Syndrome/genetics
- Sjogren's Syndrome/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Xerostomia/immunology
Collapse
Affiliation(s)
- Joanna Kern
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| | - Robert Drutel
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| | - Silvia Leanhart
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| | - Marek Bogacz
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| | - Rafal Pacholczyk
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| |
Collapse
|
22
|
Trott DW, Thabet SR, Kirabo A, Saleh MA, Itani H, Norlander AE, Wu J, Goldstein A, Arendshorst WJ, Madhur MS, Chen W, Li CI, Shyr Y, Harrison DG. Oligoclonal CD8+ T cells play a critical role in the development of hypertension. Hypertension 2014; 64:1108-15. [PMID: 25259750 DOI: 10.1161/hypertensionaha.114.04147] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent studies have emphasized a role of adaptive immunity, and particularly T cells, in the genesis of hypertension. We sought to determine the T-cell subtypes that contribute to hypertension and renal inflammation in angiotensin II-induced hypertension. Using T-cell receptor spectratyping to examine T-cell receptor usage, we demonstrated that CD8(+) cells, but not CD4(+) cells, in the kidney exhibited altered T-cell receptor transcript lengths in Vβ3, 8.1, and 17 families in response to angiotensin II-induced hypertension. Clonality was not observed in other organs. The hypertension caused by angiotensin II in CD4(-/-) and MHCII(-/-) mice was similar to that observed in wild-type mice, whereas CD8(-/-) mice and OT1xRAG-1(-/-) mice, which have only 1 T-cell receptor, exhibited a blunted hypertensive response to angiotensin II. Adoptive transfer of pan T cells and CD8(+) T cells but not CD4(+)/CD25(-) cells conferred hypertension to RAG-1(-/-) mice. In contrast, transfer of CD4(+)/CD25(+) cells to wild-type mice receiving angiotensin II decreased blood pressure. Mice treated with angiotensin II exhibited increased numbers of kidney CD4(+) and CD8(+) T cells. In response to a sodium/volume challenge, wild-type and CD4(-/-) mice infused with angiotensin II retained water and sodium, whereas CD8(-/-) mice did not. CD8(-/-) mice were also protected against angiotensin-induced endothelial dysfunction and vascular remodeling in the kidney. These data suggest that in the development of hypertension, an oligoclonal population of CD8(+) cells accumulates in the kidney and likely contributes to hypertension by contributing to sodium and volume retention and vascular rarefaction.
Collapse
Affiliation(s)
- Daniel W Trott
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Salim R Thabet
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Annet Kirabo
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Mohamed A Saleh
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Hana Itani
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Allison E Norlander
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Jing Wu
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Anna Goldstein
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - William J Arendshorst
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Meena S Madhur
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Wei Chen
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Chung-I Li
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - Yu Shyr
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.)
| | - David G Harrison
- From the Division of Clinical Pharmacology, Department of Medicine (D.W.T., S.R.T., A.K., M.A.S., H.I., J.W., A.G., M.S.M., W.C., D.G.H.) and Department of Pharmacology and Toxicology, Faculty of Pharmacy (M.A.S.), Mansoura University, Mansoura, Egypt; Departments of Molecular Physiology and Biophysics (A.E.N.) and Biostatistics (C.-I.L., Y.S.), Vanderbilt University School of Medicine, Nashville, TN; and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill (W.J.A.).
| |
Collapse
|