1
|
James EA, Joglekar AV, Linnemann AK, Russ HA, Kent SC. The beta cell-immune cell interface in type 1 diabetes (T1D). Mol Metab 2023; 78:101809. [PMID: 37734713 PMCID: PMC10622886 DOI: 10.1016/j.molmet.2023.101809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND T1D is an autoimmune disease in which pancreatic islets of Langerhans are infiltrated by immune cells resulting in the specific destruction of insulin-producing islet beta cells. Our understanding of the factors leading to islet infiltration and the interplay of the immune cells with target beta cells is incomplete, especially in human disease. While murine models of T1D have provided crucial information for both beta cell and autoimmune cell function, the translation of successful therapies in the murine model to human disease has been a challenge. SCOPE OF REVIEW Here, we discuss current state of the art and consider knowledge gaps concerning the interface of the islet beta cell with immune infiltrates, with a focus on T cells. We discuss pancreatic and immune cell phenotypes and their impact on cell function in health and disease, which we deem important to investigate further to attain a more comprehensive understanding of human T1D disease etiology. MAJOR CONCLUSIONS The last years have seen accelerated development of approaches that allow comprehensive study of human T1D. Critically, recent studies have contributed to our revised understanding that the pancreatic beta cell assumes an active role, rather than a passive position, during autoimmune disease progression. The T cell-beta cell interface is a critical axis that dictates beta cell fate and shapes autoimmune responses. This includes the state of the beta cell after processing internal and external cues (e.g., stress, inflammation, genetic risk) that that contributes to the breaking of tolerance by hyperexpression of human leukocyte antigen (HLA) class I with presentation of native and neoepitopes and secretion of chemotactic factors to attract immune cells. We anticipate that emerging insights about the molecular and cellular aspects of disease initiation and progression processes will catalyze the development of novel and innovative intervention points to provide additional therapies to individuals affected by T1D.
Collapse
Affiliation(s)
- Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Alok V Joglekar
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amelia K Linnemann
- Center for Diabetes and Metabolic Diseases, and Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sally C Kent
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
2
|
Li J, Xiao Z, Wang D, Jia L, Nie S, Zeng X, Hu W. The screening, identification, design and clinical application of tumor-specific neoantigens for TCR-T cells. Mol Cancer 2023; 22:141. [PMID: 37649123 PMCID: PMC10466891 DOI: 10.1186/s12943-023-01844-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
Recent advances in neoantigen research have accelerated the development of tumor immunotherapies, including adoptive cell therapies (ACTs), cancer vaccines and antibody-based therapies, particularly for solid tumors. With the development of next-generation sequencing and bioinformatics technology, the rapid identification and prediction of tumor-specific antigens (TSAs) has become possible. Compared with tumor-associated antigens (TAAs), highly immunogenic TSAs provide new targets for personalized tumor immunotherapy and can be used as prospective indicators for predicting tumor patient survival, prognosis, and immune checkpoint blockade response. Here, the identification and characterization of neoantigens and the clinical application of neoantigen-based TCR-T immunotherapy strategies are summarized, and the current status, inherent challenges, and clinical translational potential of these strategies are discussed.
Collapse
Affiliation(s)
- Jiangping Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Zhiwen Xiao
- Department of Otolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, People's Republic of China
| | - Donghui Wang
- Department of Radiation Oncology, The Third Affiliated Hospital Sun Yat-Sen University, Guangzhou, 510630, People's Republic of China
| | - Lei Jia
- International Health Medicine Innovation Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Shihong Nie
- Department of Radiation Oncology, West China Hospital, Sichuan University, Cancer Center, Chengdu, 610041, People's Republic of China
| | - Xingda Zeng
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Wei Hu
- Division of Vascular Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| |
Collapse
|
3
|
Xia M, Blazevic A, Fiore-Gartland A, Hoft DF. Impact of BCG vaccination on the repertoire of human γδ T cell receptors. Front Immunol 2023; 14:1100490. [PMID: 37056780 PMCID: PMC10089282 DOI: 10.3389/fimmu.2023.1100490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Introduction Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) infection is a serious threat to human health. Vaccination with BCG prevents the development of the most severe forms of TB disease in infants and was recently shown to prevent Mtb infection in previously uninfected adolescents. γδ T cells play a major role in host defense at mucosal sites and are known to respond robustly to mycobacterial infection. However, our understanding of the effects of BCG vaccination on γδ T cell responses is incomplete. Methods In this study we performed γδ T cell receptor (TCR) repertoire sequencing of samples provided pre- and post-BCG vaccination from 10 individuals to identify specific receptors and TCR clones that are induced by BCG. Results Overall, there was no change in the diversity of γTCR or δTCR clonotypes in post- vs pre-BCG samples. Furthermore, the frequencies of TCR variable and joining region genes were minimally modulated by BCG vaccination at either the γTCR or δTCR loci. However, the γTCR and δTCR repertoires of individuals were highly dynamic; a median of ~1% of γTCR and ~6% of δTCR in the repertoire were found to significantly expand or contract in post- vs pre-BCG comparisons (FDR-q < 0.05). While many of the clonotypes whose frequency changed after BCG vaccination were not shared among multiple individuals in the cohort, several shared (i.e., "public") clonotypes were identified with a consistent increase or decrease in frequency across more than one individual; the degree of sharing of these clonotypes was significantly greater than the minimal sharing that would be expected among γTCR and δTCR repertoires. An in vitro analysis of Mtb antigen-reactive γδ T cells identified clonotypes that were similar or identical to the single-chain γTCRs and δTCRs that changed consistently after BCG vaccination; pairings of γTCRs and δTCRs that increased after BCG vaccination were significantly over-represented among the Mtb-reactive γδ T cells (p = 1.2e-6). Discussion These findings generate hypotheses about specific γδTCR clonotypes that may expand in response to BCG vaccination and may recognize Mtb antigens. Future studies are required to validate and characterize these clonotypes, with an aim to better understand the role of γδ T cells in Mtb immunity.
Collapse
Affiliation(s)
- Mei Xia
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Azra Blazevic
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Daniel F. Hoft
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, United States
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
4
|
Choi DW, Cho KA, Lee HJ, Kim YH, Woo KJ, Park JW, Ryu KH, Woo SY. Co‑transplantation of tonsil‑derived mesenchymal stromal cells in bone marrow transplantation promotes thymus regeneration and T cell diversity following cytotoxic conditioning. Int J Mol Med 2020; 46:1166-1174. [PMID: 32582998 PMCID: PMC7387097 DOI: 10.3892/ijmm.2020.4657] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Bone marrow (BM) transplantation (BMT) represents a curative treatment for various hematological disorders. Prior to BMT, a large amount of the relevant anticancer drug needed to be administered to eliminate cancer cells. However, during this pre-BMT cytotoxic conditioning regimen, hematopoietic stem cells in the BM and thymic epithelial cells were also destroyed. The T cell receptor (TCR) recognizes diverse pathogen, tumor and environmental antigens, and confers immunological memory and self-tolerance. Delayed thymus reconstitution following pre-BMT cytotoxic conditioning impedes de novo thymopoiesis and limits T cell-mediated immunity. Several cytokines, such as RANK ligand, interleukin (IL)-7, IL-22 and stem cell factor, were recently reported to improve thymopoiesis and immune function following BMT. In the present study, it was found that the co-transplantation of tonsil-derived mesenchymal stromal cells (T-MSCs) with BM-derived cells (BMCs) accelerated the recovery of involuted thymuses in mice following partial pre-BMT conditioning with busulfan-cyclophosphamide treatment, possibly by inducing FMS-like tyrosine kinase 3 ligand (FLT3L) and fibroblast growth factor 7 (FGF7) production in T-MSCs. The co-transplantation of T-MSCs with BMCs also replenished the CD3+ cell population by inhibiting thymocyte apoptosis following pre-BMT cytotoxic conditioning. Furthermore, T-MSC co-transplantation improved the recovery of the TCR repertoire and led to increased thymus-generated T cell diversity.
Collapse
Affiliation(s)
- Da-Won Choi
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Hyun-Ji Lee
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Yu-Hee Kim
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Kyong-Je Woo
- Department of Plastic and Reconstructive Surgery, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Joo-Won Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| |
Collapse
|
5
|
Cook DP, Cunha JPMCM, Martens PJ, Sassi G, Mancarella F, Ventriglia G, Sebastiani G, Vanherwegen AS, Atkinson MA, Van Huynegem K, Steidler L, Caluwaerts S, Rottiers P, Teyton L, Dotta F, Gysemans C, Mathieu C. Intestinal Delivery of Proinsulin and IL-10 via Lactococcus lactis Combined With Low-Dose Anti-CD3 Restores Tolerance Outside the Window of Acute Type 1 Diabetes Diagnosis. Front Immunol 2020; 11:1103. [PMID: 32582188 PMCID: PMC7295939 DOI: 10.3389/fimmu.2020.01103] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 05/06/2020] [Indexed: 01/07/2023] Open
Abstract
A combination treatment (CT) of proinsulin and IL-10 orally delivered via genetically modified Lactococcus lactis bacteria combined with low-dose anti-CD3 (aCD3) therapy successfully restores glucose homeostasis in newly diagnosed non-obese diabetic (NOD) mice. Tolerance is accompanied by the accumulation of Foxp3+ regulatory T cells (Tregs) in the pancreas. To test the potential of this therapy outside the window of acute diabetes diagnosis, we substituted autoimmune diabetic mice, with disease duration varying between 4 and 53 days, with syngeneic islets at the time of therapy initiation. Untreated islet recipients consistently showed disease recurrence after 8.2 ± 0.7 days, while 32% of aCD3-treated and 48% of CT-treated mice remained normoglycemic until 6 weeks after therapy initiation (P < 0.001 vs. untreated controls for both treatments, P < 0.05 CT vs. aCD3 therapy). However, mice that were diabetic for more than 2 weeks before treatment initiation were less efficient at maintaining normoglycemia than those treated within 2 weeks of diabetes diagnosis, particularly in the aCD3-treated group. The complete elimination of endogenous beta cell mass with alloxan at the time of diabetes diagnosis pointed toward the significance of continuous feeding of the islet antigen proinsulin at the time of aCD3 therapy for treatment success. The CT providing proinsulin protected 69% of mice, compared to 33% when an irrelevant antigen (ovalbumin) was combined with aCD3 therapy, or to 27% with aCD3 therapy alone. Sustained tolerance was accompanied with a reduction of IGRP+CD8+ autoreactive T cells and an increase in insulin-reactive (InsB12-20 or InsB13-2) Foxp3+CD4+ Tregs, with a specific accumulation of Foxp3+ Tregs around the insulin-containing islet grafts after CT with proinsulin. The combination of proinsulin and IL-10 via oral Lactococcus lactis with low-dose aCD3 therapy can restore tolerance to beta cells in autoimmune diabetic mice, also when therapy is started outside the window of acute diabetes diagnosis, providing persistence of insulin-containing islets or prolonged beta cell function.
Collapse
Affiliation(s)
- Dana P Cook
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing, Campus Gasthuisberg O&N 1, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - João Paulo Monteiro Carvalho Mori Cunha
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing, Campus Gasthuisberg O&N 1, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Pieter-Jan Martens
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing, Campus Gasthuisberg O&N 1, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Gabriele Sassi
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing, Campus Gasthuisberg O&N 1, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Francesca Mancarella
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena and Fondazione Umberto Di Mario ONLUS-Toscana Life Science Park, Siena, Italy
| | - Giuliana Ventriglia
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena and Fondazione Umberto Di Mario ONLUS-Toscana Life Science Park, Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena and Fondazione Umberto Di Mario ONLUS-Toscana Life Science Park, Siena, Italy
| | - An-Sofie Vanherwegen
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing, Campus Gasthuisberg O&N 1, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Mark A Atkinson
- Immunology and Laboratory Medicine, Department of Pathology, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| | | | | | | | | | - Luc Teyton
- The Teyton Lab, Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, United States
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena and Fondazione Umberto Di Mario ONLUS-Toscana Life Science Park, Siena, Italy
| | - Conny Gysemans
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing, Campus Gasthuisberg O&N 1, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing, Campus Gasthuisberg O&N 1, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
6
|
Song Z, Chen X, Shi Y, Huang R, Wang W, Zhu K, Lin S, Wang M, Tian G, Yang J, Chen G. Evaluating the Potential of T Cell Receptor Repertoires in Predicting the Prognosis of Resectable Non-Small Cell Lung Cancers. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:73-83. [PMID: 32995352 PMCID: PMC7488751 DOI: 10.1016/j.omtm.2020.05.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/19/2020] [Indexed: 01/07/2023]
Abstract
For resectable cancer patients, a method that could precisely predict the risk of postoperative recurrence would be crucial for guiding adjuvant treatment. Since T cell receptor (TCR) repertoires had been shown to be closely related to the dynamics of cancers, here we enrolled a cohort of patients to evaluate the potential of TCR repertoires in predicting the prognosis of resectable non-small cell lung cancers. Specifically, TCRβ repertoires were analyzed in surgical tumor tissues and matched adjacent non-tumor tissues from 39 patients enrolled with resectable non-small cell lung cancer, through target enrichment and high-throughput sequencing. As a result, there are significant differences between the TCR repertories of tumor samples and those of matched adjacent non-tumor samples as evaluated by criteria like the number of clonotypes. In addition, TCR repertoires were significantly associated with a few clinical features, as well as somatic mutations. Finally, certain TCRβ variable-joining (V-J) pairings were featured to build a logistic regression model in predicting postoperative recurrence of resectable non-small cell lung cancers with a testing area under the receiver operating characteristic curve (AUC) of around 0.9. Thus, we hypothesize that TCR repertoires could be potentially used to predict prognosis after curative surgery for non-small cell lung cancer patients.
Collapse
Affiliation(s)
- Zhengbo Song
- Department of Medical Oncology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | | | - Yi Shi
- Department of Molecular Pathology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou 350014, China
| | - Rongfang Huang
- Department of Pathology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou 350014, China
| | - Wenxian Wang
- Department of Medical Oncology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Kunshou Zhu
- Department of Thoracic Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Shaofeng Lin
- Department of Thoracic Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Minxian Wang
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA
| | - Geng Tian
- Geneis Beijing, Beijing 100102, China
| | | | - Gang Chen
- Department of Pathology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou 350014, China.,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| |
Collapse
|
7
|
Multiple sevoflurane exposures don't disturb the T-cell receptor repertoire in infant rhesus monkeys' thymus. Life Sci 2020; 248:117457. [PMID: 32092334 DOI: 10.1016/j.lfs.2020.117457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/10/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022]
Abstract
AIMS Multiple surgical procedures and anesthesia increase the risk of the development in children. However, the influence of such exposures on the developing childhood immunity organs is rarely reported. MATERIALS AND METHODS High-throughput sequencing of T-cell receptor (TCR) repertoires (TCRseq) from rhesus monkeys' thymus was performed to investigate whether anesthetics could induce de novo antigen recognition via TCR or TCR development impairments. KEY FINDINGS No significant difference between sevoflurane and control groups regarding VJ gene combinations and diversity of V and J gene was seen, nor was there an obvious change in similar average number of Complementarity Determining Region 3 (CDR3) aa clonotypes. Our analysis of Rank abundance, Gini coefficient, Simpson index, Normalized Shannon Diversity Entropy (NSDE), Morisita-Horn Similarity Index (MHSI) and Bhattacharyya Distance (BD) indicated there is no difference in TCR diversity and similarity. SIGNIFICANCE These results suggest early events in thymic T cell development and repertoire generation are not abnormality after multiple sevoflurane exposure during childhood. The stabilization of the immune repertoires suggested the safety of sevoflurane in host immune response in children.
Collapse
|
8
|
Fu Y, Li B, Li Y, Wang M, Yue Y, Xu L, Li S, Huang Q, Liu S, Dai Y. A comprehensive immune repertoire study for patients with pulmonary tuberculosis. Mol Genet Genomic Med 2019; 7:e00792. [PMID: 31173489 PMCID: PMC6625341 DOI: 10.1002/mgg3.792] [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: 08/16/2018] [Revised: 02/25/2019] [Accepted: 04/08/2019] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Tuberculosis (TB) is a major global health problem and has replaced HIV as the leading cause of death from a single infectious agent. METHODS Here, we applied high throughput sequencing to study the immune repertoire of nine pulmonary tuberculosis patients and nine healthy control samples. RESULTS Tuberculosis patients and healthy controls displayed significantly different high express clones and distinguishable sharing of CDR3 sequences. The TRBV and TRBJ gene usage showed higher expression clones in patients than in controls and we also found specific high express TRBV and TRBJ gene clones in different groups. In addition, six highly expressed TRBV/TRBJ combinations were detected in the CD4 group, 21 in the CD8 group and 32 in the tissue group. CONCLUSION In conclusion, we studied the patients with tuberculosis as well as healthy control individuals in order to understand the characteristics of immune repertoire. Sharing of CDR3 sequences and differential expression of genes was found among the patients with tuberculosis which could be used for the development of potential vaccine and targets treatment.
Collapse
Affiliation(s)
- Yingyun Fu
- Department of Respiratory and Critical Care Medicine, The Second Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, China.,Shenzhen Key Laboratory of Respiratory Disease, Shenzhen, China
| | - Bo Li
- Department of Pediatrics, The Second Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Yazhen Li
- Department of Respiratory and Critical Care Medicine, The Second Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Minlian Wang
- Shenzhen Key Laboratory of Respiratory Disease, Shenzhen, China
| | - Yongjian Yue
- Shenzhen Key Laboratory of Respiratory Disease, Shenzhen, China
| | - Lan Xu
- Department of Respiratory and Critical Care Medicine, The Second Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Shulin Li
- Department of Respiratory and Critical Care Medicine, The Second Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Qijun Huang
- Department of Respiratory and Critical Care Medicine, The Second Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Song Liu
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, China
| |
Collapse
|
9
|
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
|
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
|
Dai YD, Sheng H, Dias P, Jubayer Rahman M, Bashratyan R, Regn D, Marquardt K. Autoimmune Responses to Exosomes and Candidate Antigens Contribute to Type 1 Diabetes in Non-Obese Diabetic Mice. Curr Diab Rep 2017; 17:130. [PMID: 29080983 DOI: 10.1007/s11892-017-0962-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW The initial autoimmune trigger of type 1 diabetes (T1D) remains unclear. In non-obese diabetic (NOD) mice, islet inflammation starts early in life, suggesting the presence of an endogenous trigger for the spontaneous autoimmune response in this T1D mouse model. In this review, we argue that abnormal release of exosomes might be the trigger of the early inflammatory and autoimmune responses in the islets. RECENT FINDINGS Exosomes are nano-sized membrane complexes that are secreted by cells following fusion of late endosomes and/or multivesicular bodies with the plasma membrane. They are known extracellular messengers, communicating among neighboring cells via transporting large molecules from parent cells to recipient cells. Recent evidence demonstrates that these extracellular vesicles can modulate immune responses. It has been shown that insulinoma and islet mesenchymal stem cell-released exosomes are potent immune stimuli that can induce autoreactive B and T cells. Searching for candidate antigens in the exosomes identified endogenous retrovirus (ERV) Env and Gag antigens, which are homologous to an endogenous murine leukemia retrovirus. Autoantibodies and autoreactive T cells spontaneously developed in NOD mice can react to these retroviral antigens. More importantly, expression of the retroviral antigens in the islet mesenchymal stem cells is associated with disease susceptibility, and the expression is restricted to T1D-susceptible but not resistant mouse strains. Exosomes are novel autoimmune targets, carrying autoantigens that can stimulate innate and adaptive immune responses. An abnormal or excess release of exosomes, particularly those ones containing endogenous retroviral antigens might be responsible for triggering tissue-specific inflammatory and autoimmune responses.
Collapse
Affiliation(s)
- Yang D Dai
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA.
- Biomedical Research Institute of Southern California, San Diego, CA, USA.
| | - Huiming Sheng
- Torrey Pines Institute for Molecular Studies, San Diego, CA, USA
- Tongren Hospital Affiliated to SJTU, School of Medicine, Shanghai, China
| | - Peter Dias
- Biomedical Research Institute of Southern California, San Diego, CA, USA
| | - M Jubayer Rahman
- Torrey Pines Institute for Molecular Studies, San Diego, CA, USA
| | - Roman Bashratyan
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Danielle Regn
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Kristi Marquardt
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW The genetic susceptibility and dominant protection for type 1 diabetes (T1D) associated with human leukocyte antigen (HLA) haplotypes, along with minor risk variants, have long been thought to shape the T cell receptor (TCR) repertoire and eventual phenotype of autoreactive T cells that mediate β-cell destruction. While autoantibodies provide robust markers of disease progression, early studies tracking autoreactive T cells largely failed to achieve clinical utility. RECENT FINDINGS Advances in acquisition of pancreata and islets from T1D organ donors have facilitated studies of T cells isolated from the target tissues. Immunosequencing of TCR α/β-chain complementarity determining regions, along with transcriptional profiling, offers the potential to transform biomarker discovery. Herein, we review recent studies characterizing the autoreactive TCR signature in T1D, emerging technologies, and the challenges and opportunities associated with tracking TCR molecular profiles during the natural history of T1D.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Amanda Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Howard R Seay
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Michael J Haller
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA.
| |
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
|
Takeda Y, Suto Y, Ito K, Hashimoto W, Nishiya T, Ueda K, Narushima T, Takahashi T, Ogasawara K. TRAV7-2*02 Expressing CD8⁺ T Cells Are Responsible for Palladium Allergy. Int J Mol Sci 2017; 18:ijms18061162. [PMID: 28561797 PMCID: PMC5485986 DOI: 10.3390/ijms18061162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 12/16/2022] Open
Abstract
While metallic biomaterials have led to an improvement in the quality of life, metal allergies, especially to palladium (Pd), has caused a recent increase in allergic patients. Metal allergy is known to be a T cell-mediated delayed-type hypersensitivity (DTH); however, the pathogenic T cell subsets and the specific T cell receptor (TCR) have not been identified. Therefore, we attempted to identify the pathogenic T cells responsible for Pd allergy. We found that activating CD8+ T cells significantly increased and that the TRAV (TCRα variable) 7-2*02 chain skewed in Pd allergic mice. Furthermore, adoptive transfer experiments revealed that in vitro-cultured Pd-stimulated antigen presenting cells (APCs) function as memory APCs with recipient mice developing Pd allergy and that the frequency of TRAV7-2*02 increases the same as conventional Pd allergic mice. In contrast, neither proliferation of CD8+ T cells nor increasing of TRAV7-2*02 was observed in major histocompatibility complex I (MHC I)-deficient Pd-APCs transferred to mice. Taken together, we revealed that TRAV7-2*02-expressing CD8+ T cells are the pathogenic T cells for the development of Pd allergy. We also identified the CDR3 consensus motif of pathogenic TCRs as CAAXSGSWQLIF in TRAV7-2*02/TRAJ (TCRα junction)22*01 positive cells. These results suggest that the specific TCRs represent novel targets for the development of diagnostics and treatments for metal allergy.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/transplantation
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Hypersensitivity/genetics
- Hypersensitivity/immunology
- Hypersensitivity/metabolism
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Lymphocyte Activation/immunology
- Mice, Inbred C57BL
- Mice, Knockout
- Palladium/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
Collapse
Affiliation(s)
- Yuri Takeda
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
- Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan.
| | - Yoshiko Suto
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Koyu Ito
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Wataru Hashimoto
- Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan.
| | - Tadashi Nishiya
- Department of Pharmacology, School of Pharmaceutical Sciences, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan.
| | - Kyosuke Ueda
- Department of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02 Aza Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8579, Japan.
| | - Takayuki Narushima
- Department of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02 Aza Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8579, Japan.
| | - Tetsu Takahashi
- Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan.
| | - Kouetsu Ogasawara
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| |
Collapse
|
15
|
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
|
16
|
TCR sequencing of single cells reactive to DQ2.5-glia-α2 and DQ2.5-glia-ω2 reveals clonal expansion and epitope-specific V-gene usage. Mucosal Immunol 2016; 9:587-96. [PMID: 26838051 DOI: 10.1038/mi.2015.147] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/29/2015] [Indexed: 02/04/2023]
Abstract
CD4+ T cells recognizing dietary gluten epitopes in the context of disease-associated human leukocyte antigen (HLA)-DQ2 or HLA-DQ8 molecules are the key players in celiac disease pathogenesis. Here, we conducted a large-scale single-cell paired T-cell receptor (TCR) sequencing study to characterize the TCR repertoire for two homologous immunodominant gluten epitopes, DQ2.5-glia-α2 and DQ2.5-glia-ω2, in blood of celiac disease patients after oral gluten challenge. Despite sequence similarity of the epitopes, the TCR repertoires are unique but shared several overall features. We demonstrate that clonally expanded T cells dominate the T-cell responses to both epitopes. Moreover, we find V-gene bias of TRAV26, TRAV4, and TRBV7 in DQ2.5-glia-α2 reactive TCRs, while DQ2.5-glia-ω2 TCRs displayed significant bias toward TRAV4 and TRBV4. The knowledge that antigen-specific TCR repertoire in chronic inflammatory diseases tends to be dominated by a few expanded clones that use the same TCR V-gene segments across patients is important information for HLA-associated diseases where the antigen is unknown.
Collapse
|
17
|
Jain S, Chen J, Nicolae A, Wang H, Shin DM, Adkins EB, Sproule TJ, Leeth CM, Sakai T, Kovalchuk AL, Raffeld M, Ward JM, Rehg JE, Waldmann TA, Jaffe ES, Roopenian DC, Morse HC. IL-21-driven neoplasms in SJL mice mimic some key features of human angioimmunoblastic T-cell lymphoma. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:3102-14. [PMID: 26363366 DOI: 10.1016/j.ajpath.2015.07.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/24/2015] [Accepted: 07/30/2015] [Indexed: 12/12/2022]
Abstract
SJL/J mice exhibit a high incidence of mature B-cell lymphomas that require CD4(+) T cells for their development. We found that their spleens and lymph nodes contained increased numbers of germinal centers and T follicular helper (TFH) cells. Microarray analyses revealed high levels of transcripts encoding IL-21 associated with high levels of serum IL-21. We developed IL-21 receptor (IL21R)-deficient Swiss Jim Lambart (SJL) mice to determine the role of IL-21 in disease. These mice had reduced numbers of TFH cells, lower serum levels of IL-21, and few germinal center B cells, and they did not develop B-cell tumors, suggesting IL-21-dependent B-cell lymphomagenesis. We also noted a series of features common to SJL disease and human angioimmunoblastic T-cell lymphoma (AITL), a malignancy of TFH cells. Gene expression analyses of AITL showed that essentially all cases expressed elevated levels of transcripts for IL21, IL21R, and a series of genes associated with TFH cell development and function. These results identify a mouse model with features of AITL and suggest that patients with the disease might benefit from therapeutic interventions that interrupt IL-21 signaling.
Collapse
Affiliation(s)
- Shweta Jain
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Jing Chen
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Alina Nicolae
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Hongsheng Wang
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Dong-Mi Shin
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland; Department of Food and Nutrition, Seoul National University, Seoul, Republic of Korea
| | - Elisabeth B Adkins
- Jackson Laboratory, Bar Harbor, Maine; Genetics Program, Tufts University Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts
| | | | - Caroline M Leeth
- Genetics Program, Tufts University Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts; Department of Animal and Poultry Sciences, College of Agriculture and Life Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Tomomi Sakai
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Alexander L Kovalchuk
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Mark Raffeld
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jerrold M Ward
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Jerold E Rehg
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elaine S Jaffe
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Derry C Roopenian
- Jackson Laboratory, Bar Harbor, Maine; Genetics Program, Tufts University Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts
| | - Herbert C Morse
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland.
| |
Collapse
|