1
|
Muthana MM, Du X, Liu M, Wang X, Wu W, Ai C, Su L, Zheng P, Liu Y. CTLA-4 antibody-drug conjugate reveals autologous destruction of B-lymphocytes associated with regulatory T cell impairment. eLife 2023; 12:RP87281. [PMID: 38127423 PMCID: PMC10735222 DOI: 10.7554/elife.87281] [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] [Indexed: 12/23/2023] Open
Abstract
Germline CTLA-4 deficiency causes severe autoimmune diseases characterized by dysregulation of Foxp3+ Tregs, hyper-activation of effector memory T cells, and variable forms autoimmune cytopenia including gradual loss of B cells. Cancer patients with severe immune-related adverse events (irAE) after receiving anti-CTLA-4/PD-1 combination immunotherapy also have markedly reduced peripheral B cells. The immunological basis for B cell loss remains unexplained. Here, we probe the decline of B cells in human CTLA-4 knock-in mice by using anti-human CTLA-4 antibody Ipilimumab conjugated to a drug payload emtansine (Anti-CTLA-4 ADC). The anti-CTLA-4 ADC-treated mice have T cell hyper-proliferation and their differentiation into effector cells which results in B cell depletion. B cell depletion is mediated by both CD4 and CD8 T cells and at least partially rescued by anti-TNF-alpha antibody. These data revealed an unexpected antagonism between T and B cells and the importance of regulatory T cells in preserving B cells.
Collapse
Affiliation(s)
- Musleh M Muthana
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Pharmacology, University of Maryland School of MedicineBaltimoreUnited States
| | - Xuexiang Du
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong UniversityJinanChina
| | - Mingyue Liu
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Pharmacology, University of Maryland School of MedicineBaltimoreUnited States
| | - Xu Wang
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Pharmacology, University of Maryland School of MedicineBaltimoreUnited States
| | - Wei Wu
- OncoC4, IncRockvilleUnited States
| | - Chunxia Ai
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong UniversityJinanChina
| | - Lishan Su
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Pharmacology, University of Maryland School of MedicineBaltimoreUnited States
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Microbiology & Immunology, University of Maryland School of MedicineBaltimoreUnited States
| | | | - Yang Liu
- OncoC4, IncRockvilleUnited States
| |
Collapse
|
2
|
Liu Y, Zhang Z, Kang Z, Zhou XJ, Liu S, Guo S, Jin Q, Li T, Zhou L, Wu X, Wang YN, Lu L, He Y, Li F, Zhang H, Liu Y, Xu H. Interleukin 4-driven reversal of self-reactive B cell anergy contributes to the pathogenesis of systemic lupus erythematosus. Ann Rheum Dis 2023; 82:1444-1454. [PMID: 37567607 DOI: 10.1136/ard-2023-224453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/18/2023] [Indexed: 08/13/2023]
Abstract
OBJECTIVES Reactivation of anergic autoreactive B cells (BND cells) is a key aetiological process in systemic lupus erythematosus (SLE), yet the underlying mechanism remains largely elusive. This study aimed to investigate how BND cells participate in the pathogenesis of SLE and the underlying mechanism. METHODS A combination of phenotypical, large-scale transcriptome and B cell receptor (BCR) repertoire profiling were employed at molecular and single cell level on samples from healthy donors and patients with SLE. Isolated naïve B cells from human periphery blood were treated with anti-CD79b mAb in vitro to induce anergy. IgM internalisation was tracked by confocal microscopy and was qualified by flow cytometer. RESULTS We characterised the decrease and disruption of BND cells in SLE patients and demonstrated IL-4 as an important cytokine to drive such pathological changes. We then elucidated that IL-4 reversed B cell anergy by promoting BCR recycling to the cell surface via STAT6 signalling. CONCLUSIONS We demonstrated the significance of IL-4 in reversing B cell anergy and established the scientific rationale to treat SLE via blocking IL-4 signalling, also providing diagnostic and prognostic biomarkers to identify patients who are most likely going to benefit from such treatments.
Collapse
Affiliation(s)
- Yaoyang Liu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhiguo Zhang
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zijian Kang
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xu-Jie Zhou
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) Ministry of Education, Beijing, China
| | - Shujun Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shicheng Guo
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Qianmei Jin
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ting Li
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ling Zhou
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Wu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yan-Na Wang
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) Ministry of Education, Beijing, China
| | - Liangjing Lu
- Department of Rheumatology and Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanran He
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Fubin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) Ministry of Education, Beijing, China
| | - Yuncai Liu
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
3
|
Muthana MM, Du X, Liu M, Wang X, Wu W, Ai C, Su L, Zheng P, Liu Y. CTLA-4 antibody-drug conjugate reveals autologous destruction of B-lymphocytes associated with regulatory T cell impairment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530608. [PMID: 36909522 PMCID: PMC10002750 DOI: 10.1101/2023.03.01.530608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Germline CTLA-4 deficiency causes severe autoimmune diseases characterized by dysregulation of Foxp3+ Tregs, hyper-activation of effector memory T cells, and variable forms autoimmune cytopenia including gradual loss of B cells. Cancer patients with severe immune-related adverse events (irAE) after receiving anti-CTLA-4/PD-1 combination immunotherapy also have markedly reduced peripheral B cells. The immunological basis for B cell loss remains unexplained. Here we probe the decline of B cells in human CTLA-4 knock-in mice by using antihuman CTLA-4 antibody Ipilimumab conjugated to a drug payload emtansine (Anti-CTLA-4 ADC). The anti-CTLA-4 ADC-treated mice have T cell hyper-proliferation and their differentiation into effector cells which results in B cell depletion. B cell depletion is mediated by both CD4 and CD8 T cells and at least partially rescued by anti-TNF-alpha antibody. These data revealed an unexpected antagonism between T and B cells and the importance of regulatory T cells in preserving B cells.
Collapse
Affiliation(s)
- Musleh M. Muthana
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Xuexiang Du
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Mingyue Liu
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Xu Wang
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Wei Wu
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- OncoC4, Inc.; Rockville, MD 20805, USA
| | - Chunxia Ai
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Lishan Su
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Microbiology & Immunology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Pan Zheng
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Surgery, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- OncoC4, Inc.; Rockville, MD 20805, USA
| | - Yang Liu
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Surgery, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- OncoC4, Inc.; Rockville, MD 20805, USA
| |
Collapse
|
4
|
Gupta S, Demirdag Y, Gupta AA. Members of the Regulatory Lymphocyte Club in Common Variable Immunodeficiency. Front Immunol 2022; 13:864307. [PMID: 35669770 PMCID: PMC9164302 DOI: 10.3389/fimmu.2022.864307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/30/2022] [Indexed: 12/29/2022] Open
Abstract
The role of CD4 T regulatory cells is well established in peripheral tolerance and the pathogenesis of the murine model and human autoimmune diseases. CD4 T regulatory cells (CD4 Tregs) have been investigated in common variable immunodeficiency (CVID). Recently, additional members have been added to the club of regulatory lymphocytes. These include CD8 T regulatory (CD8 Tregs), B regulatory (Bregs), and T follicular helper regulatory (TFR) cells. There are accumulating data to suggest their roles in both human and experimental models of autoimmune disease. Their phenotypic characterization and mechanisms of immunoregulation are evolving. Patients with CVID may present or are associated with an increased frequency of autoimmunity and autoimmune diseases. In this review, we have primarily focused on the characteristics of CD4 Tregs and new players of the regulatory club and their changes in patients with CVID in relation to autoimmunity and emphasized the complexity of interplay among various regulatory lymphocytes. We suggest future careful investigations of phenotypic and functional regulatory lymphocytes in a large cohort of phenotypic and genotypically defined CVID patients to define their role in the pathogenesis of CVID and autoimmunity associated with CVID.
Collapse
|
5
|
Wing JB, Lim EL, Sakaguchi S. Control of foreign Ag-specific Ab responses by Treg and Tfr. Immunol Rev 2020; 296:104-119. [PMID: 32564426 DOI: 10.1111/imr.12888] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/11/2020] [Accepted: 05/21/2020] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Tregs) expressing the transcription factor Foxp3 play a critical role in the control of immune homeostasis including the regulation of humoral immunity. Recently, it has become clear that a specialized subset of Tregs, T-follicular regulatory cells (Tfr), have a particular role in the control of T-follicular helper (Tfh) cell-driven germinal center (GC) responses. Following similar differentiation signals as received by Tfh, Tfr gain expression of characteristic chemokine receptors and transcription factors such as CXCR5 and BCL6 allowing them to travel to the B-cell follicle and deliver in situ suppression. It seems clear that Tfr are critical for the prevention of autoimmune antibody induction. However, their role in the control of foreign antigen-specific antibody responses appears more complex with various reports demonstrating either increased or decreased antigen-specific antibody responses following inhibition of Tfr function. Due to their recent discovery, our understanding of Tfr formation and function still has many gaps. In this review, we discuss our current knowledge of both Tregs and Tfr in the context of humoral immunity and how these cells might be manipulated in order to better control vaccine responses.
Collapse
Affiliation(s)
- James B Wing
- Laboritory of Human Immunology (Single Cell Immunology), Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ee Lyn Lim
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
6
|
Gong F, Zheng T, Zhou P. T Follicular Helper Cell Subsets and the Associated Cytokine IL-21 in the Pathogenesis and Therapy of Asthma. Front Immunol 2019; 10:2918. [PMID: 31921177 PMCID: PMC6923700 DOI: 10.3389/fimmu.2019.02918] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Abstract
For many decades, T helper 2 (TH2) cells have been considered to predominantly regulate the pathogenic manifestations of allergic asthma, such as IgE-mediated sensitization, airway hyperresponsiveness, and eosinophil infiltration. However, recent discoveries have significantly shifted our understanding of asthma from a simple TH2 cell-dependent disease to a heterogeneous disease regulated by multiple T cell subsets, including T follicular helper (TFH) cells. TFH cells, which are a specialized cell population that provides help to B cells, have attracted intensive attention in the past decade because of their crucial role in regulating antibody response in a broad range of diseases. In particular, TFH cells are essential for IgE antibody class-switching. In this review, we summarize the recent progress regarding the role of TFH cells and their signature cytokine interleukin (IL)-21 in asthma from mouse studies and clinical reports. We further discuss future therapeutic strategies to treat asthma by targeting TFH cells and IL-21.
Collapse
Affiliation(s)
- Fang Gong
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ting Zheng
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Pengcheng Zhou
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
7
|
Getahun A, Cambier JC. Non-Antibody-Secreting Functions of B Cells and Their Contribution to Autoimmune Disease. Annu Rev Cell Dev Biol 2019; 35:337-356. [PMID: 30883216 DOI: 10.1146/annurev-cellbio-100617-062518] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
B cells play multiple important roles in the pathophysiology of autoimmune disease. Beyond producing pathogenic autoantibodies, B cells can act as antigen-presenting cells and producers of cytokines, including both proinflammatory and anti-inflammatory cytokines. Here we review our current understanding of the non-antibody-secreting roles that B cells may play during development of autoimmunity, as learned primarily from reductionist preclinical models. Attention is also given to concepts emerging from clinical studies using B cell depletion therapy, which shed light on the roles of these mechanisms in human autoimmune disease.
Collapse
Affiliation(s)
- Andrew Getahun
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA; .,Department of Biomedical Research, National Jewish Health, Denver, Colorado 80206, USA
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA; .,Department of Biomedical Research, National Jewish Health, Denver, Colorado 80206, USA
| |
Collapse
|
8
|
Li W, Gauthier JM, Higashikubo R, Hsiao HM, Tanaka S, Vuong L, Ritter JH, Tong AY, Wong BW, Hachem RR, Puri V, Bharat A, Krupnick AS, Hsieh CS, Baldwin WM, Kelly FL, Palmer SM, Gelman AE, Kreisel D. Bronchus-associated lymphoid tissue-resident Foxp3+ T lymphocytes prevent antibody-mediated lung rejection. J Clin Invest 2019; 129:556-568. [PMID: 30561386 PMCID: PMC6355223 DOI: 10.1172/jci122083] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/06/2018] [Indexed: 12/19/2022] Open
Abstract
Antibody-mediated rejection (AMR) is a principal cause of acute and chronic failure of lung allografts. However, mechanisms mediating this oftentimes fatal complication are poorly understood. Here, we show that Foxp3+ T cells formed aggregates in rejection-free human lung grafts and accumulated within induced bronchus-associated lymphoid tissue (BALT) of tolerant mouse lungs. Using a retransplantation model, we show that selective depletion of graft-resident Foxp3+ T lymphocytes resulted in the generation of donor-specific antibodies (DSA) and AMR, which was associated with complement deposition and destruction of airway epithelium. AMR was dependent on graft infiltration by B and T cells. Depletion of graft-resident Foxp3+ T lymphocytes resulted in prolonged interactions between B and CD4+ T cells within transplanted lungs, which was dependent on CXCR5-CXCL13. Blockade of CXCL13 as well as inhibition of the CD40 ligand and the ICOS ligand suppressed DSA production and prevented AMR. Thus, we have shown that regulatory Foxp3+ T cells residing within BALT of tolerant pulmonary allografts function to suppress B cell activation, a finding that challenges the prevailing view that regulation of humoral responses occurs peripherally. As pulmonary AMR is largely refractory to current immunosuppression, our findings provide a platform for developing therapies that target local immune responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ramsey R. Hachem
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Ankit Bharat
- Department of Surgery, Northwestern University, Chicago, Illinois, USA
| | | | - Chyi S. Hsieh
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - William M. Baldwin
- Department of Immunology, Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio, USA
| | - Francine L. Kelly
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Scott M. Palmer
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | | | - Daniel Kreisel
- Department of Surgery
- Department of Pathology & Immunology, and
| |
Collapse
|
9
|
Macedo C, Hadi K, Walters J, Elinoff B, Marrari M, Zeevi A, Ramaswami B, Chalasani G, Landsittel D, Shields A, Alloway R, Lakkis FG, Woodle ES, Metes D. Impact of Induction Therapy on Circulating T Follicular Helper Cells and Subsequent Donor-Specific Antibody Formation After Kidney Transplant. Kidney Int Rep 2018; 4:455-469. [PMID: 30899873 PMCID: PMC6409398 DOI: 10.1016/j.ekir.2018.11.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/24/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023] Open
Abstract
Introduction The cellular events that contribute to generation of donor-specific anti-HLA antibodies (DSA) post-kidney transplantation (KTx) are not well understood. Characterization of such mechanisms could allow tailoring of immunosuppression to benefit sensitized patients. Methods We prospectively monitored circulating T follicular helper (cTFH) cells in KTx recipients who received T-cell depleting (thymoglobulin, n = 54) or T-cell nondepleting (basiliximab, n = 20) induction therapy from pre-KTx to 1 year post-KTx and assessed their phenotypic changes due to induction and DSA occurrence, in addition to healthy controls (n = 13), for a total of 307 blood samples. Results Before KTx, patients displayed comparable levels of resting, central memory cTFH cells with similar polarization to those of healthy controls. Unlike basiliximab induction, thymoglobulin induction significantly depleted cTFH cells, triggered lymphopenia-induced proliferation that skewed cTFH cells toward increased Th1 polarization, effector memory, and elevated programmed cell death protein 1 (PD-1)int/hi expression, resembling activated phenotypes. Regardless of induction, patients who developed DSA post-KTx, harbored pre-KTx donor-reactive memory interleukin (IL)-21+ cTFH cells and showed higher % cTFH and lower % of T regulatory (TREG) cells post-KTx resulting in elevated cTFH:TREG ratio at DSA occurrence. Conclusion Induction therapy distinctly shapes cTFH cell phenotype post-KTx. Monitoring cTFH cells before and after KTx may help detect those patients prone to DSA generation post-KTx.
Collapse
Affiliation(s)
- Camila Macedo
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin Hadi
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John Walters
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Beth Elinoff
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marilyn Marrari
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adriana Zeevi
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bala Ramaswami
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Geetha Chalasani
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Douglas Landsittel
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adele Shields
- Division of Transplantation, University of Cincinnati, Cincinnati, Ohio, USA
| | - Rita Alloway
- Division of Nephrology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Fadi G Lakkis
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - E Steve Woodle
- Division of Transplantation, University of Cincinnati, Cincinnati, Ohio, USA
| | - Diana Metes
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Wing JB, Tekgüç M, Sakaguchi S. Control of Germinal Center Responses by T-Follicular Regulatory Cells. Front Immunol 2018; 9:1910. [PMID: 30197643 PMCID: PMC6117393 DOI: 10.3389/fimmu.2018.01910] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/02/2018] [Indexed: 01/07/2023] Open
Abstract
Regulatory T-cells (Treg cells), expressing the transcription factor Foxp3, have an essential role in the control of immune homeostasis. In order to control diverse types of immune responses Treg cells must themselves show functional heterogeneity to control different types of immune responses. Recent advances have made it clear that Treg cells are able to mirror the homing capabilities of known T-helper subtypes such as Th1, Th2, Th17, and T-follicular helper cells (Tfh), allowing them to travel to the sites of inflammation and deliver suppression in situ. One of the more recent discoveries in this category is the description of T-follicular regulatory (Tfr) cells, a specialized subset of Treg cells that control Tfh and resulting antibody responses. In this review we will discuss recent advances in our understanding of Tfr biology and the role of both Tfr and activated extra-follicular Tregs (eTreg) in the control of humoral immunity.
Collapse
Affiliation(s)
- James B Wing
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Murat Tekgüç
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
11
|
Wang X, Dong Q, Li Q, Li Y, Zhao D, Sun J, Fu J, Meng F, Lin H, Luan J, Liu B, Wang M, Wang FS, He F, Tang L. Dysregulated Response of Follicular Helper T Cells to Hepatitis B Surface Antigen Promotes HBV Persistence in Mice and Associates With Outcomes of Patients. Gastroenterology 2018; 154:2222-2236. [PMID: 29544722 DOI: 10.1053/j.gastro.2018.03.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Production of neutralizing antibodies against hepatitis B surface antigen (HBsAg) is dysregulated in patients with persistent hepatitis B virus (HBV) infection. We investigated mechanisms by which this immune response to the virus is disrupted and whether it can be restored to promote clearance of HBV. METHODS Immune-competent C57BL/6N and C57BL/6J, as well as mice deficient in follicular helper T cells (Tfh-cell-deficient), B cells, or Foxp3+ T-regulatory cells (Treg cell deficient), were given hydrodynamic injections of pAAV/HBV1.2 plasmids. Some mice were given injections of sorted Tfh cells, pan-B cells, Treg cells, or a blocking antibody against CTLA4. Production of antibodies against HBsAg and clearance of HBV were assessed by flow cytometry, enzyme-linked immunosorbent assay, polymerase chain reaction, and immunohistochemical analyses. We obtained blood samples from patients with HBV infection and isolated Treg cells. We measured the ability of Treg cells to suppress production of interleukin 21 (IL21) in CD4+ T cells. RESULTS Immune-competent C57BL/6N and C57BL/6J mice transfected with the plasmid encoding HBV had features of viral clearance and viral persistence observed in humans. A Tfh-cell response to HBsAg was required for clearance of HBV and was suppressed by Treg cells in mice with persistent HBV infection. Depletion of Treg cells or inhibition of Treg-cell function (with blocking antibody against CTLA4) restored the Tfh-cell response against HBsAg and clearance of HBV in mice. Impaired Tfh-cell response to HBsAg was observed in blood from patients with chronic HBV infection, responsiveness was restored by depletion of Treg cells or blocking antibody against CTLA4. CONCLUSIONS In studies of HBV-infected mice and blood from patients with chronic HBV infection, we found a Tfh-cell response to HBsAg of to be required for HBV clearance, and that this response was blocked by Treg cells. Inhibiting Treg-cell activity using neutralizing antibody against CTLA4 restored the ability of Tfh cells to clear HBV infection; this approach might be developed for treatment of patients with chronic HBV infection.
Collapse
Affiliation(s)
- Xiaowen Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, P.R. China; School of Life Sciences, Peking University, Beijing, P.R. China
| | - Qingyang Dong
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, P.R. China
| | - Qian Li
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui Province, P.R. China; Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Yuanyuan Li
- Center of Infectious Disease, Beijing 302 Hospital, Beijing, P.R. China
| | - Dianyuan Zhao
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, P.R. China
| | - Jinjie Sun
- Air Force General Hospital, PLA, Beijing, P.R. China
| | - Junliang Fu
- Center of Infectious Disease, Beijing 302 Hospital, Beijing, P.R. China
| | - Fanping Meng
- Center of Infectious Disease, Beijing 302 Hospital, Beijing, P.R. China
| | - Hu Lin
- Center of Infectious Disease, Beijing 302 Hospital, Beijing, P.R. China
| | - Junjie Luan
- Center of Infectious Disease, Beijing 302 Hospital, Beijing, P.R. China
| | - Biao Liu
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, P.R. China
| | - Min Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, P.R. China
| | - Fu-Sheng Wang
- Center of Infectious Disease, Beijing 302 Hospital, Beijing, P.R. China
| | - Fuchu He
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, P.R. China; School of Life Sciences, Peking University, Beijing, P.R. China; Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China.
| | - Li Tang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, P.R. China; Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui Province, P.R. China.
| |
Collapse
|
12
|
Ellis JS, Braley-Mullen H. Mechanisms by Which B Cells and Regulatory T Cells Influence Development of Murine Organ-Specific Autoimmune Diseases. J Clin Med 2017; 6:jcm6020013. [PMID: 28134752 PMCID: PMC5332917 DOI: 10.3390/jcm6020013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/21/2016] [Accepted: 01/18/2017] [Indexed: 12/25/2022] Open
Abstract
Experiments with B cell-deficient (B−/−) mice indicate that a number of autoimmune diseases require B cells in addition to T cells for their development. Using B−/− Non-obese diabetic (NOD) and NOD.H-2h4 mice, we demonstrated that development of spontaneous autoimmune thyroiditis (SAT), Sjogren’s syndrome and diabetes do not develop in B−/− mice, whereas all three diseases develop in B cell-positive wild-type (WT) mice. B cells are required early in life, since reconstitution of adult mice with B cells or autoantibodies did not restore their ability to develop disease. B cells function as important antigen presenting cells (APC) to initiate activation of autoreactive CD4+ effector T cells. If B cells are absent or greatly reduced in number, other APC will present the antigen, such that Treg are preferentially activated and effector T cells are not activated. In these situations, B−/− or B cell-depleted mice develop the autoimmune disease when T regulatory cells (Treg) are transiently depleted. This review focuses on how B cells influence Treg activation and function, and briefly considers factors that influence the effectiveness of B cell depletion for treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Jason S Ellis
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA.
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA.
| | - Helen Braley-Mullen
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA.
- Department of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
13
|
Characterization of the Microenvironment of Nodular Lymphocyte Predominant Hodgkin Lymphoma. Int J Mol Sci 2016; 17:ijms17122127. [PMID: 27999289 PMCID: PMC5187927 DOI: 10.3390/ijms17122127] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 12/22/2022] Open
Abstract
Nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) is characterized by a low percentage of neoplastic lymphocyte predominant (LP) cells in a background of lymphocytes. The goal of this study is to characterize the microenvironment in NLPHL. Ten NLPHL cases and seven reactive lymph nodes (RLN) were analyzed by flow cytometry for the main immune cells and multiple specific subpopulations. To discriminate between cells in or outside the tumor cell area, we used CD26. We observed significantly lower levels of CD20+ B-cells and CD56+ NK cells and higher levels of CD4+ T-cells in NLPHL in comparison to RLN. In the subpopulations, we observed increased numbers of PD-1+CD4+ T follicular helper cells (TFH), CD69+CD4+ and CD69+CD8+ T-cells and CCR7-CD45RA-CD4+ effector memory T-cells, while FoxP3+CD4+ T regulatory cells (Tregs) and CCR7-CD45RA+ terminally differentiated CD4+ T-cells were decreased in NLPHL compared to RLN. CD69+ cells were increased in the tumor cell area in CD4+ and CD8+ T-cells, while FoxP3+CD25+CD4+ Tregs and CD25+CD8+ T-cells were significantly increased outside the tumor area. Thus, we show a markedly altered microenvironment in NLPHL, with lower numbers of NK cells and Tregs. PD-1+CD4+ and CD69+ T-cells were located inside, and Tregs and CD25+CD8+ cells outside the tumor cell area.
Collapse
|
14
|
T-helper signals restore B-cell receptor signaling in autoreactive anergic B cells by upregulating CD45 phosphatase activity. J Allergy Clin Immunol 2016; 138:839-851.e8. [DOI: 10.1016/j.jaci.2016.01.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 12/08/2015] [Accepted: 01/29/2016] [Indexed: 12/25/2022]
|
15
|
Lefebvre JS, Masters AR, Hopkins JW, Haynes L. Age-related impairment of humoral response to influenza is associated with changes in antigen specific T follicular helper cell responses. Sci Rep 2016; 6:25051. [PMID: 27109638 PMCID: PMC4842996 DOI: 10.1038/srep25051] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 04/08/2016] [Indexed: 12/17/2022] Open
Abstract
T follicular helper (TFH) cell responses are essential for generation of protective humoral immunity during influenza infection. Aging has a profound impact on CD4+ T cell function and humoral immunity, yet the impact of aging on antigen specific TFH responses remains unclear. Influenza specific TFH cells are generated in similar numbers in young and aged animals during infection, but TFH cells from aged mice exhibit significant differences, including reduced expression of ICOS and elevated production of IL-10 and IFNγ, which potentially impairs interaction with cognate B cells. Also, more influenza specific T cells in aged mice have a regulatory phenotype, which could contribute to the impaired TFH function. Adoptive transfer studies with young T cells demonstrated that TGF-β1 in the aged environment can drive increased regulatory T cell accumulation. Aging and the aged environment thus impact antigen specific TFH cell function and formation, which contribute to reduced protective humoral responses.
Collapse
Affiliation(s)
- Julie S Lefebvre
- Trudeau Institute, 154 Algonquin Ave, Saranac Lake, NY, 12983, USA
| | - April R Masters
- University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Jacob W Hopkins
- University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Laura Haynes
- Trudeau Institute, 154 Algonquin Ave, Saranac Lake, NY, 12983, USA
| |
Collapse
|
16
|
Dhaeze T, Stinissen P, Liston A, Hellings N. Humoral autoimmunity: a failure of regulatory T cells? Autoimmun Rev 2015; 14:735-41. [PMID: 25913138 DOI: 10.1016/j.autrev.2015.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 04/12/2015] [Indexed: 01/28/2023]
Abstract
Regulatory T cells (Tregs) are essential in maintaining tolerance to self. Several lines of evidence indicate that Tregs are functionally impaired in a variety of autoimmune diseases, leading to inefficient regulation of autoimmune T cells. Recent findings also suggest that Tregs are essential in controlling autoreactive B cells. The recently identified follicular regulatory T cell subset (TFR) is thought to regulate the production of autoantibodies in the germinal center (GC) response. Here we provide an update on the role of Tregs in controlling the GC response, and whether defective control over B cell tolerance contributes to autoimmunity.
Collapse
Affiliation(s)
- Tessa Dhaeze
- Hasselt University, Biomedisch Onderzoeksinstituut and Transnationale Universiteit Limburg, School of Life Sciences, Diepenbeek, Belgium
| | - Piet Stinissen
- Hasselt University, Biomedisch Onderzoeksinstituut and Transnationale Universiteit Limburg, School of Life Sciences, Diepenbeek, Belgium
| | - Adrian Liston
- Autoimmune Genetics Laboratory, VIB, Leuven, Belgium; Department of Microbiology and Immunology, University of Leuven, Belgium
| | - Niels Hellings
- Hasselt University, Biomedisch Onderzoeksinstituut and Transnationale Universiteit Limburg, School of Life Sciences, Diepenbeek, Belgium.
| |
Collapse
|
17
|
Hadaschik EN, Wei X, Leiss H, Heckmann B, Niederreiter B, Steiner G, Ulrich W, Enk AH, Smolen JS, Stummvoll GH. Regulatory T cell-deficient scurfy mice develop systemic autoimmune features resembling lupus-like disease. Arthritis Res Ther 2015; 17:35. [PMID: 25890083 PMCID: PMC4391674 DOI: 10.1186/s13075-015-0538-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/23/2015] [Indexed: 01/18/2023] Open
Abstract
Introduction Scurfy mice are deficient in regulatory T cells (Tregs), develop a severe, generalized autoimmune disorder that can affect almost every organ and die at an early age. Some of these manifestations resemble those found in systemic lupus erythematosus (SLE). In addition, active SLE is associated with low Treg numbers and reduced Treg function, but direct evidence for a central role of Treg malfunction in the pathophysiology of lupus-like manifestations is still missing. In the present study, we characterize the multiorgan pathology, autoantibody profile and blood count abnormalities in scurfy mice and show their close resemblances to lupus-like disease. Methods Scurfy mice have dysfunctional Tregs due to a genetic defect in the transcription factor Forkhead box protein 3 (Foxp3). We analyzed skin, joints, lung and kidneys of scurfy mice and wild-type (WT) controls by conventional histology and immunofluorescence (IF) performed hematological workups and tested for autoantibodies by IF, immunoblotting and enzyme-linked immunosorbent assay. We also analyzed the intestines, liver, spleen and heart, but did not analyze all organs known to be affected in scurfy mice (such as the testicle, the accessory reproductive structures, the pancreas or the eyes). We transferred CD4+ T cells of scurfy or WT mice into T cell-deficient B6/nude mice. Results We confirm previous reports that scurfy mice spontaneously develop severe pneumonitis and hematological abnormalities similar to those in SLE. We show that scurfy mice (but not controls) exhibited additional features of SLE: severe interface dermatitis, arthritis, mesangioproliferative glomerulonephritis and high titers of anti-nuclear antibodies, anti-double-stranded DNA antibodies, anti-histone antibodies and anti-Smith antibodies. Transfer of scurfy CD4+ T cells (but not of WT cells) induced autoantibodies and inflammation of lung, skin and kidneys in T cell-deficient B6/nude mice. Conclusion Our observations support the hypothesis that lupus-like autoimmune features develop in the absence of functional Tregs.
Collapse
Affiliation(s)
- Eva N Hadaschik
- Department of Dermatology, University of Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany.
| | - Xiaoying Wei
- Department of Dermatology, University of Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany. .,Department of Pathology, Affiliated Zhong-Da Hospital, Southeast University, 87 Dingjia Bridge, Gulou, 210009, Nanjing, China.
| | - Harald Leiss
- Department of Rheumatology, Medical University of Vienna, Wahringer Gurtel 18-20, 1090, Vienna, Austria.
| | - Britta Heckmann
- Department of Dermatology, University of Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany.
| | - Birgit Niederreiter
- Department of Rheumatology, Medical University of Vienna, Wahringer Gurtel 18-20, 1090, Vienna, Austria.
| | - Günter Steiner
- Department of Rheumatology, Medical University of Vienna, Wahringer Gurtel 18-20, 1090, Vienna, Austria.
| | - Walter Ulrich
- Department of Pathology, Hietzing Hospital, Wolkersbergenstrasse 1, 1130, Vienna, Austria.
| | - Alexander H Enk
- Department of Dermatology, University of Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany.
| | - Josef S Smolen
- Department of Rheumatology, Medical University of Vienna, Wahringer Gurtel 18-20, 1090, Vienna, Austria.
| | - Georg H Stummvoll
- Department of Rheumatology, Medical University of Vienna, Wahringer Gurtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
18
|
Abstract
T(reg) cells are essential for the maintenance of immune homeostasis and prevention of autoimmunity. In humoral immune responses, loss of T(reg) cell function causes increased levels of serum autoantibodies, hyper-IgE, spontaneous generation of germinal centres, and enhanced numbers of specialised T follicular helper cells (T(fh) cells) controlled by the lineage-defining transcription factor BCL-6 (B-cell lymphoma 6). Recent studies have demonstrated that a subset of T(reg) cells [T follicular regulatory (T(freg)) cells] are able to co-opt the follicular T-cell program by gaining expression of BCL-6 and travelling to the follicle where they have an important role in the control of expansion of T(fh) cells and the germinal centre reaction. However, the mechanisms by which they exert this control are still under investigation. In this review, we discuss the effects of T(reg) cells on humoral immunity and the mechanisms by which they exert their regulatory function.
Collapse
Affiliation(s)
- James B Wing
- Department of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita 565-0871, Osaka, Japan
| | | |
Collapse
|
19
|
Rapetti L, Chavele KM, Evans CM, Ehrenstein MR. B cell resistance to Fas-mediated apoptosis contributes to their ineffective control by regulatory T cells in rheumatoid arthritis. Ann Rheum Dis 2013; 74:294-302. [PMID: 24249811 DOI: 10.1136/annrheumdis-2013-204049] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate whether regulatory T cells (Treg) can control B cell function in rheumatoid arthritis (RA) and if not to explore the basis for this defect. METHODS Suppression of B cell responses by Treg was analysed in vitro by flow cytometry and ELISA using peripheral blood mononuclear cells from 65 patients with RA and 41 sex-matched and aged-matched healthy volunteers. Blocking and agonistic antibodies were used to define the role of Fas-mediated apoptosis in B cell regulation. RESULTS Treg failed to restrain B cell activation, proinflammatory cytokine and antibody production in the presence of responder T cells in RA patients. This lack of suppression was not only caused by impaired Treg function but was also due to B cell resistance to regulation. In healthy donors, control by Treg was associated with increased B cell death and relied upon Fas-mediated apoptosis. In contrast, RA B cells had reduced Fas expression compared with their healthy counterparts and were resistant to Fas-mediated apoptosis. CONCLUSIONS These studies demonstrate that Treg are unable to limit B cell responses in RA. This appears to be primarily due to B cell resistance to suppression, but Treg defects also contribute to this failure of regulation. Our data identify the Fas pathway as a novel target for Treg-mediated suppression of B cells and highlight a potential therapeutic approach to restore control of B cells by Treg in RA patients.
Collapse
Affiliation(s)
- Laetitia Rapetti
- Division of Medicine, Centre for Rheumatology, University College London, London, UK
| | | | - Catherine M Evans
- Division of Medicine, Centre for Rheumatology, University College London, London, UK
| | - Michael R Ehrenstein
- Division of Medicine, Centre for Rheumatology, University College London, London, UK
| |
Collapse
|
20
|
Banczyk D, Kalies K, Nachbar L, Bergmann L, Schmidt P, Bode U, Teegen B, Steven P, Lange T, Textor J, Ludwig RJ, Stöcker W, König P, Bell E, Westermann J. Activated CD4+ T cells enter the splenic T-cell zone and induce autoantibody-producing germinal centers through bystander activation. Eur J Immunol 2013; 44:93-102. [PMID: 24114675 PMCID: PMC4209793 DOI: 10.1002/eji.201343811] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 08/14/2013] [Accepted: 09/18/2013] [Indexed: 11/10/2022]
Abstract
CD4(+) T (helper) cells migrate in huge numbers through lymphoid organs. However, little is known about traffic routes and kinetics of CD4(+) T-cell subsets within different organ compartments. Such information is important because there are indications that CD4(+) T cells may influence the function of microenvironments depending on their developmental stage. Therefore, we investigated the migration of resting (naïve), activated, and recently activated (memory) CD4(+) T cells through the different compartments of the spleen. Resting and recently activated CD4(+) T cells were separated from thoracic duct lymph and activated CD4(+) T cells were generated in vitro by cross-linking the T-cell receptor and CD28. The present study shows that all three CD4(+) T-cell subsets selectively accumulate in the T-cell zone of the spleen. However, only activated T cells induce the formation of germinal centers (GCs) and autoantibodies in rats and mice. Our results suggest that in a two-step process they first activate B cells independent of the T-cell receptor repertoire and CD40 ligand (CD154) expression. The activated B cells then form GCs whereby CD154-dependent T-cell help is needed. Thus, activated T cells may contribute to the development of autoimmune diseases by activating autoreactive B cells in an Ag-independent manner.
Collapse
Affiliation(s)
- David Banczyk
- Center for Structural and Cell Biology in Medicine, Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Schmitt EG, Haribhai D, Jeschke JC, Co DO, Ziegelbauer J, Yan K, Iwakura Y, Mishra MK, Simpson P, Salzman NH, Williams CB. Chronic follicular bronchiolitis requires antigen-specific regulatory T cell control to prevent fatal disease progression. THE JOURNAL OF IMMUNOLOGY 2013; 191:5460-76. [PMID: 24163409 DOI: 10.4049/jimmunol.1301576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
To study regulatory T (Treg) cell control of chronic autoimmunity in a lymphoreplete host, we created and characterized a new model of autoimmune lung inflammation that targets the medium and small airways. We generated transgenic mice that express a chimeric membrane protein consisting of hen egg lysozyme and a hemoglobin epitope tag under the control of the Clara cell secretory protein promoter, which largely limited transgene expression to the respiratory bronchioles. When Clara cell secretory protein-membrane hen egg lysozyme/hemoglobin transgenic mice were crossed to N3.L2 TCR transgenic mice that recognize the hemoglobin epitope, the bigenic progeny developed dense, pseudo-follicular lymphocytic peribronchiolar infiltrates that resembled the histological pattern of follicular bronchiolitis. Aggregates of activated IFN-γ- and IL-17A-secreting CD4(+) T cells as well as B cells surrounded the airways. Lung pathology was similar in Ifng(-/-) and Il17a(-/-) mice, indicating that either cytokine is sufficient to establish chronic disease. A large number of Ag-specific Treg cells accumulated in the lesions, and Treg cell depletion in the affected mice led to an interstitial spread of the disease that ultimately proved fatal. Thus, Treg cells act to restrain autoimmune responses, resulting in an organized and controlled chronic pathological process rather than a progressive disease.
Collapse
Affiliation(s)
- Erica G Schmitt
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Gauld SB, De Santis JL, Kulinski JM, McGraw JA, Leonardo SM, Ruder EA, Maier W, Tarakanova VL. Modulation of B-cell tolerance by murine gammaherpesvirus 68 infection: requirement for Orf73 viral gene expression and follicular helper T cells. Immunology 2013; 139:197-204. [PMID: 23311955 DOI: 10.1111/imm.12069] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 12/22/2022] Open
Abstract
Viruses such as Epstein-Barr virus (EBV) have been linked to mechanisms that support autoantibody production in diseases such as systemic lupus erythematosus. However, the mechanisms by which viruses contribute to autoantibody production remain poorly defined. This stems in part, from the high level of seropositivity for EBV (> 95%) and the exquisite species specificity of EBV. In this study we overcame these problems by using murine gammaherpesvirus 68 (MHV68), a virus genetically and biologically related to EBV. We first showed that MHV68 drives autoantibody production by promoting a loss of B-cell anergy. We next showed that MHV68 infection resulted in the expansion of follicular helper T (Tfh) cells in vivo, and that these Tfh cells supported autoantibody production and a loss of B-cell anergy. Finally, we showed that the expansion of Tfh cells and autoantibody production was dependent on the establishment of viral latency and expression of a functional viral gene called Orf73. Collectively, our studies highlighted an unexpected role for viral latency in the development of autoantibodies following MHV68 infection and suggest that virus-induced expansion of Tfh cells probably plays a key role in the loss of B-cell anergy.
Collapse
Affiliation(s)
- Stephen B Gauld
- Medical College of Wisconsin, Division of Allergy/Immunology, Department of Pediatrics, The Children's Research Institute, Milwaukee, WI, USA.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Ellis JS, Wan X, Braley-Mullen H. Transient depletion of CD4+ CD25+ regulatory T cells results in multiple autoimmune diseases in wild-type and B-cell-deficient NOD mice. Immunology 2013; 139:179-86. [PMID: 23293979 DOI: 10.1111/imm.12065] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/03/2013] [Accepted: 01/03/2013] [Indexed: 01/18/2023] Open
Abstract
Approximately 80% of female wild-type non-obese diabetic (WT NOD) mice spontaneously develop diabetes, whereas B-cell-deficient (B(-/-)) NOD mice are resistant to diabetes. B(-/-) mice are also resistant to other spontaneous and experimentally induced autoimmune diseases, including arthritis, systemic lupus erythematosus, Sjögren syndrome and thyroiditis. Under normal conditions, activation of self-reactive T cells in the periphery is limited by CD4(+) CD25(+) natural regulatory T (Treg) cells. B(-/-) NOD.H-2h4 mice, normally resistant to spontaneous autoimmune thyroiditis (SAT), develop SAT when Treg cells are depleted, suggesting that Treg cells are preferentially activated when autoantigen is initially presented by non-B-cell antigen-presenting cells. To test the hypothesis that increased Treg cell activity in B(-/-) mice contributes to their resistance to other autoimmune diseases, WT and B(-/-) NOD mice were given anti-CD25 to transiently deplete CD4(+) CD25(+) Treg cells. The WT and B(-/-) NOD mice given anti-CD25 developed diabetes much earlier than WT mice given rat IgG, whereas rat IgG-treated B(-/-) mice did not develop diabetes. Treg-cell-depleted mice had increased lymphocyte infiltration of the pancreas, salivary glands and thyroid compared with controls given rat IgG. These results are consistent with the hypothesis that resistance of B-cell-deficient NOD mice to several autoimmune diseases is due to the activity of Treg cells.
Collapse
Affiliation(s)
- Jason S Ellis
- Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | | | | |
Collapse
|