1
|
Apley KD, Bass LE, King J, Downes G, Wang K, Forchetti MV, Moore DJ, Kendall P, Bonami RH, Berkland CJ. Evaluation of proinsulin(F25D) as a targeting ligand for insulin-binding B cells in autoimmune diabetes. Drug Deliv Transl Res 2025:10.1007/s13346-025-01869-x. [PMID: 40402465 DOI: 10.1007/s13346-025-01869-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2025] [Indexed: 05/23/2025]
Abstract
Insulin-binding B cells are implicated in Type 1 Diabetes (T1D) pathology. Antigen-specific immunotherapy (ASIT) holds promise in T1D. However, ASIT-targeted suppression of insulin-binding B cells is hampered by insulin's hormonal activity and the resulting binding and endocytosis of insulin by insulin receptors (INSR). To evaluate ASIT strategies that target insulin-binding B cells in vivo, non-hormonally active insulin variants are needed. In this work, we aimed to improve upon prior non-hormonal insulin variants by making mutations to the insulin precursor, proinsulin, and including a c-terminal sortase (SrtA) tag (LPETGGHG) to enable facile site-selective bioconjugation to scaffolds or payloads. Of the insulin variants investigated that retained low-nM binding to the murine-derived insulin autoantibody mAb 125, proinsulin(F25D)-SrtA had the lowest INSR binding and activity and the greatest fibrillation resistance. Compared to desoctapeptide insulin, a previously proposed non-hormonal insulin variant, proinsulin(F25D)-SrtA demonstrated 50-fold lower INSR binding and 100-fold greater fibrillation lag time. However, insulin(F25D)-SrtA bound to the anti-insulin antibody 12M4 isolated from a presymptomatic T1D individual, whereas proinsulin(F25D)-SrtA and desoctapeptide insulin did not, highlighting the potential for anti-insulin B cells to develop in human T1D that would escape this ASIT moiety. The characteristics of proinsulin(F25D)-SrtA make it a well-suited non-hormonal insulin variant for insulin-binding B cell targeting and warrants additional study with other anti-insulin B cell specificities derived from T1D individuals.
Collapse
Affiliation(s)
- Kyle D Apley
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, 63130, USA
| | - Lindsay E Bass
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jaylyn King
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Grant Downes
- Bioengineering Program, University of Kansas, Lawrence, KS, 66045, USA
| | - Kristen Wang
- Department of Chemistry, Washington University, St. Louis, MO, 63130, USA
| | - Mason V Forchetti
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Daniel J Moore
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pediatrics, Division of Endocrinology & Diabetes, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Peggy Kendall
- Department of Medicine, Division of Allergy and Immunology, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
| | - Rachel H Bonami
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Cory J Berkland
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, 63130, USA.
- Department of Chemistry, Washington University, St. Louis, MO, 63130, USA.
| |
Collapse
|
2
|
Wang YN, Li R, Huang Y, Chen H, Nie H, Liu L, Zou X, Zhong J, Zheng B, Gong Q. The role of B cells in the pathogenesis of type 1 diabetes. Front Immunol 2024; 15:1450366. [PMID: 39776900 PMCID: PMC11703732 DOI: 10.3389/fimmu.2024.1450366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Type 1 diabetes (T1D) is a metabolic disorder caused by a complete lack of insulin, primarily manifested by hyperglycemia. The mechanisms underlying the onset of T1D are complex, involving genetics, environment, and various unknown factors, leading to the infiltration of various immune components into the islets. Besides T cells, B cells are now considered important contributors to the pathogenesis of T1D, according to recent studies. In non-obese diabetic (NOD) mice, the absence of B cells prevents the development of T1D, and B-cell depletion can even restore the function of pancreatic β cells, emphasizing their involvement in the development of T1D. Naturally, besides pathogenic B cells, regulatory B cells (Bregs) might have a protective function in T1D. This article examines the mechanisms behind B-cell tolerance and the defects in B-cell tolerance checkpoints in T1D. We explored possible functions of B cells in T1D, including the role of islet autoantibodies in T1D, T-B cell interactions, and the role of Bregs in the pathogenesis of T1D. We also summarized the advances of B cell-targeted therapy, exploring new methods for intervention and treatment of T1D.
Collapse
Affiliation(s)
- Ya-nan Wang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Ruihua Li
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Yaxuan Huang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hui Chen
- Department of Laboratory Medicine, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Lian Liu
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Xiaoting Zou
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
3
|
Serreze DV, Dwyer JR, Racine JJ. Advancing Animal Models of Human Type 1 Diabetes. Cold Spring Harb Perspect Med 2024; 14:a041587. [PMID: 38886067 PMCID: PMC11444302 DOI: 10.1101/cshperspect.a041587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Multiple rodent models have been developed to study the basis of type 1 diabetes (T1D). However, nonobese diabetic (NOD) mice and derivative strains still provide the gold standard for dissecting the basis of the autoimmune responses underlying T1D. Here, we review the developmental origins of NOD mice, and how they and derivative strains have been used over the past several decades to dissect the genetic and immunopathogenic basis of T1D. Also discussed are ways in which the immunopathogenic basis of T1D in NOD mice and humans are similar or differ. Additionally reviewed are efforts to "humanize" NOD mice and derivative strains to provide improved models to study autoimmune responses contributing to T1D in human patients.
Collapse
|
4
|
Hu H, Vomund AN, Peterson OJ, Srivastava N, Li T, Kain L, Beatty WL, Zhang B, Hsieh CS, Teyton L, Lichti CF, Unanue ER, Wan X. Crinophagic granules in pancreatic β cells contribute to mouse autoimmune diabetes by diversifying pathogenic epitope repertoire. Nat Commun 2024; 15:8318. [PMID: 39333495 PMCID: PMC11437215 DOI: 10.1038/s41467-024-52619-5] [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: 12/11/2023] [Accepted: 09/13/2024] [Indexed: 09/29/2024] Open
Abstract
Autoimmune attack toward pancreatic β cells causes permanent loss of glucose homeostasis in type 1 diabetes (T1D). Insulin secretory granules store and secrete insulin but are also thought to be tissue messengers for T1D. Here, we show that the crinophagic granules (crinosome), a minor set of vesicles formed by fusing lysosomes with the conventional insulin dense-core granules (DCG), are pathogenic in T1D development in mouse models. Pharmacological inhibition of crinosome formation in β cells delays T1D progression without affecting the dominant DCGs. Mechanistically, crinophagy inhibition diminishes the epitope repertoire in pancreatic islets, including cryptic, modified and disease-relevant epitopes derived from insulin. These unconventional insulin epitopes are largely undetectable in the MHC-II epitope repertoire of the thymus, where only canonical insulin epitopes are presented. CD4+ T cells targeting unconventional insulin epitopes display autoreactive phenotypes, unlike tolerized T cells recognizing epitopes presented in the thymus. Thus, the crinophagic pathway emerges as a tissue-intrinsic mechanism that transforms insulin from a signature thymic self-protein to a critical autoantigen by creating a peripheral-thymic mismatch in the epitope repertoire.
Collapse
Affiliation(s)
- Hao Hu
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anthony N Vomund
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Orion J Peterson
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Neetu Srivastava
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Tiandao Li
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lisa Kain
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Wandy L Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bo Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Chyi-Song Hsieh
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Emil R Unanue
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoxiao Wan
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
5
|
Bass LE, Bonami RH. Factors Governing B Cell Recognition of Autoantigen and Function in Type 1 Diabetes. Antibodies (Basel) 2024; 13:27. [PMID: 38651407 PMCID: PMC11036271 DOI: 10.3390/antib13020027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Islet autoantibodies predict type 1 diabetes (T1D) but can be transient in murine and human T1D and are not thought to be directly pathogenic. Rather, these autoantibodies signal B cell activity as antigen-presenting cells (APCs) that present islet autoantigen to diabetogenic T cells to promote T1D pathogenesis. Disrupting B cell APC function prevents T1D in mouse models and has shown promise in clinical trials. Autoantigen-specific B cells thus hold potential as sophisticated T1D biomarkers and therapeutic targets. B cell receptor (BCR) somatic hypermutation is a mechanism by which B cells increase affinity for islet autoantigen. High-affinity B and T cell responses are selected in protective immune responses, but immune tolerance mechanisms are known to censor highly autoreactive clones in autoimmunity, including T1D. Thus, different selection rules often apply to autoimmune disease settings (as opposed to protective host immunity), where different autoantigen affinity ceilings are tolerated based on variations in host genetics and environment. This review will explore what is currently known regarding B cell signaling, selection, and interaction with T cells to promote T1D pathogenesis.
Collapse
Affiliation(s)
- Lindsay E. Bass
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Rachel H. Bonami
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
6
|
Alleva DG, Delpero AR, Sathiyaseelan T, Murikipudi S, Lancaster TM, Atkinson MA, Wasserfall CH, Yu L, Ragupathy R, Bonami RH, Zion TC. An antigen-specific immunotherapeutic, AKS-107, deletes insulin-specific B cells and prevents murine autoimmune diabetes. Front Immunol 2024; 15:1367514. [PMID: 38515750 PMCID: PMC10954819 DOI: 10.3389/fimmu.2024.1367514] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Introduction The antigen-presenting cell function of insulin-reactive B cells promotes type 1 diabetes (T1D) in non-obese diabetic (NOD) mice by stimulating pathogenic T cells leading to destruction of insulin-producing β-cells of pancreatic islets. Methods/Results To target insulin-reactive B cells, AKS-107, a human IgG1 Fc molecule fused with human insulin A and B chains, was engineered to retain conformational insulin epitopes that bound mouse and human B cell receptors but prevented binding to the insulin metabolic receptor. AKS-107 Fc-mediated deletion of insulin-reactive B cells was demonstrated via ex vivo and in vivo experiments with insulin-reactive B cell receptor transgenic mouse strains, VH125Tg/NOD and Tg125(H+L)/NOD. As an additional immune tolerance feature, the Y16A mutation of the insulin B(9-23) dominant T cell epitope was engineered into AKS-107 to suppress activation of insulin-specific T cells. In mice and non-human primates, AKS-107 was well-tolerated, non-immunogenic, did not cause hypoglycemia even at high doses, and showed an expectedly protracted pharmacokinetic profile. AKS-107 reproducibly prevented spontaneous diabetes from developing in NOD and VH125Tg/NOD mice that persisted for months after cessation of treatment, demonstrating durable immune tolerance. Discussion These preclinical outcomes position AKS-107 for clinical development in T1D prevention settings.
Collapse
Affiliation(s)
- David G. Alleva
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | - Andrea R. Delpero
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | | | - Sylaja Murikipudi
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | - Thomas M. Lancaster
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | - Mark A. Atkinson
- Departments of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and Diabetes Institute, The University of Florida, Gainesville, FL, United States
| | - Clive H. Wasserfall
- Departments of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and Diabetes Institute, The University of Florida, Gainesville, FL, United States
| | - Liping Yu
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Ramya Ragupathy
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Todd C. Zion
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| |
Collapse
|
7
|
Lee I, Ling I, Verma S, Blanche W, Pham CTN, Kendall PL. The complement regulator CD55 modulates TLR9 signaling and supports survival in marginal zone B cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582833. [PMID: 38496511 PMCID: PMC10942343 DOI: 10.1101/2024.03.01.582833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Marginal zone (MZ) B cells bridge innate and adaptive immunity by sensing bloodborne antigens and producing rapid antibody and cytokine responses. CD55 is a membrane-bound complement regulator that interferes with complement activation, an important component of innate immunity. CD55 also regulates adaptive immunity-CD55 downregulation is critical for germinal center reactions. MZ B cells also express low CD55, but its role in MZ B cell function is unknown. Using germline knockout mice, we found that similar numbers of MZ B cells are initially established in 3-week-old CD55-deficient mice compared to wild-type (WT) mice. However, MZ B cells fail to accumulate as mice age and undergo increased apoptosis. Following ex vivo stimulation of MZ B cells through Toll-like receptor 9, we observed a proinflammatory phenotype with increased IL-6 expression. These findings demonstrate a critical role for CD55 in supporting MZ B cell survival while also regulating cellular function.
Collapse
|
8
|
McNitt DH, Joosse BA, Thomas JW, Bonami RH. Productive Germinal Center Responses Depend on the Nature of Stimuli Received by Anti-Insulin B Cells in Type 1 Diabetes-Prone Mice. Immunohorizons 2023; 7:384-397. [PMID: 37261716 PMCID: PMC10448785 DOI: 10.4049/immunohorizons.2300036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023] Open
Abstract
Islet autoantibodies, including those directed at insulin, predict type 1 diabetes (T1D) in mice and humans and signal immune tolerance breach by B lymphocytes. High-affinity insulin autoantibodies and T follicular helper cell involvement implicate germinal centers (GCs) in T1D. The VH125SD BCR transgenic model, in which 1-2% of peripheral B lymphocytes recognize insulin, enables direct study of insulin-binding B cells. Our prior studies showed that anti-insulin B cell receptor transgene site-directed to H chain locus mice fail to generate insulin Ab following T-dependent immunization, but it was unclear whether anti-insulin B cells were blocked for GC initiation, survival, or differentiation into Ab-secreting cells. Here, we show that insulin-binding B cells in T1D-prone anti-insulin B cell receptor transgene site-directed to H chain locus mice can spontaneously adopt a GC phenotype and undergo class switching to the IgG1 isotype, with little if any switching to IgG2b. T-dependent immunizations with insulin SRBC or insulin CFA drove anti-insulin B lymphocytes to adopt a GC phenotype, despite blunted insulin Ab production. Dual immunization against self (insulin) and foreign (4-hydroxy-3-nitrophenylacetyl hapten conjugated to keyhole limpet hemocyanin) Ags showed an anti-insulin (but not anti-4-hydroxy-3-nitrophenylacetyl) Ab block that tracked with increased expression of the apoptosis marker, activated caspase 3, in self-reactive GC B cells. Finally, T-independent immunization with insulin conjugated to Brucella abortus ring test Ag released immune tolerance to allow robust expansion of anti-insulin GC B cells and IgG-switched insulin Ab production. Overall, these data pinpoint GC survival and Ab-secreting cell differentiation as immune tolerance blocks that limit T-dependent, but not T-independent, stimulation of anti-insulin B cell responses.
Collapse
Affiliation(s)
- Dudley H. McNitt
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Bryan A. Joosse
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - James W. Thomas
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and
Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and
Immunology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
9
|
Wilfong EM, Vowell KN, Crofford LJ, Kendall PL. Multiparameter analysis of human B lymphocytes identifies heterogeneous CD19 + CD21 lo subsets. Cytometry A 2023; 103:283-294. [PMID: 36281747 PMCID: PMC10085822 DOI: 10.1002/cyto.a.24699] [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: 03/14/2022] [Revised: 07/21/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
Abstract
Autoreactive B cell subsets have been described in a variety of settings, using multiple classification schemes and cell surface markers also found on healthy cells. CD19+ CD21lo B cells have been identified as an autoreactive-prone subset of B cells, although the downregulation of CD21 has been observed on a variety of B cell subsets in health and disease. This variation has led to confusion regarding the meaning and applicability of the loss or reduction of CD21 in peripheral B cells. To better understand the relationships between commonly used B cell markers and their associated characteristics, we analyzed human B cells from healthy participants using multiparameter flow cytometry and the visualization algorithm, tSNE. This approach revealed significant phenotypic overlap amongst five previously described autoimmune-prone B cell subsets, including CD19+ CD10- CD27- CD21lo B cells. Interestingly, 12 different subpopulations of CD19+ CD21lo B cells were identified, some of which mapped to previously described autoreactive populations, while others were consistent with healthy B cells. This suggests that CD21 is downregulated in a variety of circumstances involving B cell activation, all of which are present in low numbers even in healthy individuals. These findings describe the utility of unbiased multiparameter analysis using a relatively limited panel of flow cytometry markers to analyze autoreactive-prone and normal activated B cells.
Collapse
Affiliation(s)
- Erin M. Wilfong
- Division of Allergy, Pulmonary and Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Katherine N. Vowell
- Division of Allergy, Pulmonary and Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Leslie J. Crofford
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Microbiology, Pathology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Peggy L. Kendall
- Division of Allergy, Pulmonary and Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Microbiology, Pathology and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Division of Allergy/Immunology, Department of Medicine, Washington University, St. Louis, MO
| |
Collapse
|
10
|
Harley ITW, Allison K, Scofield RH. Polygenic autoimmune disease risk alleles impacting B cell tolerance act in concert across shared molecular networks in mouse and in humans. Front Immunol 2022; 13:953439. [PMID: 36090990 PMCID: PMC9450536 DOI: 10.3389/fimmu.2022.953439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Most B cells produced in the bone marrow have some level of autoreactivity. Despite efforts of central tolerance to eliminate these cells, many escape to periphery, where in healthy individuals, they are rendered functionally non-responsive to restimulation through their antigen receptor via a process termed anergy. Broad repertoire autoreactivity may reflect the chances of generating autoreactivity by stochastic use of germline immunoglobulin gene segments or active mechanisms may select autoreactive cells during egress to the naïve peripheral B cell pool. Likewise, it is unclear why in some individuals autoreactive B cell clones become activated and drive pathophysiologic changes in autoimmune diseases. Both of these remain central questions in the study of the immune system(s). In most individuals, autoimmune diseases arise from complex interplay of genetic risk factors and environmental influences. Advances in genome sequencing and increased statistical power from large autoimmune disease cohorts has led to identification of more than 200 autoimmune disease risk loci. It has been observed that autoantibodies are detectable in the serum years to decades prior to the diagnosis of autoimmune disease. Thus, current models hold that genetic defects in the pathways that control autoreactive B cell tolerance set genetic liability thresholds across multiple autoimmune diseases. Despite the fact these seminal concepts were developed in animal (especially murine) models of autoimmune disease, some perceive a disconnect between human risk alleles and those identified in murine models of autoimmune disease. Here, we synthesize the current state of the art in our understanding of human risk alleles in two prototypical autoimmune diseases - systemic lupus erythematosus (SLE) and type 1 diabetes (T1D) along with spontaneous murine disease models. We compare these risk networks to those reported in murine models of these diseases, focusing on pathways relevant to anergy and central tolerance. We highlight some differences between murine and human environmental and genetic factors that may impact autoimmune disease development and expression and may, in turn, explain some of this discrepancy. Finally, we show that there is substantial overlap between the molecular networks that define these disease states across species. Our synthesis and analysis of the current state of the field are consistent with the idea that the same molecular networks are perturbed in murine and human autoimmune disease. Based on these analyses, we anticipate that murine autoimmune disease models will continue to yield novel insights into how best to diagnose, prognose, prevent and treat human autoimmune diseases.
Collapse
Affiliation(s)
- Isaac T. W. Harley
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
- Rheumatology Section, Medicine Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Kristen Allison
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
| | - R. Hal Scofield
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Medical/Research Service, US Department of Veterans Affairs Medical Center, Oklahoma City, OK, United States
| |
Collapse
|
11
|
Lombard-Vadnais F, Collin R, Daudelin JF, Chabot-Roy G, Labrecque N, Lesage S. The Idd2 Locus Confers Prominent Resistance to Autoimmune Diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:898-909. [PMID: 35039332 DOI: 10.4049/jimmunol.2100456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/02/2021] [Indexed: 06/14/2023]
Abstract
Type 1 diabetes is an autoimmune disease characterized by pancreatic β cell destruction. It is a complex genetic trait driven by >30 genetic loci with parallels between humans and mice. The NOD mouse spontaneously develops autoimmune diabetes and is widely used to identify insulin-dependent diabetes (Idd) genetic loci linked to diabetes susceptibility. Although many Idd loci have been extensively studied, the impact of the Idd2 locus on autoimmune diabetes susceptibility remains to be defined. To address this, we generated a NOD congenic mouse bearing B10 resistance alleles on chromosome 9 in a locus coinciding with part of the Idd2 locus and found that NOD.B10-Idd2 congenic mice are highly resistant to diabetes. Bone marrow chimera and adoptive transfer experiments showed that the B10 protective alleles provide resistance in an immune cell-intrinsic manner. Although no T cell-intrinsic differences between NOD and NOD.B10-Idd2 mice were observed, we found that the Idd2 resistance alleles limit the formation of spontaneous and induced germinal centers. Comparison of B cell and dendritic cell transcriptome profiles from NOD and NOD.B10-Idd2 mice reveal that resistance alleles at the Idd2 locus affect the expression of specific MHC molecules, a result confirmed by flow cytometry. Altogether, these data demonstrate that resistance alleles at the Idd2 locus impair germinal center formation and influence MHC expression, both of which likely contribute to reduced diabetes incidence.
Collapse
Affiliation(s)
- Félix Lombard-Vadnais
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Roxanne Collin
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and
| | - Jean-François Daudelin
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Geneviève Chabot-Roy
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Nathalie Labrecque
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and
- Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
| | - Sylvie Lesage
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada;
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and
| |
Collapse
|
12
|
Nyhoff LE, Griffith AS, Clark ES, Thomas JW, Khan WN, Kendall PL. Btk Supports Autoreactive B Cell Development and Protects against Apoptosis but Is Expendable for Antigen Presentation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2922-2932. [PMID: 34799428 PMCID: PMC9117567 DOI: 10.4049/jimmunol.2000558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/29/2021] [Indexed: 11/19/2022]
Abstract
Bruton's tyrosine kinase (Btk) propagates B cell signaling, and BTK inhibitors are in clinical trials for autoimmune disease. Although autoreactive B cells fail to develop in the absence of Btk, its role in mature cells is unknown. To address this issue, a model of conditional removal (Btk flox/Cre-ERT2 ) was used to excise Btk from mature transgenic B cells that recognize the pathophysiologic autoantigen insulin. Anti-insulin B cells escape central tolerance and promote autoimmune diabetes, mimicking human autoreactive cells. Lifelong Btk deficiency was previously shown to eliminate 95% of anti-insulin B cells, but in this model, mature anti-insulin B cells survived for weeks after targeted Btk deletion, even when competing with a polyclonal repertoire. BCR-stimulated cells could still signal via Syk, PLCy2, and CD22, but failed to upregulate the antiapoptotic protein Bcl-xL, and proliferation was impaired. Surprisingly, Btk-depleted anti-insulin B cells could still present Ag and activate T cells, a critical function in promoting T cell-mediated islet cell destruction. Thus, pharmacologic targeting of Btk may be most effective by blocking expansion of established autoreactive cells, and preventing emergence of new ones.
Collapse
Affiliation(s)
- Lindsay E Nyhoff
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
- Division of Allergy, Pulmonary and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Amber S Griffith
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Emily S Clark
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL; and
| | - James W Thomas
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
- Division of Rheumatology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Wasif N Khan
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL; and
| | - Peggy L Kendall
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN;
- Division of Allergy, Pulmonary and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
13
|
Joosse BA, Jackson JH, Cisneros A, Santhin AB, Smith SA, Moore DJ, Crofford LJ, Wilfong EM, Bonami RH. High-Throughput Detection of Autoantigen-Specific B Cells Among Distinct Functional Subsets in Autoimmune Donors. Front Immunol 2021; 12:685718. [PMID: 34234784 PMCID: PMC8256427 DOI: 10.3389/fimmu.2021.685718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
Antigen-specific B cells (ASBCs) can drive autoimmune disease by presenting autoantigen to cognate T cells to drive their activation, proliferation, and effector cell differentiation and/or by differentiating into autoantibody-secreting cells. Autoantibodies are frequently used to predict risk and diagnose several autoimmune diseases. ASBCs can drive type 1 diabetes even when immune tolerance mechanisms block their differentiation into antibody-secreting cells. Furthermore, anti-histidyl tRNA synthetase syndrome patients have expanded IgM+ Jo-1-binding B cells, which clinically diagnostic IgG Jo-1 autoantibodies may not fully reflect. Given the potential disconnect between the pathologic function of ASBCs and autoantibody secretion, direct study of ASBCs is a necessary step towards developing better therapies for autoimmune diseases, which often have no available cure. We therefore developed a high-throughput screening pipeline to 1) phenotypically identify specific B cell subsets, 2) expand them in vitro, 3) drive them to secrete BCRs as antibody, and 4) identify wells enriched for ASBCs through ELISA detection of antibody. We tested the capacity of several B cell subset(s) to differentiate into antibody-secreting cells following this robust stimulation. IgM+ and/or IgD+, CD27- memory, memory, switched memory, and BND B cells secreted B cell receptor (BCR) as antibody following in vitro stimulation, whereas few plasmablasts responded. Bimodal responses were observed across autoimmune donors for IgM+ CD21lo and IgM- CD21lo B cells, consistent with documented heterogeneity within the CD21lo subset. Using this approach, we detected insulin-binding B cell bias towards CD27- memory and CD27+ memory subsets in pre-symptomatic type 1 diabetes donors. We took advantage of routine detection of Jo-1-binding B cells in Jo-1+ anti-histidyl tRNA synthetase syndrome patients to show that Jo-1-binding B cells and total B cells expanded 20-30-fold using this culture system. Overall, these studies highlight technology that is amenable to small numbers of cryopreserved peripheral blood mononuclear cells that enables interrogation of phenotypic and repertoire attributes of ASBCs derived from autoimmune patients.
Collapse
Affiliation(s)
- Bryan A Joosse
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James H Jackson
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Biomedical Sciences, School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Alberto Cisneros
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Austin B Santhin
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Scott A Smith
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Nashville, TN, United States
| | - Daniel J Moore
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Nashville, TN, United States.,Department of Pediatrics, Division of Endocrinology & Diabetes, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Leslie J Crofford
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Nashville, TN, United States
| | - Erin M Wilfong
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Medicine, Allergy, Pulmonary, and Critical Care, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rachel H Bonami
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Nashville, TN, United States
| |
Collapse
|
14
|
Vomund AN, Lichti CF, Peterson OJ, Arbelaez AM, Wan X, Unanue ER. Blood leukocytes recapitulate diabetogenic peptide-MHC-II complexes displayed in the pancreatic islets. J Exp Med 2021; 218:211955. [PMID: 33822842 PMCID: PMC8034384 DOI: 10.1084/jem.20202530] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/29/2021] [Accepted: 03/09/2021] [Indexed: 12/28/2022] Open
Abstract
Assessing the self-peptides presented by susceptible major histocompatibility complex (MHC) molecules is crucial for evaluating the pathogenesis and therapeutics of tissue-specific autoimmune diseases. However, direct examination of such MHC-bound peptides displayed in the target organ remains largely impractical. Here, we demonstrate that the blood leukocytes from the nonobese diabetic (NOD) mice presented peptide epitopes to autoreactive CD4 T cells. These peptides were bound to the autoimmune class II MHC molecule (MHC-II) I-Ag7 and originated from insulin B-chain and C-peptide. The presentation required a glucose challenge, which stimulated the release of the insulin peptides from the pancreatic islets. The circulating leukocytes, especially the B cells, promptly captured and presented these peptides. Mass spectrometry analysis of the leukocyte MHC-II peptidome revealed a series of β cell–derived peptides, with identical sequences to those previously identified in the islet MHC-II peptidome. Thus, the blood leukocyte peptidome echoes that found in islets and serves to identify immunogenic peptides in an otherwise inaccessible tissue.
Collapse
Affiliation(s)
- Anthony N Vomund
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Cheryl F Lichti
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO.,Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| | - Orion J Peterson
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Ana Maria Arbelaez
- Division of Endocrinology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Xiaoxiao Wan
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO.,Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| | - Emil R Unanue
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO.,Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
15
|
Young-Glazer J, Cisneros A, Wilfong EM, Smith SA, Crofford LJ, Bonami RH. Jo-1 autoantigen-specific B cells are skewed towards distinct functional B cell subsets in anti-synthetase syndrome patients. Arthritis Res Ther 2021; 23:33. [PMID: 33468230 PMCID: PMC7814460 DOI: 10.1186/s13075-020-02412-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background Anti-Jo-1 autoantibodies which recognize histidyl-tRNA synthetase identify patients with the rare rheumatologic disease, anti-histidyl-tRNA synthetase syndrome (Jo-1 ARS), a phenotypically distinct subset of idiopathic inflammatory myopathies (IIM). Jo-1-binding B cells (JBCs) are implicated in disease pathogenesis, yet they have not been studied directly. We therefore aimed to characterize JBCs to better understand how they expand and function in Jo-1 ARS. Methods We enrolled 10 IIM patients diagnosed with Jo-1 ARS, 4 patients with non-Jo-1 IIM, and 8 age- and sex-matched healthy controls. We phenotypically characterized peripheral blood mononuclear cells (PBMCs) ex vivo using flow cytometry to define the B cell subsets in which JBCs reside. We further tested their ability to differentiate into antibody-secreting cells following stimulation in vitro. Results The majority of JBCs were IgM+ (not class-switched). Compared to non-JBCs in the same donors, JBCs contained a higher percentage of autoimmune-prone CD21lo cells and were increased in the CD21lo IgM+ IgD− CD27+ memory subset relative to healthy donor B cells. Whereas non-JBCs were present in the anergic BND B cell subset, JBCs were nearly absent from this compartment. JBCs were detected among plasmablasts in some donors, but a reduced frequency of JBCs differentiated into CD38hi24− plasmablasts compared to non-JBCs present in the same wells following in vitro stimulation. Conclusions JBCs are enriched for autoimmune-prone CD21lo B cells, some of which exhibit a memory phenotype in the peripheral repertoire of Jo-1 ARS patients. JBCs undergo limited class switch and show reduced capacity to differentiate into antibody-secreting cells. This suggests complex B cell biology exists beyond class-switched cells that differentiate to secrete anti-Jo-1 autoantibody (i.e., what is captured through serum autoantibody studies). New Jo-1 ARS therapies should thus ideally target non-class-switched JBCs in addition to those that have undergone IgG class-switching to most effectively block cross-talk with autoreactive T cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-020-02412-8.
Collapse
Affiliation(s)
- Jennifer Young-Glazer
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Medical Center North T3113, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | - Alberto Cisneros
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Medical Center North T3113, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | - Erin M Wilfong
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Medical Center North T3113, 1161 21st Avenue South, Nashville, TN, 37232, USA.,Department of Medicine, Division of Allergy, Pulmonary, and Critical Care, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Scott A Smith
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Leslie J Crofford
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Medical Center North T3113, 1161 21st Avenue South, Nashville, TN, 37232, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Rachel H Bonami
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Medical Center North T3113, 1161 21st Avenue South, Nashville, TN, 37232, USA. .,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
16
|
Felton JL, Conway H, Bonami RH. B Quiet: Autoantigen-Specific Strategies to Silence Raucous B Lymphocytes and Halt Cross-Talk with T Cells in Type 1 Diabetes. Biomedicines 2021; 9:biomedicines9010042. [PMID: 33418839 PMCID: PMC7824835 DOI: 10.3390/biomedicines9010042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 01/10/2023] Open
Abstract
Islet autoantibodies are the primary biomarkers used to predict type 1 diabetes (T1D) disease risk. They signal immune tolerance breach by islet autoantigen-specific B lymphocytes. T-B lymphocyte interactions that lead to expansion of pathogenic T cells underlie T1D development. Promising strategies to broadly prevent this T-B crosstalk include T cell elimination (anti-CD3, teplizumab), B cell elimination (anti-CD20, rituximab), and disruption of T cell costimulation/activation (CTLA-4/Fc fusion, abatacept). However, global disruption or depletion of immune cell subsets is associated with significant risk, particularly in children. Therefore, antigen-specific therapy is an area of active investigation for T1D prevention. We provide an overview of strategies to eliminate antigen-specific B lymphocytes as a means to limit pathogenic T cell expansion to prevent beta cell attack in T1D. Such approaches could be used to prevent T1D in at-risk individuals. Patients with established T1D would also benefit from such targeted therapies if endogenous beta cell function can be recovered or islet transplant becomes clinically feasible for T1D treatment.
Collapse
Affiliation(s)
- Jamie L. Felton
- Department of Pediatrics, Division of Pediatric Endocrinology and the Herman B. Wells Center for Pediatric Research, Indianapolis, IN 46202, USA; (J.L.F.); (H.C.)
| | - Holly Conway
- Department of Pediatrics, Division of Pediatric Endocrinology and the Herman B. Wells Center for Pediatric Research, Indianapolis, IN 46202, USA; (J.L.F.); (H.C.)
| | - Rachel H. Bonami
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
17
|
Xiao Y, Deng C, Zhou Z. The Multiple Roles of B Lymphocytes in the Onset and Treatment of Type 1 Diabetes: Interactions between B Lymphocytes and T Cells. J Diabetes Res 2021; 2021:6581213. [PMID: 34778464 PMCID: PMC8580688 DOI: 10.1155/2021/6581213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/21/2021] [Indexed: 01/10/2023] Open
Abstract
Although type 1 diabetes is thought to be an organ-specific autoimmune disease, mediated by effective CD4+ and CD8+ T cells, it has recently become clear that B cells participate in the initiation and progress of this disease. Indeed, B cell deletion can prevent or reverse autoimmune diabetes in nonobese diabetic mice and even result in partially remaining β cell function in patients with new-onset type 1 diabetes. This review summarizes the dual role of B cells in this process not only of pathogenic effect but also of immunoregulatory function in type 1 diabetes. We focus on the impact that B cells have on regulating the activation, proliferation, and cytokine production of self-reactive T cells along with regulatory T cells, with the aim of providing a better understanding of the interactions between T and B cells in immunopathogenesis and improving the efficacy of interventions for clinical practice.
Collapse
Affiliation(s)
- Yangfan Xiao
- Clinical Nursing Teaching and Research Section, Department of Anesthesiology, and Anesthesia Medical Research Center, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Chao Deng
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, and Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, and Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
18
|
Bonami RH, Nyhoff LE, McNitt DH, Hulbert C, Felton JL, Kendall PL, Thomas JW. T-B Lymphocyte Interactions Promote Type 1 Diabetes Independently of SLAM-Associated Protein. THE JOURNAL OF IMMUNOLOGY 2020; 205:3263-3276. [PMID: 33199538 DOI: 10.4049/jimmunol.1900464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/15/2020] [Indexed: 01/05/2023]
Abstract
Signaling lymphocytic activation molecule-associated protein (SAP), a critical intracellular signaling molecule for T-B lymphocyte interactions, drives T follicular helper (Tfh) cell development in germinal centers (GCs). High-affinity islet autoantibodies predict type 1 diabetes (T1D) but do not cause β cell destruction. This paradox intimates Tfh cells as key pathologic effectors, consistent with an observed Tfh signature in T1D. To understand how fully developed Tfh (GC Tfh) contribute to different autoimmune processes, we investigated the role of SAP in T1D and autoantibody-mediated arthritis. Whereas spontaneous arthritis depended on SAP in the autoantibody-mediated K/BxN model, organized insulitis and diabetes onset were unabated, despite a blocked anti-insulin vaccine response in SAP-deficient NOD mice. GC Tfh and GC B cell development were blocked by loss of SAP in K/BxN mice. In contrast, although GC B cell formation was markedly reduced in SAP-deficient NOD mice, T cells with a GC Tfh phenotype were found at disease sites. CXCR3+ CCR6- (Tfh1) subset bias was observed among GC Tfh cells infiltrating the pancreas of NOD mice, which was enhanced by loss of SAP NOD T cells override SAP requirement to undergo activation and proliferation in response to Ag presentation, demonstrating the potential for productive cognate T-B lymphocyte interactions in T1D-prone mice. We find that SAP is essential when autoantibody-driven immune complexes promote inflammation but is not required for effective organ-specific autoimmune attack. Thus, Tfh induced in classic GC reactions are dispensable for T1D, but the autoimmune process in the NOD model retains pathogenic Tfh without SAP.
Collapse
Affiliation(s)
- Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232; .,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Lindsay E Nyhoff
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232.,Division of Allergy, Pulmonary, and Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - Dudley H McNitt
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Chrys Hulbert
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jamie L Felton
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Peggy L Kendall
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232.,Division of Allergy, Pulmonary, and Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - James W Thomas
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232; .,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
19
|
Boldison J, Da Rosa LC, Buckingham L, Davies J, Wen L, Wong FS. Phenotypically distinct anti-insulin B cells repopulate pancreatic islets after anti-CD20 treatment in NOD mice. Diabetologia 2019; 62:2052-2065. [PMID: 31444529 PMCID: PMC6805803 DOI: 10.1007/s00125-019-04974-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/08/2019] [Indexed: 01/10/2023]
Abstract
AIMS/HYPOTHESIS Autoreactive B cells escape immune tolerance and contribute to the pathogenesis of type 1 diabetes. While global B cell depletion is a successful therapy for autoimmune disease, the fate of autoreactive cells during this treatment in autoimmune diabetes is unknown. We aimed to identify and track anti-insulin B cells in pancreatic islets and understand their repopulation after anti-CD20 treatment. METHODS We generated a double transgenic system, the VH125.hCD20/NOD mouse. The VH125 transgenic mouse, expressing an increased frequency of anti-insulin B cells, was crossed with a human CD20 (hCD20) transgenic mouse, to facilitate B cell depletion using anti-CD20. B cells were analysed using multiparameter and ImageStream flow cytometry. RESULTS We demonstrated that anti-insulin B cells were recruited to the pancreas during disease progression in VH125.hCD20/NOD mice. We identified two distinct populations of anti-insulin B cells in pancreatic islets, based on CD19 expression, with both populations enriched in the CD138int fraction. Anti-insulin B cells were not identified in the plasma-cell CD138hi fraction, which also expressed the transcription factor Blimp-1. After anti-CD20 treatment, anti-insulin B cells repopulated the pancreatic islets earlier than non-specific B cells. Importantly, we observed that a CD138intinsulin+CD19- population was particularly enriched after B cell depletion, possibly contributing to the persistence of disease still observed in some mice after anti-CD20 treatment. CONCLUSIONS/INTERPRETATION Our observations may indicate why the loss of C-peptide is only temporarily delayed following anti-CD20 treatment in human type 1 diabetes.
Collapse
Affiliation(s)
- Joanne Boldison
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Larissa C Da Rosa
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Lucy Buckingham
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Joanne Davies
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| | - F Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK.
| |
Collapse
|
20
|
Egia-Mendikute L, Arpa B, Rosell-Mases E, Corral-Pujol M, Carrascal J, Carrillo J, Mora C, Chapman H, Panosa A, Vives-Pi M, Stratmann T, Serreze D, Verdaguer J. B-Lymphocyte Phenotype Determines T-Lymphocyte Subset Differentiation in Autoimmune Diabetes. Front Immunol 2019; 10:1732. [PMID: 31428087 PMCID: PMC6689997 DOI: 10.3389/fimmu.2019.01732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/09/2019] [Indexed: 11/17/2022] Open
Abstract
Previous studies indicate that B-lymphocytes play a key role activating diabetogenic T-lymphocytes during the development of autoimmune diabetes. Recently, two transgenic NOD mouse models were generated: the NOD-PerIg and the 116C-NOD mice. In NOD-PerIg mice, B-lymphocytes acquire an activated proliferative phenotype and support accelerated autoimmune diabetes development. In contrast, in 116C-NOD mice, B-lymphocytes display an anergic-like phenotype delaying autoimmune diabetes onset and decreasing disease incidence. The present study further evaluates the T- and B-lymphocyte phenotype in both models. In islet-infiltrating B-lymphocytes (IIBLs) from 116C-NOD mice, the expression of H2-Kd and H2-Ag7 is decreased, whereas that of BAFF, BAFF-R, and TACI is increased. In contrast, IIBLs from NOD-PerIg show an increase in CD86 and FAS expression. In addition, islet-infiltrating T-lymphocytes (IITLs) from NOD-PerIg mice exhibit an increase in PD-1 expression. Moreover, proliferation assays indicate a high capacity of B-lymphocytes from NOD-PerIg mice to secrete high amounts of cytokines and induce T-lymphocyte activation compared to 116C B-lymphocytes. This functional variability between 116C and PerIg B-lymphocytes ultimately results in differences in the ability to shape T-lymphocyte phenotype. These results support the role of B-lymphocytes as key regulators of T-lymphocytes in autoimmune diabetes and provide essential information on the phenotypic characteristics of the T- and B-lymphocytes involved in the autoimmune response in autoimmune diabetes.
Collapse
Affiliation(s)
- Leire Egia-Mendikute
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain
| | - Berta Arpa
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain
| | - Estela Rosell-Mases
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain
| | - Marta Corral-Pujol
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain
| | - Jorge Carrascal
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain
| | - Jorge Carrillo
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain
| | - Conchi Mora
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain
| | | | - Anaïs Panosa
- Microscopy and Flow Cytometry Facility, IRBLleida, Universitat de Lleida, Lleida, Spain
| | - Marta Vives-Pi
- Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Barcelona, Spain.,CIBER of Diabetes and Associated Metabolic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Thomas Stratmann
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - David Serreze
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Joan Verdaguer
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain.,CIBER of Diabetes and Associated Metabolic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
21
|
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: 3.2] [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
|
22
|
Smith MJ, Hinman RM, Getahun A, Kim S, Packard TA, Cambier JC. Silencing of high-affinity insulin-reactive B lymphocytes by anergy and impact of the NOD genetic background in mice. Diabetologia 2018; 61:2621-2632. [PMID: 30255377 PMCID: PMC6219930 DOI: 10.1007/s00125-018-4730-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS Previous studies have demonstrated that high-affinity insulin-binding B cells (IBCs) silenced by anergy in healthy humans lose their anergy in islet autoantibody-positive individuals with recent-onset type 1 diabetes, and in autoantibody-negative first-degree relatives carrying certain risk alleles. Here we explore the hypothesis that IBCs are found in the immune periphery of disease-resistant C57BL/6-H2g7 mice, where, as in healthy humans, they are anergic, but that in disease-prone genetic backgrounds (NOD) they become activated and migrate to the pancreas and pancreatic lymph nodes, where they participate in the development of type 1 diabetes. METHODS We compared the status of high-affinity IBCs in disease-resistant VH125.C57BL/6-H2g7 and disease-prone VH125.NOD mice. RESULTS Consistent with findings in healthy humans, high-affinity IBCs reach the periphery in disease-resistant mice and are anergic, as indicated by a reduced expression of membrane IgM, unresponsiveness to antigen and failure to become activated or accumulate in the pancreatic lymph nodes or pancreas. In NOD mice, high-affinity IBCs reach the periphery early in life and increase in number prior to the onset of hyperglycaemia. These cells are not anergic; they become activated, produce autoantibodies and accumulate in the pancreas and pancreatic lymph nodes prior to disease development. CONCLUSIONS/INTERPRETATION These findings are consistent with genetic determination of the escape of high-affinity IBCs from anergy and their early contribution to the development of type 1 diabetes.
Collapse
Affiliation(s)
- Mia J Smith
- Department of Immunology and Microbiology, University of Colorado School of Medicine, P18-8100, RC1 North, 12800 East 19th Avenue, Aurora, CO, 80045-2537, USA
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Rochelle M Hinman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, P18-8100, RC1 North, 12800 East 19th Avenue, Aurora, CO, 80045-2537, USA
| | - Andrew Getahun
- Department of Immunology and Microbiology, University of Colorado School of Medicine, P18-8100, RC1 North, 12800 East 19th Avenue, Aurora, CO, 80045-2537, USA
| | - Soojin Kim
- Department of Immunology and Microbiology, University of Colorado School of Medicine, P18-8100, RC1 North, 12800 East 19th Avenue, Aurora, CO, 80045-2537, USA
| | - Thomas A Packard
- Department of Immunology and Microbiology, University of Colorado School of Medicine, P18-8100, RC1 North, 12800 East 19th Avenue, Aurora, CO, 80045-2537, USA
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, P18-8100, RC1 North, 12800 East 19th Avenue, Aurora, CO, 80045-2537, USA.
| |
Collapse
|
23
|
Felton JL, Maseda D, Bonami RH, Hulbert C, Thomas JW. Anti-Insulin B Cells Are Poised for Antigen Presentation in Type 1 Diabetes. THE JOURNAL OF IMMUNOLOGY 2018; 201:861-873. [PMID: 29950508 DOI: 10.4049/jimmunol.1701717] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/23/2018] [Indexed: 12/29/2022]
Abstract
Early breaches in B cell tolerance are central to type 1 diabetes progression in mouse and man. Conventional BCR transgenic mouse models (VH125.Tg NOD) reveal the power of B cell specificity to drive disease as APCs. However, in conventional fixed IgM models, comprehensive assessment of B cell development is limited. To provide more accurate insight into the developmental and functional fates of anti-insulin B cells, we generated a new NOD model (VH125SDNOD) in which anti-insulin VDJH125 is targeted to the IgH chain locus to generate a small (1-2%) population of class switch-competent insulin-binding B cells. Tracking of this rare population in a polyclonal repertoire reveals that anti-insulin B cells are preferentially skewed into marginal zone and late transitional subsets known to have increased sensitivity to proinflammatory signals. Additionally, IL-10 production, characteristic of regulatory B cell subsets, is increased. In contrast to conventional models, class switch-competent anti-insulin B cells proliferate normally in response to mitogenic stimuli but remain functionally silent for insulin autoantibody production. Diabetes development is accelerated, which demonstrates the power of anti-insulin B cells to exacerbate disease without differentiation into Ab-forming or plasma cells. Autoreactive T cell responses in VH125SDNOD mice are not restricted to insulin autoantigens, as evidenced by increased IFN-γ production to a broad array of diabetes-associated epitopes. Together, these results independently validate the pathogenic role of anti-insulin B cells in type 1 diabetes, underscore their diverse developmental fates, and demonstrate the pathologic potential of coupling a critical β cell specificity to predominantly proinflammatory Ag-presenting B cell subsets.
Collapse
Affiliation(s)
- Jamie L Felton
- Division of Pediatric Endocrinology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Damian Maseda
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; and
| | - Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Chrys Hulbert
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - James W Thomas
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; and .,Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
24
|
Circulating B cells in type 1 diabetics exhibit fewer maturation-associated phenotypes. Clin Immunol 2017; 183:336-343. [PMID: 28951327 DOI: 10.1016/j.clim.2017.09.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/16/2017] [Accepted: 09/22/2017] [Indexed: 01/05/2023]
Abstract
Although autoantibodies have been used for decades as diagnostic and prognostic markers in type 1 diabetes (T1D), further analysis of developmental abnormalities in B cells could reveal tolerance checkpoint defects that could improve individualized therapy. To evaluate B cell developmental progression in T1D, immunophenotyping was used to classify circulating B cells into transitional, mature naïve, mature activated, and resting memory subsets. Then each subset was analyzed for the expression of additional maturation-associated markers. While the frequencies of B cell subsets did not differ significantly between patients and controls, some T1D subjects exhibited reduced proportions of B cells that expressed transmembrane activator and CAML interactor (TACI) and Fas receptor (FasR). Furthermore, some T1D subjects had B cell subsets with lower frequencies of class switching. These results suggest circulating B cells exhibit variable maturation phenotypes in T1D. These phenotypic variations may correlate with differences in B cell selection in individual T1D patients.
Collapse
|
25
|
Unique features in the presentation of insulin epitopes in autoimmune diabetes: an update. Curr Opin Immunol 2017; 46:30-37. [PMID: 28456018 DOI: 10.1016/j.coi.2017.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/18/2017] [Accepted: 04/04/2017] [Indexed: 01/12/2023]
Abstract
Although an autoimmune disease involves diverse self-antigens, the initiation stage may require recognition of a limited number. This concept is verified in the non-obese diabetic (NOD) mouse model of autoimmune diabetes, in which strong evidence points to insulin as the prime antigen. The NOD mouse bears the I-Ag7 class II-MHC molecules (MHCII) that share common biochemical features and peptidome selection with the human diabetes-susceptible HLA-DQ8. Furthermore, both NOD mice and patients with type 1 diabetes (T1D) display an early appearance of insulin autoantibodies (IAAs) and subsequent insulin-reactive T cell infiltration into the islets. Therefore, a better understanding of insulin presentation is crucial for assessing disease pathogenesis and therapeutic intervention. Here, we summarize recent advances in insulin presentation events that underlie the essential role of this autoantigen in driving autoimmune diabetes.
Collapse
|
26
|
Packard TA, Smith MJ, Conrad FJ, Johnson SA, Getahun A, Lindsay RS, Hinman RM, Friedman RS, Thomas JW, Cambier JC. B Cell Receptor Affinity for Insulin Dictates Autoantigen Acquisition and B Cell Functionality in Autoimmune Diabetes. J Clin Med 2016; 5:E98. [PMID: 27834793 PMCID: PMC5126795 DOI: 10.3390/jcm5110098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/24/2016] [Accepted: 11/03/2016] [Indexed: 11/30/2022] Open
Abstract
B cells have been strongly implicated in the development of human type 1 diabetes and are required for disease in the NOD mouse model. These functions are dependent on B cell antigen receptor (BCR) specificity and expression of MHC, implicating linked autoantigen recognition and presentation to effector T cells. BCR-antigen affinity requirements for participation in disease are unclear. We hypothesized that BCR affinity for the autoantigen insulin differentially affects lymphocyte functionality, including tolerance modality and the ability to acquire and become activated in the diabetogenic environment. Using combined transgenic and retrogenic heavy and light chain to create multiple insulin-binding BCRs, we demonstrate that affinity for insulin is a critical determinant of the function of these autoreactive cells. We show that both BCR affinity for insulin and genetic background affect tolerance induction in immature B cells. We also find new evidence that may explain the enigmatic ability of B cells expressing 125 anti-insulin BCR to support development of TID in NOD mice despite a reported affinity beneath requirements for binding insulin at in vivo concentrations. We report that when expressed as an antigen receptor the affinity of 125 is much higher than determined by measurements of the soluble form. Finally, we show that in vivo acquisition of insulin requires both sufficient BCR affinity and permissive host/tissue environment. We propose that a confluence of BCR affinity, pancreas environment, and B cell tolerance-regulating genes in the NOD animal allows acquisition of insulin and autoimmunity.
Collapse
Affiliation(s)
- Thomas A Packard
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
| | - Mia J Smith
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| | - Francis J Conrad
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
| | - Sara A Johnson
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
| | - Andrew Getahun
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
| | - Robin S Lindsay
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
| | - Rochelle M Hinman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
| | - Rachel S Friedman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
| | - James W Thomas
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
27
|
Leeth CM, Racine J, Chapman HD, Arpa B, Carrillo J, Carrascal J, Wang Q, Ratiu J, Egia-Mendikute L, Rosell-Mases E, Stratmann T, Verdaguer J, Serreze DV. B-lymphocytes expressing an Ig specificity recognizing the pancreatic ß-cell autoantigen peripherin are potent contributors to type 1 diabetes development in NOD mice. Diabetes 2016; 65:1977-1987. [PMID: 26961115 PMCID: PMC4915583 DOI: 10.2337/db15-1606] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
While the autoimmune destruction of pancreatic ß-cells underlying type 1 diabetes (1D) development is ultimately mediated by T-cells in NOD mice and also likely humans, B-lymphocytes play an additional key pathogenic role. It appears expression of plasma membrane bound immunoglobulin (Ig) molecules that efficiently capture ß-cell antigens allows autoreactive B-lymphocytes bypassing normal tolerance induction processes to be the subset of antigen presenting cells most efficiently activating diabetogenic T-cells. NOD mice transgenically expressing Ig molecules recognizing antigens that are (insulin) or not (hen egg lysozyme; HEL) expressed by ß-cells have proven useful in dissecting the developmental basis of diabetogenic B-lymphocytes. However, these transgenic Ig specificities were originally selected for their ability to recognize insulin or HEL as foreign, rather than autoantigens. Thus, we generated and characterized NOD mice transgenically expressing an Ig molecule representative of a large proportion of naturally occurring islet-infiltrating B-lymphocytes in NOD mice recognizing the neuronal antigen peripherin. Transgenic peripherin autoreactive B-lymphocytes infiltrate NOD pancreatic islets, acquire an activated proliferative phenotype, and potently support accelerated T1D development. These results support the concept of neuronal autoimmunity as a pathogenic feature of T1D, and targeting such responses could ultimately provide an effective disease intervention approach.
Collapse
Affiliation(s)
- Caroline M Leeth
- The Jackson Laboratory, Bar Harbor, Maine, USA Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | | | | | - Berta Arpa
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida & IRBLleida, Lleida, Spain
| | - Jorge Carrillo
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida & IRBLleida, Lleida, Spain
| | - Jorge Carrascal
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida & IRBLleida, Lleida, Spain
| | - Qiming Wang
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | | | | | | | - Thomas Stratmann
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Spain
| | - Joan Verdaguer
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida & IRBLleida, Lleida, Spain
| | | |
Collapse
|
28
|
|
29
|
Wan X, Thomas JW, Unanue ER. Class-switched anti-insulin antibodies originate from unconventional antigen presentation in multiple lymphoid sites. J Exp Med 2016; 213:967-78. [PMID: 27139492 PMCID: PMC4886365 DOI: 10.1084/jem.20151869] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 03/09/2016] [Indexed: 01/03/2023] Open
Abstract
Unanue and colleagues show that activation of anti-insulin lymphocytes can occur at diverse anatomical sites in response to circulating insulin and may be driven by unconventional antigen presentation by germinal center B cells. Autoantibodies to insulin are a harbinger of autoimmunity in type 1 diabetes in humans and in non-obese diabetic mice. To understand the genesis of these autoantibodies, we investigated the interactions of insulin-specific T and B lymphocytes using T cell and B cell receptor transgenic mice. We found spontaneous anti-insulin germinal center (GC) formation throughout lymphoid tissues with GC B cells binding insulin. Moreover, because of the nature of the insulin epitope recognized by the T cells, it was evident that GC B cells presented a broader repertoire of insulin epitopes. Such broader recognition was reproduced by activating naive B cells ex vivo with a combination of CD40 ligand and interleukin 4. Thus, insulin immunoreactivity extends beyond the pancreatic lymph node–islets of Langerhans axis and indicates that circulating insulin, despite its very low levels, can have an influence on diabetogenesis.
Collapse
Affiliation(s)
- Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - James W Thomas
- Department of Medicine, Vanderbilt University Medical School, Nashville, TN 37232
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
30
|
Crofford LJ, Nyhoff LE, Sheehan JH, Kendall PL. The role of Bruton's tyrosine kinase in autoimmunity and implications for therapy. Expert Rev Clin Immunol 2016; 12:763-73. [PMID: 26864273 DOI: 10.1586/1744666x.2016.1152888] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bruton's tyrosine kinase (BTK) mediates B cell signaling and is also present in innate immune cells but not T cells. BTK propagates B cell receptor (BCR) responses to antigen-engagement as well as to stimulation via CD40, toll-like receptors (TLRs), Fc receptors (FCRs) and chemokine receptors. Importantly, BTK can modulate signaling, acting as a "rheostat" rather than an "on-off" switch; thus, overexpression leads to autoimmunity while decreased levels improve autoimmune disease outcomes. Autoreactive B cells depend upon BTK for survival to a greater degree than normal B cells, reflected as loss of autoantibodies with maintenance of total antibody levels when BTK is absent. This review describes contributions of BTK to immune tolerance, including studies testing BTK-inhibitors for treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Leslie J Crofford
- a Division of Rheumatology & Immunology, Department of Medicine , Vanderbilt University , Nashville , TN , USA.,b Department of Pathology, Microbiology & Immunology , Vanderbilt University , Nashville , TN , USA
| | - Lindsay E Nyhoff
- b Department of Pathology, Microbiology & Immunology , Vanderbilt University , Nashville , TN , USA
| | - Jonathan H Sheehan
- c Center for Structural Biology, Department of Biochemistry , Vanderbilt University , Nashville , TN , USA
| | - Peggy L Kendall
- b Department of Pathology, Microbiology & Immunology , Vanderbilt University , Nashville , TN , USA.,d Division of Allergy, Pulmonary and Critical Care, Department of Medicine , Vanderbilt University , Nashville , TN , USA
| |
Collapse
|
31
|
Wilson CS, Elizer SK, Marshall AF, Stocks BT, Moore DJ. Regulation of B lymphocyte responses to Toll-like receptor ligand binding during diabetes prevention in non-obese diabetic (NOD) mice. J Diabetes 2016; 8:120-31. [PMID: 25564999 PMCID: PMC4598313 DOI: 10.1111/1753-0407.12263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 11/12/2014] [Accepted: 12/23/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Interactions between genetic risk factors and the environment drive type 1 diabetes (T1D). The system of Toll-like receptors (TLR) detects these environmental triggers; however, the target cell that intermediates these interactions to drive T1D remains unknown. METHODS We investigated the effect of TLR pathway activation (myeloid differentiation primary response 88 [MyD88] vs TIR-domain-containing adapter-inducing interferon-β [TRIF]) on B cell subsets via flow cytometry, including their activation, survival, proliferation, and cytoskeletal mobilization. The effect of polyinosinic-polycytidylic acid (poly(I:C)) on diabetes development was addressed, including the B cell-dependent activation of diabetes-protective DX5+ cells, using genetic models and adoptive transfer. RESULTS B lymphocytes from non-obese diabetic (NOD) mice expressed enhanced levels of TLR-responsive proteins. Ex vivo analysis of B lymphocyte subsets demonstrated that TLR3 stimulation via TRIF deletes cells exhibiting a marginal zone phenotype, whereas MyD88-dependent ligands enhance their survival. In vivo, marginal zone B cells were activated by poly(I:C) and were unexpectedly retained in the spleen of NOD mice, in contrast with the mobilization of these cells in non-autoimmune mice, a phenotype we traced to defective actin cytoskeletal dynamics. These activated B cells mediated TLR3-induced diabetes protection. CONCLUSIONS Immunotherapies must account for both B cell location and activation, and these properties may differ in autoimmune and healthy settings.
Collapse
Affiliation(s)
- Christopher S. Wilson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, School of Medicine, 1161 21st Ave South. Nashville, TN 37232-2363
| | - Sydney K. Elizer
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University, School of Medicine, 2200 Children's Way. Nashville, TN 37232-2363
| | - Andrew F. Marshall
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University, School of Medicine, 2200 Children's Way. Nashville, TN 37232-2363
| | - Blair T. Stocks
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, School of Medicine, 1161 21st Ave South. Nashville, TN 37232-2363
- Vanderbilt Medical Scientist Training Program
| | - Daniel J. Moore
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, School of Medicine, 1161 21st Ave South. Nashville, TN 37232-2363
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University, School of Medicine, 2200 Children's Way. Nashville, TN 37232-2363
| |
Collapse
|
32
|
Carrascal J, Carrillo J, Arpa B, Egia-Mendikute L, Rosell-Mases E, Pujol-Autonell I, Planas R, Mora C, Mauricio D, Ampudia RM, Vives-Pi M, Verdaguer J. B-cell anergy induces a Th17 shift in a novel B lymphocyte transgenic NOD mouse model, the 116C-NOD mouse. Eur J Immunol 2015; 46:593-608. [PMID: 26639224 DOI: 10.1002/eji.201445376] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 11/09/2015] [Accepted: 12/01/2015] [Indexed: 11/06/2022]
Abstract
Autoreactive B lymphocytes play a key role as APCs in diaebetogenesis. However, it remains unclear whether B-cell tolerance is compromised in NOD mice. Here, we describe a new B lymphocyte transgenic NOD mouse model, the 116C-NOD mouse, where the transgenes derive from an islet-infiltrating B lymphocyte of a (8.3-NODxNOR) F1 mouse. The 116C-NOD mouse produces clonal B lymphocytes with pancreatic islet beta cell specificity. The incidence of T1D in 116C-NOD mice is decreased in both genders when compared with NOD mice. Moreover, several immune selection mechanisms (including clonal deletion and anergy) acting on the development, phenotype, and function of autoreactive B lymphocytes during T1D development have been identified in the 116C-NOD mouse. Surprisingly, a more accurate analysis revealed that, despite their anergic phenotype, 116C B cells express some costimulatory molecules after activation, and induce a T-cell shift toward a Th17 phenotype. Furthermore, this shift on T lymphocytes seems to occur not only when both T and B cells contact, but also when helper T (Th) lineage is established. The 116C-NOD mouse model could be useful to elucidate the mechanisms involved in the generation of Th-cell lineages.
Collapse
Affiliation(s)
- Jorge Carrascal
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Jorge Carrillo
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Berta Arpa
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Leire Egia-Mendikute
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Estela Rosell-Mases
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Irma Pujol-Autonell
- Immunology Department, Institut d'Investigacio Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Raquel Planas
- Immunology Department, Institut d'Investigacio Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Conchi Mora
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Dídac Mauricio
- Department of Endocrinology, Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Maria Ampudia
- Immunology Department, Institut d'Investigacio Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Marta Vives-Pi
- Immunology Department, Institut d'Investigacio Germans Trias i Pujol, Badalona, Barcelona, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Verdaguer
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
33
|
Williams JM, Bonami RH, Hulbert C, Thomas JW. Reversing Tolerance in Isotype Switch-Competent Anti-Insulin B Lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:853-64. [PMID: 26109644 PMCID: PMC4506889 DOI: 10.4049/jimmunol.1403114] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 05/30/2015] [Indexed: 12/22/2022]
Abstract
Autoreactive B lymphocytes that escape central tolerance and mature in the periphery are a liability for developing autoimmunity. IgG insulin autoantibodies that predict type 1 diabetes and complicate insulin therapies indicate that mechanisms for tolerance to insulin are flawed. To examine peripheral tolerance in anti-insulin B cells, we generated C57BL/6 mice that harbor anti-insulin VDJH-125 site directed to the native IgH locus (VH125(SD)). Class switch-competent anti-insulin B cells fail to produce IgG Abs following T cell-dependent immunization of VH125(SD) mice with heterologous insulin, and they exhibit markedly impaired proliferation to anti-CD40 plus insulin in vitro. In contrast, costimulation with LPS plus insulin drives robust anti-insulin B cell proliferation. Furthermore, VH125(SD) mice produce both IgM and IgG2a anti-insulin Abs following immunization with insulin conjugated to type 1 T cell-independent Brucella abortus ring test Ag (BRT). Anti-insulin B cells undergo clonal expansion in vivo and emerge as IgM(+) and IgM(-) GL7(+)Fas(+) germinal center (GC) B cells following immunization with insulin-BRT, but not BRT alone. Analysis of Igκ genes in VH125(SD) mice immunized with insulin-BRT reveals that anti-insulin Vκ from the preimmune repertoire is selected into GCs. These data demonstrate that class switch-competent anti-insulin B cells remain functionally silent in T cell-dependent immune responses, yet these B cells are vulnerable to reversal of anergy following combined BCR/TLR engagement that promotes Ag-specific GC responses and Ab production. Environmental factors that lead to infection and inflammation could play a critical yet underappreciated role in driving loss of tolerance and promoting autoimmune disease.
Collapse
Affiliation(s)
- Jonathan M Williams
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; and Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Chrys Hulbert
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - James W Thomas
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; and Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
34
|
Fillatreau S. Pathogenic functions of B cells in autoimmune diseases: IFN-γ production joins the criminal gang. Eur J Immunol 2015; 45:966-70. [DOI: 10.1002/eji.201545544] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 02/23/2015] [Accepted: 02/26/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Simon Fillatreau
- Deutsches Rheuma-Forschungszentrum; a Leibniz Institute; Berlin Germany
| |
Collapse
|
35
|
Garabatos N, Alvarez R, Carrillo J, Carrascal J, Izquierdo C, Chapman HD, Presa M, Mora C, Serreze DV, Verdaguer J, Stratmann T. In vivo detection of peripherin-specific autoreactive B cells during type 1 diabetes pathogenesis. THE JOURNAL OF IMMUNOLOGY 2014; 192:3080-90. [PMID: 24610011 DOI: 10.4049/jimmunol.1301053] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autoreactive B cells are essential for the pathogenesis of type 1 diabetes. The genesis and dynamics of autoreactive B cells remain unknown. In this study, we analyzed the immune response in the NOD mouse model to the neuronal protein peripherin (PRPH), a target Ag of islet-infiltrating B cells. PRPH autoreactive B cells recognized a single linear epitope of this protein, in contrast to the multiple epitope recognition commonly observed during autoreactive B cell responses. Autoantibodies to this epitope were also detected in the disease-resistant NOR and C57BL/6 strains. To specifically detect the accumulation of these B cells, we developed a novel approach, octameric peptide display, to follow the dynamics and localization of anti-PRPH B cells during disease progression. Before extended insulitis was established, anti-PRPH B cells preferentially accumulated in the peritoneum. Anti-PRPH B cells were likewise detected in C57BL/6 mice, albeit at lower frequencies. As disease unfolded in NOD mice, anti-PRPH B cells invaded the islets and increased in number at the peritoneum of diabetic but not prediabetic mice. Isotype-switched B cells were only detected in the peritoneum. Anti-PRPH B cells represent a heterogeneous population composed of both B1 and B2 subsets. In the spleen, anti-PRPH B cell were predominantly in the follicular subset. Therefore, anti-PRPH B cells represent a heterogeneous population that is generated early in life but proliferates as diabetes is established. These findings on the temporal and spatial progression of autoreactive B cells should be relevant for our understanding of B cell function in diabetes pathogenesis.
Collapse
Affiliation(s)
- Nahir Garabatos
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
NFATc2 (NFAT1) assists BCR-mediated anergy in anti-insulin B cells. Mol Immunol 2014; 62:321-8. [PMID: 24507801 DOI: 10.1016/j.molimm.2014.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/16/2013] [Accepted: 01/06/2014] [Indexed: 01/03/2023]
Abstract
NFAT transcription factors play critical roles in both the activation and repression of T and B lymphocyte responses. To understand the role of NFATc2 (NFAT1) in the maintenance of tolerance for anti-insulin B cells, functionally inactive NFATc2 (NFATc2(-/-)) was introduced into C57BL/6 mice that harbor anergic anti-insulin 125Tg B cells. The production and peripheral maturation of anti-insulin B cells into follicular and marginal zone subsets was not altered by the absence of functional NFATc2. Surface B cell receptor expression levels, important for tonic signaling and altered by anergy, were not altered in any spleen B cell subset. The levels of anti-insulin antibodies were not different in 125Tg/B6/NFATc2(-/-) mice and the anti-insulin response remained silenced following T cell dependent immunization. However, studies addressing in vitro proliferation reveal the anergic state of 125Tg B cells is relieved in 125Tg/B6/NFATc2(-/-) B cells in response to BCR stimulation. In contrast, anergy is not released in 125Tg/B6/NFATc2(-/-) B cells following stimulation with anti-CD40. The relief of anergy to BCR stimulation in 125Tg/B6/NFATc2(-/-) B cells is associated with increased transcription of both NFATc1 and NFATc3 while expression of these NFATs does not change in anti-IgM stimulated 125Tg/B6/NFATc2(+/+) B cells. The data suggest that NFATc2 plays a subtle and selective role in maintaining anergy for BCR stimulation by repressing the transcription of other NFAT family members.
Collapse
|
37
|
Hinman RM, Smith MJ, Cambier JC. B cells and type 1 diabetes ...in mice and men. Immunol Lett 2014; 160:128-32. [PMID: 24472603 DOI: 10.1016/j.imlet.2014.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 01/14/2014] [Indexed: 12/25/2022]
Abstract
Nearly 70% of newly produced B cells express autoreactive antigen receptors and must be silenced to prevent autoimmunity. Failure of silencing mechanisms is apparent in type 1 diabetes (T1D), where islet antigen-specific B cells appear critical for development of disease. Evidence for a B cell role in T1D includes success of B cell targeted anti-CD20 therapy, which delays T1D progression in both NOD mice and new onset patients. Demonstrating the importance of specificity, NOD mice whose B cell repertoire is biased toward insulin reactivity show increased disease development, while bias away from insulin reactivity largely prevents disease. Finally, though not required for illness, high affinity insulin autoantibodies are often the first harbingers of T1D. B cell cytokine production and auto-antigen presentation to self-reactive T cells are likely important in pathogenesis. Here we review B cell function, as described above, in T1D in humans and the non-obese diabetic (NOD) mouse. We will discuss recent broad-based B cell depletion studies and how they may provide the basis for refinement of future treatments for the disorder.
Collapse
Affiliation(s)
- Rochelle M Hinman
- University of Colorado Denver and National Jewish Health, Denver, CO, United States.
| | - Mia J Smith
- University of Colorado Denver and National Jewish Health, Denver, CO, United States.
| | - John C Cambier
- University of Colorado Denver and National Jewish Health, Denver, CO, United States; Department of Immunology, National Jewish Health, Rm 803A, Goodman Building, 1400 Jackson Street, Denver, CO 80206, United States.
| |
Collapse
|
38
|
Bonami RH, Sullivan AM, Case JB, Steinberg HE, Hoek KL, Khan WN, Kendall PL. Bruton's tyrosine kinase promotes persistence of mature anti-insulin B cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:1459-70. [PMID: 24453243 DOI: 10.4049/jimmunol.1300125] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Autoreactive B lymphocytes are essential for the development of T cell-mediated type 1 diabetes (T1D). Cytoplasmic Bruton's tyrosine kinase (BTK) is a key component of B cell signaling, and its deletion in T1D-prone NOD mice significantly reduces diabetes. However, the role of BTK in the survival and function of autoreactive B cells is not clear. To evaluate the contributions of BTK, we used mice in which B cells express an anti-insulin BCR (125Tg) and promote T1D, despite being anergic. Crossing Btk deficiency onto 125Tg mice reveals that, in contrast to immature B cells, mature anti-insulin B cells are exquisitely dependent upon BTK, because their numbers are reduced by 95%. BTK kinase domain inhibition reproduces this effect in mature anti-insulin B cells, with less impact at transitional stages. The increased dependence of anti-insulin B cells on BTK became particularly evident in an Igκ locus site-directed model, in which 50% of B cells edit their BCRs to noninsulin specificities; Btk deficiency preferentially depletes insulin binders from the follicular and marginal zone B cell subsets. The persistent few Btk-deficient anti-insulin B cells remain competent to internalize Ag and invade pancreatic islets. As such, loss of BTK does not significantly reduce diabetes incidence in 125Tg/NOD mice as it does in NOD mice with a normal B cell repertoire. Thus, BTK targeting may not impair autoreactive anti-insulin B cell function, yet it may provide protection in an endogenous repertoire by decreasing the relative availability of mature autoreactive B cells.
Collapse
Affiliation(s)
- Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Though type 1 diabetes (T1D) is considered a T cell-mediated autoimmune disorder, recent evidence indicates that B cells play a critical role in disease. This conclusion is based in part on the success of anti-CD20 (rituximab) therapy, which by broadly depleting B cells delays disease progression in non-obese diabetic (NOD) mice and new-onset patients. B cell receptor (BCR) specificity to islet autoantigen is key. NOD mice whose B cell repertoire is biased toward insulin reactivity show increased disease development, while bias away from insulin reactivity largely prevents disease. Although the operative disease-promoting B cell effector function remains undefined, islet-antigen reactive B cells function in antigen presentation to diabetogenic CD4 T cells. Other studies implicate B cells in antigen presentation to CD8 T cells. B cell participation in TID appears predicated on faulty B cell tolerance. Here, we review extant findings implicating B cells in T1D in mice and men.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Autoantibodies/blood
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/physiopathology
- Disease Progression
- Humans
- Immune Tolerance/drug effects
- Immune Tolerance/immunology
- Immunologic Factors/pharmacology
- Lymphocyte Depletion
- Mice
- Mice, Inbred NOD
- Molecular Targeted Therapy
- Receptors, Antigen, B-Cell/antagonists & inhibitors
- Receptors, Antigen, B-Cell/immunology
- Rituximab
Collapse
Affiliation(s)
- Rochelle M Hinman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E 19th Avenue, P18-8100, Mail Stop 8333, RC1 N, Aurora, CO, 80045-2537, USA,
| | | |
Collapse
|
40
|
Khan WN, Wright JA, Kleiman E, Boucher JC, Castro I, Clark ES. B-lymphocyte tolerance and effector function in immunity and autoimmunity. Immunol Res 2013; 57:335-53. [DOI: 10.1007/s12026-013-8466-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
41
|
Henry-Bonami RA, Williams JM, Rachakonda AB, Karamali M, Kendall PL, Thomas JW. B lymphocyte "original sin" in the bone marrow enhances islet autoreactivity in type 1 diabetes-prone nonobese diabetic mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:5992-6003. [PMID: 23677466 PMCID: PMC3679359 DOI: 10.4049/jimmunol.1201359] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Effective central tolerance is required to control the large extent of autoreactivity normally present in the developing B cell repertoire. Insulin-reactive B cells are required for type 1 diabetes in the NOD mouse, because engineered mice lacking this population are protected from disease. The Cg-Tg(Igh-6/Igh-V125)2Jwt/JwtJ (VH125Tg) model is used to define this population, which is found with increased frequency in the periphery of NOD mice versus nonautoimmune C57BL/6 VH125Tg mice; however, the ontogeny of this disparity is unknown. To better understand the origins of these pernicious B cells, anti-insulin B cells were tracked during development in the polyclonal repertoire of VH125Tg mice. An increased proportion of insulin-binding B cells is apparent in NOD mice at the earliest point of Ag commitment in the bone marrow. Two predominant L chains were identified in B cells that bind heterologous insulin. Interestingly, Vκ4-57-1 polymorphisms that confer a CDR3 Pro-Pro motif enhance self-reactivity in VH125Tg/NOD mice. Despite binding circulating autoantigen in vivo, anti-insulin B cells transition from the parenchyma to the sinusoids in the bone marrow of NOD mice and enter the periphery unimpeded. Anti-insulin B cells expand at the site of autoimmune attack in the pancreas and correlate with increased numbers of IFN-γ-producing cells in the repertoire. These data identify the failure to cull autoreactive B cells in the bone marrow as the primary source of anti-insulin B cells in NOD mice and suggest that dysregulation of central tolerance permits their escape into the periphery to promote disease.
Collapse
Affiliation(s)
- Rachel A. Henry-Bonami
- Vanderbilt University, Department of Medicine, Division of Rheumatology and Immunology, Nashville TN
| | - Jonathan M. Williams
- Vanderbilt University, Department of Pathology, Microbiology and Immunology, Nashville, TN
| | - Amita B. Rachakonda
- Vanderbilt University, Department of Medicine, Division of Rheumatology and Immunology, Nashville TN
| | - Mariam Karamali
- Vanderbilt University, Department of Medicine, Division of Rheumatology and Immunology, Nashville TN
| | - Peggy L. Kendall
- Vanderbilt University, Department of Medicine, Division of Allergy, Pulmonary, and Critical Care, Nashville TN
| | - James W. Thomas
- Vanderbilt University, Department of Medicine, Division of Rheumatology and Immunology, Nashville TN
- Vanderbilt University, Department of Pathology, Microbiology and Immunology, Nashville, TN
| |
Collapse
|
42
|
The role of the complement system in metabolic organs and metabolic diseases. Semin Immunol 2013; 25:47-53. [PMID: 23684628 DOI: 10.1016/j.smim.2013.04.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/13/2013] [Indexed: 12/20/2022]
Abstract
Emerging evidence points to a close crosstalk between metabolic organs and innate immunity in the course of metabolic disorders. In particular, cellular and humoral factors of innate immunity are thought to contribute to metabolic dysregulation of the adipose tissue or the liver, as well as to dysfunction of the pancreas; all these conditions are linked to the development of insulin resistance and diabetes mellitus. A central component of innate immunity is the complement system. Interestingly, the classical view of complement as a major system of host defense that copes with infections is changing to that of a multi-functional player in tissue homeostasis, degeneration, and regeneration. In the present review, we will discuss the link between complement and metabolic organs, focusing on the pancreas, adipose tissue, and liver and the diverse effects of complement system on metabolic disorders.
Collapse
|