1
|
Chen M, Zhang Q, Wei Y, Wan Q, Xu M, Chen X. Anti-CD20 therapy ameliorates β cell function and rebalances Th17/Treg cells in NOD mice. Endocrine 2022; 76:44-52. [PMID: 35067899 DOI: 10.1007/s12020-021-02965-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/12/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Anti-CD20 therapy delays type 1 diabetes mellitus (T1DM) progression in both nonobese diabetic (NOD) mice and new-onset patients. The mechanism is not completely defined. This study aimed to investigate the effects of anti-CD20 therapy on T helper 17 (Th17) cells and regulatory T cells (Tregs) in NOD mice. The role of B cell depletion in T1DM development was also examined. METHODS NOD mice were randomly divided into two groups. The mice in the experimental group were treated with an anti-CD20 antibody, while the control mice were treated with an isotype-matched control antibody. After treatment, islet morphology and inflammation, Th17 and Treg cell frequencies in the pancreas and spleen, serum cytokine and anti-glutamic acid decarboxylase (GAD) antibody levels, interleukin (IL)-17A levels in the pancreas and spleen, insulin expression in islet cells and islet β cell function were measured. RESULTS Decreased blood glucose and increased insulin secretion were found in the exprimental group compared with the CON group. A lower islet inflammation score was also found in the experimental group. Decreased Th17 cell and IL-17A levels and augmented Treg cell levels were found in the spleen and pancreas after anti-CD20 treatment. The serum levels of B cell activating factor (BAFF), IL-17A, IL-17F, IL-23 and anti-GAD autoantibodies were decreased in the experimental group, while higher serum levels of IL-10 and transforming growth factor (TGF)-β were found. CONCLUSION Anti-CD20 therapy might have some beneficial effects that improve β cell function by relieving islet inflammation through regulation of Th17/Treg cells and the proinflammatory/anti-inflammatory balance.
Collapse
Affiliation(s)
- Min Chen
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Qianhui Zhang
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Yanhong Wei
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Qianqian Wan
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Min Xu
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Xiaoqi Chen
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China.
| |
Collapse
|
2
|
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
|
3
|
Habib T, Long SA, Samuels PL, Brahmandam A, Tatum M, Funk A, Hocking AM, Cerosaletti K, Mason MT, Whalen E, Rawlings DJ, Greenbaum C, Buckner JH. Dynamic Immune Phenotypes of B and T Helper Cells Mark Distinct Stages of T1D Progression. Diabetes 2019; 68:1240-1250. [PMID: 30894366 PMCID: PMC6610015 DOI: 10.2337/db18-1081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/15/2019] [Indexed: 01/01/2023]
Abstract
Multiple studies of B- and T-cell compartments and their response to stimuli demonstrate alterations in established type 1 diabetes (T1D). Yet it is not known whether these alterations reflect immune mechanisms that initiate islet autoimmunity, promote disease progression, or are secondary to disease. To address these questions, we used samples from the TrialNet Pathway to Prevention study to investigate T-cell responses to interleukin (IL)-2 and regulatory T cell-mediated suppression, the composition of the B-cell compartment, and B-cell responses to B-cell receptor and IL-21 receptor engagement. These studies revealed stage-dependent T- and B-cell functional and immune phenotypes; namely, early features that differentiate autoantibody-positive at-risk first-degree relatives (FDRs) from autoantibody-negative FDRs and persisted through clinical diagnosis; late features that arose at or near T1D diagnosis; and dynamic features that were enhanced early and blunted at later disease stages, indicating evolving responses along the continuum of T1D. We further explored how these specific phenotypes are influenced by therapeutic interventions. Our integrated studies provide unique insights into stable and dynamic stage-specific immune states and define novel immune phenotypes of potential clinical relevance.
Collapse
Affiliation(s)
- Tania Habib
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - S Alice Long
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Peter L Samuels
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Archana Brahmandam
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Megan Tatum
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Andrew Funk
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Anne M Hocking
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Karen Cerosaletti
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Michael T Mason
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | - Elizabeth Whalen
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA
| | - David J Rawlings
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
- Departments of Pediatrics and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Carla Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA
| | - Jane H Buckner
- Translational Research Program, Benaroya Research Institute, Seattle, WA
| | | |
Collapse
|
4
|
Manoylov IK, Boneva GV, Doytchinova IA, Mihaylova NM, Tchorbanov AI. Protein-engineered molecules carrying GAD65 epitopes and targeting CD35 selectively down-modulate disease-associated human B lymphocytes. Clin Exp Immunol 2019; 197:329-340. [PMID: 31009057 DOI: 10.1111/cei.13305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2019] [Indexed: 12/29/2022] Open
Abstract
Type 1 diabetes mellitus is an autoimmune metabolic disorder characterized by chronic hyperglycemia, the presence of autoreactive T and B cells and autoantibodies against self-antigens. A membrane-bound enzyme on the pancreatic beta-cells, glutamic acid decarboxylase 65 (GAD65), is one of the main autoantigens in type 1 diabetes. Autoantibodies against GAD65 are potentially involved in beta-cell destruction and decline of pancreatic functions. The human complement receptor type 1 (CD35) on B and T lymphocytes has a suppressive activity on these cells. We hypothesized that it may be possible to eliminate GAD65-specific B cells from type 1 diabetes patients by using chimeric molecules, containing an anti-CD35 antibody, coupled to peptides resembling GAD65 B/T epitopes. These molecules are expected to selectively bind the anti-GAD65 specific B cells by the co-cross-linking of the immunoglobulin receptor and CD35 and to deliver a suppressive signal. Two synthetic peptides derived from GAD65 protein (GAD65 epitopes) and anti-CD35 monoclonal antibody were used for the construction of two chimeras. The immunomodulatory activity of the engineered antibodies was tested in vitro using peripheral blood mononuclear cells (PBMCs) from type 1 diabetes patients. A reduction in the number of anti-GAD65 IgG antibody-secreting plasma cells and increased percentage of apoptotic B lymphocytes was observed after treatment of these PBMCs with the engineered antibodies. The constructed chimeric molecules are able to selectively modulate the activity of GAD65-specific B lymphocytes and the production of anti-GAD65 IgG autoantibodies by co-cross-linking of the inhibitory CD35 and the B cell antigen receptor (BCR). This treatment presents a possible way to alter the autoimmune nature of these cells.
Collapse
Affiliation(s)
- I K Manoylov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - G V Boneva
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - I A Doytchinova
- Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - N M Mihaylova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - A I Tchorbanov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.,National Institute of Immunology, Sofia, Bulgaria
| |
Collapse
|
5
|
Epigenetic regulation in B-cell maturation and its dysregulation in autoimmunity. Cell Mol Immunol 2018; 15:676-684. [PMID: 29375128 PMCID: PMC6123482 DOI: 10.1038/cmi.2017.133] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/22/2017] [Accepted: 10/23/2017] [Indexed: 12/11/2022] Open
Abstract
B cells have a critical role in the initiation and acceleration of autoimmune diseases, especially those mediated by autoantibodies. In the peripheral lymphoid system, mature B cells are activated by self or/and foreign antigens and signals from helper T cells for differentiating into either memory B cells or antibody-producing plasma cells. Accumulating evidence has shown that epigenetic regulations modulate somatic hypermutation and class switch DNA recombination during B-cell activation and differentiation. Any abnormalities in these complex regulatory processes may contribute to aberrant antibody production, resulting in autoimmune pathogenesis such as systemic lupus erythematosus. Newly generated knowledge from advanced modern technologies such as next-generation sequencing, single-cell sequencing and DNA methylation sequencing has enabled us to better understand B-cell biology and its role in autoimmune development. Thus this review aims to summarize current research progress in epigenetic modifications contributing to B-cell activation and differentiation, especially under autoimmune conditions such as lupus, rheumatoid arthritis and type 1 diabetes.
Collapse
|
6
|
Liu R, Zhai J, Liu L, Wang Y, Wei Y, Jiang X, Gao L, Zhu H, Zhao Y, Chai Z, Gao X. Spatially marking and quantitatively counting membrane immunoglobulin M in live cells via Ag cluster-aptamer probes. Chem Commun (Camb) 2014; 50:3560-3. [PMID: 24563906 DOI: 10.1039/c3cc49036j] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A probe composed of an aptamer and a silver cluster, where the aptamer targets mIgM of live cells and the silver cluster provides fluorescent imaging and mass quantification of mIgM of live cells, is presented. This new probe simultaneously provides accurate spatial and mass information of mIgM in live cells.
Collapse
Affiliation(s)
- Ru Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, P. R. China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Markle JGM, Frank DN, Adeli K, von Bergen M, Danska JS. Microbiome manipulation modifies sex-specific risk for autoimmunity. Gut Microbes 2014; 5:485-93. [PMID: 25007153 DOI: 10.4161/gmic.29795] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Despite growing evidence for a causal role of environmental factors in autoimmune diseases including the rise in disease frequencies over the past several decades we lack an understanding of how particular environmental exposures modify disease risk. In addition, many autoimmune diseases display sex-biased incidence, with females being disproportionately affected but the mechanisms underlying this sex bias remain elusive. Emerging evidence suggests that both host metabolism and immune function is crucially regulated by the intestinal microbiome. Recently, we showed that in the non-obese diabetic (NOD) mouse model of Type 1 Diabetes (T1D), the gut commensal microbial community strongly impacts the pronounced sex bias in T1D risk by controlling serum testosterone and metabolic phenotypes (1). Here we present new data in the NOD model that explores the correlations between microbial phylogeny, testosterone levels, and metabolic phenotypes, and discuss the future of microbiome-centered analysis and microbe-based therapeutic approaches in autoimmune diseases.
Collapse
Affiliation(s)
- Janet G M Markle
- Department of Immunology; University of Toronto; Toronto, ON Canada; Program in Genetics and Genomic Biology; The Hospital for Sick Children; Toronto, ON Canada
| | - Daniel N Frank
- Division of Infectious Diseases; University of Colorado; Aurora, CO USA
| | - Khosrow Adeli
- Department of Laboratory Medicine; The Hospital for Sick Children; Toronto, ON Canada; Department of Biochemistry; University of Toronto; Toronto, ON Canada
| | - Martin von Bergen
- Department of Metabolomics and Department of Proteomics; Helmholtz Center for Environmental Research; Leipzig, Germany; Department of Biotechnology, Chemistry, and Environmental Engineering; Aalborg University; Aalborg, Denmark
| | - Jayne S Danska
- Department of Immunology; University of Toronto; Toronto, ON Canada; Program in Genetics and Genomic Biology; The Hospital for Sick Children; Toronto, ON Canada; Department of Medical Biophysics; University of Toronto; Toronto, ON Canada
| |
Collapse
|
8
|
Li M, Song LJ, Qin XY. Advances in the cellular immunological pathogenesis of type 1 diabetes. J Cell Mol Med 2014; 18:749-58. [PMID: 24629100 PMCID: PMC4119381 DOI: 10.1111/jcmm.12270] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/30/2014] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease caused by the immune-mediated destruction of insulin-producing pancreatic β cells. In recent years, the incidence of type 1 diabetes continues to increase. It is supposed that genetic, environmental and immune factors participate in the damage of pancreatic β cells. Both the immune regulation and the immune response are involved in the pathogenesis of type 1 diabetes, in which cellular immunity plays a significant role. For the infiltration of CD4(+) and CD8(+) T lymphocyte, B lymphocytes, natural killer cells, dendritic cells and other immune cells take part in the damage of pancreatic β cells, which ultimately lead to type 1 diabetes. This review outlines the cellular immunological mechanism of type 1 diabetes, with a particular emphasis to T lymphocyte and natural killer cells, and provides the effective immune therapy in T1D, which is approached at three stages. However, future studies will be directed at searching for an effective, safe and long-lasting strategy to enhance the regulation of a diabetogenic immune system with limited toxicity and without global immunosuppression.
Collapse
Affiliation(s)
- Min Li
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Lu-Jun Song
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Xin-Yu Qin
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| |
Collapse
|
9
|
Mizrahi M, Cal P, Rosenthal M, Ochayon D, Shahaf G, Kaner Z, Kachker P, Lewis EC. Human α1-antitrypsin modifies B-lymphocyte responses during allograft transplantation. Immunology 2013; 140:362-73. [PMID: 23829472 DOI: 10.1111/imm.12149] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 06/29/2013] [Accepted: 07/01/2013] [Indexed: 12/19/2022] Open
Abstract
B-lymphocyte activities are associated with allograft rejection. Interleukin-10 (IL-10) -expressing B cells, however, exhibit regulatory attributes. Human α1-antitrypsin (hAAT), a clinically available anti-inflammatory circulating glycoprotein that rises during acute-phase responses, promotes semi-mature dendritic cells and regulatory T (Treg) cells during alloimmune responses. Whether B lymphocytes are also targets of hAAT activity has yet to be determined. Here, we examine whether hAAT modulates B-cell responses. In culture, hAAT reduced the lipopolysaccharide-stimulated Ki-67(+) B-cell population, IgM release and surface CD40 levels, but elevated IL-10-producing cells 1.5-fold. In CD40 ligand-stimulated cultures, hAAT promoted a similar trend; reduction in the Ki-67(+) B-cell population and in surface expression of CD86, CD80 and MHCII. hAAT increased interferon-γ-stimulated macrophage B-cell activating factor (BAFF) secretion, and reduced BAFF-receptor levels. Draining lymph nodes of transgenic mice that express circulating hAAT (C57BL/6 background) and that received skin allografts exhibited reduced B-lymphocyte activation compared with wild-type recipients. BSA-vaccinated hAAT transgenic mice exhibited 2.9-fold lower BSA-specific IgG levels, but 2.3-fold greater IgM levels, compared with wild-type mice. Circulating Treg cells were 1.3-fold greater in transgenic hAAT mice, but lower in B-cell knockout (BKO) and chimeric hAAT-BKO mice, compared with wild-type mice. In conclusion, B cells are cellular targets of hAAT. hAAT-induced Treg cell expansion appears to be B-cell-dependent. These changes support the tolerogenic properties of hAAT during immune responses, and suggest that hAAT may be beneficial in pathologies that involve excessive B-cell responses.
Collapse
Affiliation(s)
- Mark Mizrahi
- Faculty of Health Sciences, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Ahmed ST, Akirav E, Bradshaw E, Buckner J, McKinney E, Quintana FJ, Waldron-Lynch F, Nepom J. Immunological biomarkers: catalysts for translational advances in autoimmune diabetes. Clin Exp Immunol 2013; 172:178-85. [PMID: 23574315 DOI: 10.1111/cei.12063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2012] [Indexed: 12/20/2022] Open
Abstract
In a recent workshop organized by the JDRF focused on the 'Identification and Utilization of Robust Biomarkers in Type1 Diabetes', leaders in the field of type 1 diabetes (T1D)/autoimmunity and assay technology came together from academia, government and industry to assess the current state of the field, evaluate available resources/technologies and identify gaps that need to be filled for moving the field of T1D research forward. The highlights of this workshop are discussed in this paper, as well as the proposal for a larger, planned consortium effort, incorporating a JDRF Biomarker Core, to foster collaboration and accelerate progress in this critically needed area of T1D research.
Collapse
Affiliation(s)
- S T Ahmed
- JDRF, 26, Broadway, 14th Floor, New York, NY 10004, USA.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
It is widely accepted that Type 1 diabetes is a complex disease. Genetic predisposition and environmental factors favour the triggering of autoimmune responses against pancreatic β-cells, eventually leading to β-cell destruction. Over 40 susceptibility loci have been identified, many now mapped to known genes, largely supporting a dominant role for an immune-mediated pathogenesis. This role is also supported by the identification of several islet autoantigens and antigen-specific responses in patients with recent onset diabetes and subjects with pre-diabetes. Increasing evidence suggests certain viruses as a common environmental factor, together with diet and the gut microbiome. Inflammation and insulin resistance are emerging as additional cofactors, which might be interrelated with environmental factors. The heterogeneity of disease progression and clinical manifestations is likely a reflection of this multifactorial pathogenesis. So far, clinical trials have been mostly ineffective in delaying progression to overt diabetes in relatives at increased risk, or in reducing further loss of insulin secretion in patients with new-onset diabetes. This limited success may reflect, in part, our incomplete understanding of key pathogenic mechanisms, the lack of truly robust biomarkers of both disease activity and β-cell destruction, and the inability to assess the relative contributions of various pathogenic mechanisms at various time points during the course of the natural history of Type 1 diabetes. Emerging data and a re-evaluation of histopathological, immunological and metabolic findings suggest the hypothesis that unknown mechanisms of β-cell dysfunction may be present at diagnosis, and may contribute to the development of hyperglycaemia and clinical symptoms.
Collapse
Affiliation(s)
- A Pugliese
- Diabetes Research Institute, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
12
|
Han D, Cai X, Wen J, Kenyon NS, Chen Z. From biomarkers to a clue of biology: a computation-aided perspective of immune gene expression profiles in human type 1 diabetes. Front Immunol 2012; 3:320. [PMID: 23112798 PMCID: PMC3480653 DOI: 10.3389/fimmu.2012.00320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 10/02/2012] [Indexed: 01/25/2023] Open
Abstract
Dysregulated expression of key immune genes may cause breakdown of immunological tolerance and development of autoimmune disorders such as type 1 diabetes (T1D). General immune insufficiencies have also been implicated as a trigger of autoimmunity, due to their potential impact on immune homeostasis. Recent studies have detected evidence of systemic reduction in immune gene expression in long-term diabetic patients but the changes were not present before or at T1D onset. The changes could not be merely correlated with alteration in metabolic parameters. The studies also identified a dynamic expression pattern of several well-known as well as little-studied, immune-related genes during the course of T1D. An intriguing “ratio profile” of immune regulatory genes, such as CTLA4 and members of the S100 family, versus “baseline” immune genes, such as CD3G, prompted us to further examine immune gene expression relationships for a set of molecules representing T cells, B cells, and myeloid cells. No evidence was found to suggest an overall breach of tolerance equilibrium in T1D. Perplexingly, patients with long-term T1D presented a gene expression profile that was surprisingly more coordinated in analyses of “networking” relationship. Computational analyses of the “ratio profiles” or “relationship profiles” of immune gene expression might provide a clue for further studies of immunobiology in human T1D and other autoimmune diseases, as to how the profiles may be related to the pathogenic cause of the disease, to the effect of the diseases on immune homeostasis, or to an immunological process associated with the course of the diseases but is neither a direct cause nor a direct effect of the diseases.
Collapse
Affiliation(s)
- Dongmei Han
- Diabetes Research Institute, University of Miami Miller School of Medicine Miami, FL, USA
| | | | | | | | | |
Collapse
|
13
|
Bonner SM, Pietropaolo SL, Fan Y, Chang Y, Sethupathy P, Morran MP, Beems M, Giannoukakis N, Trucco G, Palumbo MO, Solimena M, Pugliese A, Polychronakos C, Trucco M, Pietropaolo M. Sequence variation in promoter of Ica1 gene, which encodes protein implicated in type 1 diabetes, causes transcription factor autoimmune regulator (AIRE) to increase its binding and down-regulate expression. J Biol Chem 2012; 287:17882-17893. [PMID: 22447927 PMCID: PMC3366781 DOI: 10.1074/jbc.m111.319020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 03/15/2012] [Indexed: 12/22/2022] Open
Abstract
ICA69 (islet cell autoantigen 69 kDa) is a protein implicated in type 1 diabetes mellitus in both the non-obese diabetic (NOD) mouse model and humans. ICA69 is encoded by the Ica1 gene on mouse chromosome 6 A1-A2. We previously reported reduced ICA69 expression in the thymus of NOD mice compared with thymus of several non-diabetic mouse strains. We propose that reduced thymic ICA69 expression could result from variations in transcriptional regulation of the gene and that polymorphisms within the Ica1 core promoter may partially determine this transcriptional variability. We characterized the functional promoter of Ica1 in NOD mice and compared it with the corresponding portions of Ica1 in non-diabetic C57BL/6 mice. Luciferase reporter constructs demonstrated that the NOD Ica1 promoter region exhibited markedly reduced luciferase expression in transiently transfected medullary thymus epithelial (mTEC(+)) and B-cell (M12)-derived cell lines. However, in a non-diabetic strain, C57BL/6, the Ica1 promoter region was transcriptionally active when transiently transfected into the same cell lines. We concomitantly identified five single nucleotide polymorphisms within the NOD Ica1 promoter. One of these single nucleotide polymorphisms increases the binding affinity for the transcription factor AIRE (autoimmune regulator), which is highly expressed in thymic epithelial cells, where it is known to play a key role regulating self-antigen expression. We conclude that polymorphisms within the NOD Ica1 core promoter may determine AIRE-mediated down-regulation of ICA69 expression in medullary thymic epithelial cells, thus providing a novel mechanistic explanation for the loss of immunologic tolerance to this self-antigen in autoimmunity.
Collapse
Affiliation(s)
- Samantha M Bonner
- Laboratory of Immunogenetics, Brehm Center for Diabetes Research, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Susan L Pietropaolo
- Laboratory of Immunogenetics, Brehm Center for Diabetes Research, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Yong Fan
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Yigang Chang
- Laboratory of Immunogenetics, Brehm Center for Diabetes Research, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Praveen Sethupathy
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Michael P Morran
- Laboratory of Immunogenetics, Brehm Center for Diabetes Research, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Megan Beems
- Laboratory of Immunogenetics, Brehm Center for Diabetes Research, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Nick Giannoukakis
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Giuliana Trucco
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Michael O Palumbo
- Endocrine Genetics Laboratory, Montreal Children Hospital-Research Institute, McGill University Health Center, Montreal, Quebec H3H 1P3, Canada
| | - Michele Solimena
- Department of Molecular Diabetology, Paul Langerhans Institute Dresden, Carl Gustav Carus School of Medicine, Dresden University of Technology, 01307 Dresden, Germany
| | - Alberto Pugliese
- Immunogenetics Program, Diabetes Research Institute, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Constantin Polychronakos
- Endocrine Genetics Laboratory, Montreal Children Hospital-Research Institute, McGill University Health Center, Montreal, Quebec H3H 1P3, Canada
| | - Massimo Trucco
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Massimo Pietropaolo
- Laboratory of Immunogenetics, Brehm Center for Diabetes Research, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105.
| |
Collapse
|
14
|
Anti-idiotypic antibody specific to GAD65 autoantibody prevents type 1 diabetes in the NOD mouse. PLoS One 2012; 7:e32515. [PMID: 22384267 PMCID: PMC3286479 DOI: 10.1371/journal.pone.0032515] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 01/27/2012] [Indexed: 12/31/2022] Open
Abstract
Overt autoantibodies to the smaller isoform of glutamate decarboxylase (GAD65Ab) are a characteristic in patients with Type 1 diabetes (T1D). Anti-idiotypic antibodies (anti-Id) directed to GAD65Ab effectively prevent the binding of GAD65 to GAD65Ab in healthy individuals. Levels of GAD65Ab-specific anti-Id are significantly lower in patients with T1D, leading to overt GAD65Ab in these patients. To determine the possible protective role of GAD65Ab-specific anti-Id in T1D pathogenesis, we developed the monoclonal anti-Id MAb 8E6G4 specifically targeting human monoclonal GAD65Ab b96.11. MAb 8E6G4 was demonstrated as a specific anti-Id directed to the antigen binding site of b96.11. MAb 8E6G4 recognized human antibodies in sera from healthy individuals, T2D patients, and T1D patients as established by ELISA. We confirmed these MAb 8E6G4-bound human antibodies to contain GAD65Ab by testing the eluted antibodies for binding to GAD65 in radioligand binding assays. These findings confirm that GAD65Ab are present in sera of individuals, who test GAD65Ab-negative in conventional detection assays. To test our hypothesis that GAD65Ab-specific anti-Id have an immune modulatory role in T1D, we injected young Non Obese Diabetic (NOD) mice with MAb 8E6G4. The animals were carefully monitored for development of T1D for 40 weeks. Infiltration of pancreatic islets by mononuclear cells (insulitis) was determined to establish the extent of an autoimmune attack on the pancreatic islets. Administration of MAb 8E6G4 significantly reduced the cumulative incidence rate of T1D and delayed the time of onset. Insulitis was significantly less severe in animals that received MAb 8E6G4 as compared to control animals. These results support our hypothesis that anti-Id specific to GAD65Ab have a protective role in T1D.
Collapse
|
15
|
Abstract
The role of B cells in autoimmune diseases involves different cellular functions, including the well-established secretion of autoantibodies, autoantigen presentation and ensuing reciprocal interactions with T cells, secretion of inflammatory cytokines, and the generation of ectopic germinal centers. Through these mechanisms B cells are involved both in autoimmune diseases that are traditionally viewed as antibody mediated and also in autoimmune diseases that are commonly classified as T cell mediated. This new understanding of the role of B cells opened up novel therapeutic options for the treatment of autoimmune diseases. This paper includes an overview of the different functions of B cells in autoimmunity; the involvement of B cells in systemic lupus erythematosus, rheumatoid arthritis, and type 1 diabetes; and current B-cell-based therapeutic treatments. We conclude with a discussion of novel therapies aimed at the selective targeting of pathogenic B cells.
Collapse
Affiliation(s)
- Christiane S. Hampe
- Department of Medicine, University of Washington, SLU-276, 850 Republican, Seattle, WA 98109, USA
- *Christiane S. Hampe:
| |
Collapse
|
16
|
Spectratyping analysis of the islet-reactive T cell repertoire in diabetic NOD Igμ(null) mice after polyclonal B cell reconstitution. J Transl Med 2011; 9:101. [PMID: 21722394 PMCID: PMC3141497 DOI: 10.1186/1479-5876-9-101] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 07/02/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Non Obese Diabetic mice lacking B cells (NOD.Igμ(null) mice) do not develop diabetes despite their susceptible background. Upon reconstitution of B cells using a chimera approach, animals start developing diabetes at 20 weeks of age. METHODS We have used the spectratyping technique to follow the T cell receptor (TCR) V beta repertoire of NOD.Igμ(null) mice following B cell reconstitution. This technique provides an unbiased approach to understand the kinetics of TCR expansion. We have also analyzed the TCR repertoire of reconstituted animals receiving cyclophosphamide treatment and following tissue transplants to identify common aggressive clonotypes. RESULTS We found that B cell reconstitution of NOD.Igμ(null) mice induces a polyclonal TCR repertoire in the pancreas 10 weeks later, gradually diversifying to encompass most BV families. Interestingly, these clonotypic BV expansions are mainly confined to the pancreas and are absent from pancreatic lymph nodes or spleens. Cyclophosphamide-induced diabetes at 10 weeks post-B cell reconstitution reorganized the predominant TCR repertoires by removing potential regulatory clonotypes (BV1, BV8 and BV11) and increasing the frequency of others (BV4, BV5S2, BV9, BV16-20). These same clonotypes are more frequently present in neonatal pancreatic transplants under the kidney capsule of B-cell reconstituted diabetic NOD.Igμ(null) mice, suggesting their higher invasiveness. Phenotypic analysis of the pancreas-infiltrating lymphocytes during diabetes onset in B cell reconstituted animals show a predominance of CD19+ B cells with a B:T lymphocyte ratio of 4:1. In contrast, in other lymphoid organs (pancreatic lymph nodes and spleens) analyzed by FACS, the B:T ratio was 1:1. Lymphocytes infiltrating the pancreas secrete large amounts of IL-6 and are of Th1 phenotype after CD3-CD28 stimulation in vitro. CONCLUSIONS Diabetes in NOD.Igμ(null) mice appears to be caused by a polyclonal repertoire of T cell accumulation in pancreas without much lymphoid organ involvement and is dependent on the help by B cells.
Collapse
|
17
|
Abstract
BACKGROUND Type 1 diabetes (T1DM) results from cell-mediated autoimmune destruction of the β cells of the islets of Langerhans. Autoantibodies directed against the islets are useful clinical tools that allow the recognition and confirmation of β-cell autoimmunity. CONTENT In this review we define the term "islet autoantibody," describe the pathogenesis of autoantibody generation, and explain the uses of islet autoantibodies in clinical medicine and in research studies that concern the interruption or prevention of T1DM. We also discuss the biology of islet autoantibodies and their rates of appearance at the time of onset of T1DM and their appearance before the development of T1DM. SUMMARY The presence of islet autoantibodies in persons with diabetes confirms an autoimmune etiology. In nondiabetic individuals, islet autoantibodies are strong predictors of the later development of T1DM.
Collapse
Affiliation(s)
- William E Winter
- Department of Pathology, University of Florida, Gainesville, FL 32610-0275, USA.
| | | |
Collapse
|
18
|
Wenzlau JM, Walter M, Gardner TJ, Frisch LM, Yu L, Eisenbarth GS, Ziegler AG, Davidson HW, Hutton JC. Kinetics of the post-onset decline in zinc transporter 8 autoantibodies in type 1 diabetic human subjects. J Clin Endocrinol Metab 2010; 95:4712-9. [PMID: 20610599 PMCID: PMC3050104 DOI: 10.1210/jc.2010-0169] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CONTEXT Zinc transporter 8 (ZnT8) is a newly discovered islet autoantigen in human type 1A diabetes (T1D). OBJECTIVE The objective was to document changes in ZnT8 autoantibody (ZnT8A) titer and prevalence after onset of disease in relationship to 65 kDa glutamate decarboxylase antibody (GADA) and islet cell antigen antibody (IA2A). DESIGN/PATIENTS Autoantibody radioimmunoprecipitation assays were performed on sera from three groups: 21 individuals monitored every 3 months from diagnosis for 2.5 yr; 61 individuals monitored at six monthly intervals for 5-12 yr; and a cross-sectional study of 424 patients with T1D of 20-57 yr duration. Circulating C-peptide was determined as an index of residual β-cell function. RESULTS ZnT8A titers declined exponentially from clinical onset of T1D with a t(1/2) ranging from 26 to 530 wk, similar to C-peptide (23-300 wk). Life-table analysis of antibody prevalence to 12 yr indicated that ZnT8A measured with either Arg325 or Trp325 probes persisted for a shorter interval than IA2A. Although prevalence of ZnT8A, IA2A, and GADA were comparable at disease onset (70.4 vs. 73.4 vs. 64%), only 6.7% of individuals remained ZnT8A positive after 25 yr compared with 19.5% for IA2A and 25.9% for GADA. Titers of ZnT8A and IA2A in seropositive individuals decreased progressively, whereas GADA remained elevated consistent with periodic reactivation of GADA humoral autoimmunity. CONCLUSIONS ZnT8 humoral autoreactivity declines rapidly in the first years after disease onset and is less persistent than IA2A or GADA in the longer term. ZnT8A determination may be a useful measure of therapeutic efficacy in the context of immune-based clinical interventions.
Collapse
Affiliation(s)
- J M Wenzlau
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Sauerborn M, Schellekens H. B-1 cells and naturally occurring antibodies: influencing the immunogenicity of recombinant human therapeutic proteins? Curr Opin Biotechnol 2009; 20:715-21. [PMID: 19892544 DOI: 10.1016/j.copbio.2009.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 10/14/2009] [Indexed: 12/18/2022]
Abstract
Recombinant human therapeutic proteins are increasingly being used to treat serious and life-threatening diseases like multiple sclerosis, diabetes mellitus, and cancer. An important side effect of these proteins is the development of antidrug antibodies, which can be neutralizing and thus interfere with the efficacy and safety of the drug. Some biophysical properties, for example, aggregation, also can initiate the immunogenic response to human therapeutics. Many other factors including patients' characteristics may influence this response. Besides induced antibodies, autoantibodies (i.e. naturally occurring antibodies [NAs]) against therapeutic relevant proteins in naïve patients are increasingly being identified. The role of autoreactive B cells and their escape from deletion, production of NAs and their pivotal function in the immune system, the dualistic role of B-1 cells in autoimmunity, and the influence of NAs on disease outcome and their possible impact on the efficacy of human therapeutics will be presented and discussed.
Collapse
Affiliation(s)
- Melody Sauerborn
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, 3584 CA Utrecht, The Netherlands.
| | | |
Collapse
|
20
|
Sanz I. Indications of rituximab in autoimmune diseases. DRUG DISCOVERY TODAY. THERAPEUTIC STRATEGIES 2009; 6:13-19. [PMID: 20379381 PMCID: PMC2850537 DOI: 10.1016/j.ddstr.2009.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Iñaki Sanz
- Division of Allergy, Immunology and Rheumatology University of Rochester School of Medicine and Dentistry
| |
Collapse
|