1
|
Kim KM, D'Elia AM, Rodell CB. Hydrogel-based approaches to target hypersensitivity mechanisms underlying autoimmune disease. Adv Drug Deliv Rev 2024; 212:115395. [PMID: 39004347 DOI: 10.1016/j.addr.2024.115395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
A robust adaptive immune response is essential for combatting pathogens. In the wrong context such as due to genetic and environmental factors, however, the same mechanisms crucial for self-preservation can lead to a loss of self-tolerance. Resulting autoimmunity manifests in the development of a host of organ-specific or systemic autoimmune diseases, hallmarked by aberrant immune responses and tissue damage. The prevalence of autoimmune diseases is on the rise, medical management of which focuses primarily on pharmacological immunosuppression that places patients at a risk of side effects, including opportunistic infections and tumorigenesis. Biomaterial-based drug delivery systems confer many opportunities to address challenges associated with conventional disease management. Hydrogels, in particular, can protect encapsulated cargo (drug or cell therapeutics) from the host environment, afford their presentation in a controlled manner, and can be tailored to respond to disease conditions or support treatment via multiplexed functionality. Moreover, localized delivery to affected sites by these approaches has the potential to concentrate drug action at the site, reduce off-target exposure, and enhance patient compliance by reducing the need for frequent administration. Despite their many benefits for the management of autoimmune disease, such biomaterial-based approaches focus largely on the downstream effects of hypersensitivity mechanisms and have a limited capacity to eradicate the disease. In contrast, direct targeting of mechanisms of hypersensitivity reactions uniquely enables prophylaxis or the arrest of disease progression by mitigating the basis of autoimmunity. One promising approach is to induce self-antigen-specific tolerance, which specifically subdues damaging autoreactivity while otherwise retaining the normal immune responses. In this review, we will discuss hydrogel-based systems for the treatment of autoimmune disease, with a focus on those that target hypersensitivity mechanisms head-on. As the field continues to advance, it will expand the range of therapeutic choices for people coping with autoimmune diseases, providing fresh prospects for better clinical outcomes and improved quality of life.
Collapse
Affiliation(s)
- Kenneth M Kim
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| | - Christopher B Rodell
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Root-Bernstein R. T-Cell Receptor Sequences Identify Combined Coxsackievirus- Streptococci Infections as Triggers for Autoimmune Myocarditis and Coxsackievirus- Clostridia Infections for Type 1 Diabetes. Int J Mol Sci 2024; 25:1797. [PMID: 38339075 PMCID: PMC10855694 DOI: 10.3390/ijms25031797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/19/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Recent research suggests that T-cell receptor (TCR) sequences expanded during human immunodeficiency virus and SARS-CoV-2 infections unexpectedly mimic these viruses. The hypothesis tested here is that TCR sequences expanded in patients with type 1 diabetes mellitus (T1DM) and autoimmune myocarditis (AM) mimic the infectious triggers of these diseases. Indeed, TCR sequences mimicking coxsackieviruses, which are implicated as triggers of both diseases, are statistically significantly increased in both T1DM and AM patients. However, TCRs mimicking Clostridia antigens are significantly expanded in T1DM, whereas TCRs mimicking Streptococcal antigens are expanded in AM. Notably, Clostridia antigens mimic T1DM autoantigens, such as insulin and glutamic acid decarboxylase, whereas Streptococcal antigens mimic cardiac autoantigens, such as myosin and laminins. Thus, T1DM may be triggered by combined infections of coxsackieviruses with Clostridia bacteria, while AM may be triggered by coxsackieviruses with Streptococci. These TCR results are consistent with both epidemiological and clinical data and recent experimental studies of cross-reactivities of coxsackievirus, Clostridial, and Streptococcal antibodies with T1DM and AM antigens. These data provide the basis for developing novel animal models of AM and T1DM and may provide a generalizable method for revealing the etiologies of other autoimmune diseases. Theories to explain these results are explored.
Collapse
|
3
|
Root-Bernstein R, Chiles K, Huber J, Ziehl A, Turke M, Pietrowicz M. Clostridia and Enteroviruses as Synergistic Triggers of Type 1 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24098336. [PMID: 37176044 PMCID: PMC10179352 DOI: 10.3390/ijms24098336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/24/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
What triggers type 1 diabetes mellitus (T1DM)? One common assumption is that triggers are individual microbes that mimic autoantibody targets such as insulin (INS). However, most microbes highly associated with T1DM pathogenesis, such as coxsackieviruses (COX), lack INS mimicry and have failed to induce T1DM in animal models. Using proteomic similarity search techniques, we found that COX actually mimicked the INS receptor (INSR). Clostridia were the best mimics of INS. Clostridia antibodies cross-reacted with INS in ELISA experiments, confirming mimicry. COX antibodies cross-reacted with INSR. Clostridia antibodies further bound to COX antibodies as idiotype-anti-idiotype pairs conserving INS-INSR complementarity. Ultraviolet spectrometry studies demonstrated that INS-like Clostridia peptides bound to INSR-like COX peptides. These complementary peptides were also recognized as antigens by T cell receptor sequences derived from T1DM patients. Finally, most sera from T1DM patients bound strongly to inactivated Clostridium sporogenes, while most sera from healthy individuals did not; T1DM sera also exhibited evidence of anti-idiotype antibodies against idiotypic INS, glutamic acid decarboxylase, and protein tyrosine phosphatase non-receptor (islet antigen-2) antibodies. These results suggest that T1DM is triggered by combined enterovirus-Clostridium (and possibly combined Epstein-Barr-virus-Streptococcal) infections, and the probable rate of such co-infections approximates the rate of new T1DM diagnoses.
Collapse
Affiliation(s)
| | - Kaylie Chiles
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Jack Huber
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Alison Ziehl
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Miah Turke
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Maja Pietrowicz
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
4
|
The dark side of insulin: A primary autoantigen and instrument of self-destruction in type 1 diabetes. Mol Metab 2021; 52:101288. [PMID: 34242821 PMCID: PMC8513143 DOI: 10.1016/j.molmet.2021.101288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/27/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Background Since its discovery 100 years ago, insulin, as the ‘cure’ for type 1 diabetes, has rescued the lives of countless individuals. As the century unfolded and the autoimmune nature of type 1 diabetes was recognised, a darker side of insulin emerged. Autoimmunity to insulin was found to be an early marker of risk for type 1 diabetes in young children. In humans, it remains unclear if autoimmunity to insulin is primarily due to a defect in the beta cell itself or to dysregulated immune activation. Conversely, it may be secondary to beta-cell damage from an environmental agent (e.g., virus). Nevertheless, direct, interventional studies in non-obese diabetic (NOD) mouse models of type 1 diabetes point to a critical role for (pro)insulin as a primary autoantigen that drives beta cell pathology. Scope of review Modelled on Koch's postulates for the pathogenicity of an infectious agent, evidence for a pathogenic role of (pro)insulin as an autoantigen in type 1 diabetes, particularly applicable to the NOD mouse model, is reviewed. Evidence in humans remains circumstantial. Additionally, as (pro)insulin is a target of autoimmunity in type 1 diabetes, its application as a therapeutic tool to elicit antigen-specific immune tolerance is assessed. Major conclusions Paradoxically, insulin is both a ‘cure’ and a potential ‘cause’ of type 1 diabetes, actively participating as an autoantigen to drive autoimmune destruction of beta cells - the instrument of its own destruction.
Collapse
|
5
|
Liu M, Wang Z, Feng D, Shang Y, Li X, Liu J, Li C, Yang Z. An Insulin-Inspired Supramolecular Hydrogel for Prevention of Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003599. [PMID: 34026440 PMCID: PMC8132061 DOI: 10.1002/advs.202003599] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/11/2021] [Indexed: 05/10/2023]
Abstract
Supramolecular peptide hydrogel has shown promising potential in vaccine development largely because of its ability to function both as antigen depot and immune adjuvant. Nap-GdFdFdY, a tetrapeptide hydrogel that has been previously reported to exhibit adjuvant effect, is inadvertently found to contain conserved peptide sequence for insulin, proinsulin, and glutamic acid decarboxylase, 3 major autoantigens for the autoimmune type 1 diabetes (T1D). At present, despite being managed clinically with insulin replacement therapy, T1D remains a major health threat with rapidly increasing incidences, especially in children and young adults, and antigen-specific immune tolerance induction has been proposed as a feasible approach to prevent or delay T1D progression at an early stage. Here, it is reported that innoculation of Nap-GdFdFdY leads to complete protection of nonobese diabetic (NOD) mice from T1D development till the age of 36 weeks. Better maintenance of pancreatic islet morphology with minimal immune cell infiltration is also observed from mice exposed to Nap-GdFdFdY. This beneficial impact is mainly due to its facilitative role on enhancing peripheral T regulatory cell (Treg) population, shown as increased splenic Treg percentage, and function, demonstrated by maintenance of circulating TGF-β1 level. Serum cytokine microarray data further implicate a "buffering" role of Nap-GdFdFdY on systemic inflammatory tone in NOD mice. Thus, with its versatility, applicability, and excellent potency, Nap-GdFdFdY is posited as a novel therapeutic intervention for T1D.
Collapse
Affiliation(s)
- Mohan Liu
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhongyan Wang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Dandan Feng
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Yuna Shang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
| | - Xinxin Li
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Chen Li
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhimou Yang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu221004P. R. China
| |
Collapse
|
6
|
Finotti P, Pagetta A. A mutant α1antitrypsin in complex with heat shock proteins as the primary antigen in type 1 diabetes in silico investigation. Sci Rep 2021; 11:3002. [PMID: 33542414 PMCID: PMC7862655 DOI: 10.1038/s41598-021-82730-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/25/2021] [Indexed: 11/09/2022] Open
Abstract
Based on previous results demonstrating that complexes of a mutant α1-antitrypsin with the heat shock proteins (HSP)70 and glucose-regulated protein94 (Grp94) circulate in the blood of patients with type 1 diabetes, we raised the hypothesis that these complexes could represent the primary antigen capable of triggering the autoimmune reactions leading to overt diabetes. As a first approach to this issue, we searched whether A1AT and HSPs had a sequence similarity to major islet antigen proteins so as to identify among the similar sequences those with potential relevance for the pathogenesis of diabetes. A thorough in silico analysis was performed to establish the score of similarity of the human proteins: A1AT, pro-insulin (INS), GAD65, IAPP, IA-2, ICA69, Grp94, HSP70 and HSP60. The sequences of A1AT and HSPs with the highest score of similarity to the islet peptides reported in the literature as the main autoantigens in human diabetes were recorded. At variance with other HSPs, also including HSP90 and Grp78, Grp94 contained the highest number and the longest sequences with structural similarity to A1AT and to well-known immunogenic peptides/epitopes of INS, GAD65, and IA-2. The similarity of A1AT with Grp94 and that of Grp94 with INS also suggested a functional relationship among the proteins. Specific sequences were identified in A1AT, Grp94 and HSP70, with the highest score of cross-similarity to a pattern of eight different islet protein epitopes. The similarity also involved recently discovered autoantigens in type 1 diabetes such as a hybrid peptides of insulin and the defective ribosomal insulin gene product. The significant similarity displayed by specific sequences of Grp94 and A1AT to the islet peptides considered main antigens in human diabetes, is a strong indication for testing these sequences as new peptides of immunogenic relevance in diabetes.
Collapse
Affiliation(s)
- Paola Finotti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Building "C", Largo E. Meneghetti, 2, 35131, Padua, Italy.
| | - Andrea Pagetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Building "C", Largo E. Meneghetti, 2, 35131, Padua, Italy
| |
Collapse
|
7
|
Villalba A, Rodriguez-Fernandez S, Ampudia RM, Cano-Sarabia M, Perna-Barrull D, Bertran-Cobo C, Ehrenberg C, Maspoch D, Vives-Pi M. Preclinical evaluation of antigen-specific nanotherapy based on phosphatidylserine-liposomes for type 1 diabetes. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 48:77-83. [DOI: 10.1080/21691401.2019.1699812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Adrian Villalba
- Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain
| | - Silvia Rodriguez-Fernandez
- Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain
| | - Rosa-Maria Ampudia
- Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain
| | - Mary Cano-Sarabia
- Catalan Institute of Nanoscience and Nanotechnology, Bellaterra, Spain
| | - David Perna-Barrull
- Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain
| | - Cesc Bertran-Cobo
- Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain
| | - Clara Ehrenberg
- Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology, Bellaterra, Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Marta Vives-Pi
- Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain
- CIBER of Diabetes and Associated Metabolic Disease (CIBERDEM). ISCIII, Barcelona, Spain
| |
Collapse
|
8
|
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease that causes severe loss of pancreatic β cells. Autoreactive T cells are key mediators of β cell destruction. Studies of organ donors with T1D that have examined T cells in pancreas, the diabetogenic insulitis lesion, and lymphoid tissues have revealed a broad repertoire of target antigens and T cell receptor (TCR) usage, with initial evidence of public TCR sequences that are shared by individuals with T1D. Neoepitopes derived from post-translational modifications of native antigens are emerging as novel targets that are more likely to evade self-tolerance. Further studies will determine whether T cell responses to neoepitopes are major disease drivers that could impact prediction, prevention, and therapy. This Review provides an overview of recent progress in our knowledge of autoreactive T cells that has emerged from experimental and clinical research as well as pathology investigations.
Collapse
|
9
|
Wan X, Zaghouani H. Antigen-specific therapy against type 1 diabetes: mechanisms and perspectives. Immunotherapy 2014; 6:155-64. [PMID: 24491089 DOI: 10.2217/imt.13.172] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Type 1 diabetes (T1D) is an immune-mediated disease that occurs when the insulin-producing β‑cells of the pancreatic islets are destroyed by an inflammatory process perpetuated by cells of the immune system. The logical approach to suppress T1D is to inactivate or eliminate the lymphocytes responsible for inducing inflammation and targeting the β‑cells. Antigen-specific approaches have been devised and were able to target inflammatory lymphocytes and induce apoptosis or block trafficking to pancreatic islets. Lack of costimulation, expansion of Tregs and bystander suppression are likely mechanisms by which antigen-specific treatments modulate pathogenic T cells. This strategy, however, while prevents the onset of T1D, could not overcome overt T1D, perhaps because of collateral damage to the islet vascular network. Recent developments indicate that donor endothelial stem cell precursors can repair the islets' vascular niche and assist antigen-specific therapy against overt T1D.
Collapse
Affiliation(s)
- Xiaoxiao Wan
- Department of Pathology & Immunology. Washington University School of Medicine, St Louis, MO 63110, USA
| | | |
Collapse
|
10
|
Oldstone MBA. Molecular mimicry: its evolution from concept to mechanism as a cause of autoimmune diseases. Monoclon Antib Immunodiagn Immunother 2014; 33:158-65. [PMID: 24694269 DOI: 10.1089/mab.2013.0090] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
On a clonal level, certain antibodies and T cells can interact with dissimilar antigens found in microbes and in host cells. More than 5% of over 800 monoclonal antibodies derived from multiple RNA and DNA viruses, as well as from a large number of T cell clones, engage in such interactions. Several of these cross-reactions, which we termed molecular mimicry, are against unique host proteins involved in autoimmune responses and diseases. Thus, molecular mimicry initiated as a host response to a virus or a microbial infection, but alternatively cross-reacting with an appropriate host-antigen, can be a mechanism for instigating an autoimmune disease. Molecular mimicry provides an explanation for the genetic observation that identical twins rarely manifest the same autoimmune disease and the documented epidemiologic evidence that microbial and/or viral infections often precede autoimmune disorders.
Collapse
Affiliation(s)
- Michael B A Oldstone
- The Scripps Research Institute , Department of Immunology and Microbial Science, La Jolla, California
| |
Collapse
|
11
|
Wright GP, Ehrenstein MR, Stauss HJ. Regulatory T-cell adoptive immunotherapy: potential for treatment of autoimmunity. Expert Rev Clin Immunol 2014; 7:213-25. [DOI: 10.1586/eci.10.96] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Abstract
Type 1 diabetes (T1D) represents 10 to 15% of all forms of diabetes. Its incidence shows a consistent rise in all countries under survey. Evidence for autoimmunity in human T1D relies on the detection of insulitis, of islet cell antibodies, of activated β-cell-specific T lymphocytes and on the association of T1D with a restricted set of class II major histocompatibility complex (MHC) alleles. However, mechanisms that initiate the failure of immune tolerance to β-cell autoantigens remain elusive in common forms of T1D. T1D commonly develop as a multifactorial disease in which environmental factors concur with a highly multigenic background. The disease is driven by the activation of T-lymphocytes against pancreatic β-cells. Several years elapse between initial triggering of the autoimmune response to β cells, as evidenced by the appearance or islet cell autoantibodies, and the onset of clinical diabetes, defining a prediabetes stage. Active mechanisms hold back autoreactive effector T-cells in prediabetes, in particular a subset of CD4+ T-cells (T(reg)) and other regulatory T-cells, such as invariant NKT cells. There is evidence in experimental models that systemic or local infections can trigger autoimmune reactions to β-cells. However, epidemiological observations that have accumulated over years have failed to identify undisputable environmental factors that trigger T1D. Moreover, multiple environmental factors may intervene in the disease evolution and protective as weel as triggering environmental factors may be involved. Available models also indicate that local signals within the islets are required for full-blown diabetes to develop. Many autoantigens that are expressed by β-cells but also by the other endocrine islet cells and by neurons are recognized by lymphocytes along the development of T1D. The immune image of β-cells is that of native components of the β-cell membrane, as seen by B-lymphocytes, and of fragments of intracellular β-cell proteins in the form of peptides loaded onto class I MHC molecules on the β-cell surface and class I and class II molecules onto professional antigen presenting cells. Given the key role of T lymphocytes in T1D, the cartography of autoantigen-derived peptides that are presented to class I-restricted CD8(+) T-cells and class II-restricted CD4(+) T-cells is of outmost importance and is a necessary step in the development of diagnostic T-cell assays and of immunotherapy of T1D.
Collapse
|
13
|
T cell recognition of autoantigens in human type 1 diabetes: clinical perspectives. Clin Dev Immunol 2011; 2011:513210. [PMID: 21785617 PMCID: PMC3140193 DOI: 10.1155/2011/513210] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 03/18/2011] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease driven by the activation of lymphocytes against pancreatic β-cells. Among β-cell autoantigens, preproinsulin has been ascribed a key role in the T1D process. The successive steps that control the activation of autoreactive lymphocytes have been extensively studied in animal models of T1D, but remains ill defined in man. In man, T lymphocytes, especially CD8+ T cells, are predominant within insulitis. Developing T-cell assays in diabetes autoimmunity is, thus, a major challenge. It is expected to help defining autoantigens and epitopes that drive the disease process, to pinpoint key functional features of epitope-specific T lymphocytes along the natural history of diabetes and to pave the way towards therapeutic strategies to induce immune tolerance to β-cells. New T-cell technologies will allow defining autoreactive T-cell differentiation programs and characterizing autoimmune responses in comparison with physiologically appropriate immune responses. This may prove instrumental in the discovery of immune correlates of efficacy in clinical trials.
Collapse
|
14
|
Chaparro RJ, Dilorenzo TP. An update on the use of NOD mice to study autoimmune (Type 1) diabetes. Expert Rev Clin Immunol 2011; 6:939-55. [PMID: 20979558 DOI: 10.1586/eci.10.68] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The widely used nonobese diabetic (NOD) mouse model of autoimmune (Type 1) diabetes mellitus shares multiple characteristics with the human disease, and studies employing this model continue to yield clinically relevant and important information. Here, we review some of the recent key findings obtained from NOD mouse investigations that have both advanced our understanding of disease pathogenesis and suggested new therapeutic targets and approaches. Areas discussed include antigen discovery, identification of genes and pathways contributing to disease susceptibility, development of strategies to image islet inflammation and the testing of therapeutics. We also review recent technical advances that, combined with an improved understanding of the NOD mouse model's limitations, should work to ensure its popularity, utility and relevance in the years ahead.
Collapse
Affiliation(s)
- Rodolfo José Chaparro
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
15
|
[Antitumor effects of raddeanin A on S180, H22 and U14 cell xenografts in mice]. Curr Opin Immunol 2010; 20:111-8. [PMID: 18799026 DOI: 10.1016/j.coi.2007.11.005] [Citation(s) in RCA: 154] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 11/19/2007] [Accepted: 11/19/2007] [Indexed: 01/19/2023]
Abstract
BACKGROUND & OBJECTIVE Raddeanin A, a triterpenoid saponin from Anemone raddeana Regel, has good antitumor activity in vitro. This study was to investigate its antitumor effects on tumor cell xenografts in mice. METHODS The inhibitory effects of raddeanin A on the proliferation of human nasopharyngeal carcinoma KB cells and ovarian cancer SKOV3 cells were measured by MTT assay. The inhibitory effects of raddeanin A injection on the growth of sarcoma S180, liver cancer H22 and cervical carcinoma U14 cell xenografts in mice and the effect of raddeanin A lavage on the growth of S180 cell xenografts were measured. The acute toxicity of raddeanin A was also measured. RESULTS The 50% inhibition concentration (IC(50)) of raddeanin A was 4.64 microg/mL for KB cells and 1.40 microg/mL for SKOV3 cells. When injected with raddeanin A at a dose of 4.5 mg/kg, the growth inhibition rates of S180, H22 and U14 cell xenografts were 60.5%, 36.2% and 61.8%, respectively. When lavaged with raddeanin A at a dose of 200 mg/kg, the growth inhibition rate of S180 cell xenografts was 64.7%. The median lethal dose (LD50) of raddeanin A lavage was 1.1 g/kg and that of raddeanin A injection was 16.1 mg/kg. CONCLUSION Raddeanin A has good antitumor activity both in vitro and in vivo, and would be a potential antitumor medicine.
Collapse
|
16
|
Pugliese A. Insulin: a critical autoantigen and potential therapeutic agent in Type 1 diabetes. Expert Rev Clin Immunol 2010; 2:419-31. [PMID: 20476913 DOI: 10.1586/1744666x.2.3.419] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Insulin is a polypeptide hormone secreted by pancreatic beta-cells and is critical for glucose homeostasis. Abnormalities in insulin secretion result in various forms of diabetes. Type 1A diabetes is an autoimmune form in which insulin has been identified as a critical autoantigen. Recent studies have identified genetic determinants of insulin-specific autoimmune responses and insulin epitopes targeted by autoreactive T lymphocytes. The study of insulin as an autoantigen has also led to discoveries about basic mechanisms of immunological tolerance and autoimmunity. Experimental and clinical evidence suggests that insulin and insulin-derived peptides may delay and perhaps prevent the development of diabetes. Further clinical trials may identify effective treatment modalities for inhibiting diabetogenic autoimmunity and preventing disease development.
Collapse
Affiliation(s)
- Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136, USA.
| |
Collapse
|
17
|
Shehadeh N, Pollack S, Wildbaum G, Zohar Y, Shafat I, Makhoul R, Daod E, Hakim F, Perlman R, Karin N. Selective autoantibody production against CCL3 is associated with human type 1 diabetes mellitus and serves as a novel biomarker for its diagnosis. THE JOURNAL OF IMMUNOLOGY 2009; 182:8104-9. [PMID: 19494336 DOI: 10.4049/jimmunol.0803348] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have recently demonstrated that patients suffering from chronic autoimmune diseases develop an autoantibody response against key mediators that participate in the initiation and progression of these diseases. In this paper, we show that patients with type 1 diabetes mellitus (T1DM), but not those suffering from several other inflammatory autoimmune diseases, display a selective autoantibody titer to a single CC chemokine named CCL3. From the diagnostic point we show that this response could be used as a biomarker for diagnosis of T1DM, a disease that is currently diagnosed by autoantibodies to competitive anti-insulin Abs, islet cell Abs, and glutamic acid decarboxylase Abs. We show that our currently suggested biomarker is more reliable than each of the above alone, including diagnosis of T1DM at its preclinical stage, and could therefore be used as a novel way for diagnosis of T1DM. These Abs were found to be neutralizing Abs. It is possible, though hard to prove, that these Abs participate in the natural regulation of the human disease. Hence, it has previously been shown by others that selective neutralization of CCL3 suppresses T1DM in NOD mice. Theses results together with ours suggest CCL3 as a preferential target for therapy of T1DM.
Collapse
Affiliation(s)
- Naim Shehadeh
- Department of Immunology, Rambam Medical Center, Technion, Haifa, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Toma A, Laïka T, Haddouk S, Luce S, Briand JP, Camoin L, Connan F, Lambert M, Caillat-Zucman S, Carel JC, Muller S, Choppin J, Lemonnier F, Boitard C. Recognition of human proinsulin leader sequence by class I-restricted T-cells in HLA-A*0201 transgenic mice and in human type 1 diabetes. Diabetes 2009; 58:394-402. [PMID: 19011169 PMCID: PMC2628613 DOI: 10.2337/db08-0599] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE A restricted region of proinsulin located in the B chain and adjacent region of C-peptide has been shown to contain numerous candidate epitopes recognized by CD8(+) T-cells. Our objective is to characterize HLA class I-restricted epitopes located within the preproinsulin leader sequence. RESEARCH DESIGN AND METHODS Seven 8- to 11-mer preproinsulin peptides carrying anchoring residues for HLA-A1, -A2, -A24, and -B8 were selected from databases. HLA-A2-restricted peptides were tested for immunogenicity in transgenic mice expressing a chimeric HLA-A*0201/beta2-microglobulin molecule. The peptides were studied for binding to purified HLA class I molecules, selected for carrying COOH-terminal residues generated by proteasome digestion in vitro and tested for recognition by human lymphocytes using an ex vivo interferon-gamma (IFN-gamma) ELISpot assay. RESULTS Five HLA-A2-restricted peptides were immunogenic in transgenic mice. Murine T-cell clones specific for these peptides were cytotoxic against cells transfected with the preproinsulin gene. They were recognized by peripheral blood mononuclear cells (PBMCs) from 17 of 21 HLA-A2 type 1 diabetic patients. PBMCs from 25 of 38 HLA-A1, -A2, -A24, or -B8 patients produced IFN-gamma in response to six preproinsulin peptides covering residues 2-25 within the preproinsulin region. In most patients, the response was against several class I-restricted peptides. T-cells recognizing preproinsulin peptide were characterized as CD8(+) T-cells by staining with peptide/HLA-A2 tetramers. CONCLUSIONS We defined class I-restricted epitopes located within the leader sequence of human preproinsulin through in vivo (transgenic mice) and ex vivo (diabetic patients) assays, illustrating the possible role of preproinsulin-specific CD8(+) T-cells in human type 1 diabetes.
Collapse
Affiliation(s)
- Andréa Toma
- Institut National de Santé et de Recherche Médicale U561 et Université Paris N, Hôpital Cochin-Saint Vincent de Paul, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
T-cell reactivity to insulin peptide A1-12 in children with recently diagnosed type 1 diabetes or multiple beta-cell autoantibodies. J Autoimmun 2008; 31:142-8. [PMID: 18565729 DOI: 10.1016/j.jaut.2008.04.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Revised: 04/14/2008] [Accepted: 04/24/2008] [Indexed: 11/20/2022]
Abstract
Insulin-specific immune responses appear early in preclinical type 1 diabetes (T1D), and bovine insulin in cow's milk-based infant formulas has been suggested to be of importance in induction of the primary response to insulin in humans. To characterize insulin-specific T-cell reactivity we studied T-cell responses to 10 insulin peptides derived from bovine (BI) and human insulin (HI) in 42 children with recently diagnosed T1D, 47 children with multiple autoantibodies and 111 autoantibody-negative control children with risk-associated HLA alleles. Proliferation responses detected in antigen-stimulated peripheral blood mononuclear cells did not differ between the three groups when the comparison was performed without considering HLA genotypes. However, significant differences were observed, when children with the high-risk genotype HLA (DRB1*03)-DQA1*05-DQB1*02/DRB1*0401-DQA1*03-DQB1*0302 were analyzed separately. The responses to the peptides including amino acids A1-12 derived from B1 and H1 were significantly higher in children with T1D (P=0.008, P=0.004, for B1 and H1, respectively) and in children with diabetes-associated autoantibodies (P=0.002 and P=0.001, respectively) than in control children. Positive responses (stimulation indices SI> or =3) were seen more frequently in T1D children than in controls (4/7 vs 2/19; P=0.03 and 4/7 vs 1/19; P=0.01 for B1 and H1, respectively). T-cell response to the insulin peptide A1-12 is enhanced in clinical and preclinical T1D associated with the high-risk HLA-genotype emphasizing the importance of this epitope.
Collapse
|
20
|
Vaccination against self to prevent autoimmune disease: the type 1 diabetes model. Immunol Cell Biol 2008; 86:139-45. [PMID: 18180798 DOI: 10.1038/sj.icb.7100151] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immune tolerance to self-antigens is physiological. Given a repertoire of self-reactive, potentially pathogenic lymphocytes, therapeutic options to diminish autoimmune disease risk include deletion, reduced activation or increased regulation of self-reactive lymphocytes by means that mimic or promote physiological mechanisms of immunity. Vaccination with self-antigen to promote self-antigen-specific tolerance, 'negative vaccination', may represent the most specific and potentially safest means of averting autoimmune disease. This strategy is therapeutically effective in inbred rodent models but its translation in humans has failed to meet expectations. This failure can be attributed to the use of suboptimal dosage regimens in end-stage disease, as well as other factors. This review focuses on vaccination against self-antigen in type 1 diabetes, an autoimmune disease unique in that individuals at risk can be identified years before clinical presentation. Moreover, the spontaneously diabetic non-obese diabetic mouse, which mimics human type 1 diabetes in many ways, has provided 'proof-of-concept' for negative vaccination. Recent trials of a nasal insulin vaccine in humans at risk of type 1 diabetes provide evidence of tolerance induction as a basis for clinical efficacy.
Collapse
|
21
|
Di Lorenzo TP, Peakman M, Roep BO. Translational mini-review series on type 1 diabetes: Systematic analysis of T cell epitopes in autoimmune diabetes. Clin Exp Immunol 2007; 148:1-16. [PMID: 17349009 PMCID: PMC1868845 DOI: 10.1111/j.1365-2249.2006.03244.x] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
T cell epitopes represent the molecular code words through which the adaptive immune system communicates. In the context of a T cell-mediated autoimmune disease such as type 1 diabetes, CD4 and CD8 T cell recognition of islet autoantigenic epitopes is a key step in the autoimmune cascade. Epitope recognition takes place during the generation of tolerance, during its loss as the disease process is initiated, and during epitope spreading as islet cell damage is perpetuated. Epitope recognition is also a potentially critical element in therapeutic interventions such as antigen-specific immunotherapy. T cell epitope discovery, therefore, is an important component of type 1 diabetes research, in both human and murine models. With this in mind, in this review we present a comprehensive guide to epitopes that have been identified as T cell targets in autoimmune diabetes. Targets of both CD4 and CD8 T cells are listed for human type 1 diabetes, for humanized [human leucocyte antigen (HLA)-transgenic] mouse models, and for the major spontaneous disease model, the non-obese diabetic (NOD) mouse. Importantly, for each epitope we provide an analysis of the relative stringency with which it has been identified, including whether recognition is spontaneous or induced and whether there is evidence that the epitope is generated from the native protein by natural antigen processing. This analysis provides an important resource for investigating diabetes pathogenesis, for developing antigen-specific therapies, and for developing strategies for T cell monitoring during disease development and therapeutic intervention.
Collapse
Affiliation(s)
- T P Di Lorenzo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | |
Collapse
|
22
|
Chan J, Clements W, Field J, Nasa Z, Lock P, Yap F, Toh BH, Alderuccio F. Transplantation of bone marrow genetically engineered to express proinsulin II protects against autoimmune insulitis in NOD mice. J Gene Med 2007; 8:1281-90. [PMID: 16989008 DOI: 10.1002/jgm.968] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a T-cell-dependent autoimmune disease resulting from destructive inflammation (insulitis) of the insulin-producing pancreatic beta-cells. Transgenic expression of proinsulin II by a MHC class II promoter or transfer of bone marrow from these transgenic mice protects NOD mice from insulitis and diabetes. We assessed the feasibility of gene therapy in the NOD mouse as an approach to treat T1D by ex vivo genetic manipulation of normal hematopoietic stem cells (HSCs) with proinsulin II followed by transfer to recipient mice. METHODS HSCs were isolated from 6-8-week-old NOD female mice and transduced in vitro with retrovirus encoding enhanced green fluorescent protein (EGFP) and either proinsulin II or control autoantigen. Additional control groups included mice transferred with non-manipulated bone marrow and mice which did not receive bone marrow transfer. EGFP-sorted or non-sorted HSCs were transferred into pre-conditioned 3-4-week-old female NOD mice and insulitis was assessed 8 weeks post-transfer. RESULTS Chimerism was established in all major lymphoid tissues, ranging from 5-15% in non-sorted bone marrow transplants to 20-45% in EGFP-sorted bone marrow transplants. The incidence and degree of insulitis was significantly reduced in mice receiving proinsulin II bone marrow compared to controls. However, the incidence of sialitis in mice receiving proinsulin II bone marrow and control mice was not altered, indicating protection from insulitis was antigen specific. CONCLUSIONS We show for the first time that ex vivo genetic manipulation of HSCs to express proinsulin II followed by transplantation to NOD mice can establish molecular chimerism and protect from destructive insulitis in an antigen-specific manner.
Collapse
Affiliation(s)
- James Chan
- Department of Immunology, Monash University, Commercial Road, Prahran, Victoria 3181, Australia
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Narendran P, Neale AM, Lee BH, Ngui K, Steptoe RJ, Morahan G, Madsen O, Dromey JA, Jensen KP, Harrison LC. Proinsulin is encoded by an RNA splice variant in human blood myeloid cells. Proc Natl Acad Sci U S A 2006; 103:16430-5. [PMID: 17053071 PMCID: PMC1618309 DOI: 10.1073/pnas.0607380103] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Genes for peripheral tissue-restricted self-antigens are expressed in thymic and hematopoietic cells. In thymic medullary epithelial cells, self-antigen expression imposes selection on developing autoreactive T cells and regulates susceptibility to autoimmune disease in mouse models. Less is known about the role of self-antigen expression by hematopoietic cells. Here we demonstrate that one of the endocrine self-antigens expressed by human blood myeloid cells, proinsulin, is encoded by an RNA splice variant. The surface expression of immunoreactive proinsulin was significantly decreased after transfection of monocytes with small interfering RNA to proinsulin. Furthermore, analogous to proinsulin transcripts in the thymus, the abundance of the proinsulin RNA splice variant in blood cells corresponded with the length of the variable number of tandem repeats 5' of the proinsulin gene, known to be associated with type 1 diabetes susceptibility. Self-antigen expression by peripheral myeloid cells extends the umbrella of "immunological self" and, by analogy with the thymus, may be implicated in peripheral immune tolerance.
Collapse
Affiliation(s)
- Parth Narendran
- *Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Alana M. Neale
- *Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Bo Han Lee
- *Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Katrina Ngui
- *Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Raymond J. Steptoe
- *Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Grant Morahan
- The Western Australian Institute of Medical Research, Perth, Western Australia 6000, Australia; and
| | - Ole Madsen
- Hagedorn Research Institute, DK-2820 Copenhagen, Denmark
| | - James A. Dromey
- *Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Kent P. Jensen
- *Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Leonard C. Harrison
- *Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
- To whom correspondence should be addressed at:
The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia. E-mail:
| |
Collapse
|
24
|
Higashide T, Kawamura T, Nagata M, Kotani R, Kimura K, Hirose M, Inada H, Niihira S, Yamano T. T cell epitope mapping study with insulin overlapping peptides using ELISPOT assay in Japanese children and adolescents with type 1 diabetes. Pediatr Res 2006; 59:445-50. [PMID: 16492987 DOI: 10.1203/01.pdr.0000200803.72985.3c] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease. Insulin seems to be a critical antigen recognized by autoreactive T cells. In this study, we performed T cell epitope mapping of insulin using serial overlapping peptides in Japanese patients with T1D. Serial overlapping insulin peptides comprising 23 peptides, which were each 15-amino acid long, were prepared based on insulin sequence. Cytokine secretion from peripheral T cells against these peptides was studied by enzyme-linked immunospot (ELISPOT) assay in 18 patients with recent-onset T1D and 12 patients with established T1D, and compared with 17 healthy control subjects. In ELISPOT assay, IFN-gamma-secreting T cells, but not IL-4, against several insulin peptides were observed in 77.8% of patients with recent-onset T1D, 50.0% of patients with established T1D, and 0% of healthy control subjects. All epitopes recognized by T cells were identified in the B-chain of insulin. The most frequent epitope existed at the B10-24 region (9/18), followed by B1-15 and B11-25 regions (6/18, each), with B4-18, B9-23, and B12-26 identified in some patients. These data did not correlate with insulin autoantibodies or HLA-DRB1 of the patients. This is the first report of T cell epitope mapping using one amino acid serial overlapping peptides of insulin in T1D. ELISPOT assay revealed the frequent existence of insulin peptide-specific T cells in patients with recent-onset and established T1D. The T cell epitopes of insulin were similar but not identical in our cohort, which probably explains the difficulty encountered in prevention of human T1D by using insulin.
Collapse
Affiliation(s)
- Takashi Higashide
- Department of Pediatrics, Osaka City University Graduate School of Medicien, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Oling V, Marttila J, Ilonen J, Kwok WW, Nepom G, Knip M, Simell O, Reijonen H. GAD65- and proinsulin-specific CD4+ T-cells detected by MHC class II tetramers in peripheral blood of type 1 diabetes patients and at-risk subjects. J Autoimmun 2005; 25:235-43. [PMID: 16263242 DOI: 10.1016/j.jaut.2005.09.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Revised: 06/10/2005] [Accepted: 09/07/2005] [Indexed: 10/25/2022]
Abstract
In type 1 diabetes the major loss of insulin producing beta-cells is caused by autoreactive T-cells specific for antigens expressed by the pancreatic islets. In this study we have analyzed the prevalence of glutamate decarboxylase 65 (GAD65)- and proinsulin-specific CD4(+) T-cells in type 1 diabetes patients, at-risk subjects and in HLA-matched control children. Peripheral blood mononuclear cells were cultured in the presence of two different GAD65 peptides (555-567, 557I and 274-286) or with a proinsulin (B24-C36) peptide for 10-11days. The autoreactive T-cells were detected using antigen specific-MHC class II tetramers by flow cytometry. Our results show that 11 of 18 (61%) type 1 diabetes patients and 7 of the 20 (35%) at-risk subjects were positive for one of the three GAD65 or proinsulin-containing tetramers, whereas only 2 of 21 (9.5%) controls had tetramer binding cells (p = 0.0007 type 1 diabetes vs. controls and p = 0.0488 at-risk subjects vs. controls, Chi-square test). Type 1 diabetes patients responded to all three peptides. At-risk subjects recognized also the GAD65 555-567 557I peptide, while none of the controls responded to it. In conclusion, type 1 diabetes patients and at-risk subjects have a significantly higher prevalence of GAD65- and proinsulin-specific CD4(+) T-cells than the control subjects.
Collapse
Affiliation(s)
- Viveka Oling
- Department of Virology, University of Turku, Medicity, Biocity 4 krs., Tykistökatu 6 A, 20520 Turku, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Danke NA, Yang J, Greenbaum C, Kwok WW. Comparative study of GAD65-specific CD4+ T cells in healthy and type 1 diabetic subjects. J Autoimmun 2005; 25:303-11. [PMID: 16249070 DOI: 10.1016/j.jaut.2005.08.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Revised: 08/03/2005] [Accepted: 08/17/2005] [Indexed: 10/25/2022]
Abstract
Glutamic acid decarboxylase 65 (GAD65) is a putative autoantigen associated with the pathogenesis of type 1 diabetes (T1D). The prevalence of autoreactive CD4+ T cells towards the immunodominant GAD65(555-567) epitope in DR4 healthy and T1D subjects was investigated with class II tetramers. A slightly higher percentage of diabetic subjects had GAD65(555-567) tetramer-positive T cells upon GAD65(555-567) peptide stimulation on the total CD4+ T-cell populations compared to healthy subjects. In contrast, three quarters of subjects in both groups had tetramer-positive T cells resulting from stimulation of the CD4+CD25+ regulatory T-cell depleted CD4+ T cells. The frequencies and TCR Vbeta gene usages of GAD65(555-567) T cells were also similar in both groups. Experiments demonstrated that GAD65(555-567)-reactive T cells in healthy and diabetic subjects had different CD45RA phenotypes. For the healthy group, GAD65(555-567)-reactive T cells were generally found in the CD45RA+ naïve T-cell pool while GAD65(555-567)-reactive T cells from T1D subjects were present in both CD45RA+ naïve and CD45RA- memory T-cell pools. These findings suggested that there is no difference in thymic selection of DR4 restricted GAD-reactive T cells amongst healthy and T1D individuals but GAD65(555-567)-reactive T cells have been preferentially activated in diabetic patients.
Collapse
Affiliation(s)
- Nancy A Danke
- Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | | | | | | |
Collapse
|
27
|
Chang Y, Yap S, Ge X, Piganelli J, Bertera S, Giannokakis N, Mathews C, Prud'homme G, Trucco M. DNA vaccination with an insulin construct and a chimeric protein binding to both CTLA4 and CD40 ameliorates type 1 diabetes in NOD mice. Gene Ther 2005; 12:1679-85. [PMID: 16107864 DOI: 10.1038/sj.gt.3302578] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Type 1 diabetes (T1D), a T-cell-mediated autoimmune disease, could be attributed to many defects in nonobese diabetic (NOD) mice, including deficient expressions of costimulatory molecules that impair antigen presentation. Thus, this deficient antigen presentation may result in a reduced ability to induce a tolerogenic response through negative selection/regulation of autoreactive T cells. Improperly activated T cells seem to be able to induce autoimmune responses causing diabetes. To re-establish tolerance to autoantigens by modulating costimulation, we constructed and tested a new type of DNA vaccine encoding a membrane-bound preproinsulin (mbPPI) and a chimeric gene vector encoding mutant B7.1/CD40L (mB7.1/CD40L) fusion protein. This mutant B7.1 binds CTLA4 but not CD28. We report that young NOD mice immunized with mbPPI along with mB7.1/CD40L DNA vectors significantly reduced diabetes incidence while treatment with CTLA4/IgG1 exacerbated diabetes. In conclusion, the combination of mbPPI and mB7.1/CD40L was able to protect against autoimmunity and diabetes in NOD mice possibly by promoting a more efficient presentation of autoantigen PPI and inducing specific tolerance to PPI by negatively regulating autoreactive T cells.
Collapse
Affiliation(s)
- Y Chang
- Division of Immunogenetics, Department of Pediatrics, University of Pittsburgh, School of Medicine, Rangos Research Center, Children's Hospital of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Garcia CA, Prabakar KR, Diez J, Cao ZA, Allende G, Zeller M, Dogra R, Mendez A, Rosenkranz E, Dahl U, Ricordi C, Hanahan D, Pugliese A. Dendritic Cells in Human Thymus and Periphery Display a Proinsulin Epitope in a Transcription-Dependent, Capture-Independent Fashion. THE JOURNAL OF IMMUNOLOGY 2005; 175:2111-22. [PMID: 16081777 DOI: 10.4049/jimmunol.175.4.2111] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The natural expression of tissue-specific genes in the thymus, e.g., insulin, is critical for self-tolerance. The transcription of tissue-specific genes is ascribed to peripheral Ag-expressing (PAE) cells, which discordant studies identified as thymic epithelial cells (TEC) or CD11c+ dendritic cells (DC). We hypothesized that, consistent with APC function, PAE-DC should constitutively display multiple self-epitopes on their surface. If recognized by Abs, such epitopes could help identify PAE cells to further define their distribution, nature, and function. We report that selected Abs reacted with self-epitopes, including a proinsulin epitope, on the surface of CD11c+ cells. We find that Proins+ CD11c+ PAE cells exist in human thymus, spleen, and also circulate in blood. Human thymic Proins+ cells appear as mature DC but express CD8alpha, CD20, CD123, and CD14; peripheral Proins+ cells appear as immature DC. However, DC derived in vitro from human peripheral blood monocytes include Proins+ cells that uniquely differentiate and mature into thymic-like PAE-DC. Critically, we demonstrate that human Proins+ CD11c+ cells transcribe the insulin gene in thymus, spleen, and blood. Likewise, we show that mouse thymic and peripheral CD11c+ cells transcribe the insulin gene and display the proinsulin epitope; moreover, by using knockout mice, we show that the display of this epitope depends upon insulin gene transcription and is independent of Ag capturing. Thus, we propose that PAE cells include functionally distinct DC displaying self-epitopes through a novel, transcription-dependent mechanism. These cells might play a role in promoting self-tolerance, not only in the thymus but also in the periphery.
Collapse
Affiliation(s)
- Carlos A Garcia
- Immunogenetics Program and Cell Transplant Center, Diabetes Research Institute, Miller School of Medicine, University of Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Toma A, Haddouk S, Briand JP, Camoin L, Gahery H, Connan F, Dubois-Laforgue D, Caillat-Zucman S, Guillet JG, Carel JC, Muller S, Choppin J, Boitard C. Recognition of a subregion of human proinsulin by class I-restricted T cells in type 1 diabetic patients. Proc Natl Acad Sci U S A 2005; 102:10581-6. [PMID: 16030147 PMCID: PMC1180789 DOI: 10.1073/pnas.0504230102] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2005] [Indexed: 11/18/2022] Open
Abstract
Proinsulin is a key autoantigen in type 1 diabetes. Evidence in the mouse has underscored the importance of the insulin B chain region in autoimmunity to pancreatic beta cells. In man, a majority of proteasome cleavage sites are predicted by proteasome cleavage algorithms within this region. To study CD8+ T cell responses to the insulin B chain and adjacent C peptide, we selected 8- to 11-mer peptides according to proteasome cleavage patterns obtained by digestion of two peptides covering proinsulin residues 28 to 64. We studied their binding to purified HLA class I molecules and their recognition by T cells from diabetic patients. Peripheral blood mononuclear cells from 17 of 19 recent-onset and 12 of 13 long-standing type 1 diabetic patients produced IFN-gamma in response to proinsulin peptides as shown by using an ELISPOT assay. In most patients, the response was against several class I-restricted peptides. Nine peptides were recognized within the proinsulin region covering residues 34 to 61. Four yielded a high frequency of recognition in HLA-A1 and -B8 patients. Three peptides located in the proinsulin region 41-51 were shown to bind several HLA molecules and to be recognized in a high percentage of diabetic patients.
Collapse
Affiliation(s)
- Andréa Toma
- Institut National de la Santé et de la Recherche Médicale U561, Hôpital Cochin-Saint Vincent de Paul, Université Paris V, 75014 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Csorba TR, Lyon AW. Abnormal proinsulin congeners as autoantigens that initiate the pathogenesis of Type 1 diabetes. Med Hypotheses 2005; 64:186-91. [PMID: 15533639 DOI: 10.1016/j.mehy.2004.04.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2004] [Accepted: 04/20/2004] [Indexed: 11/23/2022]
Abstract
Exposure of concentrated and purified monomeric insulin solutions to inorganic oxidants as iodine and chlorine lead to the appearance of a minor peak on gel chromatography that was disulfide cross-linked. It exhibited a 20-fold reduction in bioactivity, a markedly decreased immunoreactivity and a different electrophoretic pattern as compared to the starting material. The covalent dimer was formed by way of aggregation. These findings were reproduced by incubating monomeric insulin in normal blood spiked to hyperglycemic levels. In contrast, normoglycemic blood used as incubating medium failed to induce dimerization. Since the conformation of proinsulin is similar to that of insulin, involving the exposure of the anterior A7-B7 disulfide bridge, the authors hypothesize that proinsulin dimers rather than insulin dimers might be formed in Type 1 diabetes (TD1), leading to the autoimmune destruction of pancreatic B-cells. Proinsulin is present in a soluble aggregate state in the coated granule and may further accumulate allowing disulfide exchange due to abnormalities of the processing enzymes. This might be caused by inborn errors in genetically susceptible children or perhaps secondary to viral infection, whereas crystallization of insulin in mature granules is unlikely to be conducive to dimerization. Dimeric proinsulin would then migrate to pancreatic B-cell membranes to be taken up by surface immunoglobulins on B-lymphocytes that would in turn present it to cytotoxic T lymphocytes. The abnormal configuration of this congener would not be recognized as self by the immune system, triggering a selective destruction of pancreatic B-cells in the early pre-clinical development of TD1, resulting eventually in clinical disease. Since proinsulin is continuously converted to insulin in the coated granules of Type 2 diabetics, dimerization is unlikely in this condition. As pointed out by earlier investigators, TD1 is not a homogeneous disease, since several of its clinical features are different in children up to 6 years of age as compared to older patients. It is thus conceivable that there are subsets of TD1 triggered by dissimilar autoantigens and we propose in the present hypothesis that a conformationally altered congener of native proinsulin might play such a role.
Collapse
Affiliation(s)
- Thomas R Csorba
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Calgary, Foothills Medical Centre, 1403 - 29th Street N.W., Calgary, Alta., Canada T2N 2T9.
| | | |
Collapse
|
31
|
Mannering SI, Morris JS, Stone NL, Jensen KP, VAN Endert PM, Harrison LC. CD4+T Cell Proliferation in Response to GAD and Proinsulin in Healthy, Pre-diabetic, and Diabetic Donors. Ann N Y Acad Sci 2004; 1037:16-21. [PMID: 15699488 DOI: 10.1196/annals.1337.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The ability to measure proliferation of autoantigen-specific T cells is critical for the evaluation of cellular immune function. Using a novel, sensitive, CFSE-based assay, we were able to directly quantitate autoantigen-specific CD4(+) T cell proliferation. However, peripheral blood cells from healthy, pre-diabetic and diabetic donors exhibited overlap in responses to glutamic acid decarboxylase (GAD65) and proinsulin (PI). This indicates that autoantigen-induced CD4(+) T cell proliferation in a functionally complex cell population may not discriminate disease in the general population. Clear discrimination was found between diabetic and healthy sibs, suggesting the need to standardize the genetic and environmental background. In addition, the ability of the CFSE assay to allow analysis of the phenotype and function of autoantigen-responsive T cells may improve discrimination.
Collapse
Affiliation(s)
- Stuart I Mannering
- Virology, Victorian Infectious Diseases Reference Lab, 10 Wreckyn, North Melbourne, Victoria, 3051, Australia.
| | | | | | | | | | | |
Collapse
|
32
|
Judkowski VA, Allicotti GM, Sarvetnick N, Pinilla C. Peptides from common viral and bacterial pathogens can efficiently activate diabetogenic T-cells. Diabetes 2004; 53:2301-9. [PMID: 15331539 DOI: 10.2337/diabetes.53.9.2301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cross-reactivity between an autoantigen and unknown microbial epitopes has been proposed as a molecular mechanism involved in the development of insulin-dependent diabetes (type 1 diabetes). Type 1 diabetes is an autoimmune disease that occurs in humans and the nonobese diabetic (NOD) mouse. BDC2.5 is an islet-specific CD4+ T-cell clone derived from the NOD mouse whose natural target antigen is unknown. A biometrical analysis of screening data from BDC2.5 T-cells and a positional scanning synthetic combinatorial library (PS-SCL) was used to analyze and rank all peptides in public viral and bacterial protein databases and identify potential molecular mimic sequences with predicted reactivity. Selected sequences were synthesized and tested for stimulatory activity with BDC2.5 T-cells. Active peptides were identified, and some of them were also able to stimulate spontaneously activated T-cells derived from young, pre-diabetic NOD mice, indicating that the reactivity of the BDC2.5 T-cell is directed at numerous mouse peptides. Our results provide evidence for their possible role as T-cell ligands involved in the activation of diabetogenic T-cells.
Collapse
Affiliation(s)
- Valeria A Judkowski
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Ct., San Diego, CA 92121, USA
| | | | | | | |
Collapse
|
33
|
Achenbach P, Koczwara K, Knopff A, Naserke H, Ziegler AG, Bonifacio E. Mature high-affinity immune responses to (pro)insulin anticipate the autoimmune cascade that leads to type 1 diabetes. J Clin Invest 2004. [DOI: 10.1172/jci200421307] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
34
|
Lieberman SM, DiLorenzo TP. A comprehensive guide to antibody and T-cell responses in type 1 diabetes. ACTA ACUST UNITED AC 2004; 62:359-77. [PMID: 14617043 DOI: 10.1034/j.1399-0039.2003.00152.x] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease in which the insulin-producing beta cells in the pancreatic islets are selectively eliminated. T cells specific for beta-cell antigens are the mediators of this precise cellular destruction. However, antibodies to beta-cell proteins are also generated and may be used for predicting disease in at-risk populations. Over the past two decades, numerous beta-cell proteins and lipids have been implicated as autoantigens in patients or in non-obese diabetic (NOD) mice, a well-studied animal model of T1D. Here, we present a review of these antigens, accompanied by their T-cell epitopes, where known, and a discussion of our current understanding of why particular self-proteins become disease-inciting antigens. Although two dozen beta-cell antigens have been identified to date, few of these have been confirmed to be recognized by pathogenic T cells early in the disease process. Further identification and characterization of initiating beta-cell antigens targeted by pathogenic T cells should be a priority for future studies.
Collapse
Affiliation(s)
- S M Lieberman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
35
|
Narendran P, Elsegood K, Leech NJ, Macindoe WM, Boons GJ, Dayan CM. Dendritic cell-based assays, but not mannosylation of antigen, improves detection of T-cell responses to proinsulin in type 1 diabetes. Immunology 2004; 111:422-9. [PMID: 15056379 PMCID: PMC1782449 DOI: 10.1111/j.1365-2567.2004.01825.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
In vitro detection of T-cell responses to autoantigens in type 1 diabetes is recognized as being technically challenging. We aimed to accurately measure cellular responses to proinsulin in patients with diabetes, and speculated that presentation of antigen by dendritic cells (DCs) would enhance the sensitivity of the peripheral blood assay. Antigen was mannosylated to facilitate uptake through DC surface mannose receptors to further improve the assay. Whole proinsulin, as well as mannosylated peptides of proinsulin, were combined with peripheral T cells and autologous immature DCs in a proliferative assay in a panel of newly diagnosed type 1 diabetic patients. The DC-based assay detected responses to proinsulin in five of 15 diabetic patients compared to one of 15 diabetic patients detected using the standard mononuclear cell assay. When the results of all patients were combined, the DC assay, but not the mononuclear cell assay, had a proinsulin response that was significantly higher than background (P < 0.001). The DC assay was, however, associated with high autologous mixed lymphocyte reactions that possibly masked responses in individual patients. Mannosylated antigen was taken up in larger quantities than non-mannosylated antigen, but not presented any more powerfully. Our data suggest that autologous DC-based assays are more powerful than standard peripheral blood mononuclear cell assays. However, they are compromised by high autologous mixed lymphocyte reactions and this requires addressing before they can be used as a routine readout of in vitro peripheral T-cell responses.
Collapse
Affiliation(s)
- Parth Narendran
- University Division of Medicine, Bristol Royal Infirmary, Bristol, UK.
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Type 1 diabetes (TID) results from T-cell-mediated destruction of pancreatic b cells in genetically predisposed individuals. Autoreactive CD4(+) T helper cells and CD8(+) cytotoxic T lymphocytes (CTLs) recognize b-cell-derived peptides in the context of major histocompatibility complex class II and I molecules, respectively, in a process that terminates in b-cell death. Many peptide epitopes derived from b-cell proteins have been described for both humans and the nonobese diabetic (NOD) mouse, but their relative importance in disease pathogenesis is unclear. The significance of identifying key b-cell epitopes is underscored by a study showing that in the NOD mouse monitoring of a single population of b-cell-specific CTLs in the peripheral blood using a high-avidity analogue of the endogenous peptide may be used to accurately predict diabetes occurrence. Future studies focused on the discovery of immunodominant b-cell epitopes and their high-avidity analogues should have considerable implications for prediction and immunotherapy of TID.
Collapse
Affiliation(s)
- Constadina Panagiotopoulos
- British Columbia's Children's Hospital, Department of Pathology and Laboratory Medicine, 4480 Oak Street, Room 2G11, Vancouver, British Columbia V6H 3V4, Canada
| | | | | |
Collapse
|
37
|
Pugliese A. Central and peripheral autoantigen presentation in immune tolerance. Immunology 2004; 111:138-46. [PMID: 15027898 PMCID: PMC1782408 DOI: 10.1111/j.0019-2805.2003.01804.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2003] [Revised: 11/14/2003] [Accepted: 11/19/2003] [Indexed: 12/11/2022] Open
Abstract
Recent studies in both humans and experimental rodent models provide new insight into key mechanisms regulating tolerance to self-molecules. These recent advances are bringing about a paradigm shift in our views about tolerance to self-molecules with tissue-restricted expression. There is, indeed, mounting evidence that selected antigen-presenting cells (APCs) have the ability to synthesize and express self-molecules, and that such expression is critical for self-tolerance. Insulin is a key hormone produced exclusively by pancreatic beta-cells and a critical autoantigen in type 1 diabetes. It provides an excellent example of a molecule with tissue-restricted expression that is expressed ectopically by APCs. The fact that APCs expressing insulin have been demonstrated in both thymus and peripheral lymphoid tissues suggests that they may play a role in insulin presentation in both the central and peripheral immune system. Experimental mice, in which insulin expression was altered, provide functional data that help to dissect the role of insulin presentation by APCs of the immune system. This review addresses recent literature and emerging concepts about the expression of self-molecules in the thymus and peripheral lymphoid tissues and its relation to self-tolerance.
Collapse
Affiliation(s)
- Alberto Pugliese
- Diabetes Research Institute, University of Miami School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
38
|
Mannering SI, Morris JS, Jensen KP, Purcell AW, Honeyman MC, van Endert PM, Harrison LC. A sensitive method for detecting proliferation of rare autoantigen-specific human T cells. J Immunol Methods 2003; 283:173-83. [PMID: 14659909 DOI: 10.1016/j.jim.2003.09.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The ability to measure proliferation of rare antigen-specific T cells among many bystanders is critical for the evaluation of cellular immune function in health and disease. T-cell proliferation in response to antigen has been measured almost exclusively by 3H-thymidine incorporation. This method does not directly identify the phenotype of the proliferating cells and is frequently not sufficiently sensitive to detect rare autoantigen-specific T cells. To overcome these problems, we developed a novel assay for antigen-specific human T-cell proliferation. Peripheral blood mononuclear cells (PBMC) were labelled with the fluorescent dye 5,6-carboxylfluorescein diacetate succinimidyl ester (CFSE) and cells that proliferated in response to antigen, with resultant reduction in CFSE intensity, were measured directly by flow cytometry. This assay was more sensitive than 3H-thymidine incorporation and detected the proliferation of rare antigen-specific CD4(+) T cells at 10-fold lower antigen concentrations. It also allowed the phenotype of the proliferating cells to be directly determined. Using the CFSE assay we were able to measure directly the proliferation of human CD4(+) T cells from healthy donors in response to the type 1 diabetes autoantigens glutamic acid decarboxylase (GAD) and proinsulin (PI).
Collapse
Affiliation(s)
- Stuart I Mannering
- Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterised by immunity to pancreatic beta-cell autoantigens, associated with beta-cell destruction leading to insulin deficiency and hyperglycaemia. The rigorous definition of an autoimmune disease requires evidence that an autoantigen elicits pathological immune responses. Using criteria for the pathogenicity of an autoantigen, we examine the evidence for proinsulin as an autoantigen in T1D. We conclude that proinsulin satisfies these criteria. As a corollary, proinsulin is a potential immunotherapeutic tool for the prevention of T1D.
Collapse
Affiliation(s)
- Parth Narendran
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, 3050 Parkville, Australia
| | | | | |
Collapse
|
40
|
Abulafia-Lapid R, Gillis D, Yosef O, Atlan H, Cohen IR. T cells and autoantibodies to human HSP70 in type 1 diabetes in children. J Autoimmun 2003; 20:313-21. [PMID: 12791317 DOI: 10.1016/s0896-8411(03)00038-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We studied T-cell proliferative responses (stimulation index: SI) and autoantibodies to human HSP60, HSP70 and HSP90 proteins in 25 children (mean age 10.1+/-3.8 years) newly diagnosed with Type 1 diabetes. The control group for T cells included 25 adults and three pediatric donors without Type 1 diabetes. Controls for antibodies included 10 pediatric subjects. The T-cell responses to HSP70 of the test group (mean SI=4.5+/-3.1) were significantly greater than those of the control group (meanSI=1.4+/-0.6; p<0.0001); the incidence of HSP70 responders was (85%) compared to 14% in the control group. All but three of the Type 1 children who responded to HSP70 also responded to HSP60 (85%). The T-cell responses of the Type 1 group to HSP90 (mean SI=1.7+/-1.1) were similar to those of the control group (mean SI=1.5+/-0.7). We mapped HSP70 epitopes recognized by T cells in seven subjects using overlapping peptides of the molecule. Among the Type 1 subjects, IgG seropositivity was 45% to HSP60, 30% to HSP70, and 15% to HSP90. Thus, we conclude that children with newly diagnosed Type 1 diabetes manifest heightened T-cell autoimmunity to HSP70 and HSP60, but not to HSP90.
Collapse
Affiliation(s)
- Rivka Abulafia-Lapid
- HBRC Department of Biophysics and Nuclear Medicine, Hadassah Hebrew University Hospital, Ein-Karem, Jerusalem, Israel.
| | | | | | | | | |
Collapse
|
41
|
Martinez NR, Augstein P, Moustakas AK, Papadopoulos GK, Gregori S, Adorini L, Jackson DC, Harrison LC. Disabling an integral CTL epitope allows suppression of autoimmune diabetes by intranasal proinsulin peptide. J Clin Invest 2003. [DOI: 10.1172/jci200317166] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
42
|
Pugliese A. Peptide-based treatment for autoimmune diseases: learning how to handle a double-edged sword. J Clin Invest 2003; 111:1280-2. [PMID: 12727917 PMCID: PMC154453 DOI: 10.1172/jci18395] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Alberto Pugliese
- Departments of Medicine, University of Miami, Miami, Florida 33136, USA.
| |
Collapse
|
43
|
Steptoe RJ, Ritchie JM, Harrison LC. Transfer of hematopoietic stem cells encoding autoantigen prevents autoimmune diabetes. J Clin Invest 2003; 111:1357-63. [PMID: 12727927 PMCID: PMC154439 DOI: 10.1172/jci15995] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2002] [Accepted: 02/26/2003] [Indexed: 11/17/2022] Open
Abstract
Bone marrow or hematopoietic stem cell transplantation is a potential treatment for autoimmune disease. The clinical application of this approach is, however, limited by the risks associated with allogeneic transplantation. In contrast, syngeneic transplantation would be safe and have wide clinical application. Because T cell tolerance can be induced by presenting antigen on resting antigen-presenting cells (APCs), we reasoned that hematopoietic stem cells engineered to express autoantigen in resting APCs could be used to prevent autoimmune disease. Proinsulin is a major autoantigen associated with pancreatic beta cell destruction in humans with type 1 diabetes (T1D) and in autoimmune NOD mice. Here, we demonstrate that syngeneic transplantation of hematopoietic stem cells encoding proinsulin transgenically targeted to APCs totally prevents the development of spontaneous autoimmune diabetes in NOD mice. This antigen-specific immunotherapeutic strategy could be applied to prevent T1D and other autoimmune diseases in humans.
Collapse
Affiliation(s)
- Raymond J Steptoe
- Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | | | |
Collapse
|
44
|
Abstract
Type 1 (insulin-dependent) diabetes mellitus results from selective immune-mediated destruction of pancreatic islet beta cells. Strategies to prevent or reverse the development of diabetes can be divided into three groups, depending on whether they focus on beta-cell protection, regeneration or replacement. Prevention of immune beta-cell destruction involves either halting the immune attack directed against beta cells or making beta cells better able to withstand immune attack, for example, by making them resistant to free radical damage. The recent identification of beta-cell growth factors and development of stem cell technologies provides an alternative route to the reversal of diabetes, namely beta-cell regeneration. Interestingly, stem cell-derived islets appear to be less sensitive to recurrent immune destruction that is normally seen in response to islet transplantation. The last alternative is beta-cell replacement or substitution. This covers a wide range of interventions including human whole pancreas transplantation, xenotransplantation, genetically modified beta cells, mechanical insulin sensing and delivery devices, and the artificial pancreas. This review describes recent advances in each of these research areas and aims to provide clinicians with an idea of where and when an effective strategy to prevent or reverse diabetes development will become available.
Collapse
Affiliation(s)
- Nikolai Petrovsky
- Autoimmunity Research Unit, Canberra Hospital and Medical Informatics Centre, University of Canberra, ACT, Australia.
| | | | | |
Collapse
|
45
|
Abstract
Type 1 diabetes mellitus results from immune-mediated destruction of pancreatic beta-cells, leading to loss of insulin production. Strategies to prevent or reverse diabetes development include beta-cell protection, regeneration, or replacement. Recent advances in our understanding of the autoimmune process leading to diabetes has generated interest in the potential use of immunomodulatory agents that may collectively be termed vaccines, to prevent type 1 diabetes. Vaccines may work in various ways, including changing the immune response from a destructive (e.g. Th1) to a more benign (e.g. Th2) response, inducing antigen-specific regulatory T cells, deleting autoreactive T cells, or preventing immune cell interaction. To date, most diabetes vaccine development has been in animal models, with relatively few human trials having been completed. A major finding of animal models such as the non-obese diabetic (NOD) mouse is that they are extremely sensitive to diabetes protection, such that many interventions that protect mice are not successful in humans. This is particularly evident for human insulin tolerance studies, including the Diabetes Prevention Trial-1, where no human protection was seen from insulin despite positive NOD results. Further challenges are posed by the need to translate protective vaccine doses in mice to effective human doses. Despite such problems, some promising human vaccine data are beginning to emerge. Recent pilot studies have suggested a beneficial effect in recent-onset human type 1 diabetes from administration of nondepleting anti-CD3 antibodies or a peptide from heat shock protein 60. Given past experience, however, large multicenter, double-blind, controlled confirmatory studies are clearly required and longer term toxicity issues of drugs such as anti-CD3 need to be addressed.Diabetes vaccine development would benefit greatly from the development of reliable surrogate markers of immunoregulation. These would allow faster and more efficient screening of vaccine candidates, and would also assist in the translation of vaccine doses from animal to human studies. Unfortunately, research funding bodies desperate to find a cure are embarking on expensive clinical trials without first addressing important underlying issues such as animal-human dose translation and possible mechanisms of action. No doubt this is due to pressure from their constituency to rapidly find a cure, but unfortunately this approach may slow rather than speed the development of an effective vaccine cure. However, despite the significant hurdles that remain, vaccines remain one of the most promising strategies to prevent type 1 diabetes, with major advantages including convenience, safety, and long-lasting protection.
Collapse
Affiliation(s)
- Nikolai Petrovsky
- Autoimmunity Research Unit, The Canberra Hospital, Canberra, Australian Capital Territory, Australia and John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia.
| | | | | |
Collapse
|
46
|
Narendran P, Williams AJ, Elsegood K, Leech NJ, Dayan CM. Humoral and cellular immune responses to proinsulin in adults with newly diagnosed type 1 diabetes. Diabetes Metab Res Rev 2003; 19:52-9. [PMID: 12592644 DOI: 10.1002/dmrr.332] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease characterized by immunity against pancreatic islet-derived proteins. The object of this study was to measure antibody and T-cell responses against proinsulin (PI), an islet-derived protein, and to map its dominant T-cell epitopes. METHODS Antibody responses to proinsulin, insulin, glutamic acid decarboxylase (GAD), protein tyrosine phosphatase IA-2 and islet-cell antigen were measured in 116 newly diagnosed diabetic subjects aged 16 to 40 years. T-cell proliferative responses to proinsulin and proinsulin peptides were measured in 33 of these diabetic subjects and in 21 healthy control subjects. RESULTS 22% of diabetic subjects but no control subjects expressed antibodies to proinsulin. A strong correlation existed between antibody levels to proinsulin and insulin within diabetic subjects. Similar proportions of diabetic (12%) and healthy (9.5%) subjects displayed T-cell responses to proinsulin. There was no correlation between antibody and T-cell responses to proinsulin within subjects. Amino acid region 56 to 72 was identified as the major T-cell epitope of proinsulin, though significant responses to region 14 to 37 were also present. CONCLUSION Elevated proinsulin autoantibodies in diabetic subjects confirm proinsulin is an important autoantigen in type 1 diabetes. Though elevated cellular immunity to proinsulin protein was not detected, two dominant T-cell epitopes of proinsulin were identified that span the C-peptide and insulin junctions. Immunity to proinsulin was lower than that reported for childhood-onset type 1 diabetes and we propose that, like insulin, proinsulin may be targeted less frequently in adulthood.
Collapse
Affiliation(s)
- Parth Narendran
- University Division of Medicine, Bristol Royal Infirmary, Bristol BS2 8HW, United Kingdom
| | | | | | | | | |
Collapse
|
47
|
Rosmalen JGM, Leenen PJM, Pelegri C, Drexhage HA, Homo-Delarche F. Islet abnormalities in the pathogenesis of autoimmune diabetes. Trends Endocrinol Metab 2002; 13:209-14. [PMID: 12185667 DOI: 10.1016/s1043-2760(02)00600-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Type 1 diabetes mellitus is a T-cell-mediated autoimmune disease that results in the destruction of the insulin-producing beta cells in the pancreatic islets of Langerhans. In spite of extensive genetic and immunological studies, mainly performed in the non-obese diabetic (NOD) spontaneous mouse model, the etiology of the autoimmune attack remains unknown. Several autoantigens have been identified and numerous studies have suggested a role for defective regulation of immune function. However, this account does not explain why the autoimmune process specifically affects the insulin-producing beta cells. Thus, abnormal immune regulation might explain the predisposition to autoimmunity in general, but additional factors should then determine the target of the autoimmune attack. Here, we review the evidence that abnormalities in islet cell differentiation and function exist that might trigger the immune system towards beta-cell autoimmunity in humans and NOD mice.
Collapse
Affiliation(s)
- Judith G M Rosmalen
- Dept Immunology, Erasmus MC, University Medical Center, PO Box 1738, 3000 DR Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
48
|
Durinovic-Belló I, Boehm BO, Ziegler AG. Predominantly recognized proinsulin T helper cell epitopes in individuals with and without islet cell autoimmunity. J Autoimmun 2002; 18:55-66. [PMID: 11869047 DOI: 10.1006/jaut.2001.0566] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Antibody response to insulin and to its precursor ProInsulin is associated with increased risk for type 1 diabetes (T1D), though little is known about T cell reactivity to this molecule. In the present study from peripheral blood mononuclear cells (PBMC), in vivo primed CD45RO+ memory T helper (Th) cells were enriched and their reactivity to eight overlapping ProInsulin peptides and to protein was analyzed. Individuals with high risk HLA-DRB1*04, DQB1*0302 alleles were investigated: relatives of patients with T1D having humoral markers of islet cell autoimmunity (autoantibody positive, Ab+; n=11), patients with T1D (n=8), and healthy control individuals (n=16). The ProInsulin epitope which was most frequently recognized in all the tested individuals was C-peptide (C) 18-A-chain (A)1. In Ab+ relatives the responses to this epitope and to two additional parts of the ProInsulin, B-chain (B) 11-C24 and C28-A21, was observed. In T1D patients who have already been treated with insulin, response to peptide B20-C4 and to the entire insulin molecule predominates. Our findings suggest that the spontaneous memory Th cell response to ProInsulin in individuals with high risk HLA alleles is predominantly directed to one epitope which maps to the central, C-peptide region. In individuals with humoral markers of islet cell autoimmunity and in patients with T1D, spread response to distinct ProInsulin regions was observed.
Collapse
Affiliation(s)
- Ivana Durinovic-Belló
- Diabetes Research Institute of the Academic Hospital München-Schwabing, Koelner Platz 1, 80804 Munich, Germany.
| | | | | |
Collapse
|
49
|
Abstract
At present it is possible to predict the development of type 1A diabetes (immune-mediated diabetes) in man and prevent the disorder in animals. Studies of immunity to insulin play a prominent role in both disease prediction and disease prevention. For both man and the NOD mouse, insulin autoantibodies usually precede the development of diabetes and can be utilized to assist in disease prediction. T cells clones recognizing insulin, both CD4 and CD8, can transfer disease to young mice or immunodeficient animals. Specific insulin peptides reacting with these clones have been identified, their crystal structure when bound to a human "diabetogenic" MHC allele has been determined, and specific peptides can be used either to induce or to prevent disease. Clinical trials of both insulin and an altered peptide ligand of insulin to prevent islet beta-cell destruction are underway. Insulin is one of a number of islet autoantigens, but it is likely that immune responses to insulin will be central to both pathogenesis and immunologic protection.
Collapse
Affiliation(s)
- Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Denver, CO 80262, USA
| | | |
Collapse
|
50
|
Chen W, Bergerot I, Elliott JF, Harrison LC, Abiru N, Eisenbarth GS, Delovitch TL. Evidence that a peptide spanning the B-C junction of proinsulin is an early Autoantigen epitope in the pathogenesis of type 1 diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4926-35. [PMID: 11673498 DOI: 10.4049/jimmunol.167.9.4926] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The expression of pro(insulin) in the thymus may lead to the negative selection of pro(insulin) autoreactive T cells and peripheral tolerance to this autoantigen in type 1 diabetes (T1D). We investigated whether proinsulin is expressed in the thymus of young nonobese diabetic (NOD) mice, whether T cells from naive NOD female mice at weaning are reactive to mouse proinsulin, and the role of proinsulin as a pathogenic autoantigen in T1D. Proinsulin II mRNA transcripts were detected in the thymus of 2-wk-old NOD mice at similar levels to other control strains. Despite this expression, proinsulin autoreactive T cells were detected in the periphery of 2- to 3-wk-old naive NOD mice. Peripheral T cells reactive to the insulin, glutamic acid decarboxylase 65 (GAD65), GAD67, and islet cell Ag p69 autoantigens were also detected in these mice, indicating that NOD mice are not tolerant to any of these islet autoantigens at this young age. T cell reactivities to proinsulin and islet cell Ag p69 exceeded those to GAD67, and T cell reactivity to proinsulin in the spleen and pancreatic lymph nodes was directed mainly against a p24-33 epitope that spans the B chain/C peptide junction. Intraperitoneal immunization with proinsulin perinatally beginning at 18 days of age delayed the onset and reduced the incidence of T1D. However, s.c. immunization with proinsulin initiated at 5 wk of age accelerated diabetes in female NOD mice. Our findings support the notion that proinsulin p24-33 may be a primary autoantigen epitope in the pathogenesis of T1D in NOD mice.
Collapse
Affiliation(s)
- W Chen
- Autoimmunity/Diabetes Group, The John P. Robarts Research Institute and Department of Microbiology, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|