1
|
Liu X, Zhang S, Li X, Zheng P, Hu F, Zhou Z. Vaccination with a co-expression DNA plasmid containing GAD65 fragment gene and IL-10 gene induces regulatory CD4(+) T cells that prevent experimental autoimmune diabetes. Diabetes Metab Res Rev 2016; 32:522-33. [PMID: 26797873 DOI: 10.1002/dmrr.2780] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 12/11/2015] [Accepted: 01/15/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND The non-obese diabetic (NOD) mouse is a commonly used animal model for studying type 1 diabetes (T1D). The aims of our study were to explore the diabetes-preventive effect in NOD mice and the potential mechanisms of an optimized co-expression DNA vaccine containing GAD65 fragment gene with the IL-10 gene (SGAD65190-315 /IL-10). METHODS Female NOD mice at the age of 3-4 weeks old were randomly divided into two groups and received intra-muscular injection of either blank pBudCE4.l vector (n = 34) or pBudCE4.l carrying the SGAD65190-315 /IL-10 (n = 32). The incidence of diabetes was monitored up to 30 weeks of age. The severity of insulitis, apoptosis rate of β cells and relevant mechanisms were examined. RESULTS Administration with SGAD65190-315 /IL-10 blocked the onset of autoimmune diabetes in NOD mice, significantly suppressed islet inflammation, inhibited the apoptosis of islet β cells, induced immune tolerance to autoantigen GAD65 and proinsulin and shifted the Th1/Th2 balance towards Th2. More importantly, the frequencies of CD4(+) CD25(+) Foxp3(+) regulatory T cells (Tregs) in the spleen and pancreatic lymph nodes in vaccine-immunized mice were significantly increased, and these Tregs were GAD65-reactive. In addition, Treg depletion by anti-CD25 mAb administration abolished the protective effects of SGAD65190-315 /IL-10 on diabetes and insulitis. Moreover, depletion of CD4(+) CD25(+) T cells using magnetic-activated cell sorting impaired the protective effect of SGAD65190-315 /IL-10 vaccination on adoptive transfer of diabetes. CONCLUSIONS Our data suggested that SGAD65190-315 /IL-10 DNA vaccine had protective effects on T1D by upregulating autoantigen-reactive Tregs. Our findings may provide a novel preventive therapy for T1D. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Xinyuan Liu
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Song Zhang
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Xia Li
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Peilin Zheng
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Fang Hu
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| |
Collapse
|
2
|
Stifter K, Schuster C, Schlosser M, Boehm BO, Schirmbeck R. Exploring the induction of preproinsulin-specific Foxp3(+) CD4(+) Treg cells that inhibit CD8(+) T cell-mediated autoimmune diabetes by DNA vaccination. Sci Rep 2016; 6:29419. [PMID: 27406624 PMCID: PMC4942695 DOI: 10.1038/srep29419] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/14/2016] [Indexed: 12/26/2022] Open
Abstract
DNA vaccination is a promising strategy to induce effector T cells but also regulatory Foxp3+ CD25+ CD4+ Treg cells and inhibit autoimmune disorders such as type 1 diabetes. Little is known about the antigen requirements that facilitate priming of Treg cells but not autoreactive effector CD8+ T cells. We have shown that the injection of preproinsulin (ppins)-expressing pCI/ppins vector into PD-1- or PD-L1-deficient mice induced Kb/A12-21-monospecific CD8+ T cells and autoimmune diabetes. A pCI/ppinsΔA12-21 vector (lacking the critical Kb/A12-21 epitope) did not induce autoimmune diabetes but elicited a systemic Foxp3+ CD25+ Treg cell immunity that suppressed diabetes induction by a subsequent injection of the diabetogenic pCI/ppins. TGF-β expression was significantly enhanced in the Foxp3+ CD25+ Treg cell population of vaccinated/ppins-primed mice. Ablation of Treg cells in vaccinated/ppins-primed mice by anti-CD25 antibody treatment abolished the protective effect of the vaccine and enabled diabetes induction by pCI/ppins. Adoptive transfer of Treg cells from vaccinated/ppins-primed mice into PD-L1−/− hosts efficiently suppressed diabetes induction by pCI/ppins. We narrowed down the Treg-stimulating domain to a 15-residue ppins76–90 peptide. Vaccine-induced Treg cells thus play a crucial role in the control of de novo primed autoreactive effector CD8+ T cells in this diabetes model.
Collapse
Affiliation(s)
- Katja Stifter
- Department of Internal Medicine I, Ulm University Medical Center, Ulm, Germany
| | - Cornelia Schuster
- Department of Internal Medicine I, Ulm University Medical Center, Ulm, Germany
| | - Michael Schlosser
- Department of Medical Biochemistry and Molecular Biology, Research Group of Predictive Diagnostics, University Medical Centre Greifswald, Karlsburg, Germany
| | - Bernhard Otto Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore, Singapore.,Imperial College London, London, UK
| | - Reinhold Schirmbeck
- Department of Internal Medicine I, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
3
|
Krishnamurthy B, Selck C, Chee J, Jhala G, Kay TWH. Analysis of antigen specific T cells in diabetes - Lessons from pre-clinical studies and early clinical trials. J Autoimmun 2016; 71:35-43. [PMID: 27083395 DOI: 10.1016/j.jaut.2016.03.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 01/06/2023]
Abstract
Antigen-specific immune tolerance promises to provide safe and effective therapies to prevent type 1 diabetes (T1D). Antigen-specific therapy requires two components: well-defined, clinically relevant autoantigens; and safe approaches to inducing tolerance in T cells specific for these antigens. Proinsulin is a critical autoantigen in both NOD mice, based on knockout mouse studies and induction of immune tolerance to proinsulin preventing disease whereas most antigens cannot, and also in human T1D based on proinsulin-specific T cells being found in the islets of affected individuals and the early appearance of insulin autoantibodies. Effective antigen-specific therapies that prevent T1D in humans have not yet been developed although doubt remains about the best molecular form of the antigen, the dose and the route of administration. Preclinical studies suggest that antigen specific therapy is most useful when administered before onset of autoimmunity but this time-window has not been tested in humans until the recent "pre-point" study. There may be a 'window of opportunity' during the neonatal period when 'vaccine' like administration of proinsulin for a short period may be sufficient to prevent diabetes. After the onset of autoimmunity, naive antigen-specific T cells have differentiated into antigen-experienced memory cells and the immune responses have spread to multiple antigens. Induction of tolerance at this stage becomes more difficult although recent studies have suggested generation of antigen-specific TR1 cells can inhibit memory T cells. Preclinical studies are required to identify additional 'help' that is required to induce tolerance to memory T cells and develop protocols for effective therapy in individuals with established autoimmunity.
Collapse
Affiliation(s)
- Balasubramanian Krishnamurthy
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia
| | - Claudia Selck
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia
| | - Jonathan Chee
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia
| | - Guarang Jhala
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia
| | - Thomas W H Kay
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia.
| |
Collapse
|
4
|
Hassan GA, Sliem HA, Ellethy AT, Salama MES. Role of immune system modulation in prevention of type 1 diabetes mellitus. Indian J Endocrinol Metab 2012; 16:904-909. [PMID: 23226634 PMCID: PMC3510959 DOI: 10.4103/2230-8210.102989] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
An increased incidence of Type 1 diabetes mellitus (T1DM) is expected worldwide. Eventually, T1DM is fatal unless treated with insulin. The expansion of interventions to prevent diabetes and the use of alternative treatments to insulin is a dream to be fulfilled. The pathophysiology in T1DM is basically a destruction of beta cells in the pancreas, regardless of which risk factors or causative entities have been present. Individual risk factors can have separate patho-physiological processes to, in turn, cause this beta cell destruction. Currently, autoimmunity is considered the major factor in the pathophysiology of T1DM. In a genetically susceptible individual, viral infection may stimulate the production of antibodies against a viral protein that trigger an autoimmune response against antigenically similar beta cell molecules. Many components of the immune system have been implicated in autoimmunity leading to β-cell destruction, including cytotoxic and helper T-cells, B-cells, macrophages, and dendritic cells. The inflammatory process in early diabetes is thought to be initiated and propagated by the effect of Th1-secreted cytokines (e.g. g interferon) and suppressed by Th2-secreted antiinflammatory cytokines (interleukins). Structure and function of β-cell may be modulated by using Th1/Th2-secreted cytokines. Several experimental and clinical trials of applying GAD65, Hsp60, peptide-MHC, pepetide-277 immunization, anti-CD3 infusion, and interleukins to modulate immune response in T1DM were done. Applying such trials in patients with prediabetes, will most likely be the future key in preventing Type 1 autoimmune diabetes.
Collapse
Affiliation(s)
- Gamal Abdulrhman Hassan
- Department of Anatomy and Genetics, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Hamdy Ahmad Sliem
- Department of Internal Medicine, College of Dentistry, Qassim University, Saudi Arabia
| | | | - Mahmoud El-Sawy Salama
- Department of Basic Oral and Medical Science, College of Dentistry, Qassim University, Saudi Arabia
| |
Collapse
|
5
|
Affiliation(s)
- Chantal Mathieu
- Laboratory and Clinic of Experimental Medicine and Endocrinology, University Hospital Leuven, Catholic University of Leuven, Leuven 3000, Belgium.
| | | |
Collapse
|
6
|
Johnson MC, Wang B, Tisch R. Genetic vaccination for re-establishing T-cell tolerance in type 1 diabetes. HUMAN VACCINES 2011; 7:27-36. [PMID: 21157183 DOI: 10.4161/hv.7.1.12848] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes (T1D) is a T-cell mediated autoimmune disease resulting in the destruction of the insulin-secreting β cells. Currently, there is no established clinical approach to effectively suppress long-term the diabetogenic response. Genetic-based vaccination offers a general strategy to reestablish β-cell specific tolerance within the T-cell compartment. The transfer of genes encoding β-cell autoantigens, anti-inflammatory cytokines and/or immunomodulatory proteins has proven to be effective at preventing and suppressing the diabetogenic response in animal models of T1D. The current review will discuss genetic approaches to prevent and treat T1D with an emphasis on plasmid DNA- and adeno-associated virus-based vaccines.
Collapse
Affiliation(s)
- Mark C Johnson
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, North Carolina, USA
| | | | | |
Collapse
|
7
|
Abstract
Autoimmune diseases represent a group of disorders in which there exists a large unmet medical need for effective treatments, but also where there exists a tremendous responsibility among physicians and drug developers to maintain adequate and acceptable patient safety. Several drugs have been approved and many others are about to be approved for the treatment of autoimmune diseases, but in pushing the envelope of therapeutic efficacy, concerns have been raised about the long-term safety of these new therapies. DNA vaccines provide a method of treating autoimmune diseases in a highly specific manner, and could therefore overcome these safety concerns while still maintaining comparable efficacy. The numerous reports of DNA vaccines in animal models of autoimmune diseases and results from three recent human trials of DNA vaccines in autoimmune diseases are reviewed here.
Collapse
Affiliation(s)
- Hideki Garren
- Bayhill Therapeutics, Inc., Suite 150, San Mateo, CA 94404, USA.
| |
Collapse
|
8
|
Eldor R, Cohen IR, Raz I. Innovative Immune-Based Therapeutic Approaches for the Treatment of Type 1 Diabetes Mellitus. Int Rev Immunol 2009; 24:327-39. [PMID: 16318985 DOI: 10.1080/08830180500379697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Type 1 diabetes mellitus is an autoimmune disease caused by a culmination of noxious processes of autoimmunity composed of various components of the innate and adaptive immune systems. Current treatment of type 1 diabetes focuses on restraining the endocrine disease without affecting the autoimmune process that underlies it. Prevention of this disease requires immune modulation and early intervention. New therapeutic approaches can be classified on the basis of the immunological arm targeted, that is, T-cell immune modulation (using cytokines, anti-CD3 monoclonal antibodies, and peptide MHC class II dimers), innate immune system modulation (using alpha-galactosylceramide or peptide 277), or specific antigen vaccination (glutamic acid decarboxylase and insulin). Here we review the most promising therapies developed based on these targets and emphasize those that have reached human phase clinical investigation.
Collapse
Affiliation(s)
- Roy Eldor
- Diabetes Research Center, Department of Medicine, Hadassah-Hebrew University Hospital, Jerusalem, Israel.
| | | | | |
Collapse
|
9
|
Ho PP, Higgins JP, Kidd BA, Tomooka B, Digennaro C, Lee LY, de Vegvar HEN, Steinman L, Robinson WH. Tolerizing DNA vaccines for autoimmune arthritis. Autoimmunity 2009; 39:675-82. [PMID: 17178564 DOI: 10.1080/08916930601061603] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Current therapies for rheumatoid arthritis (RA) and other autoimmune diseases non-specifically suppress immune function, and there is great need for fundamental approaches such as antigen-specific tolerizing therapy. In this paper we describe development of antigen-specific tolerizing DNA vaccines to treat collagen-induced arthritis (CIA) in mice, and use of protein microarrays to monitor response to therapy and to identify potential additional autoimmune targets for next generation vaccines. We demonstrate that tolerizing DNA vaccines encoding type II collagen (CII) reduced the incidence and severity of CIA. Atorvastatin, a statin drug found to reduce the severity of autoimmunity, potentiated the effect of DNA vaccines encoding CII. Analysis of cytokines produced by collagen-reactive T cells derived from mice receiving tolerizing DNA encoding CII, as compared to control vaccines, revealed reduced production of the pro-inflammatory cytokines IFN-gamma and TNF-alpha. Arthritis microarray analysis demonstrated reduced spreading of autoantibody responses in mice treated with DNA encoding CII. The development of tolerizing DNA vaccines, and the use of antibody profiling to guide design of and to monitor therapeutic responses to such vaccines, represents a promising approach for the treatment of RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Peggy P Ho
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Much emphasis has been placed on the so-called "biologics" in the treatment of immune disorders within the last few years. Here we discuss the expanding horizon of potential strategies for immunotherapies targeting T lymphocytes as key effectors and regulators of autoimmunity. We review emerging reagents in a variety of animal models and human disorders that may offer new therapeutic options in current or modified iterations.
Collapse
Affiliation(s)
- Erica Lee
- Department of Dermatology, Weill Medical College of Cornell University, New York, NY, USA
| | | |
Collapse
|
11
|
Boehm BO, Rosinger S, Sauer G, Manfras BJ, Palesch D, Schiekofer S, Kalbacher H, Burster T. Protease-resistant human GAD-derived altered peptide ligands decrease TNF-alpha and IL-17 production in peripheral blood cells from patients with type 1 diabetes mellitus. Mol Immunol 2009; 46:2576-84. [PMID: 19505724 DOI: 10.1016/j.molimm.2009.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 05/11/2009] [Indexed: 10/20/2022]
Abstract
Glutamic acid decarboxylase 65 (GAD) and proinsulin are major diabetes-associated autoantigens that drive autoreactive T cells. Altered peptide ligands (APL) have been proposed as reagents for the modification of autoimmune reactions. Here, we have prepared GAD-derived protease-resistant APL (prAPL) by cleavage site-directed modification. The resulting prAPL are resistant to lysosomal and serum proteases, bind with high-affinity to HLA-DRB1(*)0401 and have a prolonged half-life in the serum. GAD-derived prAPL significantly decreased the secretion of proinflammatory cytokines by a GAD-specific human T cell clone. Likewise, the production of IL-17, TNF-alpha, and secretion of IL-6 by peripheral blood lymphocytes from patients with type 1 diabetes mellitus (T1D) was reduced, when stimulated with both GAD and GAD-derived prAPL. Thus, prAPL with high affinity for HLA-DRB1(*)0401 mitigate the response of GAD-reactive human Th17 cells. The strategy of designing specific immunomodulatory protease-resistant altered peptide ligands provides the basis for novel avenues of therapeutic intervention.
Collapse
Affiliation(s)
- Bernhard O Boehm
- Department of Internal Medicine I, University Medical Center Ulm and Center of Excellence, Germany
| | | | | | | | | | | | | | | |
Collapse
|
12
|
A novel mechanism for GABA synthesis and packaging into synaptic vesicles. Neurochem Int 2009; 55:9-12. [PMID: 19428801 DOI: 10.1016/j.neuint.2009.01.020] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 01/20/2009] [Accepted: 01/21/2009] [Indexed: 11/24/2022]
Abstract
This review focuses on the recent advances that were made in understanding the fundamental mechanisms of the regulation of l-glutamic acid decarboxylase (GAD; E.C. 4.1.1.15), the enzyme responsible for the synthesis of the major inhibitory neurotransmitter gamma-amino butyric acid (GABA). In the brain, there are two isoforms of GAD- GAD67 and GAD65, where 67 and 65 refer to their respective molecular weights in kDa. A number of neurodegenerative diseases are known to occur as a result of insufficient inhibition due to failure of GABA neurotransmission. Since the rate-limiting step in GABA biosynthesis is the decarboxylation of glutamate by GAD, it is important to understand how GAD is regulated. So far, we know that GAD is regulated at the transcriptional level by alternate splicing and at the post-translational level by protein phosphorylation, palmitoylation and activity-dependent cleavage. Here, we present new evidence of the presence of GAD65 associated with mitochondria in the axon terminal and project a model in which ATP generated by mitochondrial GAD65 may serve an important function in providing energy for GAD65 mediated GABA biosynthesis and packaging into synaptic vesicles by vesicular GABA transporter (VGAT).
Collapse
|
13
|
Song X, Liang F, Liu N, Luo Y, Xue H, Yuan F, Tan L, Sun Y, Xi C, Xi Y. Construction and characterization of a novel DNA vaccine that is potent antigen-specific tolerizing therapy for experimental arthritis by increasing CD4+CD25+Treg cells and inducing Th1 to Th2 shift in both cells and cytokines. Vaccine 2008; 27:690-700. [PMID: 19095031 DOI: 10.1016/j.vaccine.2008.11.090] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 10/31/2008] [Accepted: 11/12/2008] [Indexed: 01/04/2023]
Abstract
Currently available treatments for rheumatoid arthritis (RA) are often ineffective in ameliorating the progression of disease, particularly the invasive destruction of articular cartilage and bone, and RA remains incurable. Therefore, vaccinotherapy of RA with an antigen-specific tolerizing DNA vaccine may offer new promise for overcoming this difficulty. Using recombinant technology, the DNA sequences encoding chicken type II collagen (CCOL2A1) with deleted N-propeptides were obtained from the plasmid pPIC9K/pCalpha(1)(II), and then cloned into pcDNA3.1(+). The resulting recombinant plasmid pcDNA-CCOL2A1 was produced in Escherichia coli, purified, characterized and used as a tolerizing DNA vaccine for the treatment of collagen-induced arthritis (CIA). Therapeutic efficacy and potential action mechanisms of pcDNA-CCOL2A1 tolerizing DNA vaccine against CIA were studied. Here we demonstrate that a single intravenous treatment with novel tolerizing DNA vaccine pcDNA-CCOL2A1 can induce potent immune tolerance against CIA. The efficacy of this therapy was verified by clinical visual scoring, radiographic X-ray, histopathological examination, and anti-CII IgG levels. Furthermore, the action mechanism behind this efficacy can be at least partially attributed to increased CD4(+)CD25(+) T regulatory cells, which specifically down-modulate the T lymphocyte proliferative response to CCII, induce a shift of Th1 to Th2 cells, as well as down-regulate Th1-cytokine TNF-alpha, while up-regulating both Th2-cytokine IL-10 and Th3-cytokine TGF-beta. More importantly, pcDNA-CCOL2A1 alone seems to be as effective as the current "golden standard" treatment, methotrexate (MTX). Taken together, these results suggest that we have successfully developed a novel tolerizing DNA vaccine encoding CCII, which is the first description of a tolerizing DNA vaccine encoding CCII for antigen-specific tolerizing therapy but not prophylactic against CIA.
Collapse
Affiliation(s)
- Xinqiang Song
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital Affiliated to Academy of Medical Sciences, No. 8 Dongda Ave., Fengtai District, Beijing 100071, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jégou JF, Chan P, Schouft MT, Gasque P, Vaudry H, Fontaine M. Protective DNA vaccination against myelin oligodendrocyte glycoprotein is overcome by C3d in experimental autoimmune encephalomyelitis. Mol Immunol 2007; 44:3691-701. [PMID: 17521729 DOI: 10.1016/j.molimm.2007.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Accepted: 04/04/2007] [Indexed: 01/26/2023]
Abstract
Complement receptor 2 (CR2) and its physiological ligand, C3d, known for its molecular adjuvant property on the immune response, exhibit opposite effects with regard to autoimmunity. Although CR2 has been implicated in maintaining self-tolerance, recent studies reported a role for C3d signaling to CR2 in tolerance breakdown to self-antigens and the initiation of inflammatory autoimmune pathologies. In the present study, we have investigated the effect of C3d in a model of tolerogenic DNA vaccination encoding the myelin oligodendrocyte glycoprotein (MOG-DNA) which protected mice from the induction of an experimental autoimmune encephalomyelitis (EAE). We show that fusing two or three copies of C3d to MOG overcomes the protective effect of DNA vaccination. Multimeric C3d was able to revert the unresponsiveness state of specific T cells induced by MOG-DNA, independently of a modification in the Th1/Th2 cytokine pattern. Interestingly, the adjuvant effect of C3d was not sufficient to boost the anti-MOG antibody response after DNA vaccination. These findings suggest that C3d might be involved in self-tolerance breakdown and could contribute to the pathogenesis of central nervous system autoimmune disorders.
Collapse
Affiliation(s)
- Jean-François Jégou
- INSERM U413, IFRMP 23, Laboratory of Cellular and Molecular Neuroendocrinology, University of Rouen, 76821 Mont Saint-Aignan, France.
| | | | | | | | | | | |
Collapse
|
15
|
Abstract
Type I diabetes mellitus (T1D) is due to a loss of immune tolerance to islet antigen and thus, there is intense interest in developing therapies that can re-establish it. Tolerance is maintained by complex mechanisms that include inhibitory molecules and several types of regulatory T cells (Tr). A major historical question is whether gene therapy can be employed to generate Tr cells. This review shows that gene transfer of immunoregulatory molecules can prevent T1D and other autoimmune diseases. In our studies, non-viral gene transfer is enhanced by in vivo electroporation (EP). This technique can be used to perform DNA vaccination against islet cell antigens and when combined with appropriate immune ligands results in the generation of Tr cells and protection against T1D. In vivo EP can also be applied for non-immune therapy of diabetes. It can be used to deliver protein drugs such as glucagon-like peptide 1 (GLP-1), leptin or transforming growth factor beta (TGF-beta). These act in T1D or type II diabetes (T2D) by restoring glucose homeostasis, promoting islet cell survival and growth or improving wound healing and other complications. Furthermore, we show that in large animals EP can deliver peptide hormones, such as growth hormone releasing hormone (GHRH). We conclude that the non-viral gene therapy and EP represent a safe and efficacious approach with clinical potential.
Collapse
Affiliation(s)
- G J Prud'homme
- Department of Laboratory Medicine, St Michael's Hospital and University of Toronto, Ontario, Canada.
| | | | | |
Collapse
|
16
|
Glinka Y, Chang Y, Prud'homme GJ. Protective Regulatory T Cell Generation in Autoimmune Diabetes by DNA Covaccination with Islet Antigens and a Selective CTLA-4 Ligand. Mol Ther 2006; 14:578-87. [PMID: 16790365 DOI: 10.1016/j.ymthe.2006.03.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2005] [Revised: 03/16/2006] [Accepted: 03/24/2006] [Indexed: 01/11/2023] Open
Abstract
DNA vaccination of autoimmune diabetes-prone NOD mice with unmodified target islet antigens, i.e., preproinsulin (PPIns) or glutamic acid decarboxylase 65 (GAD65), is poorly protective. However, in this study, we demonstrate protection against disease by covaccination with a mutant B7-1 molecule (B7-1wa) that binds the negative T cell regulator CTLA-4 (CD152), but not CD28. Codelivery of plasmids encoding a PPIns-GAD65 fusion construct and B7-1wa protected against both insulitis and diabetes. In vitro, the T cells of covaccinated mice had negative responses to both insulin and GAD65, and this was restored by adding blocking antibodies to transforming growth factor beta1 (TGF-beta1), suggesting a role for this cytokine. Adoptive transfer experiments revealed that DNA vaccination generated protective CD4(+) regulatory T cells (Tr) of either CD25(+) or CD25(-) phenotype. Furthermore, vaccinated mice had increased numbers of T cells with Tr-associated markers, such as CTLA-4, Foxp3, and membrane-bound TGF-beta1. Tr cells inhibited the responses of diabetogenic T cells to islet antigens, and depletion of T cells expressing membrane-bound TGF-beta1 abolished the suppressive effect. Thus, selective engagement of CTLA-4 during islet-antigen DNA vaccination induces Tr cells that protect against this autoimmune disease.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, CD
- Antigens, Differentiation/metabolism
- Biomarkers
- CTLA-4 Antigen
- Cell Differentiation
- Cytokines/biosynthesis
- DNA/genetics
- DNA/immunology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Female
- Immunotherapy
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Ligands
- Mice
- Mice, Inbred NOD
- Phenotype
- Receptors, Interleukin-2/metabolism
- Substrate Specificity
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Yelena Glinka
- Department of Laboratory Medicine and Pathobiology, University of Toronto and St. Michael's Hospital, Toronto, Ontario, Canada M5B 1W8
| | | | | |
Collapse
|
17
|
Li A, Ojogho O, Franco E, Baron P, Iwaki Y, Escher A. Pro-apoptotic DNA vaccination ameliorates new onset of autoimmune diabetes in NOD mice and induces foxp3+ regulatory T cells in vitro. Vaccine 2006; 24:5036-46. [PMID: 16621191 DOI: 10.1016/j.vaccine.2006.03.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 03/10/2006] [Accepted: 03/16/2006] [Indexed: 11/24/2022]
Abstract
We have shown previously that incorporation of a cDNA coding for the pro-apoptotic protein BAX into plasmid DNA coding for a secreted form of the pancreatic beta-cell antigen glutamic acid decarboxylase (GAD) promotes prevention of type 1 diabetes in non-obese diabetic (NOD) mice. Here we present evidence indicating that injection of the same vaccine at time of early diabetes onset could ameliorate the disease with efficacy, with 42% of mice overtly diabetic by 40 weeks of age compared to 92% in control groups. In addition, immunological analysis revealed that the DNA vaccine induced CD4(+)CD25(+) T cells cultured from draining lymph nodes that had immunosuppressive function in vitro. The induced regulatory T cells (Tregs) expressed the foxp3 gene and showed cell-contact-dependent as well as TGF-beta- and IL-10-independent immunosuppressive activity. Data also revealed that CD4(+)CD25(-) T cells from mice immunized with the DNA vaccine yielded a cell population that was foxp3(+), showed increased expression of CD25 compared to control, and had immunosuppressive function in vitro, indicating that Tregs could have developed from antigen-induced, peripheral T lymphocytes. In contrast, injection of DNA coding for SGAD55 or BAX alone did not induce Tregs. Altogether, our data confirm that pro-apoptotic DNA vaccination can be used as an immunosuppressive strategy and demonstrate its potential for therapy of pathological autoimmunity.
Collapse
Affiliation(s)
- Alice Li
- Center for Transplant Immunology Research, Loma Linda University and Medical Center, CA 92354, USA
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
The most intensively studied autoimmune disorder, type 1 diabetes mellitus (DM1), has attracted perhaps the greatest interest for gene-based therapeutic and prophylactic interventions. The final clinical manifestation of this immunologically and genetically complex disease, the absence of insulin, is the major starting point for almost all the gene therapy modalities attempted to date. Insulin replacement by transplantation of islets of Langerhans or surrogate beta cells is the obvious choice, but the allogeneic nature of the transplants activates potent antidonor immunoreactivity necessitating gene and cell-based immunosuppressive strategies as an alternative to the toxic pharmacologic immunosuppressives indicated for classic solid organ transplants. Accumulating knowledge of the cellular mechanisms involved in onset, however, have yielded promising tolerance induction prophylactic approaches using genes and cells. Despite the early successes in a number of animal models, the true test of efficacy in humans remains to be demonstrated.
Collapse
Affiliation(s)
- Nick Giannoukakis
- Diabetes Institute, Pediatric Research Section, Children's Hospital of Pittsburgh and University of Pittsburgh, Rangos Research Center, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
19
|
Abstract
Whether autoimmunity results primarily from a defect of the immune system, target organ dysfunction, or both remains an open issue in most human autoimmune diseases. The highly multigenic background on which diabetes develops in the NOD mouse and in the human suggests that numerous gene variants associate in contributing to activation of autoimmunity to beta-cells. Both immune genes and islet-related genes are involved. The presence of beta-cells is required for initiation of diabetes autoimmunity to proceed. Available experiments in the NOD mouse and epidemiological evidence in the human point to proinsulin as a key autoantigen in diabetes. The functional importance of insulin, the high number of autoantigens characterized at different stages of diabetes, and their clustering within beta-cell subparticles point to the islet as a starting point in the initiation phase of the disease. Genes that direct the autoimmune reaction toward the beta-cell target, autoantigens that are recognized by autoreactive B- and T-cells along the autoimmune process, the importance of beta-cells in the activation of autoreactive lymphocytes, and the expression level of key beta-cell molecules along diabetes development are successively considered in this review.
Collapse
Affiliation(s)
- Béatrice Faideau
- INSERM U561, Hôpital Cochin-Saint Vincent de Paul, 82 Avenue Denfert Rochereau, 75014 Paris, France
| | | | | | | | | |
Collapse
|
20
|
Ho PP, Fontoura P, Platten M, Sobel RA, DeVoss JJ, Lee LY, Kidd BA, Tomooka BH, Capers J, Agrawal A, Gupta R, Zernik J, Yee MK, Lee BJ, Garren H, Robinson WH, Steinman L. A suppressive oligodeoxynucleotide enhances the efficacy of myelin cocktail/IL-4-tolerizing DNA vaccination and treats autoimmune disease. THE JOURNAL OF IMMUNOLOGY 2005; 175:6226-34. [PMID: 16237121 DOI: 10.4049/jimmunol.175.9.6226] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Targeting pathogenic T cells with Ag-specific tolerizing DNA vaccines encoding autoantigens is a powerful and feasible therapeutic strategy for Th1-mediated autoimmune diseases. However, plasmid DNA contains abundant unmethylated CpG motifs, which induce a strong Th1 immune response. We describe here a novel approach to counteract this undesired side effect of plasmid DNA used for vaccination in Th1-mediated autoimmune diseases. In chronic relapsing experimental autoimmune encephalomyelitis (EAE), combining a myelin cocktail plus IL-4-tolerizing DNA vaccine with a suppressive GpG oligodeoxynucleotide (GpG-ODN) induced a shift of the autoreactive T cell response toward a protective Th2 cytokine pattern. Myelin microarrays demonstrate that tolerizing DNA vaccination plus GpG-ODN further decreased anti-myelin autoantibody epitope spreading and shifted the autoreactive B cell response to a protective IgG1 isotype. Moreover, the addition of GpG-ODN to tolerizing DNA vaccination therapy effectively reduced overall mean disease severity in both the chronic relapsing EAE and chronic progressive EAE mouse models. In conclusion, suppressive GpG-ODN effectively counteracted the undesired CpG-induced inflammatory effect of a tolerizing DNA vaccine in a Th1-mediated autoimmune disease by skewing both the autoaggressive T cell and B cell responses toward a protective Th2 phenotype. These results demonstrate that suppressive GpG-ODN is a simple and highly effective novel therapeutic adjuvant that will boost the efficacy of Ag-specific tolerizing DNA vaccines used for treating Th1-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Peggy P Ho
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Prevention of type 1 diabetes mellitus requires early intervention in the autoimmune process directed against beta cells of the pancreatic islets of Langerhans. This autoimmune inflammatory process is thought to be caused by the effect of Th1 cells and their secreted cytokines (e.g. interferon) and to be suppressed by Th2-secreted anti-inflammatory cytokines (e.g. IL-4, IL-10). Various methods aimed specifically at halting or modulating this response have been attempted. An alternative method is the re-induction of tolerance towards the putative self antigen that causes the disease. Proposed antigens such as insulin, glutamic acid decarboxilase (GAD) and the heat shock protein 60 (Hsp60)-derived peptide 277 have been used successfully in murine diabetes models and in initial clinical trials in early diabetes patients. Here, we review the results of these trials.
Collapse
Affiliation(s)
- Itamar Raz
- Diabetes Research Center, Department of Medicine, Hadassah-Hebrew University Hospital, Jerusalem 91120, Israel
| | | | | |
Collapse
|
22
|
Han G, Li Y, Wang J, Wang R, Chen G, Song L, Xu R, Yu M, Wu X, Qian J, Shen B. Active Tolerance Induction and Prevention of Autoimmune Diabetes by Immunogene Therapy Using Recombinant Adenoassociated Virus Expressing Glutamic Acid Decarboxylase 65 Peptide GAD500–585. THE JOURNAL OF IMMUNOLOGY 2005; 174:4516-24. [PMID: 15814672 DOI: 10.4049/jimmunol.174.8.4516] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tolerance induction of autoreactive T cells against pancreatic beta cell-specific autoantigens such as glutamic acid decarboxylase 65 (GAD65) and insulin has been attempted as a method to prevent autoimmune diabetes. In this study, we investigate whether adenoassociated virus (AAV) gene delivery of multiple immunodominant epitopes expressing GAD(500-585) could induce potent immune tolerance and persistently suppress autoimmune diabetes in NOD mice. A single muscle injection of 7-wk-old female NOD mice with rAAV/GAD(500-585) (3 x 10(11) IU/mouse) quantitatively reduced pancreatic insulitis and efficiently prevented the development of overt type I diabetes. This prevention was marked by the inactivation of GAD(500-585)-responsive T lymphocytes, the enhanced GAD(500-585)-specific Th2 response (characterized by increased IL-4, IL-10 production, and decreased IFN-gamma production; especially elevated anti-GAD(500-585) IgG1 titer; and relatively unchanged anti-GAD(500-585) IgG2b titer), the increased secretion of TGF-beta, and the production of protective regulatory cells. Our studies also revealed that peptides 509-528, 570-585, and 554-546 in the region of GAD(500-585) played important roles in rAAV/GAD(500-585) immunization-induced immune tolerance. These data indicate that using AAV, a vector with advantage for therapeutic gene delivery, to transfer autoantigen peptide GAD(500-585), can induce immunological tolerance through active suppression of effector T cells and prevent type I diabetes in NOD mice.
Collapse
Affiliation(s)
- Gencheng Han
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Type 1 diabetes is a T-cell-mediated autoimmune disease, resulting in destruction of the insulin-producing beta cells in the pancreas. Disease progression is thought to involve the action of T-cells, particularly those producing Th1-type cytokines. Given the complexity in understanding the precise etiology of autoimmune diseases, the diversity of autoantigens, and the variability that exists between individual patients, it might be very difficult to eliminate autoaggressive T-cell responses without resorting to generalized means of immunosuppression. However, recent evidence shows that autoimmune processes are composed not only of autoaggressive T-cell responses but also of autoreactive regulatory components. Enhancing regulatory T-cell responses, therefore, has become an area of intense focus as a means of treating autoimmune diseases like type 1 diabetes. This review will concentrate on two different types of regulatory T-cells, the naturally occurring ('professional') CD4+CD25+ T-cells and antigen-induced ('adaptive') CD4+ Th2-like regulatory T-cells.
Collapse
Affiliation(s)
- Amy E Juedes
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, San Diego, CA, USA
| | | |
Collapse
|
24
|
Gauvrit A, Debailleul M, Vu AT, Sai P, Bach JM. DNA vaccination encoding glutamic acid decarboxylase can enhance insulitis and diabetes in correlation with a specific Th2/3 CD4 T cell response in non-obese diabetic mice. Clin Exp Immunol 2004; 137:253-62. [PMID: 15270841 PMCID: PMC1809102 DOI: 10.1111/j.1365-2249.2004.02546.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
DNA vaccination encoding beta cell autoantigens has been shown very recently to prevent type I diabetes in non-obese diabetic (NOD) mice. However, DNA vaccination encoding microbial or reporter antigens is known to induce specific long-lasting CD4 Th1 and strong cytolytic CD8 T cell responses. As this immune phenotype is associated strongly with beta cell destruction leading to diabetes, we have chosen to study the effects of plasmids encoding glutamic acid decarboxylase (GAD), a crucial beta cell autoantigen, in female NOD mice that developed a 'moderate' diabetes incidence. In the present study, 3-week-old female NOD mice were vaccinated twice in tibialis muscles with plasmid-DNA encoding 65-kDa GAD or betagalactosidase. In GAD-DNA immunized mice, diabetes cumulative incidence (P < 3.10(-3)) and insulitis (P < 7.10(-3)) increased significantly. Simultaneously, DNA immunization induced GAD-specific CD4 T cells secreting interleukin (IL)-4 (P < 0.05) and transforming growth factor (TGF)-beta (P = 0.03). These cells were detected in spleen and in pancreatic lymph nodes. Furthermore, vaccination produced high amounts of Th2 cytokine-related IgG1 (P < 3.10(-3)) and TGF-beta-related IgG2b to GAD (P = 0.015). Surprisingly, diabetes onset was correlated positively with Th2-related GAD-specific IgG1 (P < 10(-4)) and TGF-beta-related IgG2b (P < 3.10(-3)). Moreover, pancreatic lesions resembled Th2-related allergic inflammation. These results indicate, for the first time, that GAD-DNA vaccination could increase insulitis and diabetes in NOD mice. In addition, our study suggests that Th2/3 cells may have potentiated beta cell injury.
Collapse
Affiliation(s)
- A Gauvrit
- Immuno-Endocrinology Unit, ENVN/INRA/University, Nantes, France
| | | | | | | | | |
Collapse
|
25
|
Li AF, Hough J, Henderson D, Escher A. Co-delivery of pro-apoptotic BAX with a DNA vaccine recruits dendritic cells and promotes efficacy of autoimmune diabetes prevention in mice. Vaccine 2004; 22:1751-63. [PMID: 15068859 DOI: 10.1016/j.vaccine.2003.10.049] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2003] [Accepted: 10/09/2003] [Indexed: 12/14/2022]
Abstract
Genetic vaccines encoding pancreatic beta cell antigens can prevent autoimmune (type 1) diabetes when delivered into murine model systems, but there is a need to improve their efficacy. Here, we investigated the effects of intramuscular delivery of DNA coding for the pro-apoptotic protein BAX together with an intracellular or a secreted form of the beta cell antigen glutamic acid decarboxylase (GAD) on diabetes onset and immune responses in non-obese diabetic (NOD) mice. We hypothesized that induction of apoptosis in vaccine-containing cells could lead to GAD tolerance and disease suppression. Remarkably, monitoring of spontaneous diabetes onset indicated that only delivery of DNA coding for secreted GAD and BAX resulted in significant prevention of the disease. Using GFP as a model plasmid-encoded antigen revealed that co-delivery of BAX resulted in the recruitment of GFP-containing dendritic cells (DCs) in the draining lymph nodes and spleen of NOD mice. Furthermore, data indicated that subcellular localization of GAD had an effect on both the number and function of antigen presenting cells (APCs) recruited by BAX as well as on IFN-gamma secretion, and that diabetes suppression was unlikely to be caused by increased T helper 2 (Th2)-like activity. Our results indicate that, under certain conditions, co-delivery of DNA encoding BAX can improve the efficacy of genetic vaccination for prevention of pathogenic autoimmunity via a mechanism likely to involve modulation of antigen presenting cell function. In addition, our data also suggest that properties associated with subcellular localization of an antigen in apoptotic cells can have a significant effect on induced immune responses.
Collapse
MESH Headings
- Animals
- Antibodies/analysis
- Apoptosis/genetics
- Apoptosis/immunology
- Autoimmune Diseases/prevention & control
- Blood Glucose/metabolism
- Cytokines/biosynthesis
- Cytokines/genetics
- DNA, Complementary/genetics
- DNA, Complementary/immunology
- Dendritic Cells/immunology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/prevention & control
- Enzyme-Linked Immunosorbent Assay
- Female
- Fluorescent Antibody Technique
- Genes, bcl-2/genetics
- Genes, bcl-2/immunology
- Glutamate Decarboxylase/genetics
- Glutamate Decarboxylase/immunology
- Humans
- Immunoblotting
- Injections, Intramuscular
- Isoenzymes/genetics
- Isoenzymes/immunology
- Luciferases/biosynthesis
- Luciferases/genetics
- Luciferases/immunology
- Lymphocyte Culture Test, Mixed
- Mice
- Mice, Inbred NOD
- Plasmids/genetics
- Plasmids/immunology
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/immunology
- Proto-Oncogene Proteins c-bcl-2
- Subcellular Fractions/metabolism
- Th1 Cells/immunology
- Th2 Cells/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- bcl-2-Associated X Protein
Collapse
Affiliation(s)
- Alice F Li
- Department of Biochemistry and Microbiology, Center for Molecular Biology and Gene Therapy, 11085 Campus Street, Loma Linda University, Loma Linda, CA 92350, USA
| | | | | | | |
Collapse
|
26
|
Giannoukakis N, Trucco M. Current status and prospects for gene and cell therapeutics for type 1 diabetes mellitus. Rev Endocr Metab Disord 2003; 4:369-80. [PMID: 14618022 DOI: 10.1023/a:1027306213563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Nick Giannoukakis
- Department of Pathology and Diabetes Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
27
|
Robinson WH, Fontoura P, Lee BJ, de Vegvar HEN, Tom J, Pedotti R, DiGennaro CD, Mitchell DJ, Fong D, Ho PPK, Ruiz PJ, Maverakis E, Stevens DB, Bernard CCA, Martin R, Kuchroo VK, van Noort JM, Genain CP, Amor S, Olsson T, Utz PJ, Garren H, Steinman L. Protein microarrays guide tolerizing DNA vaccine treatment of autoimmune encephalomyelitis. Nat Biotechnol 2003; 21:1033-9. [PMID: 12910246 DOI: 10.1038/nbt859] [Citation(s) in RCA: 199] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2003] [Accepted: 06/25/2003] [Indexed: 11/09/2022]
Abstract
The diversity of autoimmune responses poses a formidable challenge to the development of antigen-specific tolerizing therapy. We developed 'myelin proteome' microarrays to profile the evolution of autoantibody responses in experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis (MS). Increased diversity of autoantibody responses in acute EAE predicted a more severe clinical course. Chronic EAE was associated with previously undescribed extensive intra- and intermolecular epitope spreading of autoreactive B-cell responses. Array analysis of autoantigens targeted in acute EAE was used to guide the choice of autoantigen cDNAs to be incorporated into expression plasmids so as to generate tolerizing vaccines. Tolerizing DNA vaccines encoding a greater number of array-determined myelin targets proved superior in treating established EAE and reduced epitope spreading of autoreactive B-cell responses. Proteomic monitoring of autoantibody responses provides a useful approach to monitor autoimmune disease and to develop and tailor disease- and patient-specific tolerizing DNA vaccines.
Collapse
Affiliation(s)
- William H Robinson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Seifarth C, Pop S, Liu B, Wong CP, Tisch R. More stringent conditions of plasmid DNA vaccination are required to protect grafted versus endogenous islets in nonobese diabetic mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:469-76. [PMID: 12817032 DOI: 10.4049/jimmunol.171.1.469] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recurrent autoimmune destruction of the insulin-producing beta cells is a key factor limiting successful islet graft transplantation in type I diabetic patients. In this study, we investigated the feasibility of using an Ag-specific plasmid DNA (pDNA)-based strategy to protect pro-islets that had developed from a neonatal pancreas implanted under the kidney capsule of nonobese diabetic (NOD) mice. NOD recipient mice immunized with pDNA encoding a glutamic acid decarboxylase 65 (GAD65)-IgFc fusion protein (JwGAD65), IL-4 (JwIL4), and IL-10 (pIL10) exhibited an increased number of intact pro-islets expressing high levels of insulin 15 wk posttransplant, relative to NOD recipient mice immunized with pDNA encoding a hen egg lysozyme (HEL)-IgFc fusion protein (JwHEL)+JwIL4 and pIL10 or left untreated. Notably, the majority of grafted pro-islets detected in JwGAD65+JwIL4- plus pIL10-treated recipients was free of insulitis. In addition, administration of JwGAD65+JwIL4+pIL10 provided optimal protection for engrafted islets compared with recipient NOD mice treated with JwGAD65+JwIL4 or JwGAD65+pIL10, despite effective protection of endogenous islets mediated by the respective pDNA treatments. Efficient protection of pro-islet grafts correlated with a marked reduction in GAD65-specific IFN-gamma reactivity and an increase in IL-10-secreting T cells. These results demonstrate that pDNA vaccination can be an effective strategy to mediate long-term protection of pro-islet grafts in an Ag-specific manner and that conditions are more stringent to suppress autoimmune destruction of grafted vs endogenous islets.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Female
- Glutamate Decarboxylase/administration & dosage
- Glutamate Decarboxylase/genetics
- Glutamate Decarboxylase/therapeutic use
- Immunoglobulin Fc Fragments/administration & dosage
- Immunoglobulin Fc Fragments/genetics
- Immunoglobulin Fc Fragments/therapeutic use
- Injections, Intramuscular
- Interferon-gamma/antagonists & inhibitors
- Interferon-gamma/metabolism
- Interleukin-10/administration & dosage
- Interleukin-10/genetics
- Interleukin-10/therapeutic use
- Interleukin-4/administration & dosage
- Interleukin-4/genetics
- Interleukin-4/metabolism
- Interleukin-4/therapeutic use
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Islets of Langerhans Transplantation/immunology
- Islets of Langerhans Transplantation/methods
- Islets of Langerhans Transplantation/pathology
- Isoenzymes/administration & dosage
- Isoenzymes/genetics
- Isoenzymes/therapeutic use
- Mice
- Mice, Inbred NOD
- Plasmids/administration & dosage
- Plasmids/immunology
- Plasmids/therapeutic use
- Transplantation, Isogeneic
- Up-Regulation/genetics
- Up-Regulation/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Vaccines, DNA/therapeutic use
Collapse
Affiliation(s)
- Christian Seifarth
- Department of Microbiology and Immunology, School of Medicine, Curriculum in Oral Biology, and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
29
|
Bottino R, Lemarchand P, Trucco M, Giannoukakis N. Gene- and cell-based therapeutics for type I diabetes mellitus. Gene Ther 2003; 10:875-89. [PMID: 12732873 DOI: 10.1038/sj.gt.3302015] [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: 12/29/2022]
Abstract
Type 1 diabetes mellitus, an autoimmune disorder is an attractive candidate for gene and cell-based therapy. From the use of gene-engineered immune cells to induce hyporesponsiveness to autoantigens to islet and beta cell surrogate transplants expressing immunoregulatory genes to provide a local pocket of immune privilege, these strategies have demonstrated proof of concept to the point where translational studies can be initiated. Nonetheless, along with the proof of concept, a number of important issues have been raised by the choice of vector and expression system as well as the point of intervention; prophylactic or therapeutic. An assessment of the current state of the science and potential leads to the conclusion that some strategies are ready for safety trials while others require varying degrees of technical and conceptual refinement.
Collapse
Affiliation(s)
- R Bottino
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
30
|
Affiliation(s)
- Gerald T Nepom
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA.
| |
Collapse
|
31
|
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
|
32
|
Abstract
A precise knowledge of the defects underlying type 1 and type 2 diabetes is essential for designing appropriate therapeutic strategies. Because experiments in humans are limited, naturally occurring, and especially genetically engineered rodent models, have revolutionized research in diabetes. We review some of the models created recently and discuss their impact on human diabetes.
Collapse
Affiliation(s)
- Rohit N Kulkarni
- Joslin Diabetes Center, One Joslin Place, Room 602, Boston, MA 02215, USA.
| | | |
Collapse
|
33
|
Glinka Y, De Pooter R, Croze F, Prud'homme GJ. Regulatory cytokine production stimulated by DNA vaccination against an altered form of glutamic acid decarboxylase 65 in nonobese diabetic mice. J Mol Med (Berl) 2003; 81:175-84. [PMID: 12682726 DOI: 10.1007/s00109-002-0412-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2002] [Accepted: 12/03/2002] [Indexed: 10/20/2022]
Abstract
Nonobese diabetic (NOD) mice develop a T-cell dependent autoimmune form of diabetes, in which glutamic acid decarboxylase 65 (GAD65) is an important islet target antigen. Intramuscular DNA vaccination with a plasmid encoding native GAD65 (a cytosolic antigen) did not significantly alter the incidence of diabetes, but vaccination against an altered form of GAD65 with a signal peptide (spGAD), which is secreted in vitro, was protective. The preventive effect was further enhanced by repeated injections of the spGAD plasmid. Following DNA injection into muscle GAD65 was expressed for several months, and this was not accompanied by an inflammatory response. Immunization against GAD65 was not associated with substantial alterations in cytokine production by splenic lymphocytes stimulated with immunogenic GAD65 peptides. In contrast, spGAD induced increased secretion of both interleukin 10 and interferon gamma and a striking decrease in the interferon gamma/interleukin 10 ratio in culture supernatants. Similarly, spGAD-immunized mice had higher serum interleukin 10 levels and lower serum interferon gamma levels than other groups, suggesting a systemic effect. In nondiabetic mice there was increased basal production of transforming growth factor beta(1), which was enhanced by antigenic stimulation. These alterations in regulatory cytokine production were apparent both early and late after the treatment was initiated. These findings suggest that DNA vaccination against spGAD protects NOD mice by increasing regulatory cytokine production.
Collapse
Affiliation(s)
- Yelena Glinka
- Department of Pathology, McGill University, Montreal, Qc H3A2B4 Canada
| | | | | | | |
Collapse
|
34
|
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
|
35
|
Hänninen A, Hamilton-Williams E, Kurts C. Development of new strategies to prevent type 1 diabetes: the role of animal models. Ann Med 2003; 35:546-63. [PMID: 14708966 DOI: 10.1080/07853890310014597] [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: 02/05/2023] Open
Abstract
Type 1 diabetes is an immune-mediated disease typically preceded by a long preclinical stage during which a growing number of islet-cell-specific autoantibodies appear in the serum. Although antigen-specific T lymphocytes and cytokines rather than these autoantibodies are the likely executors of beta-cell-destruction, these autoantibodies reflect the existence of autoimmunity that targets islet beta-cells. Abrogation of this autoimmunity during the preclinical stage would be the key to the prevention of type 1 diabetes. However, the quest of protecting islet-cells from the immune attack requires detailed knowledge of mechanisms that control islet-inflammation and beta-cell-destruction, and of mechanisms that control immune tolerance to peripheral self-antigens in general. This knowledge can only be obtained through further innovative research in experimental animal models. In this review, we will first examine how research in non-obese diabetic mice has already led to promising new strategies of diabetes prevention now being tested in human clinical trials. Thereafter, we will discuss how recent advances in understanding the mechanisms that control immune response to peripheral self-antigens such as beta-cell antigens may help to develop even more selective and effective strategies to prevent diabetes in the future.
Collapse
Affiliation(s)
- Arno Hänninen
- MediCity Research Laboratory, Turku University, Finland,
| | | | | |
Collapse
|
36
|
Karges W, Pechhold K, Al Dahouk S, Riegger I, Rief M, Wissmann A, Schirmbeck R, Barth C, Boehm BO. Induction of autoimmune diabetes through insulin (but not GAD65) DNA vaccination in nonobese diabetic and in RIP-B7.1 mice. Diabetes 2002; 51:3237-44. [PMID: 12401715 DOI: 10.2337/diabetes.51.11.3237] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Insulin has been used to modify T-cell autoimmunity in experimental models of type 1 diabetes. In a large clinical trial, the effect of insulin to prevent type 1 diabetes is currently investigated. We here show that insulin can adversely trigger autoimmune diabetes in two mouse models of type 1 diabetes, using intramuscular DNA vaccination for antigen administration. In female nonobese diabetic (NOD) mice, diabetes development was enhanced after preproinsulin (ppIns) DNA treatment, and natural diabetes resistance in male NOD mice was diminished by ppIns DNA vaccination. In contrast, GAD65 DNA conferred partial diabetes protection, and empty DNA plasmid was without effect. In RIP-B7.1 C57BL/6 mice (expressing the T-cell costimulatory molecule B7.1 in pancreatic beta-cells), autoimmune diabetes occurred in 70% of animals after ppIns vaccination, whereas diabetes did not develop spontaneously in RIP-B7.1 mice or after GAD65 or control DNA treatment. Diabetes was characterized by diffuse CD4(+)CD8(+) T-cell infiltration of pancreatic islets and severe insulin deficiency, and ppIns, proinsulin, and insulin DNA were equally effective for disease induction. Our work provides a new model of experimental autoimmune diabetes suitable to study mechanisms and outcomes of insulin-specific T-cell reactivity. In antigen-based prevention of type 1 diabetes, diabetes acceleration should be considered as a potential adverse result.
Collapse
Affiliation(s)
- Wolfram Karges
- Division of Endocrinology, Department of Internal Medicine, University of Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Halbout P, Briand JP, Bécourt C, Muller S, Boitard C. T cell response to preproinsulin I and II in the nonobese diabetic mouse. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2436-43. [PMID: 12193712 DOI: 10.4049/jimmunol.169.5.2436] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunization against insulin, insulin B chain, or B chain peptide B(9-23) (preproinsulin peptide II(33-47)) prevents diabetes in the nonobese diabetic (NOD) mouse. Whether or not peptide II(33-47) is the only proinsulin determinant recognized by CD4 T cells remains unclear. Using two peptide libraries spanning the entire sequence of preproinsulin I and preproinsulin II, respectively, we identified T cells specific for four proinsulin epitopes within the islet cell infiltrate of prediabetic female NOD mice. These epitopes were among immunogenic epitopes to which a T cell response was detected after immunization of NOD mice with individual peptides in CFA. Immunogenic epitopes were found on both isoforms of insulin, especially proinsulin II, which is the isoform expressed in the thymus. The autoimmune response to proinsulin represented only part of the immune response to islet cells within the islet cell infiltrate in 15-wk-old NOD mice. This is the first systematic study of preproinsulin T cell epitopes in the NOD mouse model.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cell Movement/immunology
- Diabetes Mellitus, Type 1/immunology
- Epitope Mapping
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Female
- Hybridomas/immunology
- Hybridomas/metabolism
- Injections, Subcutaneous
- Insulin
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Lymphocyte Count
- Male
- Mice
- Mice, Inbred NOD
- Molecular Sequence Data
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Prediabetic State/immunology
- Proinsulin/administration & dosage
- Proinsulin/immunology
- Protein Precursors/administration & dosage
- Protein Precursors/immunology
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Philippe Halbout
- Institut National de la Santé et de la Recherche Médicale Unité 561, Hôpital Cochin-Saint Vincent de Paul, Paris, France
| | | | | | | | | |
Collapse
|
38
|
Urbanek-Ruiz I, Ruiz PJ, Steinman L, Fathman CG. Immunomodulatory vaccination in autoimmune disease. Endocrinol Metab Clin North Am 2002; 31:441-56, viii-ix. [PMID: 12092460 DOI: 10.1016/s0889-8529(01)00021-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The development of vaccines is arguably the most significant achievement in medicine to date. The practice of innoculation with the fluid from a sore to protect from a disease actually dates back to ancient China; however, with the introduction of Jenner's smallpox vaccine, and greater understanding of the immune system, vaccines have become specific and systematic. Traditional vaccines have used killed pathogens (hepatitis A and the Salk polio vaccines), immunogenic subunits of a given pathogen (hepatitis B subunit vaccine), or live attenuated pathogens (measles, mumps, rubella, Sabin polio vaccines) to generate protective immunity. Currently, a new generation of vaccines that use the genetic material of a pathogen to elicit protective immunity are being developed. Although the most widespread and successful use of vaccines today remains in the arena of infectious diseases, manipulations of immune responses to protect against cancers, neurologic diseases, and autoimmunity are being explored rigorously.
Collapse
Affiliation(s)
- Irene Urbanek-Ruiz
- Department of Medicine, Division of Immunology, Center for Clinical Immunology at Stanford, Stanford University School of Medicine, 269 Campus Drive, Rm 2240, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
39
|
Robinson WH, Garren H, Utz PJ, Steinman L. Millennium Award. Proteomics for the development of DNA tolerizing vaccines to treat autoimmune disease. Clin Immunol 2002; 103:7-12. [PMID: 11987980 DOI: 10.1006/clim.2002.5185] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Autoimmune disease affects 3% of the world population, yet current therapies that globally suppress immune function are inadequate. Tremendous need exists for specific and curative therapies, and we describe a strategy for development of antigen-specific therapies that inactivate pathogenic lymphocytes causing tissue injury. Major barriers to development of antigen-specific therapies for T-cell-mediated autoimmune diseases, such as multiple sclerosis, rheumatoid arthritis, and autoimmune diabetes, include (i) lack of knowledge of the specificity of autoimmune responses, for which proteomic technologies represent powerful tools to identify the self-protein targets of the autoimmune response, and (ii) lack of methods to induce specific immune tolerance, for which DNA tolerizing vaccines represent a promising strategy. We termed our approach Reverse Genomics: use of the proteomics-determined specificity of the autoantibody response to develop and select DNA tolerizing vaccines. Studies performed using animal models for multiple sclerosis and autoimmune diabetes support our Reverse Genomics approach. Through integration of proteomics with specific tolerizing therapies, we are developing a comprehensive approach to treat human autoimmune disease.
Collapse
Affiliation(s)
- William H Robinson
- Department of Neurology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | |
Collapse
|
40
|
Prud'homme GJ, Chang Y, Li X. Immunoinhibitory DNA vaccine protects against autoimmune diabetes through cDNA encoding a selective CTLA-4 (CD152) ligand. Hum Gene Ther 2002; 13:395-406. [PMID: 11860706 DOI: 10.1089/10430340252792521] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Cytotoxic T lymphocyte antigen 4 (CTLA-4 or CD152) is a strong negative regulator of T cell activity. Like CD28 (a positive regulator) it binds to B7-1 and B7-2, and there is no known natural selective ligand. Monoclonal antibodies to CTLA-4 generally have a masking effect, enhancing rather than suppressing responses. However, a single amino acid substitution in B7-1 (W88 > A; denoted B7-1wa) abrogates binding to CD28 but not to CTLA-4. We constructed plasmids encoding B7-1 or B7-1wa, as cell-surface or Ig fusion proteins. In a bound state, B7-1-Ig enhanced CD3-mediated T cell activation, but B7-1wa-Ig was inhibitory, as expected of a CTLA-4 ligand. To alter immunity in vivo, we inoculated mice intramuscularly (i.m.) with a carcinoembryonic antigen (CEA) plasmid. Gene transfer was amplified by electroporation. Co-injection of a B7-1wa (membrane-bound form) plasmid blocked induction of anti-CEA immunity, whereas a B7-1 plasmid was stimulatory. We studied this DNA covaccination method in nonobese diabetic (NOD) mice with autoimmune diabetes. Delivery of either preproinsulin I (PPIns) or B7-1wa cDNA alone did not suppress the autoimmune anti-insulin response of spleen cells. However, co-delivery of B7-1wa and PPIns cDNA abrogated reactivity to insulin and ameliorated disease. Interferon-gamma and interleukin-4 were both depressed, arguing against a Th2 bias. Reactivity to glutamic acid decarboxylase 65, another major islet autoantigen, was not altered and suppressor cells were not identified, suggesting induction of tolerance to insulin by either T cell anergy or deletion. Selective engagement of CTLA-4 through gene transfer represents a novel and powerful way to block autoimmunity specifically.
Collapse
MESH Headings
- Abatacept
- Animals
- Antigens, CD
- Antigens, Differentiation/genetics
- Antigens, Differentiation/immunology
- B7-1 Antigen/administration & dosage
- B7-1 Antigen/genetics
- CTLA-4 Antigen
- Cells, Cultured
- DNA, Complementary/administration & dosage
- DNA, Complementary/immunology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/prevention & control
- Female
- Immunoconjugates
- Immunosuppression Therapy/methods
- Insulin/metabolism
- Ligands
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Spleen
- T-Lymphocytes/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Gerald J Prud'homme
- The Department of Pathology, McGill University, Montreal, Qc, Canada H3A2B4.
| | | | | |
Collapse
|