151
|
Creusot RJ, Chang P, Healey DG, Tcherepanova IY, Nicolette CA, Fathman CG. A short pulse of IL-4 delivered by DCs electroporated with modified mRNA can both prevent and treat autoimmune diabetes in NOD mice. Mol Ther 2010; 18:2112-20. [PMID: 20628358 DOI: 10.1038/mt.2010.146] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Bone marrow-derived dendritic cells (DCs) are cells of the immune system that have been used as a tool to boost, modulate, or dampen immune responses. In the context of autoimmunity, DCs can be modified to express immunoregulatory products encoded by transgenes, and used therapeutically in adoptive cellular therapy. DCs that were lentivirally transduced (lt) to express interleukin 4 (IL-4) can significantly delay or prevent the onset of autoimmune diabetes in nonobese diabetic (NOD) mice. However, modifying cells using viral vectors carries the dual risk of oncogenicity or immunogenicity. This study demonstrates that NOD DCs, electroporated with "translationally enhanced" IL-4 mRNA (eDC/IL-4), can be equally efficient therapeutically, despite the reduced amount and shorter duration of IL-4 secretion. Moreover, a single injection of eDC/IL-4 in NOD mice shortly after the onset of hyperglycemia was able to maintain stable glycemia for up to several months in a significant fraction of treated mice. Treatment with eDC/IL-4 boosted regulatory T (Tregs) cell functions and modulated T helper responses to reduce pathogenicity. Thus, treatment with DCs, electroporated with modified IL-4 mRNA to express IL-4 for up to 24 hours, constitutes a viable cellular therapy approach for the regulation of autoimmune diabetes, as a preferred alternative to the use of viral vectors.
Collapse
Affiliation(s)
- Rémi J Creusot
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California 94305-5166, USA
| | | | | | | | | | | |
Collapse
|
152
|
Koulmanda M, Strom TB. T-cell-directed treatment strategies for Type 1 diabetes and the confounding role of inflammation. Immunotherapy 2010; 2:431-6. [PMID: 20635995 PMCID: PMC3070950 DOI: 10.2217/imt.10.40] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Maria Koulmanda
- Harvard Medical School, Beth Israel Deaconess Medical Center, Transplant Institute; E/CLS 609, 330 Brookline Ave, Boston, MA, USA, Tel.: +1 617 735 2882, Fax: +1 617 735 2903,
| | | |
Collapse
|
153
|
Petrich de Marquesini LG, Fu J, Connor KJ, Bishop AJ, McLintock NE, Pope C, Wong FS, Dayan CM. IFN-gamma and IL-10 islet-antigen-specific T cell responses in autoantibody-negative first-degree relatives of patients with type 1 diabetes. Diabetologia 2010; 53:1451-60. [PMID: 20369219 DOI: 10.1007/s00125-010-1739-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 02/08/2010] [Indexed: 01/13/2023]
Abstract
AIMS/HYPOTHESIS Islet antibody-negative first-degree relatives of type 1 diabetes patients have a very low risk of developing diabetes. We studied the balance between IFN-gamma (proinflammatory) and IL-10 (regulatory) T cell responses in these participants. METHODS Peripheral blood T cells from adult (18-50 years old, n = 40) DRB1*0401-positive first-degree relatives negative for GAD and tyrosine phosphatase-like insulinoma antigen 2 (IA-2) antibodies were tested for IFN-gamma and IL-10 responses in a sensitive cytokine enzyme-linked immunospot assay against a panel of seven peptide epitopes derived from IA-2 and proinsulin. Comparison was made with HLA-matched newly diagnosed type 1 diabetic patients (n = 42) and healthy controls (n = 39). RESULTS First-degree relatives and newly diagnosed type 1 diabetic patients displayed a similar frequency of IFN-gamma responses to the peptide panel and both were significantly greater than in healthy controls (relatives 9.6%, patients 11.8%, controls 4.0%, p = 0.003). First-degree relatives and newly diagnosed type 1 diabetic patients also showed similar frequencies of IL-10 responses, which were significantly lower than in healthy controls (relatives 7.1%, patients 9.0%, controls 15.8%, p = 0.003). However, individual IL-10 responses of first-degree relatives were similar in size to those in healthy controls and larger than those in newly diagnosed type 1 diabetic patients (relatives median 29 spot-forming cells/1 x 10(6) peripheral blood mononuclear cells, controls 33, patients 11, p = 0.02). CONCLUSIONS/INTERPRETATION Taken together, these results suggest that antibody-negative first-degree relatives have a balance of proinflammatory and regulatory T cells, which is intermediate between that of newly diagnosed type 1 diabetic patients and healthy controls. This suggests that even a moderate regulatory response may be sufficient to prevent the development of clinical type 1 diabetes in genetically predisposed individuals.
Collapse
Affiliation(s)
- L G Petrich de Marquesini
- Henry Wellcome Laboratory for Integrative Neuroscience and Endocrinology, Dorothy Hodgkin Building, Clinical Science at South Bristol, University of Bristol, Whitson St, Bristol, BS1 3NY, UK
| | | | | | | | | | | | | | | |
Collapse
|
154
|
Cox SL, Silveira PA. Emerging roles for B lymphocytes in Type 1 diabetes. Expert Rev Clin Immunol 2010; 5:311-24. [PMID: 20477009 DOI: 10.1586/eci.09.4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Self-reactive B lymphocytes play two main pathological roles in autoimmune diseases: as secretors of autoantibodies and as specialized antigen-presenting cells that present self-components to autoreactive T lymphocytes. In recognition of these roles, recent clinical trials have utilized B-lymphocyte-depleting monoclonal antibodies to treat various autoimmune diseases, with encouraging results in those where humoral autoimmunity is clearly important. Surprisingly, recent results in animal models suggest that B-lymphocyte depletion may also be effective in the treatment of T-lymphocyte-mediated autoimmune diseases, such as Type 1 diabetes (T1D). This article reviews the experimental evidence that has uncovered pathogenic as well as regulatory roles for B lymphocytes in the prodrome of T1D and how this information is being used to develop novel therapeutic strategies to treat the disease.
Collapse
Affiliation(s)
- S Lewis Cox
- Immunology Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia.
| | | |
Collapse
|
155
|
Wicklow BA, Polychronakos C. Insulin auto-immunity: implications for the prevention of Type 1 diabetes mellitus. Expert Rev Clin Immunol 2010; 5:55-62. [PMID: 20476900 DOI: 10.1586/1744666x.5.1.55] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mounting evidence suggests insulin is an important and potentially initiating antigen in the pathogenesis of Type 1 diabetes. High-affinity insulin antibodies are found early in disease development and appear to predict progression. Insulin is the only Type 1 diabetes auto-antigen with exclusive pancreatic expression and the only one whose gene maps to a major susceptibility locus. Preclinical studies in rodent models of immune-mediated diabetes show great promise for the possibility of preventing disease by peripheral tolerization. Translation of this evidence to clinical trials of oral, intranasal and parenteral insulin to invoke immune tolerance and prevent diabetes has not proven successful to date, but promising results in a small subset of highest-risk individuals have maintained enthusiasm for this promising prevention strategy. Currently, studies of oral and intranasal insulin are ongoing to determine the optimal dose, timing and target population for Type 1 diabetes prevention.
Collapse
Affiliation(s)
- Brandy A Wicklow
- Department of Pediatrics, Division of Pediatric Endocrinology, Montreal Children's Hospital, McGill University Health Center, 2300 rue Tupper, Montreal, QC H3H1P3, Canada
| | | |
Collapse
|
156
|
Lin M, Stoica-Nazarov C, Surls J, Kehl M, Bona C, Olsen C, Brumeanu TD, Casares S. Reversal of type 1 diabetes by a new MHC II-peptide chimera: “Single-epitope-mediated suppression” to stabilize a polyclonal autoimmune T-cell process. Eur J Immunol 2010; 40:2277-88. [DOI: 10.1002/eji.200940094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
157
|
Mannering SI, Brodnicki TC. Recent insights into CD4+ T-cell specificity and function in type 1 diabetes. Expert Rev Clin Immunol 2010; 3:557-64. [PMID: 20477160 DOI: 10.1586/1744666x.3.4.557] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Type 1 diabetes (T1D) is caused by T-cell-mediated destruction of the insulin-producing beta-cells in the pancreas. Genetic and immunological evidence from humans and mouse models indicates that CD4(+) T cells play a crucial role in the development and prevention of T1D. The dichotomy between CD4(+) T regulatory and effector T cells has encouraged research into the role of these cell subsets in T1D. New antigens and epitopes recognized by CD4(+) T cells in affected individuals have been identified. Growing knowledge of T-cell specificity and function is helping to develop new assays for analyzing islet antigen-specific CD4(+) T cells from human blood. Here we discuss, with particular reference to human studies, advances in our understanding of CD4(+) T-cell responses in T1D.
Collapse
Affiliation(s)
- Stuart I Mannering
- Autoimmunity & Transplantation Division, The Walter & Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia.
| | | |
Collapse
|
158
|
Tracy S, Drescher KM, Jackson JD, Kim K, Kono K. Enteroviruses, type 1 diabetes and hygiene: a complex relationship. Rev Med Virol 2010; 20:106-16. [PMID: 20049905 PMCID: PMC7169204 DOI: 10.1002/rmv.639] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which the immune system mounts an attack on the host's insulin‐producing β cells. Because most cases of T1D cannot be attributed only to individual genetics, it is strongly inferred that there is a significant environmental contribution, such as infection, impacting disease development. The human enteroviruses (HEV) are common picornaviruses often implicated as triggers of human T1D, although precisely which of the numerous HEV may be involved in human T1D development is unknown. Experiments using non‐obese diabetic (NOD) mice, commonly used to model T1D, show that induction of T1D by HEV infection in NOD mice is a multifactorial process involving both the virus and the host. Interestingly, results demonstrate that HEV infection of NOD mice can also induce long‐term protection from T1D under certain conditions, suggesting that a similar mechanism may occur in humans. Based upon both experimental animal and observational human studies, we postulate that HEV have a dual role in T1D development and can either cause or prevent autoimmune disease. Whichever outcome occurs depends upon multiple variables in the host‐virus equation, many of which can be deduced from results obtained from NOD mouse studies. We propose that the background to the sharply rising T1D incidences observed in the 20th century correlates with increased levels of hygiene in human societies. Viewing T1D in this perspective suggests that potential preventative options could be developed. Copyright © 2009 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- S Tracy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA.
| | | | | | | | | |
Collapse
|
159
|
Shoda L, Kreuwel H, Gadkar K, Zheng Y, Whiting C, Atkinson M, Bluestone J, Mathis D, Young D, Ramanujan S. The Type 1 Diabetes PhysioLab Platform: a validated physiologically based mathematical model of pathogenesis in the non-obese diabetic mouse. Clin Exp Immunol 2010; 161:250-67. [PMID: 20491795 DOI: 10.1111/j.1365-2249.2010.04166.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Type 1 diabetes is an autoimmune disease whose clinical onset signifies a lifelong requirement for insulin therapy and increased risk of medical complications. To increase the efficiency and confidence with which drug candidates advance to human type 1 diabetes clinical trials, we have generated and validated a mathematical model of type 1 diabetes pathophysiology in a well-characterized animal model of spontaneous type 1 diabetes, the non-obese diabetic (NOD) mouse. The model is based on an extensive survey of the public literature and input from an independent scientific advisory board. It reproduces key disease features including activation and expansion of autoreactive lymphocytes in the pancreatic lymph nodes (PLNs), islet infiltration and beta cell loss leading to hyperglycaemia. The model uses ordinary differential and algebraic equations to represent the pancreas and PLN as well as dynamic interactions of multiple cell types (e.g. dendritic cells, macrophages, CD4+ T lymphocytes, CD8+ T lymphocytes, regulatory T cells, beta cells). The simulated features of untreated pathogenesis and disease outcomes for multiple interventions compare favourably with published experimental data. Thus, a mathematical model reproducing type 1 diabetes pathophysiology in the NOD mouse, validated based on accurate reproduction of results from multiple published interventions, is available for in silico hypothesis testing. Predictive biosimulation research evaluating therapeutic strategies and underlying biological mechanisms is intended to deprioritize hypotheses that impact disease outcome weakly and focus experimental research on hypotheses likely to provide insight into the disease and its treatment.
Collapse
Affiliation(s)
- L Shoda
- Entelos Inc., Foster City, CA 94404, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Luo X, Herold KC, Miller SD. Immunotherapy of type 1 diabetes: where are we and where should we be going? Immunity 2010; 32:488-99. [PMID: 20412759 PMCID: PMC2860878 DOI: 10.1016/j.immuni.2010.04.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 03/22/2010] [Accepted: 03/31/2010] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disorder characterized by destruction of insulin-producing pancreatic beta cells. Many broad-based immunosuppressive and antigen-specific immunoregulatory therapies have been and are currently being evaluated for their utility in the prevention and treatment of T1D. Looking forward, this review discusses the potential therapeutic use of antigen-specific tolerance strategies, including tolerance induced by "tolerogenic" antigen-presenting cells pulsed with diabetogenic antigens and transfer of induced or expanded regulatory T cells, which have demonstrated efficacy in nonobese diabetic (NOD) mice. Depending on the time of therapeutic intervention in the T1D disease process, antigen-specific immunoregulatory strategies may be employed as monotherapies, or in combination with short-term tolerance-promoting immunoregulatory drugs and/or drugs promoting differentiation of insulin-producing beta cells from endogenous progenitors.
Collapse
Affiliation(s)
- Xunrong Luo
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Kevan C. Herold
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Stephen D. Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
161
|
Yi W, Seth NP, Martillotti T, Wucherpfennig KW, Sant'Angelo DB, Denzin LK. Targeted regulation of self-peptide presentation prevents type I diabetes in mice without disrupting general immunocompetence. J Clin Invest 2010; 120:1324-36. [PMID: 20200448 DOI: 10.1172/jci40220] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Accepted: 01/06/2010] [Indexed: 01/06/2023] Open
Abstract
Peptide loading of MHC class II (MHCII) molecules is directly catalyzed by the MHCII-like molecule HLA-DM (DM). Another MHCII-like molecule, HLA-DO (DO), associates with DM, thereby modulating DM function. The biological role of DO-mediated regulation of DM activity in vivo remains unknown; however, it has been postulated that DO expression dampens presentation of self antigens, thereby preventing inappropriate T cell activation that ultimately leads to autoimmunity. To test the idea that DO modulation of the MHCII self-peptide repertoire mediates self tolerance, we generated NOD mice that constitutively overexpressed DO in DCs (referred to herein as NOD.DO mice). NOD mice are a mouse model for type 1 diabetes, an autoimmune disease mediated by the destruction of insulin-secreting pancreatic beta cells. Our studies showed that diabetes development was completely blocked in NOD.DO mice. Similar to NOD mice, NOD.DO animals selected a diabetogenic T cell repertoire, and the numbers and function of Tregs were normal. Indeed, immune system function in NOD.DO mice was equivalent to that in NOD mice. NOD.DO DCs, however, presented an altered MHCII-bound self-peptide repertoire, thereby preventing the activation of diabetogenic T cells and subsequent diabetes development. These studies show that DO expression can shape the overall MHCII self-peptide repertoire to promote T cell tolerance.
Collapse
Affiliation(s)
- Woelsung Yi
- Immunology Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | | | | |
Collapse
|
162
|
Rigby MR. The role of the physician-scientist in bridging basic and clinical research in type 1 diabetes. Curr Opin Endocrinol Diabetes Obes 2010; 17:131-42. [PMID: 20160647 DOI: 10.1097/med.0b013e32833759d2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE OF REVIEW In a relatively short time, advances in both basic science and clinical medicine have revolutionized the way we understand disease processes and suggested novel approaches that may be able to be used to treat or cure some of the most relevant human afflictions. In type 1 diabetes, one unintended consequence of this has been the polarization of the investigational groups (i.e., immunologists and endocrinologists) interested in developing novel therapies for this condition. This review will examine how and why such polarization exists, and why past and current approaches to develop critically needed translational investigators may be falling short. RECENT FINDINGS Despite significant efforts to increase the number of individuals trained in both basic science and clinical medicine, the number of academic physician-scientists is on the decline. Increased demands from academic institutions coupled with severe difficulty in securing extramural funding are probably playing important roles in this concerning trend. SUMMARY Type 1 diabetes will continue to be a significant strain on individuals, their families and society until a cure is found. More than ever, there is a critical need to support appropriately trained translational investigators who can best facilitate bringing the promise of basic research to clinical reality.
Collapse
Affiliation(s)
- Mark R Rigby
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| |
Collapse
|
163
|
Customized Cell-Based Treatment Options to Combat Autoimmunity and Restore β-Cell Function in Type 1 Diabetes Mellitus: Current Protocols and Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:641-65. [DOI: 10.1007/978-90-481-3271-3_28] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
164
|
Abstract
The health-promoting effects of various constituents of the olive tree (Olea europaea) are mainly associated with hypoglycaemic and insulin-sensitising activities and have been widely demonstrated in the metabolic syndrome and type 2 diabetes. However, their biological activity in autoimmune type 1 diabetes (T1D) is poorly characterised. Therefore, the influence of O. europaea-derived components present in dry olive leaf extract (DOLE) was examined in two established preclinical models of human T1D, which differ in some aspects of diabetogenesis: multiple low-dose streptozotocin-induced diabetes in susceptible C57BL/6 and CBA/H mouse strains; cyclophosphamide-accelerated diabetes in non-obese diabetic mice. In both T1D models, in vivo administration of DOLE significantly reduced clinical signs of diabetes (hyperglycaemia and body weight loss) and led to complete suppression of histopathological changes in pancreatic islets. In line with these, insulin expression and release were restored in DOLE-treated mice. Interestingly, inducible NO synthase expression and NO production were significantly elevated in peripheral tissues but were down-regulated within the local environment of the endocrine pancreas. This interference was reflected in NO-mediated suppression of T lymphocyte proliferation and lower production of the proinflammatory cytokines interferon-gamma, IL-17 and TNF-alpha in the spleen, with subsequent blockade of beta-cell destruction. The results suggest that DOLE interferes with development of autoimmune diabetes by down-regulating production of proinflammatory and cytotoxic mediators. Therefore, the potential use of a DOLE-enriched diet for prophylaxis/treatment of human T1D, and possibly other autoimmune diseases, is worthy of further investigation.
Collapse
|
165
|
Marin-Gallen S, Clemente-Casares X, Planas R, Pujol-Autonell I, Carrascal J, Carrillo J, Ampudia R, Verdaguer J, Pujol-Borrell R, Borràs FE, Vives-Pi M. Dendritic cells pulsed with antigen-specific apoptotic bodies prevent experimental type 1 diabetes. Clin Exp Immunol 2009; 160:207-14. [PMID: 20030670 DOI: 10.1111/j.1365-2249.2009.04082.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Dendritic cells (DCs) are powerful antigen-presenting cells capable of maintaining peripheral tolerance. The possibility to generate tolerogenic DCs opens new therapeutic approaches in the prevention or remission of autoimmunity. There is currently no treatment inducing long-term tolerance and remission in type 1 diabetes (T1D), a disease caused by autoimmunity towards beta cells. An ideal immunotherapy should inhibit the autoimmune attack, avoid systemic side effects and allow islet regeneration. Apoptotic cells--a source of autoantigens--are cleared rapidly by macrophages and DCs through an immunologically silent process that contributes to maintaining tolerance. Our aims were to prevent T1D and to evaluate the re-establishment of peripheral tolerance using autologous DCs pulsed in vitro with apoptotic bodies from beta cells. Immature DCs derived from bone marrow of non-obese diabetic (NOD) mice were obtained and pulsed with antigen-specific apoptotic bodies from the beta cell line NIT-1. Those DCs that phagocytosed apoptotic cells diminished the expression of co-stimulatory molecules CD40 and CD86 and reduced secretion of proinflammatory cytokines. Moreover, these cells were resistant to increase the expression of co-stimulatory molecules after lipopolysaccharide activation. The administration of these cells to NOD transgenic mice expressing interferon-beta in their insulin-producing cells, a model of accelerated autoimmune diabetes, decreased diabetes incidence significantly and correlated positively with insulitis reduction. DCs pulsed with apoptotic cells that express disease-associated antigens constitutes a promising strategy to prevent T1D.
Collapse
Affiliation(s)
- S Marin-Gallen
- Laboratory of Immunobiology for Research and Diagnosis-Blood and Tissue Bank (LIRAD-BST), Germans Trias i Pujol Research Institute, Department of Cellular Biology, Physiology and Immunology, Autonomous University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Van Belle TL, Juntti T, Liao J, von Herrath MG. Pre-existing autoimmunity determines type 1 diabetes outcome after Flt3-ligand treatment. J Autoimmun 2009; 34:445-52. [PMID: 20004555 DOI: 10.1016/j.jaut.2009.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 11/13/2009] [Accepted: 11/16/2009] [Indexed: 12/11/2022]
Abstract
Redirection of immune responses by manipulation of antigen-presenting cells is an emerging strategy for immunosuppressive treatment of autoimmune diseases. In vivo expansion of dendritic cells (DC) by Fms-like tyrosine kinase-3 (Flt3)-Ligand (FL) treatment was shown to delay diabetes onset in the NOD model of autoimmune diabetes. However, we show here that Flt3 stimulation actually accelerates autoimmunity when autoreactive CD8 T cells are detectable in blood prior to treatment. With autoreactive CD8 cells present, the capacity of FL to expand DCs and induce Treg remained intact, but both numbers and the functional response of islet-specific CD8s were boosted. Also, the inhibitory receptor PD-1 on (autoreactive) CD8 T cells and its ligand PD-L1 on Treg were no longer upregulated. These data highlight the need to pre-screen for T cell autoreactivity prior to generalized DC expansion and illustrate how accelerated disease can occur when the intended initiation of regulatory mechanisms is impaired later in diabetogenesis.
Collapse
Affiliation(s)
- Tom L Van Belle
- Diabetes Center at San Diego, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
167
|
Bresson D, von Herrath M. Immunotherapy for the prevention and treatment of type 1 diabetes: optimizing the path from bench to bedside. Diabetes Care 2009; 32:1753-68. [PMID: 19794001 PMCID: PMC2752914 DOI: 10.2337/dc09-0373] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Damien Bresson
- From the Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Matthias von Herrath
- From the Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| |
Collapse
|
168
|
Norquay LD, D'Aquino KE, Opare-Addo LM, Kuznetsova A, Haas M, Bluestone JA, White MF. Insulin receptor substrate-2 in beta-cells decreases diabetes in nonobese diabetic mice. Endocrinology 2009; 150:4531-40. [PMID: 19574401 PMCID: PMC2754683 DOI: 10.1210/en.2009-0395] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Insulin receptor substrate-2 (Irs2) integrates insulin-like signals with glucose and cAMP agonists to regulate beta-cell growth, function, and survival. This study investigated whether increased Irs2 concentration in beta-cells could reduce beta-cell destruction and the incidence of type 1 diabetes in nonobese diabetic (NOD) mice. NOD mice were intercrossed with C57BL/6 mice overexpressing Irs2 specifically in beta-cells to create NOD(Irs2) mice. After backcrossing NOD(Irs2) mice for 12 generations, glucose homeostasis and diabetes incidence were compared against NOD littermates. Compared with 12-wk-old NOD mice, the progression of severe insulitis was reduced and islet mass was increased in NOD(Irs2) mice. Moreover, the risk of diabetes decreased 50% in NOD(Irs2) mice until the experiment was terminated at 40 wk of age. Nondiabetic NOD(Irs2) mice displayed better glucose tolerance than nondiabetic NOD mice throughout the duration of the study and up to the age of 18 months. The effect of Irs2 to increase islet mass and improve glucose tolerance raised the possibility that NOD(Irs2) mice might have an increased capacity to respond to anti-CD3 antibody, which can induce remission of overt diabetes in some NOD mice. Anti-CD3 antibody injections restored glucose tolerance in newly diabetic NOD and NOD(Irs2) mice; however, anti-CD3-treated NOD(Irs2) mice were less likely than NOD mice to relapse during the experimental period because they displayed 10-fold greater beta-cell mass and mitogenesis. In conclusion, increased Irs2 attenuated the progression of beta-cell destruction, promoted beta-cell mitogenesis, and reduced diabetes incidence in NOD(Irs2) mice.
Collapse
Affiliation(s)
- Lisa D Norquay
- Division of Endocrinology, Children's Hospital Boston, Harvard Medical School, Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
169
|
Rewers M, Gottlieb P. Immunotherapy for the prevention and treatment of type 1 diabetes: human trials and a look into the future. Diabetes Care 2009; 32:1769-82. [PMID: 19794002 PMCID: PMC2752911 DOI: 10.2337/dc09-0374] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Marian Rewers
- From the Barbara Davis Center, University of Colorado, Health Sciences Center, Denver, Colorado
| | - Peter Gottlieb
- From the Barbara Davis Center, University of Colorado, Health Sciences Center, Denver, Colorado
| |
Collapse
|
170
|
Filippi CM, von Herrath MG. Good and bad sides of viruses in Type 1 diabetes. Future Virol 2009. [DOI: 10.2217/fvl.09.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
171
|
Kaminitz A, Mizrahi K, Yaniv I, Farkas DL, Stein J, Askenasy N. Low levels of allogeneic but not syngeneic hematopoietic chimerism reverse autoimmune insulitis in prediabetic NOD mice. J Autoimmun 2009; 33:83-91. [DOI: 10.1016/j.jaut.2009.07.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 06/29/2009] [Accepted: 07/01/2009] [Indexed: 12/29/2022]
|
172
|
von Herrath M, Nepom GT. Remodeling rodent models to mimic human type 1 diabetes. Eur J Immunol 2009; 39:2049-54. [DOI: 10.1002/eji.200939429] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
173
|
Abstract
A major effort has been on-going to develop immunotherapies to prevent and/or treat type 1 diabetes (T1D). This autoimmune disease is characterized by the selective loss of the insulin-producing beta cells via the cumulative effects of autoantigen-specific CD4(+) and CD8(+) T cells, autoantibodies, and activated antigen-presenting cells. To be applicable in a clinical setting, immunotherapies must suppress established beta-cell autoimmunity. Preclinical studies and recent clinical findings suggest that antigen-specific and systemic-based strategies can be effective in this regard. However, either approach alone may not be sufficient to block the diabetogenic response and establish long-term protection in the clinic. In this review, we will discuss the importance of both strategies and how a combinatorial approach to treat T1D is appealing.
Collapse
Affiliation(s)
- Kevin S Goudy
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, 27599, USA
| | | |
Collapse
|
174
|
Rajagopalan G, Mangalam AK, Sen MM, Cheng S, Kudva YC, David CS. Autoimmunity in HLA-DQ8 transgenic mice expressing granulocyte/macrophage-colony stimulating factor in the beta cells of islets of langerhans. Autoimmunity 2009; 40:169-79. [PMID: 17453715 DOI: 10.1080/08916930701201083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Type 1 diabetes (T1D) is a polygenic autoimmune disease with a strong HLA association particularly, HLA-DQ8. We investigated whether islet-specific expression of granulocyte/macrophage colony-stimulating factor (Ins.GM-CSF) in A Beta degrees.NOD.DQ8 mice (HLA-DQ8 transgenic mice on a NOD background lacking endogenous mouse MHC class II molecules) would predispose to development of spontaneous autoimmune diabetes. A Beta degrees.NOD.DQ8 mice expressing GM-CSF in the pancreatic ss cells (8+ G+) as well as litter mates lacking either HLA-DQ8 (8 - G+) or GM-CSF (8+ G -) or both (8 - G -) exhibited insulitis and sialadenitis of varying degrees. But none of the mice progressed to develop T1D. Other than the marked mononuclear cell infiltration in livers of mice expressing GM-CSF irrespective of HLA-DQ8 expression (8+ G+ or 8 - G+), no other changes were observed in the animals. Thus, we have shown for the first time that expression of HLA-DQ8 in the diabetes-predisposing mileu of NOD genetic background is not sufficient to predispose to development of autoimmune diabetes even when the potent immunostimulatory cytokine, GM-CSF is expressed in the pancreatic islets.
Collapse
|
175
|
Abstract
Gene silencing by double-stranded RNA, denoted RNA interference, represents a new paradigm for rational drug design. However, the transformative therapeutic potential of short interfering RNA (siRNA) has been stymied by a key obstacle-safe delivery to specified target cells in vivo. Macrophages are particularly attractive targets for RNA interference therapy because they promote pathogenic inflammatory responses in diseases such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease and diabetes. Here we report the engineering of beta1,3-D-glucan-encapsulated siRNA particles (GeRPs) as efficient oral delivery vehicles that potently silence genes in mouse macrophages in vitro and in vivo. Oral gavage of mice with GeRPs containing as little as 20 microg kg(-1) siRNA directed against tumour necrosis factor alpha (Tnf-alpha) depleted its messenger RNA in macrophages recovered from the peritoneum, spleen, liver and lung, and lowered serum Tnf-alpha levels. Screening with GeRPs for inflammation genes revealed that the mitogen-activated protein kinase kinase kinase kinase 4 (Map4k4) is a previously unknown mediator of cytokine expression. Importantly, silencing Map4k4 in macrophages in vivo protected mice from lipopolysaccharide-induced lethality by inhibiting Tnf-alpha and interleukin-1beta production. This technology defines a new strategy for oral delivery of siRNA to attenuate inflammatory responses in human disease.
Collapse
|
176
|
Filippi CM, Estes EA, Oldham JE, von Herrath MG. Immunoregulatory mechanisms triggered by viral infections protect from type 1 diabetes in mice. J Clin Invest 2009; 119:1515-23. [PMID: 19478458 DOI: 10.1172/jci38503] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 03/11/2009] [Indexed: 01/07/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that is caused by the destruction of insulin-producing beta cells. Viral infections induce immune responses that can damage beta cells and promote T1D or on the other hand prevent the development of the disease. However, the opposing roles of viral infections in T1D are not understood mechanistically. We report here that viruses that do not inflict damage on beta cells provided protection from T1D by triggering immunoregulatory mechanisms. Infection of prediabetic NOD mice with Coxsackie virus B3 or lymphocytic choriomeningitis virus (LCMV) delayed diabetes onset and reduced disease incidence. Delayed T1D onset was due to transient upregulation of programmed cell death-1 ligand 1 (PD-L1) on lymphoid cells, which prevented the expansion of diabetogenic CD8+ T cells expressing programmed cell death-1 (PD-1). Reduced T1D incidence was caused by increased numbers of invigorated CD4+CD25+ Tregs, which produced TGF-beta and maintained long-term tolerance. Full protection from T1D resulted from synergy between PD-L1 and CD4+CD25+ Tregs. Our results provide what we believe to be novel mechanistic insight into the role of viruses in T1D and should be valuable for prospective studies in humans.
Collapse
|
177
|
Aouadi M, Tesz GJ, Nicoloro SM, Wang M, Chouinard M, Soto E, Ostroff GR, Czech MP. Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature 2009; 458:1180-4. [PMID: 19407801 PMCID: PMC2879154 DOI: 10.1038/nature07774] [Citation(s) in RCA: 453] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 01/06/2009] [Indexed: 12/14/2022]
Abstract
Gene silencing by double-stranded RNA, denoted RNA interference, represents a new paradigm for rational drug design. However, the transformative therapeutic potential of short interfering RNA (siRNA) has been stymied by a key obstacle-safe delivery to specified target cells in vivo. Macrophages are particularly attractive targets for RNA interference therapy because they promote pathogenic inflammatory responses in diseases such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease and diabetes. Here we report the engineering of beta1,3-D-glucan-encapsulated siRNA particles (GeRPs) as efficient oral delivery vehicles that potently silence genes in mouse macrophages in vitro and in vivo. Oral gavage of mice with GeRPs containing as little as 20 microg kg(-1) siRNA directed against tumour necrosis factor alpha (Tnf-alpha) depleted its messenger RNA in macrophages recovered from the peritoneum, spleen, liver and lung, and lowered serum Tnf-alpha levels. Screening with GeRPs for inflammation genes revealed that the mitogen-activated protein kinase kinase kinase kinase 4 (Map4k4) is a previously unknown mediator of cytokine expression. Importantly, silencing Map4k4 in macrophages in vivo protected mice from lipopolysaccharide-induced lethality by inhibiting Tnf-alpha and interleukin-1beta production. This technology defines a new strategy for oral delivery of siRNA to attenuate inflammatory responses in human disease.
Collapse
Affiliation(s)
- Myriam Aouadi
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | | | |
Collapse
|
178
|
Minor role of bystander tolerance to fetal calf serum in a peptide-specific dendritic cell vaccine model against autoimmunity: comparison with serum-free cultures. J Immunother 2009; 31:656-64. [PMID: 18600179 DOI: 10.1097/cji.0b013e31818283ef] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) are currently considered as promising tools for vaccination against tumors and also autoimmune responses. A major point of concern has been the use of fetal calf serum (FCS) as a source of heterologous antigen in DC cultures. FCS peptides can be presented by the DCs and cause T-cell responses in the recipient. We investigated the role of FCS in an autoimmune model where DC injections can prevent peptide-specifically from experimental autoimmune encephalomyelitis (EAE). We show that murine bone marrow-derived DCs generated in FCS-containing or serum-free media resulting in a similar phenotype, maturation potential, and functions. Peptide-specific protection could be achieved similarly with FCS-DC or serum-free DCs. Although FCS-DC induced strong CD4 T cell proliferation and cytokine production against FCS, these T cells lack antigenic recall during EAE. Even if FCS was reinjected, the effect on EAE resulted only in a 3-day delay of disease onset. Together, our data show that presentation of bystander antigens by peptide-specific DC vaccinations may have little influence on T-cell responses in vivo if the bystander antigen cannot be recalled by specific T cells.
Collapse
|
179
|
Valle A, Jofra T, Stabilini A, Atkinson M, Roncarolo MG, Battaglia M. Rapamycin prevents and breaks the anti-CD3-induced tolerance in NOD mice. Diabetes 2009; 58:875-81. [PMID: 19151201 PMCID: PMC2661605 DOI: 10.2337/db08-1432] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Accepted: 01/05/2009] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Non-Fc-binding anti-CD3-specific antibodies represent a promising therapy for preserving C-peptide production in subjects with recent-onset type 1 diabetes. However, the mechanisms by which anti-CD3 exerts its beneficial effect are still poorly understood, and it is questionable whether this therapeutic approach will prove durable with regard to its ability to impart metabolic preservation without additional actions designed to maintain immunological tolerance. We used the NOD mouse model to test whether rapamycin, a compound well-known for its immunomodulatory activity in mice and humans, could increase the therapeutic effectiveness of anti-CD3 treatment in type 1 diabetes. RESEARCH DESIGN AND METHODS Rapamycin was administered to diabetic NOD mice simultaneously with anti-CD3 or to NOD mice cured by anti-CD3 therapy. The ability of this combined therapy to revert type 1 diabetes and maintain a state of long-term tolerance was monitored and compared with that of anti-CD3 therapy alone. RESULTS Rapamycin inhibited the ability of anti-CD3 to revert disease without affecting the frequency/phenotype of T-cells. Rapamycin also reinstated diabetes in mice whose disease was previously reversed by anti-CD3. Withdrawal of rapamycin in these latter animals promptly restored a normoglycemic state. CONCLUSIONS Our findings indicate that, when combined with anti-CD3, rapamycin exerts a detrimental effect on the disease outcome in NOD mice for as long as it is administered. These results suggest strong caution with regard to combining these treatments in type 1 diabetic patients.
Collapse
Affiliation(s)
- Andrea Valle
- San Raffaele Diabetes Research Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| | - Tatiana Jofra
- San Raffaele Diabetes Research Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| | - Angela Stabilini
- San Raffaele Diabetes Research Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| | - Mark Atkinson
- Department of Pathology, University of Florida, Gainesville, Florida
| | - Maria-Grazia Roncarolo
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
- Università Vita-Salute San Raffaele, Milan Italy
| | - Manuela Battaglia
- San Raffaele Diabetes Research Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| |
Collapse
|
180
|
Perone MJ, Bertera S, Shufesky WJ, Divito SJ, Montecalvo A, Mathers AR, Larregina AT, Pang M, Seth N, Wucherpfennig KW, Trucco M, Baum LG, Morelli AE. Suppression of autoimmune diabetes by soluble galectin-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:2641-53. [PMID: 19234158 PMCID: PMC2929001 DOI: 10.4049/jimmunol.0800839] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease that targets the beta-cells of the pancreas. We investigated the ability of soluble galectin-1 (gal-1), an endogenous lectin that promotes T cell apoptosis, to down-regulate the T cell response that destroys the pancreatic beta-cells. We demonstrated that in nonobese diabetic (NOD) mice, gal-1 therapy reduces significantly the amount of Th1 cells, augments the number of T cells secreting IL-4 or IL-10 specific for islet cell Ag, and causes peripheral deletion of beta-cell-reactive T cells. Administration of gal-1 prevented the onset of hyperglycemia in NOD mice at early and subclinical stages of T1D. Preventive gal-1 therapy shifted the composition of the insulitis into an infiltrate that did not invade the islets and that contained a significantly reduced number of Th1 cells and a higher percentage of CD4(+) T cells with content of IL-4, IL-5, or IL-10. The beneficial effects of gal-1 correlated with the ability of the lectin to trigger apoptosis of the T cell subsets that cause beta-cell damage while sparing naive T cells, Th2 lymphocytes, and regulatory T cells in NOD mice. Importantly, gal-1 reversed beta-cell autoimmunity and hyperglycemia in NOD mice with ongoing T1D. Because gal-1 therapy did not cause major side effects or beta-cell toxicity in NOD mice, the use of gal-1 to control beta-cell autoimmunity represents a novel alternative for treatment of subclinical or ongoing T1D.
Collapse
Affiliation(s)
- Marcelo J. Perone
- T.E. Starzl Transplantation Institute and Dept. of Surgery, University of Pittsburgh Medical Center
| | | | - William J. Shufesky
- T.E. Starzl Transplantation Institute and Dept. of Surgery, University of Pittsburgh Medical Center
| | - Sherrie J. Divito
- T.E. Starzl Transplantation Institute and Dept. of Surgery, University of Pittsburgh Medical Center
| | - Angela Montecalvo
- T.E. Starzl Transplantation Institute and Dept. of Surgery, University of Pittsburgh Medical Center
| | - Alicia R. Mathers
- Dermatology, University of Pittsburgh Medical Center
- Immunology, University of Pittsburgh Medical Center
| | - Adriana T. Larregina
- Dermatology, University of Pittsburgh Medical Center
- Immunology, University of Pittsburgh Medical Center
| | - Mabel Pang
- Dept. of Pathology and Jonsson Comprehensive Cancer Center, UCLA School of Medicine, Los Angeles, CA
| | - Nilufer Seth
- Dept. of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, and Dept. of Pathology and Neurology, Harvard Medical School, Boston, MA
| | - Kai W. Wucherpfennig
- Dept. of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, and Dept. of Pathology and Neurology, Harvard Medical School, Boston, MA
| | - Massimo Trucco
- Division of Immunogenetics, Dept. of Pediatrics, Rangos Research Center, University of Pittsburgh. Pittsburgh, PA
| | - Linda G. Baum
- Dept. of Pathology and Jonsson Comprehensive Cancer Center, UCLA School of Medicine, Los Angeles, CA
| | - Adrian E. Morelli
- T.E. Starzl Transplantation Institute and Dept. of Surgery, University of Pittsburgh Medical Center
- Immunology, University of Pittsburgh Medical Center
| |
Collapse
|
181
|
Abstract
Type 1 diabetes is an immune-mediated disease in which pancreatic insulin-producing beta cells are damaged and destroyed. Animal models have served a prominent function in the development of the present ideas of pathogenesis and approaches to therapy. This commentary addresses the utility and limitations of these models for facilitating the 'translation' of immunology research into clinical applications.
Collapse
Affiliation(s)
- Matthias von Herrath
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA.
| | | |
Collapse
|
182
|
Berggreen E, Nyløkken K, Delaleu N, Hajdaragic-Ibricevic H, Jonsson MV. Impaired vascular responses to parasympathetic nerve stimulation and muscarinic receptor activation in the submandibular gland in nonobese diabetic mice. Arthritis Res Ther 2009; 11:R18. [PMID: 19200376 PMCID: PMC2688250 DOI: 10.1186/ar2609] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 01/22/2009] [Accepted: 02/06/2009] [Indexed: 12/02/2022] Open
Abstract
Introduction Decreased vascular responses to salivary gland stimulation are observed in Sjögren's syndrome patients. We investigate whether impaired vascular responses to parasympathetic stimulation and muscarinic receptor activation in salivary glands parallels hyposalivation in an experimental model for Sjögren's syndrome. Methods Blood flow responses in the salivary glands were measured by laser Doppler flowmeter. Muscarinic receptor activation was followed by saliva secretion measurements. Nitric oxide synthesis-mediated blood flow responses were studied after administration of a nitric oxide synthase inhibitor. Glandular autonomic nerves and muscarinic 3 receptor distributions were also investigated. Results Maximal blood flow responses to parasympathetic stimulation and muscarinic receptor activation were significantly lower in nonobese diabetic (NOD) mice compared with BALB/c mice, coinciding with impaired saliva secretion in nonobese diabetic mice (P < 0.005). Nitric oxide synthase inhibitor had less effect on blood flow responses after parasympathetic nerve stimulation in nonobese diabetic mice compared with BALB/c mice (P < 0.02). In nonobese diabetic mice, salivary gland parasympathetic nerve fibres were absent in areas of focal infiltrates. Muscarinic 3 receptor might be localized in the blood vessel walls of salivary glands. Conclusions Impaired vasodilatation in response to parasympathetic nerve stimulation and muscarinic receptor activation may contribute to hyposalivation observed in nonobese diabetic mice. Reduced nitric oxide signalling after parasympathetic nerve stimulation may contribute in part to the impaired blood flow responses. The possibility of muscarinic 3 receptor in the vasculature supports the notion that muscarinic 3 receptor autoantibodies present in nonobese diabetic mice might impair the fluid transport required for salivation. Parasympathetic nerves were absent in areas of focal infiltrates, whereas a normal distribution was found within glandular epithelium. Trial registration The trial registration number for the present study is 79-04/BBB, given by the Norwegian State Commission for Laboratory Animals.
Collapse
Affiliation(s)
- Ellen Berggreen
- Department of Biomedicine, Jonas Liesvei 91, Bergen 5009, Norway.
| | | | | | | | | |
Collapse
|
183
|
Van Belle TL, Taylor P, von Herrath MG. Mouse Models for Type 1 Diabetes. DRUG DISCOVERY TODAY. DISEASE MODELS 2009; 6:41-45. [PMID: 20407588 PMCID: PMC2855847 DOI: 10.1016/j.ddmod.2009.03.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Our understanding of the genetics, aetiology and pathogenesis of Type 1 Diabetes (T1D) was propelled by the discovery of animal models of T1D in the late 1970s and early 1980s, particularly the non-obese diabetic (NOD) mouse. Since then, transgenic and gene-targeting technologies allowed the generation of many models with reduced genetic and pathogenic complexity. These models allowed researchers to zoom in on specific aspects of this complex disease. In this review, we provide an overview of currently available mouse models for T1D.
Collapse
Affiliation(s)
- T L Van Belle
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
184
|
Affiliation(s)
- Clive Wasserfall
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | | |
Collapse
|
185
|
Experimental tuberculosis: the role of comparative pathology in the discovery of improved tuberculosis treatment strategies. Tuberculosis (Edinb) 2008; 88 Suppl 1:S35-47. [PMID: 18762152 DOI: 10.1016/s1472-9792(08)70035-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of laboratory animals is critical to the discovery and in vivo pre-clinical testing of new drugs and drug combinations for use in humans. M. tuberculosis infection of mice, rats, guinea pigs, rabbits and non-human primates are the most commonly used animal models of human tuberculosis. While granulomatous inflammation characterizes the most fundamental host response to M. tuberculosis aerosol infection in humans and animals, there are important species differences in pulmonary and extra-pulmonary lesion morphology which may influence responses to drug therapy. Lesions that progress to necrosis or cavitation are common, unfavorable host responses in naturally occurring tuberculosis of humans, but are not seen consistently in experimental infections in most animal model species. The importance of these unique lesion morphologies is that they represent irreversible tissue damage that can harbor persistent bacilli which are difficult to treat with standard therapies. Understanding the differences in host response to experimental tuberculosis infections may aid in selecting the most appropriate animal models to test drugs that have been rationally designed to have specific mechanisms of action in vivo. A better understanding of lesion pathogenesis across species may also aid in the identification of novel therapeutic targets or strategies that can be used alone or in combination with more conventional tuberculosis treatments in humans.
Collapse
|
186
|
Skyler JS, Greenbaum CJ, Lachin JM, Leschek E, Rafkin-Mervis L, Savage P, Spain L, Type 1 Diabetes TrialNet Study Group. Type 1 Diabetes TrialNet--an international collaborative clinical trials network. Ann N Y Acad Sci 2008; 1150:14-24. [PMID: 19120262 PMCID: PMC2918900 DOI: 10.1196/annals.1447.054] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Type 1 Diabetes TrialNet is an international consortium of clinical research centers aimed at the prevention or delay of type 1 diabetes (T1D). The fundamental goal of TrialNet is to counter the T1D disease process by immune modulation and/or enhancement of beta cell proliferation and regeneration. To achieve this goal, TrialNet researchers are working to better understand the natural history of the disease, to identify persons at risk, and to clinically evaluate novel therapies that balance potential risks and benefits. The particular focus is on studies of preventive measures. In addition, TrialNet evaluates therapies in individuals with newly diagnosed T1D with preserved beta cell function to help determine the risk/benefit profile and gain an initial assessment of potential efficacy in preservation of beta cell function, so that promising agents can be studied in prevention trials. In addition, TrialNet evaluates methodologies that enhance the conduct of its clinical trials, which includes tests of outcome assessment methodology, the evaluation of surrogate markers, and mechanistic studies laying the foundation for future clinical trials.
Collapse
Affiliation(s)
- Jay S Skyler
- Diabetes Research Institute, Miami, Florida 33101-6960, USA.
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Thrower SL, James L, Hall W, Green KM, Arif S, Allen JS, Van-Krinks C, Lozanoska-Ochser B, Marquesini L, Brown S, Wong FS, Dayan CM, Peakman M. Proinsulin peptide immunotherapy in type 1 diabetes: report of a first-in-man Phase I safety study. Clin Exp Immunol 2008; 155:156-65. [PMID: 19040615 DOI: 10.1111/j.1365-2249.2008.03814.x] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Immunotherapeutic strategies under consideration for type 1 diabetes include modification of the autoimmune response through antigen-specific routes. Administration of short peptides representing T cell epitopes targeted by patients with the disease represents one approach. This study evaluated safety and mechanistic outcomes during first-in-man intradermal administration of a human leucocyte antigen-DR4 (HLA-DR4)-restricted peptide epitope of proinsulin (C19-A3). This randomized, open-label study assessed two major theoretical risks of peptide immunotherapy, namely induction of allergic hypersensitivity and exacerbation of the proinflammatory autoimmune response, using clinical assessment and mechanistic assays in vitro. Patients with long-standing type 1 diabetes and HLA-DRB1*0401 genotype received 30 microg (n = 18) or 300 microg (n = 18) of peptide in three equal doses at 0, 1 and 2 months or no intervention (n = 12). Proinsulin peptide immunotherapy in the dosing regimen used is well tolerated and free from risk of systemic hypersensitivity and induction/reactivation of proinsulin-specific, proinflammatory T cells. Peptide-specific T cells secreting the immune suppressive cytokine interleukin (IL)-10 were observed at month 3 in four of 18 patients in the low-dose group (versus one of 12 in the control group; P = not significant). Mean IL-10 response to peptide in the low-dose group increased between 0 and 3 months (P = 0.05 after stimulation with 5 microM peptide in vitro) and then declined to baseline levels between 3 and 6 months (P = 0.01 at 10 microM peptide in vitro). These studies pave the way for future investigations in new-onset patients designed to examine whether proinsulin peptide immunotherapy has beneficial effects on markers of T cell autoimmunity and preservation of beta cell mass.
Collapse
Affiliation(s)
- S L Thrower
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Tyrosine kinase inhibitors reverse type 1 diabetes in nonobese diabetic mice. Proc Natl Acad Sci U S A 2008; 105:18895-900. [PMID: 19015530 DOI: 10.1073/pnas.0810246105] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The recent development of small-molecule tyrosine kinase (TK) inhibitors offers increasing opportunities for the treatment of autoimmune diseases. In this study, we investigated the potential of this new class of drugs to treat and cure type 1 diabetes (T1D) in the NOD mouse. Treatment of prediabetic and new onset diabetic mice with imatinib (Gleevec) prevented and reversed T1D. Similar results were observed with sunitinib (Sutent), an additional approved multikinase inhibitor, suggesting that the primary target of imatinib, c-Abl, was not essential in blocking disease in this model. Additional studies with another TK inhibitor, PLX647 (targeting c-Kit and c-Fms) or an anti-c-Kit mAb showed only marginal efficacy whereas a soluble form of platelet-derived growth factor receptor (PDGFR), PDGFRbetaIg, rapidly reversed diabetes. These findings strongly suggest that inhibition of PDGFR is critical to reverse diabetes and highlight a crucial role of inflammation in the development of T1D. These conclusions were supported by the finding that the adaptive immune system was not significantly affected by imatinib treatment. Finally, and most significantly, imatinib treatment led to durable remission after discontinuation of therapy at 10 weeks in a majority of mice. Thus, long-term efficacy and tolerance is likely to depend on inhibiting a combination of tyrosine kinases supporting the use of selective kinase inhibitors as a new, potentially very attractive approach for the treatment of T1D.
Collapse
|
189
|
Calderon B, Suri A, Pan XO, Mills JC, Unanue ER. IFN-gamma-dependent regulatory circuits in immune inflammation highlighted in diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:6964-74. [PMID: 18981116 PMCID: PMC2653256 DOI: 10.4049/jimmunol.181.10.6964] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We demonstrate diverse roles of IFN-gamma in the induction and regulation of immune-mediated inflammation using a transfer model of autoimmune diabetes. The diabetogenic CD4(+)BDC2.5 (BDC) T cell clone upon transfer into NOD.scid mice induced destruction of islets of Langerhans leading to diabetes. Administration of a neutralizing Ab to IFN-gamma (H22) resulted in long-term protection (LTP) from diabetes, with inflammation but persistence of a significant, albeit decreased, number of beta cells. BDC T cells were a mixture of cells expressing high, intermediate, and low levels of the TCR. Clonotype(low) BDC T cells were required for LTP. Furthermore, islet-infiltrating leukocytes in the LTP mice contained Foxp3(+)CD4 T cells. Islet inflammation in both diabetic and LTP mice was characterized by heavy infiltration of macrophages. Gene expression profiles indicated that macrophages in diabetic mice were M1 type, while LTP mice contained M2 differentiated. The LTP was abolished if mice were treated with either Ab-depleting CD4 T cells or a neutralizing Ab to CTLA-4, in this case, only at a late stage. Neutralization of IL-10, TGF-beta, glucocorticoid-induced TNF receptor (GITR), or CD25 had no effect. Transfer of only clonotype(high)-expressing BDC T cells induced diabetes; in contrast, H22 Abs did not inhibit diabetes. While clonotype(high) T cells induced diabetes even when IFN-gamma was neutralized, paradoxically there was reduced inflammation and no diabetes if host myeloid cells lacked IFN-gamma receptor. Hence, using monoclonal CD4 T cells, IFN-gamma can have a wide diversity of roles, depending on the setting of the immune process.
Collapse
Affiliation(s)
- Boris Calderon
- Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, MO 63110 USA
| | - Anish Suri
- Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, MO 63110 USA
| | - Xiaoou O. Pan
- Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, MO 63110 USA
| | - Jason C. Mills
- Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, MO 63110 USA
| | - Emil R. Unanue
- Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, MO 63110 USA
| |
Collapse
|
190
|
Affiliation(s)
- Li-Jun Yang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
191
|
Curative and beta cell regenerative effects of alpha1-antitrypsin treatment in autoimmune diabetic NOD mice. Proc Natl Acad Sci U S A 2008; 105:16242-7. [PMID: 18852471 DOI: 10.1073/pnas.0808031105] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Invasive insulitis is a destructive T cell-dependent autoimmune process directed against insulin-producing beta cells that is central to the pathogenesis of type 1 diabetes mellitus (T1DM) in humans and the clinically relevant nonobese diabetic (NOD) mouse model. Few therapies have succeeded in restoring long-term, drug-free euglycemia and immune tolerance to beta cells in overtly diabetic NOD mice, and none have demonstrably enabled enlargement of the functional beta cell mass. Recent studies have emphasized the impact of inflammatory cytokines on the commitment of antigen-activated T cells to various effector or regulatory T cell phenotypes and insulin resistance and defective insulin signaling. Hence, we tested the hypothesis that inflammatory mechanisms trigger insulitis, insulin resistance, faulty insulin signaling, and the loss of immune tolerance to islets. We demonstrate that treatment with alpha1-antitrypsin (AAT), an agent that dampens inflammation, does not directly inhibit T cell activation, ablates invasive insulitis, and restores euglycemia, immune tolerance to beta cells, normal insulin signaling, and insulin responsiveness in NOD mice with recent-onset T1DM through favorable changes in the inflammation milieu. Indeed, the functional mass of beta cells expands in AAT-treated diabetic NOD mice.
Collapse
|
192
|
Wong FS, Dayan CM. Regulatory T cells in autoimmune endocrine diseases. Trends Endocrinol Metab 2008; 19:292-9. [PMID: 18786834 DOI: 10.1016/j.tem.2008.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 07/10/2008] [Accepted: 07/10/2008] [Indexed: 11/15/2022]
Abstract
Regulatory T cells (Tregs) play a vital role in maintaining homeostatic balance within the immune system. Tregs are required for mediating normal immune responses against pathogens and then terminating such responses when no longer required. However, Tregs also provide important regulation of autoimmune responses, specifically in preventing autoimmunity. They are defined by expression of surface markers and production of specific cytokines that dictate their mode of action. In this review we discuss the recent advances in the understanding of the biology of Tregs, with a focus on their role(s) in human endocrine disease. In addition, recent discoveries of how hormones influence the development and function of Tregs are highlighted.
Collapse
Affiliation(s)
- F Susan Wong
- Department of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | | |
Collapse
|
193
|
Delaleu N, Madureira AC, Immervoll H, Jonsson R. Inhibition of experimental Sjögren's syndrome through immunization with HSP60 and its peptide amino acids 437-460. ACTA ACUST UNITED AC 2008; 58:2318-28. [PMID: 18668586 DOI: 10.1002/art.23656] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To investigate a potential immunomodulatory effect of the 60-kd heat-shock protein (Hsp60) on experimental spontaneous Sjögren's syndrome (SS). METHODS Seven-week-old nonobese diabetic (NOD) mice were immunized with eukaryotic Hsp60 or an Hsp60-derived peptide (amino acid residue [aa] 437-460). At 21 weeks of age, nondiabetic mice were investigated for salivary gland inflammation, exocrine function, and extraglandular disease manifestations. In addition, biomarker profiles comprising 87 analytes in serum and 75 in saliva were analyzed. RESULTS In mice immunized with Hsp60 and aa 437-460, SS-related histopathologic features were significantly reduced compared with NOD controls. In addition, 50% of Hsp60-injected mice and 33% of aa 437-460-injected mice retained normal exocrine function. Both treatments induced similar changes in biomarker profiles. Notably, levels of circulating interferon-gamma-inducible 10-kd protein (IP-10) and eotaxin were decreased significantly after treatment. Anti-type 3 muscarinic acetylcholine receptor (anti-M3R) IgG1, interleukin-10, and leptin discriminated best between the different treatment groups. Successful prevention of hyposalivation was accompanied by quantitative alterations in 36 biomarkers, of which 19 mediators of inflammation declined to levels comparable with those found in BALB/c mice. Low secreted vascular endothelial growth factor A was the most accurate predictor of successful prevention of hyposalivation. Low salivary granulocyte chemotactic protein 2 was identified as the best predictor of normal secretory function across the strains. CONCLUSION Immunization with Hsp60 and its peptide aa 437-460 led to inhibition of SS in NOD mice. Comprehensive analyses revealed specific biomarker signatures capable of predicting treatment group and treatment outcome. Molecules involved in inflammatory chemotaxis, neovascularization, and regulatory pathways caused the differences displayed by the biomarker profiles.
Collapse
|
194
|
Lu Y, Parker M, Pileggi A, Zhang B, Choi YK, Molano RD, Wasserfall C, Ricordi C, Inverardi L, Brantly M, Schatz D, Atkinson M, Song S. Human alpha 1-antitrypsin therapy induces fatal anaphylaxis in non-obese diabetic mice. Clin Exp Immunol 2008; 154:15-21. [PMID: 18759852 DOI: 10.1111/j.1365-2249.2008.03721.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Previous studies have shown that human alpha-1 antitrypsin (hAAT) gene delivery prevents type 1 diabetes (T1D) in non-obese diabetic (NOD) mice. Furthermore, hAAT protein administration prolongs acceptance of islet allografts. Therefore, we evaluated the use of purified hAAT protein therapy to prevent T1D in NOD mice. Female NOD, non-obese resistant (NOR), Balb/c and C57BL/6 mice were injected intraperitoneally with vehicle alone or vehicle containing hAAT, human albumin or mouse albumin (or mg/injection/mouse; 2x/week). Preparations of clinical-grade hAAT included API(R), Aralast, Prolastin and Zemaira. Surprisingly, hAAT administration was associated with a high rate of fatal anaphylaxis. In studies seeking T1D prevention at 4 weeks of age, 100% mice died after six injections of hAAT. When administrated at 8-10 weeks of age, most (80-100%) NOD mice died following the fourth injection of hAAT, while 0% of Balb/c and C57BL/6 mice and 10% of NOR mice died. Interestingly, repeated injections of human albumin, but not mouse albumin, also induced sudden death in NOD mice. Antibodies to hAAT were induced 2-3 weeks after hAAT administration and death was prevented by treatment with anti-platelet-activating factor along with anti-histamine. In studies of disease reversal in NOD mice, using the four pharmaceutical grade formulations of hAAT, anaphylactic deaths were observed with all hAAT preparations. The propensity for fatal anaphylaxis following antigenic administration appears to be NOD- but not hAAT-specific. The susceptibility of NOD mice to hypersensitivity provides a significant limitation for testing of hAAT. Development of strategies to avoid this unwanted response is required to use this promising therapeutic agent for T1D.
Collapse
Affiliation(s)
- Y Lu
- Department of Pharmaceutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Xiu Y, Wong CP, Bouaziz JD, Hamaguchi Y, Wang Y, Pop SM, Tisch RM, Tedder TF. B lymphocyte depletion by CD20 monoclonal antibody prevents diabetes in nonobese diabetic mice despite isotype-specific differences in Fc gamma R effector functions. THE JOURNAL OF IMMUNOLOGY 2008; 180:2863-75. [PMID: 18292508 DOI: 10.4049/jimmunol.180.5.2863] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NOD mice deficient for B lymphocytes from birth fail to develop autoimmune or type 1 diabetes. To assess whether B cell depletion influences type 1 diabetes in mice with an intact immune system, NOD female mice representing early and late preclinical stages of disease were treated with mouse anti-mouse CD20 mAbs. Short-term CD20 mAb treatment in 5-wk-old NOD female mice reduced B cell numbers by approximately 95%, decreased subsequent insulitis, and prevented diabetes in >60% of littermates. In addition, CD20 mAb treatment of 15-wk-old NOD female mice significantly delayed, but did not prevent, diabetes onset. Protection from diabetes did not result from altered T cell numbers or subset distributions, or regulatory/suppressor T cell generation. Rather, impaired CD4+ and CD8+ T cell activation in the lymph nodes of B cell-depleted NOD mice may delay diabetes onset. B cell depletion was achieved despite reduced sensitivity of NOD mice to CD20 mAbs compared with C57BL/6 mice. Decreased B cell depletion resulted from deficient FcgammaRI binding of IgG2a/c CD20 mAbs and 60% reduced spleen monocyte numbers, which in combination reduced Ab-dependent cellular cytotoxicity. With high-dose CD20 mAb treatment (250 microg) in NOD mice, FcgammaRIII and FcgammaRIV compensated for inadequate FcgammaRI function and mediated B cell depletion. Thereby, NOD mice provide a model for human FcgammaR polymorphisms that reduce therapeutic mAb efficacy in vivo. Moreover, this study defines a new, clinically relevant approach whereby B cell depletion early in the course of disease development may prevent diabetes or delay progression of disease.
Collapse
Affiliation(s)
- Yan Xiu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
196
|
Roisin-Bouffay C, Castellano R, Valéro R, Chasson L, Galland F, Naquet P. Mouse vanin-1 is cytoprotective for islet beta cells and regulates the development of type 1 diabetes. Diabetologia 2008; 51:1192-201. [PMID: 18463844 DOI: 10.1007/s00125-008-1017-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Accepted: 03/30/2008] [Indexed: 10/22/2022]
Abstract
AIMS/HYPOTHESIS Islet cell death is a key initiating and perpetuating event in type 1 diabetes and involves both immune-mediated and endogenous mechanisms. The epithelial pantetheinase vanin-1 is proinflammatory and cytoprotective via cysteamine release in some tissues. We investigated the impact of a vanin-1 deficiency on islet death and type 1 diabetes incidence. METHODS Vanin-1-deficient mice were produced and tested in drug-induced and autoimmune diabetes models. The contribution of vanin-1 to islet survival versus immune responses was evaluated using lymphocyte transfer and islet culture experiments. RESULTS The vanin-1/cysteamine pathway contributes to the protection of islet beta cells from streptozotocin-induced death in vitro and in vivo. Furthermore, vanin-1-deficient NOD mice showed a significant aggravation of diabetes, which depended upon loss of vanin-1 expression by host tissues. This increased islet fragility was accompanied by greater CD4+ insulitis without impairment of regulatory cells. Addition of cystamine, the product of pantetheinase activity, protected islets in vitro and compensated for vanin-1 deficiency in vivo. CONCLUSIONS/INTERPRETATION This study unravels a major cytoprotective role of cysteamine for islet cells and suggests that modulation of pantetheinase activity may offer alternative strategies to maintain islet cell homeostasis.
Collapse
MESH Headings
- Amidohydrolases
- Animals
- Cell Adhesion Molecules/deficiency
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Death/physiology
- Cells, Cultured
- Cystamine/pharmacology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/prevention & control
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Disease Models, Animal
- Enzyme Inhibitors/pharmacology
- Female
- GPI-Linked Proteins
- Homeostasis/physiology
- Incidence
- Insulin/metabolism
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Kaplan-Meier Estimate
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, Mutant Strains
- T-Lymphocytes, Regulatory/pathology
- Th1 Cells/pathology
Collapse
Affiliation(s)
- C Roisin-Bouffay
- Aix Marseille Université, Faculté des Sciences de Luminy, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | | | | | | | | | | |
Collapse
|
197
|
AAV8-mediated gene transfer of interleukin-4 to endogenous beta-cells prevents the onset of diabetes in NOD mice. Mol Ther 2008; 16:1409-16. [PMID: 18560422 DOI: 10.1038/mt.2008.116] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We have demonstrated the ability to deliver and express genes specifically in beta-cells for at least 6 months, using a murine insulin promoter (mIP) in a double-stranded, self-complementary AAV vector (dsAAV8-mIP). In this study, we evaluated the effects of dsAAV8-mIP-mediated delivery of interleukin 4 (mIL-4) to endogenous beta-cells in nonobese diabetic (NOD) mice. In 4-week-old NOD mice, the extent of gene transfer and expression in endogenous beta-cells after ip delivery of dsAAV8-mIP-enhanced green fluorescent protein (eGFP) was comparable to normal BALB/C mice. Further, after IP delivery of dsAAV8-mIP-IL4, expression of mIL-4 was detected in islets isolated from the treated mice and cultured. AAV8-mIP-mediated gene expression of mIL-4 in endogenous beta- cells of 4- and 8-week-old NOD mice prevented the onset of hyperglycemia in NOD mice and reduced the severity of insulitis. Moreover, expression of mIL-4 also maintained the level of CD4(+)CD25(+)FoxP3(+) cells, and adoptive transfer of splenocytes from nondiabetic dsAAV8-mIP-IL-4 mice to NODscid mice was able to block the diabetes induced by splenocytes co-adoptively transferred from nondiabetic dsAAV-mIP-eGFP mice. Taken together, these results demonstrate that local expression of mIL-4 in islets prevents islet destruction and blocks autoimmunity, partly through regulation of T-cell function.
Collapse
|
198
|
Sakata M, Yasuda H, Moriyama H, Yamada K, Kotani R, Kurohara M, Okumachi Y, Kishi M, Arai T, Hara K, Hamada H, Yokono K, Nagata M. Prevention of recurrent but not spontaneous autoimmune diabetes by transplanted NOD islets adenovirally transduced with immunomodulating molecules. Diabetes Res Clin Pract 2008; 80:352-9. [PMID: 18400329 DOI: 10.1016/j.diabres.2008.01.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Accepted: 01/21/2008] [Indexed: 11/19/2022]
Abstract
Pancreatic islet transplantation has the potential to maintain normoglycemia in patients with established type 1 diabetes, thereby obviating the need for frequent insulin injections. Our previous study showed that recombinant IL-12p40-producing islets prevented the recurrence of NOD diabetes. First, to see which immunomodulating molecule-secreting islet grafts can most powerfully prevent diabetes development in NOD mice without immunosuppressant, NOD islets were transfected with one of the following adenoviral vectors: Ad.IL-12p40, Ad.TGF-beta, Ad.CTLA4-Ig, or Ad.TNF-alpha after which they were transplanted under the renal capsule of acutely diabetic NOD mice. The immunomodulating molecules produced by these adenovirus-transfected islets in vitro were 74+/-19ng, 50+/-4ng, 821+/-31ng, and 77+/-18ng/100 islets, respectively. Transplantation of IL-12p40, TNF-alpha, and CTLA4-Ig but not TGF-beta-secreting islets displayed enhanced survival and delayed diabetes recurrence in recent-onset diabetic recipients. IL-12p40-producing islet grafts most powerfully prevented recurrent diabetes in NOD mice. In addition, local production of TNF-alpha and CTLA4-Ig significantly prolonged islet graft survival. In second series of experiment, these manipulated islets were transplanted under the renal capsule of 10-week-old NOD recipients and were also transplanted subcutaneously into 2-week-old NOD recipients. Transplantation of these islets into 2- or 10-week-old pre-diabetic mice failed to protect them from developing diabetes; in fact, transplantation of Ad.TNF-alpha-transfected islets into 2-week-old mice actually accelerated diabetes onset. Taken together, this approach was ineffectual as a prophylactic protocol. In conclusion, this study showed comparisons of the immunomodulating effects of 4 different adenoviral vectors in the same transplantation model and local production of IL-12p40, TNF-alpha and CTLA4-Ig significantly prevented recurrent diabetes in NOD mice.
Collapse
Affiliation(s)
- Muneaki Sakata
- Department of Internal and Geriatric Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Affiliation(s)
- Rebecca J Brown
- Clinical Endocrinology Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
200
|
Creusot RJ, Yaghoubi SS, Kodama K, Dang DN, Dang VH, Breckpot K, Thielemans K, Gambhir SS, Fathman CG. Tissue-targeted therapy of autoimmune diabetes using dendritic cells transduced to express IL-4 in NOD mice. Clin Immunol 2008; 127:176-87. [PMID: 18337172 PMCID: PMC2453076 DOI: 10.1016/j.clim.2007.12.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Accepted: 12/26/2007] [Indexed: 12/11/2022]
Abstract
A deficit in IL-4 production has been previously reported in both diabetic human patients and non-obese diabetic (NOD) mice. In addition, re-introducing IL-4 into NOD mice systemically, or as a transgene, led to a beneficial outcome in most studies. Here, we show that prediabetic, 12-week old female NOD mice have a deficit in IL-4 expression in the pancreatic lymph nodes (PLN) compared to age-matched diabetes-resistant NOD.B10 mice. By bioluminescence imaging, we demonstrated that the PLN was preferentially targeted by bone marrow-derived dendritic cells (DCs) following intravenous (IV) administration. Following IV injection of DCs transduced to express IL-4 (DC/IL-4) into 12-week old NOD mice, it was possible to significantly delay or prevent the onset of hyperglycemia. We then focused on the PLN to monitor, by microarray analysis, changes in gene expression induced by DC/IL-4 and observed a rapid normalization of the expression of many genes, that were otherwise under-expressed compared to NOD.B10 PLN. The protective effect of DC/IL-4 required both MHC and IL-4 expression by the DCs. Thus, adoptive cellular therapy, using DCs modified to express IL-4, offers an effective, tissue-targeted cellular therapy to prevent diabetes in NOD mice at an advanced stage of pre-diabetes, and may offer a safe approach to consider for treatment of high risk human pre-diabetic patients.
Collapse
Affiliation(s)
- Rémi J Creusot
- Department of Medicine, Division of Immunology and Rheumatology, Stanford, CA 94305-5166, USA
| | | | | | | | | | | | | | | | | |
Collapse
|