101
|
Masuda H, Atsumi T, Fujisaku A, Shimizu C, Yoshioka N, Koike T. Acute onset of type 1 diabetes accompanied by acute hepatitis C: the potential role of proinflammatory cytokine in the pathogenesis of autoimmune diabetes. Diabetes Res Clin Pract 2007; 75:357-61. [PMID: 16968656 DOI: 10.1016/j.diabres.2006.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Revised: 06/26/2006] [Accepted: 07/12/2006] [Indexed: 01/28/2023]
Abstract
A 22-year-old Japanese woman presented with general fatigue. Five days later, she demonstrated a body temperature of 39 degrees C and a loss in weight of 5kg. She thereafter became unconscious and was taken to Tomakomai City General Hospital. Urinary ketone body was positive, and plasma glucose was 1063mg/dl. The serum asparate aminotransferase and alanine aminotransferase levels were 158 and 1220IU/l, respectively. An arterial blood gas analysis showed metabolic acidosis. Glycated hemoglobin was 10.9%. Urinary C-peptide immnoreactivity was 11microg/day. Anti-glutamic acid decarboxylase antibody was 12.4U/ml. In general, islet-associated autoantibodies are detectable several years before the development of overt autoimmune diabetes, thus suggesting that an autoimmune reaction against beta-cells had already started in this case. On viral examinations, hepatitis C virus (HCV) antibody was negative, while HCV-RNA was positive. Based on these findings, she was diagnosed to have autoimmune diabetes and acute hepatitis C. In addition, her serum interleukin-18 level was elevated to 506pg/ml. The duration of diabetic characteristic symptoms before diagnosis is usually several weeks in most cases of autoimmune diabetes. However, it was extremely short in this case. Taken together, these findings suggested that the progression of autoimmune diabetes might have been accelerated due to the infection of HCV.
Collapse
Affiliation(s)
- Hajime Masuda
- Department of Internal Medicine, Tomakomai City General Hospital, 1-2-21 Honko-cho, Tomakomai 053-8567, Japan.
| | | | | | | | | | | |
Collapse
|
102
|
Uno S, Imagawa A, Okita K, Sayama K, Moriwaki M, Iwahashi H, Yamagata K, Tamura S, Matsuzawa Y, Hanafusa T, Miyagawa J, Shimomura I. Macrophages and dendritic cells infiltrating islets with or without beta cells produce tumour necrosis factor-alpha in patients with recent-onset type 1 diabetes. Diabetologia 2007; 50:596-601. [PMID: 17221211 DOI: 10.1007/s00125-006-0569-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Accepted: 11/17/2006] [Indexed: 10/23/2022]
Abstract
AIMS/HYPOTHESIS Type 1A diabetes results from autoimmune destruction of pancreatic beta cells. We examined the involvement of TNF-alpha and IL-1beta, as well as of T cells, macrophages and dendritic cells, in the destruction of beta cells in patients with recent-onset type 1 diabetes. MATERIALS AND METHODS We obtained pancreatic biopsy specimens from six patients with recent-onset type 1 diabetes and analysed these by immunohistochemistry. RESULTS T cell infiltration was less common in islets without beta cells (12.5 [0-33.3]%) than in those with beta cells (46.0 [17.4-83.3]%), while macrophages and dendritic cells showed a similar extent of infiltration into islets both with or without beta cells. TNF-alpha was detected in 25.0 (4.3-46.9)% of macrophages and 11.8 (0-40.0)% of dendritic cells infiltrating the islets in samples from each patient, but not at all in T cells. IL-1beta was detected in 1.8 (0-11.3)% of T cells infiltrating the islets with beta cells, while it was found in 19.2 (0-35.3)% of macrophages or 10.7 (0-31.3)% of dendritic cells infiltrating the islets in samples from each patient (all values median [range]). CONCLUSIONS/INTERPRETATION Macrophages and dendritic cells infiltrate the islets and produce inflammatory cytokines (TNF-alpha and IL-1beta) during the development of type 1A diabetes.
Collapse
Affiliation(s)
- S Uno
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, and Department of Internal Medicine, Mino City Hospital, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Chamberlain G, Wållberg M, Rainbow D, Hunter K, Wicker LS, Green EA. A 20-Mb region of chromosome 4 controls TNF-alpha-mediated CD8+ T cell aggression toward beta cells in type 1 diabetes. THE JOURNAL OF IMMUNOLOGY 2007; 177:5105-14. [PMID: 17015694 DOI: 10.4049/jimmunol.177.8.5105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Identification of candidate genes and their immunological mechanisms that control autoaggressive T cells in inflamed environments, may lead to novel therapies for autoimmune diseases, like type 1 diabetes (T1D). In this study, we used transgenic NOD mice that constitutively express TNF-alpha in their islets from neonatal life (TNF-alpha-NOD) to identify protective alleles that control T1D in the presence of a proinflammatory environment. We show that TNF-alpha-mediated breakdown in T cell tolerance requires recessive NOD alleles. To identify some of these recessive alleles, we crossed TNF-alpha-NOD mice to diabetes-resistant congenic NOD mice having protective alleles at insulin-dependent diabetes (Idd) loci that control spontaneous T1D at either the preinsulitis (Idd3.Idd5) or postinsulitis (Idd9) phases. No protection from TNF-alpha-accelerated T1D was afforded by resistance alleles at Idd3.Idd5. Lack of protection was not at the level of T cell priming, the efficacy of islet-infiltrating APCs to present islet peptides, nor the ability of high levels of CD4+ Foxp3+ T cells to accumulate in the islets. In contrast, protective alleles at Idd9 significantly increased the age at which TNF-alpha-NOD mice developed T1D. Disease delay was associated with a decreased ability of CD8+ T cells to respond to islet Ags presented by islet-infiltrating APCs. Finally, we demonstrate that the protective region on chromosome 4 that controls T1D in TNF-alpha-Idd9 mice is restricted to the Idd9.1 region. These data provide new evidence of the mechanisms by which selective genetic loci control autoimmune diseases in the presence of a strong inflammatory assault.
Collapse
Affiliation(s)
- Giselle Chamberlain
- Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | | | | | | | | |
Collapse
|
104
|
Tse HM, Milton MJ, Schreiner S, Profozich JL, Trucco M, Piganelli JD. Disruption of Innate-Mediated Proinflammatory Cytokine and Reactive Oxygen Species Third Signal Leads to Antigen-Specific Hyporesponsiveness. THE JOURNAL OF IMMUNOLOGY 2007; 178:908-17. [PMID: 17202352 DOI: 10.4049/jimmunol.178.2.908] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Successful Ag activation of naive T helper cells requires at least two signals consisting of TCR and CD28 on the T cell interacting with MHC II and CD80/CD86, respectively, on APCs. Recent evidence demonstrates that a third signal consisting of proinflammatory cytokines and reactive oxygen species (ROS) produced by the innate immune response is important in arming the adaptive immune response. In an effort to curtail the generation of an Ag-specific T cell response, we targeted the synthesis of innate immune response signals to generate Ag-specific hyporesponsiveness. We have reported that modulation of redox balance with a catalytic antioxidant effectively inhibited the generation of third signal components from the innate immune response (TNF-alpha, IL-1beta, ROS). In this study, we demonstrate that innate immune-derived signals are necessary for adaptive immune effector function and disruption of these signals with in vivo CA treatment conferred Ag-specific hyporesponsiveness in BALB/c, NOD, DO11.10, and BDC-2.5 mice after immunization. Modulating redox balance led to decreased Ag-specific T cell proliferation and IFN-gamma synthesis by diminishing ROS production in the APC, which affected TNF-alpha levels produced by CD4(+) T cells and impairing effector function. These results demonstrate that altering redox status can be effective in T cell-mediated diseases such as autoimmune diabetes to generate Ag-specific immunosuppression because it inhibits the third signal necessary for CD4(+) T cells to transition from expansion to effector function.
Collapse
Affiliation(s)
- Hubert M Tse
- Diabetes Institute, Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
105
|
Chen K, Wei Y, Sharp GC, Braley-Mullen H. Decreasing TNF-alpha results in less fibrosis and earlier resolution of granulomatous experimental autoimmune thyroiditis. J Leukoc Biol 2006; 81:306-14. [PMID: 17046971 PMCID: PMC1748426 DOI: 10.1189/jlb.0606402] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Granulomatous experimental autoimmune thyroiditis (G-EAT) is induced in DBA/1 mice by adoptive transfer of mouse thyroglobulin (MTg)-primed spleen cells. TNF-alpha is an important proinflammatory cytokine and apoptotic molecule involved in many autoimmune diseases. To study its role in G-EAT, anti-TNF-alpha mAb was given to recipient mice. Disease severity was comparable between mice with or without anti-TNF-alpha treatment at days 19-21, the time of maximal severity of G-EAT, suggesting TNF-alpha is not essential for development of thyroid inflammation. However, thyroid lesions resolved at day 48 in anti-TNF-alpha-treated mice, while thyroids of rat Ig-treated controls had fibrosis. These results suggested that reducing TNF-alpha contributed to resolution of inflammation and inhibited fibrosis. Gene and protein expression of inflammatory molecules was examined by RT-PCR and immunostaining, and apoptosis was detected using TUNEL staining and an apoptosis kit. Thyroids of anti-TNF-alpha-treated controls had reduced proinflammatory and profibrotic molecules, e.g., IFN-gamma, IL-1beta, IL-17, inducible NOS and MCP-1, at day 19 compared with thyroids of rat Ig-treated mice. There were more apoptotic thyrocytes in rat Ig-treated controls than in anti-TNF-alpha-treated mice. The site of expression of the anti-apoptotic molecule FLIP also differed between rat Ig-treated and anti-TNF-alpha-treated mice. FLIP was predominantly expressed by inflammatory cells of rat Ig-treated mice and by thyrocytes of anti-TNF-alpha-treated mice. These results suggest that anti-TNF-alpha may regulate expression of proinflammatory cytokines and apoptosis in thyroids, resulting in less inflammation, earlier resolution, and reduced fibrosis.
Collapse
Affiliation(s)
| | | | | | - Helen Braley-Mullen
- Departments of Internal Medicine
- Molecular Microbiology & Immunology, University of Missouri School of Medicine, and
- VA Research Service, Columbia, MO 65212
- Corresponding Author: Helen Braley-Mullen, or Kemin Chen, Division of Immunology & Rheumatology, Dept. of Medicine, University of Missouri, M306 Medical Sciences, One Hospital Dr., Columbia, MO 65212. Tel: 573-882-4325, Fax: 573-882-1380. , or
| |
Collapse
|
106
|
Moore DJ, Markmann JF, Deng S. Avenues for immunomodulation and graft protection by gene therapy in transplantation. Transpl Int 2006; 19:435-45. [PMID: 16771864 DOI: 10.1111/j.1432-2277.2006.00314.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Organ transplantation represents the only definitive therapy for many causes of end-organ failure. However, the universal success of this therapy is limited by chronic allograft rejection, the side effects of chronic immunosuppressive therapy, and a severe shortage of donor organs. Presently, the success of solid-organ transplantation depends on the continuous administration of toxic and nonspecific immunosuppressive agents, therapies that present risks for opportunistic infection, malignancy, and a variety of agent-specific side effects. To promote the use of transplantation with limited risk of long-term sequelae, three dominant research challenges emerge: (i) elimination of the need for exogenous immunosuppression by immunological tolerance induction; (ii) prevention of chronic rejection/graft dysfunction; and (iii) expansion of available organs for transplantation. Gene therapy may provide significant advances and solutions in each of these areas. Rejection of the graft in the immediate post-transplant period has been attacked through the transfer of immunomodulatory molecules in addition to tolerance inducing approaches. Chronic graft rejection may be similarly addressed through permanent tolerance induction or alternatively through the introduction of molecules to resist chronic graft damage. Genetic manipulation of stem cells may ultimately produce transgenic animals to serve as tissue donors to overcome the limited donor organ supply. This review will highlight ongoing developments in the translation of gene therapy approaches to the challenges inherent in transplantation.
Collapse
Affiliation(s)
- Daniel J Moore
- Department of Pediatrics, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
107
|
Narang AS, Mahato RI. Biological and biomaterial approaches for improved islet transplantation. Pharmacol Rev 2006; 58:194-243. [PMID: 16714486 DOI: 10.1124/pr.58.2.6] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Islet transplantation may be used to treat type I diabetes. Despite tremendous progress in islet isolation, culture, and preservation, the clinical use of this modality of treatment is limited due to post-transplantation challenges to the islets such as the failure to revascularize and immune destruction of the islet graft. In addition, the need for lifelong strong immunosuppressing agents restricts the use of this option to a limited subset of patients, which is further restricted by the unmet need for large numbers of islets. Inadequate islet supply issues are being addressed by regeneration therapy and xenotransplantation. Various strategies are being tried to prevent beta-cell death, including immunoisolation using semipermeable biocompatible polymeric capsules and induction of immune tolerance. Genetic modification of islets promises to complement all these strategies toward the success of islet transplantation. Furthermore, synergistic application of more than one strategy is required for improving the success of islet transplantation. This review will critically address various insights developed in each individual strategy and for multipronged approaches, which will be helpful in achieving better outcomes.
Collapse
Affiliation(s)
- Ajit S Narang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 26 S. Dunlap St., Feurt Building, Room 413, Memphis, TN 38163, USA
| | | |
Collapse
|
108
|
Fleischer A, Ghadiri A, Dessauge F, Duhamel M, Rebollo MP, Alvarez-Franco F, Rebollo A. Modulating apoptosis as a target for effective therapy. Mol Immunol 2006; 43:1065-79. [PMID: 16099509 DOI: 10.1016/j.molimm.2005.07.013] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2005] [Indexed: 10/25/2022]
Abstract
Alterations in cell proliferation and cell death are essential determinants in the pathogenesis and progression of several diseases such as cancer, neurodegenerative disorders or autoimmune diseases among others. Complex networks of regulatory factors determine whether cells proliferate or die. Recent progress in understanding the molecular changes offer the possibility of specifically targeting molecules and pathways to achieve more effective and rational therapies. Drugs that target molecules involved in apoptosis are used as treatment against several diseases. Candidates such as TNF death receptor family, caspase inhibitors, antagonists of the p53-MDM2 interaction, NF-kappaB and PI3K pathways and Bcl-2 family members have been targeted as cancer cell killing agents. Moreover, apoptosis of tumor cells can also be achieved by targeting the inhibitor of apoptosis proteins, IAPs, in addition to the classical antiproliferative approach. Disruption of STAT activation and interferon beta therapy have been used as a treatment to prevent the progression of some autoimmune diseases. In models of Parkinson's, Alzheimer's and amyotrophic lateral sclerosis, blocking of Par-4 expression or function, as well as caspase activation, prevents neuronal cell death. Finally, it has been shown that gene therapy may be an encouraging approach for treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Aarne Fleischer
- Laboratoire d'Immunologie Cellulaire et Tissulaire, U543 INSERM, Hôpital Pitié Salpêtrière, Bâtiment CERVI, 83 Bd de 1'Hôpital, 75013 Paris, France
| | | | | | | | | | | | | |
Collapse
|
109
|
Tarbell KV, Yamazaki S, Steinman RM. The interactions of dendritic cells with antigen-specific, regulatory T cells that suppress autoimmunity. Semin Immunol 2006; 18:93-102. [PMID: 16469503 DOI: 10.1016/j.smim.2006.01.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Dendritic cells (DCs) are important for several aspects of the development and function of CD4(+) CD25(+) regulatory T cells (Tregs), which are critical for maintaining peripheral tolerance and preventing autoimmunity. In cultures from human thymus, dendritic cells (DCs) conditioned with thymic stromal lymphopoietin (TSLP) mediate the production of Tregs from CD4(+) CD25(-) thymocytes. In cultures from mouse lymphoid organs, CD86-rich DCs induce the proliferation and improved suppressive function of antigen-specific Tregs. DC-expanded, antigen-specific Tregs show greatly enhanced efficacy relative to polyclonal populations in blocking experimental autoimmunity. In several animal models including NOD diabetes, Tregs directed to one autoantigen are able to block autoimmunity induced by multiple antigens from the target organ. Distinct states of DC differentiation or maturation are likely to be important for the emerging roles of DCs in the biology of Tregs, particularly the control of autoimmunity in an antigen-dependent manner.
Collapse
Affiliation(s)
- Kristin V Tarbell
- Laboratory of Cellular Physiology and Immunology and the Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, Box 176, New York, NY 10021-6399, USA.
| | | | | |
Collapse
|
110
|
Long M, Slaiby AM, Hagymasi AT, Mihalyo MA, Lichtler AC, Reiner SL, Adler AJ. T-bet down-modulation in tolerized Th1 effector CD4 cells confers a TCR-distal signaling defect that selectively impairs IFN-gamma expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 176:1036-45. [PMID: 16393991 PMCID: PMC2846362 DOI: 10.4049/jimmunol.176.2.1036] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
When Th1 effector CD4 cells encounter tolerizing Ag in vivo, their capacity to express the effector cytokines IFN-gamma and TNF-alpha is lost more rapidly than noneffector functions such as IL-2 production and proliferation. To localize the relevant intracellular signaling defects, cytokine expression was compared following restimulation with Ag vs agents that bypass TCR-proximal signaling. IFN-gamma and TNF-alpha expression were both partially rescued when TCR-proximal signaling was bypassed, indicating that both TCR-proximal and -distal signaling defects impair the expression of these two effector cytokines. In contrast, bypassing TCR-proximal signaling fully rescued IL-2 expression. T-bet, a transcription and chromatin remodeling factor that is required to direct the differentiation of naive CD4 cells into IFN-gamma-expressing Th1 effectors, was partially down-modulated in tolerized Th1 effectors. Enforcing T-bet expression during tolerization selectively rescued the ability to express IFN-gamma, but not TNF-alpha. Conversely, expression of a dominant-negative T-bet in Th1 effectors selectively impaired the ability to express IFN-gamma, but not TNF-alpha. Analysis of histone acetylation at the IFN-gamma promoter further suggested that down-modulation of T-bet expression during Th1 effector CD4 cell tolerization does not impair IFN-gamma expression potential through alterations in chromatin structure.
Collapse
Affiliation(s)
- Meixiao Long
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Aaron M. Slaiby
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Adam T. Hagymasi
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Marianne A. Mihalyo
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Alexander C. Lichtler
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, CT 06030
| | - Steven L. Reiner
- Abramson Family Cancer Research Institute and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Adam J. Adler
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| |
Collapse
|
111
|
Lee LF, Xu B, Michie SA, Beilhack GF, Warganich T, Turley S, McDevitt HO. The role of TNF-alpha in the pathogenesis of type 1 diabetes in the nonobese diabetic mouse: analysis of dendritic cell maturation. Proc Natl Acad Sci U S A 2005; 102:15995-6000. [PMID: 16247001 PMCID: PMC1276103 DOI: 10.1073/pnas.0508122102] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
TNF-alpha has been linked to the development of type 1 diabetes (T1D). We previously reported that neonatal treatment of nonobese diabetic (NOD) mice with TNF-alpha accelerated the onset of T1D, whereas TNF-alpha blockade in the same time period resulted in a complete absence of diabetes. The mechanisms by which TNF-alpha modulates development of T1D in NOD mice remain unclear. Here we tested the effects of TNF-alpha on the maturation of dendritic cells (DCs) in the NOD mouse. We found that neonatal treatment with TNF-alpha caused an increase in expression of maturation markers on CD11c(+)CD11b(+) DC subpopulations, whereas treatment with anti-TNF-alpha resulted in a decrease in expression of maturation markers in the CD11c(+)CD11b(+) subset. Moreover, neonatal treatment with TNF-alpha resulted in skewed development of a CD8alpha(+)CD11b(-)CD11c(+) DC subset such that TNF-alpha decreases the CD8alpha(+)CD11c(+) DC subset, increases the CD11c(+)CD11b(+) subset, and causes an increase in the expression of CD40 and CD54 on mature DCs capable of inducing immunity. Anti-TNF-alpha-treated mice had an increase in the CD8alpha(+)CD11c(+) DCs. Notably, adoptively transferred naïve CD4(+) T cells from BDC2.5 T cell receptor transgenic mice proliferated in the pancreatic lymph nodes in TNF-alpha-treated NOD mice but not in anti-TNF-alpha-treated mice. Finally, we show that anti-TNF-alpha-treated mice showed immunological tolerance to islet cell proteins. We conclude that TNF-alpha plays an important role in the initiation of T1D in the NOD mouse by regulating the maturation of DCs and, thus, the activation of islet-specific pancreatic lymph node T cells.
Collapse
Affiliation(s)
- Li-Fen Lee
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
112
|
Shoda LKM, Young DL, Ramanujan S, Whiting CC, Atkinson MA, Bluestone JA, Eisenbarth GS, Mathis D, Rossini AA, Campbell SE, Kahn R, Kreuwel HTC. A comprehensive review of interventions in the NOD mouse and implications for translation. Immunity 2005; 23:115-26. [PMID: 16111631 DOI: 10.1016/j.immuni.2005.08.002] [Citation(s) in RCA: 231] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Type 1 diabetes (T1D) animal models such as the nonobese diabetic (NOD) mouse have improved our understanding of disease pathophysiology, but many candidate therapeutics identified therein have failed to prevent/cure human disease. We have performed a comprehensive evaluation of disease-modifying agents tested in the NOD mouse based on treatment timing, duration, study length, and efficacy. Interestingly, some popular tenets regarding NOD interventions were not confirmed: all treatments do not prevent disease, treatment dose and timing strongly influence efficacy, and several therapies have successfully treated overtly diabetic mice. The analysis provides a unique perspective on NOD interventions and suggests that the response of this model to therapeutic interventions can be a useful predictor of the human response as long as careful consideration is given to treatment dose, timing, and protocols; more thorough investigation of these parameters should improve clinical translation.
Collapse
|
113
|
Filippi C, von Herrath M. How viral infections affect the autoimmune process leading to type 1 diabetes. Cell Immunol 2005; 233:125-32. [PMID: 15963965 DOI: 10.1016/j.cellimm.2005.04.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2005] [Accepted: 04/21/2005] [Indexed: 12/31/2022]
Abstract
Despite a large body of evidence describing associations between viruses and the development of type 1 diabetes (T1D) in genetically prone individuals, clearly defining causative infectious agents has not been successful. A likely explanation is that the link between infections and autoimmunity is more multifaceted than we initially assumed. Viral footprints might be hard to detect systemically or in the target organ once autoimmunity has been initiated, and several infections might have to act in concert to precipitate clinical autoimmunity. Furthermore, cells cross-reactive between viral and self-antigens might express low avidity T cell receptors and only be present transiently in the blood of affected individuals. In addition, there are two new observations from animal models that we should take into account at this point: first, viral infections alone might not be able to induce disease in the absence of other inflammatory factors (supporting the "fertile field hypothesis" [M.G. von Herrath et al., Microorganisms and autoimmunity: making the barren field fertile? Nat. Rev. Microbiol. 1 (2003) 151-157, ]). Second, increasing evidence indicates that viruses can play a role in preventing rather than enhancing T1D development (supporting the "hygiene hypothesis" [J.F. Bach, Protective role of infections and vaccinations on autoimmune diseases. J. Autoimmun. 16 (2001) 347-353]). In this article we will present an overview of the early events and requirements that could account for T1D predisposition and development, and explain how these can be modulated by viral infections. Focusing on coxsackie B and lymphocytic choriomeningitis virus infections, we will discuss new data that can hopefully help us understand how virus-induced inflammation can positively or negatively affect the clinical outcome of islet-autoimmunity and T1D.
Collapse
Affiliation(s)
- Christophe Filippi
- La Jolla Institute for Allergy and Immunology, 10355 Science Center Drive, San Diego, CA 92121, USA.
| | | |
Collapse
|
114
|
Oikawa Y, Yamato E, Tashiro F, Yamamoto M, Uozumi N, Shimada A, Shimizu T, Miyazaki J. Protective role for cytosolic phospholipase A2alpha in autoimmune diabetes of mice. FEBS Lett 2005; 579:3975-8. [PMID: 15996660 DOI: 10.1016/j.febslet.2005.06.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2005] [Revised: 06/10/2005] [Accepted: 06/11/2005] [Indexed: 11/17/2022]
Abstract
Cytosolic phospholipase A(2)alpha (cPLA(2)alpha) plays an important role in arachidonate pathway. To investigate the contribution of cPLA(2)alpha to autoimmune diabetes, we established non-obese diabetic (NOD) mouse, an excellent model for human type 1 diabetes, deficient in cPLA(2)alpha. These mice showed severe insulitis and a higher incidence of diabetes. In their macrophages, decreased prostaglandin E(2) (PGE(2)) induced by cPLA(2)alpha deficiency, and the increase in production of tumor necrosis factor (TNF)-alpha were observed. These results suggested that cPLA(2)alpha plays a protective role in progression of insulitis and development of autoimmune diabetes by suppression of TNF-alpha production from macrophages.
Collapse
Affiliation(s)
- Y Oikawa
- Division of Stem Cell Regulation Research, G6, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
115
|
Abstract
Evidence exists for an essential role of beta-cell apoptosis in the pathology of type 1 and type 2 diabetes. Current methods for diabetes-associated apoptosis detection, however, suffer the drawbacks of relying on in situ-based strategies. In this study, we attempted to measure, both in vitro and ex vivo, levels of beta-cell apoptosis in diabetic mice using Cy5.5-labeled annexin V. We used streptozotocin-treated BALB/c mice and NOD mice of different ages as models of type 1 diabetes and db/db mice as a model of type 2 diabetes. With annexin V Cy5.5, we established differences in levels of apoptosis between diabetic and control animals. Intravenously administered annexin V Cy5.5 accumulated in pancreata of diabetic mice but not in nondiabetic controls. Furthermore, its localization was specific to apoptotic events within diabetic islets; its selectivity was supported by transferase-mediated dUTP nick-end labeling staining. Because annexin V defines an early marker of apoptosis and the developed probe is suitable for in vivo administration, it may provide a promising tool for real-time identification in intact animals of the earliest stages of diabetes-associated beta-cell death and for tracing the events that characterize the pathology of the disease.
Collapse
Affiliation(s)
- Zdravka Medarova
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | |
Collapse
|
116
|
Jaeschke A, Rincón M, Doran B, Reilly J, Neuberg D, Greiner DL, Shultz LD, Rossini AA, Flavell RA, Davis RJ. Disruption of the Jnk2 (Mapk9) gene reduces destructive insulitis and diabetes in a mouse model of type I diabetes. Proc Natl Acad Sci U S A 2005; 102:6931-5. [PMID: 15867147 PMCID: PMC1100789 DOI: 10.1073/pnas.0502143102] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The c-Jun NH(2)-terminal kinase isoform (JNK) 1 is implicated in type 2 diabetes. However, a potential role for the JNK2 protein kinase in diabetes has not been established. Here, we demonstrate that JNK2 may play an important role in type 1 (insulin-dependent) diabetes that is caused by autoimmune destruction of beta cells. Studies of nonobese diabetic mice demonstrated that disruption of the Mapk9 gene (which encodes the JNK2 protein kinase) decreased destructive insulitis and reduced disease progression to diabetes. CD4(+) T cells from JNK2-deficient nonobese diabetic mice produced less IFN-gamma but significantly increased amounts of IL-4 and IL-5, indicating polarization toward the Th2 phenotype. This role of JNK2 to control the Th1/Th2 balance of the immune response represents a mechanism of protection against autoimmune diabetes. We conclude that JNK protein kinases may have important roles in diabetes, including functions of JNK1 in type 2 diabetes and JNK2 in type 1 diabetes.
Collapse
Affiliation(s)
- Anja Jaeschke
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Abstract
The significance of type I interferons (IFN-alpha/beta) in biology and medicine renders research on their activities continuously relevant to our understanding of normal and abnormal (auto) immune responses. This relevance is bolstered by discoveries that unambiguously establish IFN-alpha/beta, among the multitude of cytokines, as dominant in defining qualitative and quantitative characteristics of innate and adaptive immune processes. Recent advances elucidating the biology of these key cytokines include better definition of their complex signaling pathways, determination of their importance in modifying the effects of other cytokines, the role of Toll-like receptors in their induction, their major cellular producers, and their broad and diverse impact on both cellular and humoral immune responses. Consequently, the role of IFN-alpha/beta in the pathogenesis of autoimmunity remains at the forefront of scientific inquiry and has begun to illuminate the mechanisms by which these molecules promote or inhibit systemic and organ-specific autoimmune diseases.
Collapse
|
118
|
Roep BO, Atkinson M, von Herrath M. Satisfaction (not) guaranteed: re-evaluating the use of animal models of type 1 diabetes. Nat Rev Immunol 2005; 4:989-97. [PMID: 15573133 DOI: 10.1038/nri1502] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Without a doubt, rodent models have been instrumental in describing pathways that lead to pancreatic beta-cell destruction, evaluating potential causes of type 1 diabetes and providing proof-of-principle for the potential of immune-based interventions. However, despite more than two decades of productive research, we are still yet to define an initiating autoantigen for the human disease, to determine the precise mechanisms of beta-cell destruction in humans and to design interventions that prevent or cure type 1 diabetes. In this Perspective article, we propose that a major philosophical change would benefit this field, a proposition that is based on evaluation of situations in which rodent models have provided useful guidance and in which they have led to disappointments.
Collapse
Affiliation(s)
- Bart O Roep
- Bart O. Roep is at the Leiden University Medical Center, Department of Immunohematology and Blood Transfusion, Leiden NL-2300 RC, The Netherlands.
| | | | | |
Collapse
|
119
|
Adler AJ. Peripheral Tolerization of Effector and Memory T Cells: Implications for Autoimmunity and Tumor-Immunity. ACTA ACUST UNITED AC 2005; 1:21-28. [PMID: 20411047 DOI: 10.2174/1573395052952879] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Due to the random generation of T cell antigen receptors, a large fraction of developing T cells have the potential to recognize self-determinants. To prevent this self-reactive T cell repertoire from mediating autoimmunity, the immune system utilizes several mechanisms to induce tolerance to self. The majority of self-reactive T cells undergo negative selection (i.e., apoptosis) during development if their antigen receptors have high affinity for MHC-self-peptide complexes present in the thymus. Nonetheless, some T cells recognize self-epitopes that are not present in the thymus, and will thus reach maturation and migrate to peripheral lymphoid organs were they can be subject to a number of peripheral tolerance mechanisms such as deletion, inactivation (i.e., anergy) or suppression. While peripheral tolerization of naive (i.e., antigen-inexperienced) T cells has been studied extensively, there are potential situations in which self-reactive T cells might first encounter immunogenic forms of antigen (deriving from pathogens or vaccines) and thus be programmed to develop effector and memory functions. This article will review recent studies that have explored the potential of effector and memory T cells to undergo peripheral tolerization, as well as potential implications of these findings for autoimmunity and tumor-immunity.
Collapse
Affiliation(s)
- Adam J Adler
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| |
Collapse
|
120
|
Lee MS, Chang I, Kim S. Death effectors of beta-cell apoptosis in type 1 diabetes. Mol Genet Metab 2004; 83:82-92. [PMID: 15464423 DOI: 10.1016/j.ymgme.2004.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2004] [Revised: 07/31/2004] [Accepted: 08/03/2004] [Indexed: 01/15/2023]
Abstract
While it is generally agreed that apoptosis of pancreatic beta-cells is the most important and final step in the progression of type 1 diabetes without which clinical diabetes does not develop, it has not been elucidated which molecule(s) are the real culprit(s) in type 1 diabetes. Perforin, FasL, TNFalpha, IL-1, IFNgamma, and NO have been claimed as the effector molecules; however, they, as a single agent, might explain only part of beta-cell death in type 1 diabetes. While FasL was initially considered as a strong candidate for the most important death effector, following experiments cast doubt on such a hypothesis. Combinations or synergism between IFNgamma and TNFalpha or IL-1beta are being revisited as the death effectors, and molecular mechanism explaining such a synergism was addressed in several recent papers. The role of NF-kappaB for pancreatic beta-cell death in type 1 diabetes is also controversial. While NF-kappaB plays anti-apoptotic roles in most other death models, its role in type 1 diabetes might be different probably due to the involvement of multiple cytokines at different stages of the disease progression and the peculiarity of pancreatic beta-cells. Recent papers also suggested a role for Ca2+ in cytokine-mediated pancreatic beta-cell death. Such participation of Ca2+ in beta-cell death appears to have a close relevance to the mitochondrial events or ER stress that constitutes an important part of cell death machinery recently identified.
Collapse
Affiliation(s)
- Myung-Shik Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong Kangnam-ku, Seoul 135-710, Republic of Korea.
| | | | | |
Collapse
|
121
|
Machen J, Bertera S, Chang Y, Bottino R, Balamurugan AN, Robbins PD, Trucco M, Giannoukakis N. Prolongation of islet allograft survival following ex vivo transduction with adenovirus encoding a soluble type 1 TNF receptor–Ig fusion decoy. Gene Ther 2004; 11:1506-14. [PMID: 15229635 DOI: 10.1038/sj.gt.3302320] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Islet transplantation is a viable long-term therapeutic alternative to daily insulin replacement for type I diabetes. The allogeneic nature of the transplants poses immunological challenges for routine clinical utility. Gene transfer of immunoregulatory molecules and those that improve insulin release kinetics provides rational approaches to facilitate allogeneic islet transplantation as a potential therapy. We have examined the efficacy of a soluble type 1 tumor necrosis factor receptor (TNFR) immunoglobulin-Fc fusion transgene (TNFR-Ig) to protect human islets from cytokine-induced apoptosis in culture, as well as in facilitating allogeneic islet transplants in diabetic mice. Cultured human islets were transduced with an adenoviral vector encoding human TNFR-Ig (Ad-TNFR-Ig). TNFR-Ig protein was secreted by cultured islets, as well as by transduced mouse islet transplants recovered from mouse recipients. Glucose-induced insulin release kinetics were comparable among untransduced, Ad-TNFR-Ig-infected human islets and vector-transduced islets exposed to cytokines. In parallel, Ad-TNFR-Ig-infected islets were protected from cytokine-induced apoptosis activation. Finally, diabetic mice transplanted with allogeneic islets expressing TNFR-Ig returned to and maintained normoglycemia significantly longer than untransduced islet recipients. These data support the potential utility of TNFR-Ig gene transfer to islets as a means of facilitating allogeneic islet transplantation.
Collapse
Affiliation(s)
- J Machen
- Diabetes Institute, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
122
|
Movahedi B, Van de Casteele M, Caluwé N, Stangé G, Breckpot K, Thielemans K, Vreugdenhil G, Mathieu C, Pipeleers D. Human pancreatic duct cells can produce tumour necrosis factor-alpha that damages neighbouring beta cells and activates dendritic cells. Diabetologia 2004; 47:998-1008. [PMID: 15184981 DOI: 10.1007/s00125-004-1426-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2003] [Accepted: 04/17/2004] [Indexed: 10/26/2022]
Abstract
AIMS/HYPOTHESIS In the human pancreas, a close topographic relationship exists between duct cells and beta cells. This explains the high proportion of duct cells in isolated human islet preparations. We investigated whether human duct cells are a source of TNFalpha-mediated interactions with beta cells and immune cells. This cytokine has been implicated in the development of autoimmune diabetes in mice. METHODS Human duct cells were isolated from donor pancreases and examined for their ability to produce TNFalpha following a stress-signalling pathway. Duct-cell-released TNFalpha was tested for its in vitro effects on survival of human beta cells and on activation of human dendritic cells. RESULTS Exposure of human pancreatic duct cells to interleukin-1beta (IL-1beta) induces TNFalpha gene expression, synthesis of the 26,000 M(r) TNFalpha precursor and conversion to the 17,000 M(r) mature form, which is rapidly released. This effect is NO-independent and involves p38 MAPK and NF-kappaB signalling. Duct-cell-released TNFalpha contributed to cytokine-induced apoptosis of isolated human beta cells. It also induced activation of human dendritic cells. CONCLUSIONS/INTERPRETATION Human pancreatic duct cells are a potential source of TNFalpha that can cause apoptosis of neighbouring beta cells and initiate an immune response through activation of dendritic cells. They may thus actively participate in inflammatory and immune processes that threaten beta cells during development of diabetes or after human islet cell grafts have been implanted.
Collapse
Affiliation(s)
- B Movahedi
- Diabetes Research Centre, Brussels Free University-VUB and JDRF Centre for Beta Cell Therapy in Europe, Laarbeeklaan 103, 1090 Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Vosters O, Beuneu C, Nagy N, Movahedi B, Aksoy E, Salmon I, Pipeleers D, Goldman M, Verhasselt V. CD40 expression on human pancreatic duct cells: role in nuclear factor-kappa B activation and production of pro-inflammatory cytokines. Diabetologia 2004; 47:660-8. [PMID: 15298343 DOI: 10.1007/s00125-004-1363-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
AIMS/HYPOTHESIS Human pancreatic duct cells are closely associated with islet beta cells, and contaminate islet suspensions transplanted in Type 1 diabetes mellitus patients. Activated duct cells produce cytotoxic mediators and possibly contribute to the pathogenesis of Type 1 diabetes mellitus or islet graft rejection. As CD40 transduces activation signals involved in inflammatory and immune disorders, we investigated CD40 expression on duct cells and their response to CD40 engagement. METHODS CD40 expression on human pancreatic duct cells was analysed by flow cytometry and immunohistochemical analyses. To assess the function of CD40 expression on duct cells, activation of the transcription factor nuclear factor-kappa B was determined using electrophoretic mobility shift assay and ELISA. Cytokine mRNA levels were quantified by real-time RT-PCR, and protein levels by Luminex technology. RESULTS Isolated human pancreatic duct cells and Capan-2 cell lines were found to express constitutively CD40. The expression of CD40 on duct cells was confirmed in vivo on human normal and pathological pancreatic specimens. CD40 ligation on Capan-2 cells induced rapid nuclear factor-kappa B activation, and supershift assays demonstrated that p50/p65 heterodimers and p50/p50 homodimers were present in the activated complexes in the nucleus. This activation was accompanied by tumour necrosis factor-a and interleukin-1beta mRNA accumulation. Tumour necrosis factor-alpha protein secretion was confirmed in CD40-activated Capan-2 cells and in isolated human pancreatic duct cells. CONCLUSIONS/INTERPRETATION Interaction between activated T lymphocytes expressing CD40 ligand and duct cells expressing CD40 may contribute to the immune responses involved in Type 1 diabetes mellitus and islet graft rejection. Interfering with CD40-mediated duct cell activation could alleviate beta cell damage of immune origin.
Collapse
Affiliation(s)
- O Vosters
- Laboratory of Experimental Immunology, Brussels Free University, Erasme Hospital, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Abstract
Tumour necrosis factor (TNF) is involved in the pathogenesis of several diseases. In mice, human TNF signals only through p55, one of two murine TNF receptors. We here report a study of growth, viability and morphological alterations in transgenic mice expressing a low constitutive and tissue-restricted level of human TNF in vivo. The transgene was expressed solely in T cells. The transgenic mice showed a marked failure to thrive and a rapid cellular depletion in spleen and thymus. Slight fibrosis was seen in most tissues investigated, in addition to immature adipose tissue and irregular lymphocytic areas. Serum levels of hTNF were only slightly increased in the transgenic mice, enough, however, to cause an inflammatory reaction. All the symptoms were abrogated by an inhibitory hTNF antibody, demonstrating the essential role of hTNF in this phenotype. Transgenic mice constitute a multidimensional system allowing observation of disease processes over time in all tissues. The effects of hTNF were seen first and foremost in the lymphoid organs of the transgenic mice, verifying their cells as major targets at low levels of hTNF expression in the T-cell compartments. Chronic, low levels of TNF expression cause profound disturbances in lymphoid tissue development resulting in cachexia and premature death.
Collapse
Affiliation(s)
- Heidi Glosli
- The Biotechnology Centre of Oslo, University of Oslo, N-0317 Oslo, Norway
| | | | | |
Collapse
|
125
|
Christen U, Juedes A, Homann D, von Herrath MG. Virally induced inflammation and therapeutic avenues in type 1 diabetes. Endocrinol Metab Clin North Am 2004; 33:45-58, viii. [PMID: 15053894 DOI: 10.1016/s0889-8529(03)00097-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Urs Christen
- Immune Regulation Laboratory, La Jolla Institute for Allergy and Immunology, 10355 Science Center Drive, San Diego, CA 92121, USA
| | | | | | | |
Collapse
|
126
|
Marshall ACJ, Toh BH, Alderuccio F. Tumor necrosis factor alpha is not implicated in the genesis of experimental autoimmune gastritis. J Autoimmun 2004; 22:1-11. [PMID: 14709408 DOI: 10.1016/j.jaut.2003.09.003] [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] [Indexed: 12/01/2022]
Abstract
Experimental autoimmune gastritis (EAG) characterised by mononuclear cell infiltrate, parietal and zymogenic cell destruction and circulating autoantibodies to gastric H(+)/K(+)ATPase is an animal model for human autoimmune gastritis, that leads to pernicious anaemia. We have previously shown that Fas has a role in initiating damage to target cells in EAG. Here we used three strategies to examine the role of TNFalpha in this disease. We administered neutralising anti-TNFalpha antibody either as a single injection or as twice weekly injections for 8 weeks to mice subjected to neonatal thymectomy-induced EAG. To address the role of apoptotic signals through TNFR1, TNFR1 deficient mice were either neonatally thymectomised or crossed to PC-GMCSF transgenic mice that spontaneously develop EAG. Neonatally thymectomised mice treated with anti-TNFalpha antibody developed destructive gastritis and autoantibodies to gastric H(+)/K(+)ATPase similar to control mice. Following either neonatal thymectomy or crossing to PC-GMCSF transgenic mice, TNFR1 deficient mice developed autoantibody-positive destructive gastritis at similar frequency compared with wild type and heterozygous littermates. Our observations that neutralisation of TNFalpha and absence of TNFR1 has no discernible effect on development of EAG suggest that TNFalpha is not required for mucosal cell damage or development of autoimmune gastritis. While blocking TNFalpha activity has therapeutic benefit in certain autoimmune diseases, this is not the case for EAG.
Collapse
Affiliation(s)
- Aiden C J Marshall
- Department of Pathology and Immunology, Central and Eastern Clinical School, Monash University, AMREP, Commercial Road, Prahran, Victoria 3181, Australia
| | | | | |
Collapse
|
127
|
Ando H, Kurita S, Takamura T. The specific p38 mitogen-activated protein kinase pathway inhibitor FR167653 keeps insulitis benign in nonobese diabetic mice. Life Sci 2004; 74:1817-27. [PMID: 14741738 DOI: 10.1016/j.lfs.2003.09.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The p38 mitogen-activated protein kinase (MAPK) pathway is important in Th1 immunity, macrophage activation, and apoptosis. Since they may be associated with beta-cell destruction during the development of type 1 diabetes, we investigated the role of the p38 MAPK pathway in female nonobese diabetic (NOD) mice. Phosphorylated p38 MAPK was observed immunohistochemically in CD4+ cells that had infiltrated into the islets and part of beta-cells, increasing in proportion to the severity of insulitis. Continuous oral administration of 0.08% FR167653, a specific p38 MAPK pathway inhibitor, significantly reduced the ex vivo production of interferon-gamma by splenic Th1 cells without affecting interleukin-4 production by Th2 cells. FR167653 administration from 4-30 weeks of age prevented NOD mice from developing diabetes without affecting the severity of insulitis. Treatment with FR167653 after insulitis had developed (i.e. from 10-30 weeks of age) also prevented diabetes, further suggesting that treatment with the p38 MAPK pathway inhibitor keeps insulitis benign in NOD mice, partly by inhibiting Th1 immunity. These findings suggest that p38 MAPK is a key mediator that switches insulitis from benign to destructive in the development of type 1 diabetes.
Collapse
Affiliation(s)
- Hitoshi Ando
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Science, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641, Japan
| | | | | |
Collapse
|
128
|
Affiliation(s)
- Michelle Solomon
- Department of Immunology, The Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
129
|
Long M, Higgins AD, Mihalyo MA, Adler AJ. Effector CD4 cell tolerization is mediated through functional inactivation and involves preferential impairment of TNF-alpha and IFN-gamma expression potentials. Cell Immunol 2003; 224:114-21. [PMID: 14609577 PMCID: PMC2846335 DOI: 10.1016/j.cellimm.2003.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
It has recently been shown that effector/memory T cells can undergo peripheral tolerization in response to self-antigen. In the present study, we found that within 24h self-antigen profoundly impairs the ability of CD4 effectors to express TNF-alpha (and to a lesser extent IFN-gamma); however, several days of self-antigen exposure is required to impair non-effector functions such as IL-2 expression and proliferation. Since only half of the initial effector CD4 cell population expresses effector cytokines following brief antigenic stimulation, tolerization might have been mediated either through functional inactivation of effector-competent cells, or alternatively by the selective deletion of competent and expansion of non-competent cells. When briefly stimulated effectors were fractionated based on their expression of IFN-gamma, the IFN-gamma(-) sub-population was able to express IFN-gamma following secondary stimulation, indicating that all effector CD4 cells are functionally competent. Furthermore, both IFN-gamma(+) and IFN-gamma(-) sub-populations underwent tolerization in response to self-HA (although the former was slightly more prone to deletion at later time points). Thus, effector CD4 cell tolerization is mediated primarily through the functional inactivation of effector-competent cells.
Collapse
Affiliation(s)
| | | | | | - Adam J. Adler
- Corresponding author. Fax: 1-860-679-1265. (A.J. Adler)
| |
Collapse
|
130
|
Yang Y, Santamaria P. Dissecting autoimmune diabetes through genetic manipulation of non-obese diabetic mice. Diabetologia 2003; 46:1447-64. [PMID: 14586501 DOI: 10.1007/s00125-003-1218-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2003] [Revised: 07/10/2003] [Indexed: 11/29/2022]
Abstract
Type 1 diabetes results from a genetically and immunologically complex autoimmune process that is specifically directed against the pancreatic beta cells. Non-obese diabetic mice spontaneously develop a form of autoimmune diabetes closely resembling the disease in humans. This happens because, like human diabetic patients, non-obese diabetic mice have an unfortunate combination of apparently normal alleles at numerous loci associated with Type 1 diabetes. In isolation, each of these allelic variants affords a small degree of susceptibility to diabetes. In combination, however, they set in motion a series of immunological events that lead to islet inflammation and overt diabetes. Type 1 diabetes is associated with defects in self-tolerance and immunoregulation. It involves presentation of beta cell antigens to autoreactive T lymphocytes by professional antigen-presenting cells, the recruitment of antigen-activated T cells into pancreatic islets, and the differentiation of these antigen-activated lymphocytes into beta cell killers. Understanding the precise sequence of events in the pathogenesis of Type 1 diabetes has been, and remains, a challenging task. Much of our understanding of the immunology of the disease stems from studies of genetically engineered, non-obese diabetic mice. These mice provide reductionist systems, with which the contribution of individual cellular elements, molecules or genes to the disease process can be dissected. This review focuses on the lessons that have been learned through studies of these mice.
Collapse
Affiliation(s)
- Y Yang
- Julia McFarlane Diabetes Research Centre, The University of Calgary, Faculty of Medicine, Calgary, Alberta, Canada
| | | |
Collapse
|
131
|
Rabinovitch A, Suarez-Pinzon WL. Role of cytokines in the pathogenesis of autoimmune diabetes mellitus. Rev Endocr Metab Disord 2003; 4:291-9. [PMID: 14501180 DOI: 10.1023/a:1025160614313] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Alex Rabinovitch
- 430 Heritage Medical Research Centre, University of Alberta, Edmonton, Alberta, Canada T6G 2S2.
| | | |
Collapse
|
132
|
Coppin H, Roth MP, Liblau RS. Cytokine and cytokine receptor genes in the susceptibility and resistance to organ-specific autoimmune diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 520:33-65. [PMID: 12613571 DOI: 10.1007/978-1-4615-0171-8_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Affiliation(s)
- Hélène Coppin
- Laboratoire d'immunologie Cellulaire INSERM CJF 97-11, Hospital Pitie-Salpetriere, Paris, France
| | | | | |
Collapse
|
133
|
Rabinovitch A. Immunoregulation by cytokines in autoimmune diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 520:159-93. [PMID: 12613578 DOI: 10.1007/978-1-4615-0171-8_10] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
134
|
Meagher C, Sharif S, Hussain S, Cameron MJ, Arreaza GA, Delovitch TL. Cytokines and chemokines in the pathogenesis of murine type 1 diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 520:133-58. [PMID: 12613577 DOI: 10.1007/978-1-4615-0171-8_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Affiliation(s)
- C Meagher
- The Robarts Research Institute and University of Western Ontario, Department of Microbiology and Immunology, and Medicine, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
135
|
Christen U, von Herrath MG. Cytokines and chemokines in virus-induced autoimmunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 520:203-20. [PMID: 12613580 DOI: 10.1007/978-1-4615-0171-8_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Urs Christen
- The Scripts Research Institute, Division of Virology, La Jolla, California, USA
| | | |
Collapse
|
136
|
Mageed RA, Isenberg DA. Tumour necrosis factor alpha in systemic lupus erythematosus and anti-DNA autoantibody production. Lupus 2003; 11:850-5. [PMID: 12529050 DOI: 10.1191/0961203302lu306rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumour necrosis factor (TNFalpha) is a cytokine with a wide range of diverse and at times paradoxical effects. These include immunoregulatory, lymphoid organogenesis and pro-inflammatory effects. In recent years, TNFalpha has become a focus of interest more for its inflammatory effects in a number of chronic autoimmune diseases. This interest culminated in the successful treatment of patients with rheumatoid arthritis, Crohn's diseases and ankylosing spondylitis with blocking antibodies or soluble TNFalpha receptors. Paradoxically, however, TNFalpha also has immunomodulatory effects in some autoimmune conditions such as lupus in some mouse models of the disease and in diabetes in the none-obese diabetic mouse. The role TNFalpha plays in human systemic lupus erythematosus is, however, controversial. In this article we review some of the studies carried out to elucidate the effects of TNFalpha in lupus disease and likely mechanisms of action. Further, we discuss recent data on the likely effects of blocking TNFalpha on anti-DNA autoantibody production.
Collapse
Affiliation(s)
- R A Mageed
- Department of Immunology and Molecular Pathology, Royal Free and University College London, The Windeyer Institute, London, UK.
| | | |
Collapse
|
137
|
Adorini L. Cytokine-based immunointervention in the treatment of autoimmune diseases. Clin Exp Immunol 2003; 132:185-92. [PMID: 12699404 PMCID: PMC1808696 DOI: 10.1046/j.1365-2249.2003.02144.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2003] [Indexed: 11/20/2022] Open
Affiliation(s)
- L Adorini
- BioXell, Via Olgettina 58, 20132 Milan, Italy.
| |
Collapse
|
138
|
Chang I, Kim S, Kim JY, Cho N, Kim YH, Kim HS, Lee MK, Kim KW, Lee MS. Nuclear factor kappaB protects pancreatic beta-cells from tumor necrosis factor-alpha-mediated apoptosis. Diabetes 2003; 52:1169-75. [PMID: 12716748 DOI: 10.2337/diabetes.52.5.1169] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recent studies incriminating tumor necrosis factor (TNF)-alpha as the final effector in pancreatic beta-cell death in type 1 diabetes underscore the potential role of TNF-alpha-dependent NF-kappaB activation as an important modulator of pancreatic beta-cell death in autoimmune diabetes. Although nuclear factor (NF)-kappaB activation has been implicated in the protection of target cells against apoptosis by a variety of death effectors, its role in pancreatic islet cell death is not clear. We studied the role of NF-kappaB activation in pancreatic islet cell death by using a gamma-interferon (IFN-gamma)/TNF-alpha synergism model we had previously reported. TNF-alpha induced inhibitor of kappaB (IkappaB) degradation and p65 translocation from cytoplasm to nuclei in MIN6N8 insulinoma cells. The NF-kappaB DNA-binding nuclear complex activated by TNF-alpha contained both the p65 and p50 subunit. IFN-gamma pretreatment did not affect TNF-alpha-induced NF-kappaB activation. Treatment with a proteasome inhibitor blocked p65 translocation and induced susceptibility to TNF-alpha in otherwise resistant insulinoma cells or primary pancreatic islet cells. Specific inhibition of NF-kappaB activation by adenoviral transduction of IkappaB "superrepressor" also sensitized insulinoma cells and primary islet beta-cells to TNF-alpha-induced apoptosis. These results suggest the protective role of NF-kappaB activation against cytokine-mediated pancreatic beta-cell death, contrary to previous reports implicating NF-kappaB as a mediator of pancreatic islet cell death.
Collapse
Affiliation(s)
- Inik Chang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-dong, Kangnam-ku, Seoul 135-710, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
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
|
140
|
Dahlén E, Dawe K, Ohlsson L, Hall H, Hedse K, Annersten K, Aström M, Gross DJ, Hedlund G. Linomide inhibits insulitis and modulates cytokine production in pancreatic islets in the nonobese diabetic mouse. Int Immunopharmacol 2003; 3:17-30. [PMID: 12538031 DOI: 10.1016/s1567-5769(02)00137-6] [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: 11/21/2022]
Abstract
Linomide is an immunomodulator which has been shown to potently inhibit autoimmunity in several animal models for human autoimmune diseases, including type I diabetes in the nonobese diabetic (NOD) mouse. In this study, we investigate the basis for Linomide's protective effects in the NOD mouse by immunohistochemical and RT-PCR analysis of the phenotype and cytokine expression by cells infiltrating the islets of Langerhans in the pancreas. Linomide treatment was found to reduce the infiltration of T cells, B cells, dendritic cells (DC) and MHC class II(+) cells into the islets, but did not reduce macrophage (MPhi) infiltration. This was seen following Linomide treatment at 3-5, 4-8 and 14-24 weeks of age and thus appears to be independent of the stage of the autoreactive process and the extent of insulitis. The reduced insulitis may be due to reduced expression of adhesion molecules since decreased numbers of islet-associated blood vessels expressing CD106 and MAdCAM-1 were detected following Linomide treatment. Furthermore, short term Linomide treatment (3 or 7 days), which did not alter the number of infiltrating cells, was found to inhibit the production of TNF-alpha which is known to induce the expression of CD106 and MAdCAM-1. These results suggest that the reduced insulitis observed in Linomide-treated animals is secondary to a functional modulation of infiltrating cells.
Collapse
Affiliation(s)
- Eva Dahlén
- Active Biotech Research AB, Box 724, SE-220 07 Lund, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Christen U, von Herrath MG. Transgenic animal models for type 1 diabetes: linking a tetracycline-inducible promoter with a virus-inducible mouse model. Transgenic Res 2002; 11:587-95. [PMID: 12509133 DOI: 10.1023/a:1021144209209] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Autoimmunity is thought to emerge as a consequence of genetic predispositions and environmental tiggering factors. Often the etiology and the mechanisms involved in the autoaggressive destruction of self-components are rather complex and in many cases poorly understood. Chemokines and cytokines are central mediators of inflammatory processes that are involved in initiation and progression of autoimmunity. Many animal models for human autoimmune diseases use transgenic technology to express chemokines and/or cytokines in an organ or tissue specific manner. However, most of these model systems express the transgene irreversibly without considering the time of expression as a very important parameter. Here, we review experiences that were made from using a tetracycline-inducible promotor system (tTA-system) to express TNFalpha at various times during an ongoing autoimmune process, such as the destruction of pancreatic beta-cells in a mouse model for human type 1 diabetes.
Collapse
Affiliation(s)
- Urs Christen
- Department of Immune Regulation, IR-3, La Jolla Institute for Allergy and Immunology, San Diego, CA, USA
| | | |
Collapse
|
142
|
Ventura-Oliveira D, Vilella CA, Zanin ME, Castro GM, Moreira Filho DC, Zollner RL. Kinetics of TNF-alpha and IFN-gamma mRNA expression in islets and spleen of NOD mice. Braz J Med Biol Res 2002; 35:1347-55. [PMID: 12426635 DOI: 10.1590/s0100-879x2002001100013] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Insulin-dependent diabetes mellitus is caused by autoimmune destruction of pancreatic beta cells. Non-obese diabetic (NOD) mice spontaneously develop diabetes similar to the human disease. Cytokines produced by islet-infiltrating mononuclear cells may be directly cytotoxic and can be involved in islet destruction coordinated by CD4+ and CD8+ cells. We utilized a semiquantitative RT-PCR assay to analyze in vitro the mRNA expression of TNF-alpha and IFN-gamma cytokine genes in isolated islets (N = 100) and spleen cells (5 x 10(5) cells) from female NOD mice during the development of diabetes and from female CBA-j mice as a related control strain that does not develop diabetes. Cytokine mRNAs were measured at 2, 4, 8, 14 and 28 weeks of age from the onset of insulitis to the development of overt diabetes. An increase in IFN-gamma expression in islets was observed for females aged 28 weeks (149 +/- 29 arbitrary units (AU), P<0.05, Student t-test) with advanced destructive insulitis when compared with CBA-j mice, while TNF-alpha was expressed in both NOD and CBA-j female islets at the same level at all ages studied. In contrast, TNF-alpha in spleen was expressed at higher levels in NOD females at 14 weeks (99 +/- 8 AU, P<0.05) and 28 weeks (144 +/- 17 AU, P<0.05) of age when compared to CBA-j mice. The data suggest that IFN-gamma and TNF-alpha expression in pancreatic islets of female NOD mice is associated with beta cell destruction and overt diabetes.
Collapse
Affiliation(s)
- D Ventura-Oliveira
- Disciplina e Laboratório de Imunologia Clínica, Departamento de Clínica Médica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | | | | | | | | | | |
Collapse
|
143
|
Wu AJ, Hua H, Munson SH, McDevitt HO. Tumor necrosis factor-alpha regulation of CD4+CD25+ T cell levels in NOD mice. Proc Natl Acad Sci U S A 2002; 99:12287-92. [PMID: 12221281 PMCID: PMC129437 DOI: 10.1073/pnas.172382999] [Citation(s) in RCA: 203] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mechanism by which tumor necrosis factor-alpha (TNF) differentially modulates type I diabetes mellitus in the nonobese diabetic (NOD) mouse is not well understood. CD4+CD25+ T cells have been implicated as mediators of self-tolerance. We show (i) NOD mice have a relative deficiency of CD4+CD25+ T cells in thymus and spleen; (ii) administration of TNF or anti-TNF to NOD mice can modulate levels of this population consistent with their observed differential age-dependent effects on diabetes in the NOD mouse; (iii) CD4+CD25+ T cells from NOD mice treated neonatally with TNF show compromised effector function in a transfer system, whereas those treated neonatally with anti-TNF show no alteration in ability to prevent diabetes; and (iv) repeated injection of CD4+CD25+ T cells into neonatal NOD mice delays diabetes onset for as long as supplementation occurred. These data suggest that alterations in the number and function of CD4+CD25+ T cells may be one mechanism by which TNF and anti-TNF modulate type I diabetes mellitus in NOD mice.
Collapse
Affiliation(s)
- Ava J Wu
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
144
|
Weiss L, Barak V, Zeira M, Abdul-Hai A, Raibstein I, Reich S, Hirschfeld E, Gross D, Slavin S. Cytokine production in Linomide-treated nod mice and the potential role of a Th (1)/Th(2) shift on autoimmune and anti-inflammatory processes. Cytokine 2002; 19:85-93. [PMID: 12182843 DOI: 10.1006/cyto.2002.1956] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Linomide prevents the development of autoimmune insulitis and insulin-deficient diabetes mellitus in female NOD mice. Linomide prevents development of autoimmune manifestations in other experimentally induced and spontaneous autoimmune diseases as well, but the mechanism of action is unknown. The present report summarizes our investigations on the effect of Linomide on different functional T cell subsets in NOD mice analyzed according to their cytokine profile. Supernatants from cultured splenocytes and peritoneal cells taken from Linomide-treated mice contained lower levels of TNFalpha, IL-1 beta, IFN gamma and IL-12 versus higher levels of IL-4, IL-6 and IL-10 in comparison with supernatants from cultures of untreated mice. Our results suggest that regulation of autoimmunity following oral Linomide administration in NOD mice induces a shift from Th(1) to Th(2) phenotype response, thereby preventing the development of diabetes by active cytokine-induced immunoregulation of T cell subsets, including downregulation of Th(1) and upregulation of Th(2).
Collapse
Affiliation(s)
- Lola Weiss
- Departments of Bone Marrow Transplantation & Cancer Immunotherapy, Hadassah University Hospital, Jerusalem, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Visser J, Groen H, Klatter F, Rozing J. Timing of pentoxifylline treatment determines its protective effect on diabetes development in the Bio Breeding rat. Eur J Pharmacol 2002; 445:133-40. [PMID: 12065204 DOI: 10.1016/s0014-2999(02)01625-4] [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: 11/17/2022]
Abstract
Diabetes-prone Bio Breeding (DP-BB) rats spontaneously develop diabetes between 60 and 120 days of age. Diabetes-resistant (DR)-BB rats can be induced to develop diabetes by poly(I:C) and anti-RT6. Here, we studied the effect of pentoxifylline, a potent anti-inflammatory agent, on diabetes development in both BB rat models of insulin-dependent diabetes mellitus and investigated whether these effects were related to differential modulation of tumour necrosis factor (TNF)-alpha and interleukin-10. When DP-BB rats received pentoxifylline from day 60 onwards, diabetes development was delayed and reduced. The other treatment protocols had no effect. In DR-BB rats, pentoxifylline treatment resulted only in a delay of diabetes development. In both BB rat models, in vivo pentoxifylline treatment potently suppressed TNF-alpha, but only moderately affected interleukin-10 production in vitro. These results show that timing of pentoxifylline treatment determines its protective effect on diabetes development in DP-BB rats. The observed pentoxifylline-induced increase of the interleukin-10/TNF-alpha ratio might be a mechanism for protection or delay of the diabetes development.
Collapse
Affiliation(s)
- Jeroen Visser
- Department of Cell Biology, Immunology Section, Faculty Medical Sciences, Groningen University, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | | | | |
Collapse
|
146
|
McDevitt H, Munson S, Ettinger R, Wu A. Multiple roles for tumor necrosis factor-alpha and lymphotoxin alpha/beta in immunity and autoimmunity. ARTHRITIS RESEARCH 2002; 4 Suppl 3:S141-52. [PMID: 12110133 PMCID: PMC3240146 DOI: 10.1186/ar570] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/29/2002] [Accepted: 03/03/2002] [Indexed: 11/10/2022]
Abstract
Tumor necrosis factor (TNF)-alpha and lymphotoxin (LT) alpha/beta play multiple roles in the development and function of the immune system. This article focuses on three important aspects of the effects of these cytokines on the immune response and on autoimmunity. In several experimental systems (Jurkat T cells, murine T-cell hybridomas), TNF-alpha appears to cause a downregulation of signaling through the TCR, revealed by changes in calcium flux, activation of p21, p23 and ZAP70, and a decrease in nuclear activation of NF-kappaB. Previous and present results suggest that TNF-alpha interferes in some manner with signaling through the TCR, at a locus yet to be delineated. Transgenic expression of LTbetaR-Fc in nonobese diabetic (NOD) transgenic mice results in prevention of type 1 diabetes in NOD mice as long as the level of expression of the fusion protein (under the control of the cytomegalovirus promoter) remains above a level of 2-3 microg/ml. Once the expression levels of the fusion protein have dropped below this critical level, the diabetic process resumes and the animals become diabetic at 40-50 weeks of age, whereas nontransgenic littermates develop diabetes by 25-30 weeks of age. The paradoxical effects of neonatal TNF-alpha administration in NOD mice in increasing incidence of and hastening onset of type 1 diabetes, while neonatal anti-TNF administration completely prevents all signs of islet cell autoimmunity, are due partly to the low levels of CD4+CD25+ T cells in NOD mice. These low levels are reduced by a further 50% on neonatal administration of nontoxic levels of TNF-alpha. In contrast, neonatal administration of anti-TNF-alpha results in a dramatic increase in the levels of CD4+CD25+ regulatory T cells, to levels beyond those seen in wild-type untreated NOD mice. TNF-alpha and LTalpha/beta thus have pleomorphic regulatory effects on the development and expression of autoimmunity.
Collapse
Affiliation(s)
- Hugh McDevitt
- Department of Microbiology and Immunology, Stanford University Medical Center, California 94305, USA.
| | | | | | | |
Collapse
|
147
|
Casares S, Hurtado A, McEvoy RC, Sarukhan A, von Boehmer H, Brumeanu TD. Down-regulation of diabetogenic CD4+ T cells by a soluble dimeric peptide-MHC class II chimera. Nat Immunol 2002; 3:383-91. [PMID: 11862219 DOI: 10.1038/ni770] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Type 1 diabetes is an organ-specific autoimmune disease that is mediated by autoreactive T cells. We show here that administration of a soluble dimeric peptide-major histocompatibility complex (pMHC) class II chimera (DEF) to prediabetic double-transgenic mice prevents the onset of disease or, in animals that are already diabetic, restores normoglycemia. The antidiabetogenic effects of DEF rely on the induction of anergy in splenic autoreactive CD4+ T cells via alteration of early T cell receptor signaling and stimulation of interleukin 10-secreting T regulatory type 1 cells in the pancreas. Soluble dimeric pMHC class II may be useful in the development of immunospecific therapies for type 1 diabetes.
Collapse
Affiliation(s)
- Sofia Casares
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | | | |
Collapse
|
148
|
Mabley JG, Pacher P, Southan GJ, Salzman AL, Szabó C. Nicotine reduces the incidence of type I diabetes in mice. J Pharmacol Exp Ther 2002; 300:876-881. [PMID: 11861793 DOI: 10.1124/jpet.300.3.876] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Nicotine has been previously shown to have immunosuppressive actions. Type I diabetes is an autoimmune disease resulting from the specific destruction of the insulin-producing pancreatic beta-cells. Thus, we hypothesized that nicotine may exert protective effects against type I diabetes. The multiple low-dose streptozotocin (MLDS)-induced model and spontaneous nonobese diabetic (NOD) mouse model of type I diabetes were used to assess whether nicotine could prevent this autoimmune disease. Blood glucose levels, diabetes incidence, pancreas insulin content, and cytokine levels were measured in both models of diabetes, both to asses the level of protection exerted by nicotine and to further investigate its mechanism of action. Nicotine treatment reduced the hyperglycemia and incidence of disease in both the MLDS and NOD mouse models of diabetes. Nicotine also protected against the diabetes-induced decrease in pancreatic insulin content observed in both animal models. The pancreatic levels of the Th1 cytokines interleukin (IL)-12, IL-1, tumor necrosis factor (TNF)-alpha, and interferon (IFN)-gamma were increased in both MLDS-induced and spontaneous NOD diabetes, an effect prevented by nicotine treatment. Nicotine treatment increased the pancreatic levels of the Th2 cytokines IL-4 and IL-10. Nicotine treatment reduces the incidence of type I diabetes in two animal models by changing the profile of pancreatic cytokine expression from Th1 to Th2.
Collapse
Affiliation(s)
- J G Mabley
- Inotek Corporation, Beverly, Massachusetts 01915, USA.
| | | | | | | | | |
Collapse
|
149
|
Abstract
A better understanding of the basic mechanisms of uveitis and of the role of cytokines in experimental ocular inflammation autoimmune diseases should allow us to define new approaches for therapy. Modulation of the cytokine network by either blocking cytokine activity or administration of regulatory Th2 cytokines has shown its efficacy in several experimental autoimmune diseases including uveitis. However, cytokines present pleiotropic activities and thus may exert different effects depending on the autoimmune diseases, making interventions on their production complex. Anti-cytokine therapy or a combination of anti-cytokine drugs, antibodies, and cytokine gene therapy to synergize the therapeutical effects of other treatments appear to be of interest. Improvements in drug delivery and in biotechnology will also allow us to elaborate new and safe immunomodulatory strategies.
Collapse
|
150
|
Bach JF. Immunotherapy of type 1 diabetes: lessons for other autoimmune diseases. ARTHRITIS RESEARCH 2002; 4 Suppl 3:S3-15. [PMID: 12110118 PMCID: PMC3240130 DOI: 10.1186/ar554] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2002] [Revised: 02/27/2002] [Accepted: 03/03/2002] [Indexed: 02/07/2023]
Abstract
The nonobese diabetic (NOD) mouse is a well-recognised animal model of spontaneous autoimmune insulin-dependent diabetes mellitus. The disease is T-cell mediated, involving both CD4 and CD8 cells. Its progress is controlled by a variety of regulatory T cells. An unprecedented number of immunological treatments have been assessed in this mouse strain. This chapter systematically reviews most of these therapeutic manoeuvres, discussing them in the context of their significance with regard to the underlying mechanisms and the potential clinical applications. The contrast between the surprisingly high rate of success found for a multitude of treatments (more than 160) administered early in the natural history of the disease and the few treatments active at a late stage is discussed in depth. Most of the concepts and strategies derived from this model apply to other autoimmune diseases, for which no such diversified data are available.
Collapse
|