1
|
Locker KC, Kachapati K, Wu Y, Bednar KJ, Adams D, Patel C, Tsukamoto H, Heuer LS, Aronow BJ, Herr AB, Ridgway WM. Endosomal Sequestration of TLR4 Antibody Induces Myeloid-Derived Suppressor Cells and Reverses Acute Type 1 Diabetes. Diabetes 2022; 71:470-482. [PMID: 35040474 PMCID: PMC8893939 DOI: 10.2337/db21-0426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 12/08/2021] [Indexed: 11/13/2022]
Abstract
We previously showed that treating NOD mice with an agonistic monoclonal anti-TLR4/MD2 antibody (TLR4-Ab) reversed acute type 1 diabetes (T1D). Here, we show that TLR4-Ab reverses T1D by induction of myeloid-derived suppressor cells (MDSCs). Unbiased gene expression analysis after TLR4-Ab treatment demonstrated upregulation of genes associated with CD11b+Ly6G+ myeloid cells and downregulation of T-cell genes. Further RNA sequencing of purified, TLR4-Ab-treated CD11b+ cells showed significant upregulation of genes associated with bone marrow-derived CD11b+ cells and innate immune system genes. TLR4-Ab significantly increased percentages and numbers of CD11b+ cells. TLR4-Ab-induced CD11b+ cells, derived ex vivo from TLR4-Ab-treated mice, suppress T cells, and TLR4-Ab-conditioned bone marrow cells suppress acute T1D when transferred into acutely diabetic mice. Thus, the TLR4-Ab-induced CD11b+ cells, by the currently accepted definition, are MDSCs able to reverse T1D. To understand the TLR4-Ab mechanism, we compared TLR4-Ab with TLR4 agonist lipopolysaccharide (LPS), which cannot reverse T1D. TLR4-Ab remains sequestered at least 48 times longer than LPS within early endosomes, alters TLR4 signaling, and downregulates inflammatory genes and proteins, including nuclear factor-κB. TLR4-Ab in the endosome, therefore, induces a sustained, attenuated inflammatory response, providing an ideal "second signal" for the activation/maturation of MDSCs that can reverse acute T1D.
Collapse
Affiliation(s)
- Kathryn C.S. Locker
- Division of Immunobiology, Cincinnati Children’s Hospital, Cincinnati, OH
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center, and University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kritika Kachapati
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Yuehong Wu
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kyle J. Bednar
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - David Adams
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Caroline Patel
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Hiroki Tsukamoto
- Department of Pharmaceutical Sciences, School of Pharmacy at Fukuoka International University of Health and Welfare, Okawa, Fukuoka, Japan
| | - Luke S. Heuer
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA
| | - Bruce J. Aronow
- Division of Bioinformatics, Cincinnati Children’s Hospital, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Andrew B. Herr
- Division of Immunobiology, Cincinnati Children’s Hospital, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children’s Hospital, Cincinnati, OH
| | - William M. Ridgway
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA
| |
Collapse
|
2
|
MBD2 acts as a repressor to maintain the homeostasis of the Th1 program in type 1 diabetes by regulating the STAT1-IFN-γ axis. Cell Death Differ 2022; 29:218-229. [PMID: 34420035 PMCID: PMC8738722 DOI: 10.1038/s41418-021-00852-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 08/01/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
The methyl-CpG-binding domain 2 (MBD2) interprets DNA methylome-encoded information through binding to the methylated CpG DNA, by which it regulates target gene expression at the transcriptional level. Although derailed DNA methylation has long been recognized to trigger or promote autoimmune responses in type 1 diabetes (T1D), the exact role of MBD2 in T1D pathogenesis, however, remains poorly defined. Herein, we generated an Mbd2 knockout model in the NOD background and found that Mbd2 deficiency exacerbated the development of spontaneous T1D in NOD mice. Adoptive transfer of Mbd2-/- CD4 T cells into NOD.scid mice further confirmed the observation. Mechanistically, Th1 stimulation rendered the Stat1 promoter to undergo a DNA methylation turnover featured by the changes of DNA methylation levels or patterns along with the induction of MBD2 expression, which then bound to the methylated CpG DNA within the Stat1 promoter, by which MBD2 maintains the homeostasis of Th1 program to prevent autoimmunity. As a result, ectopic MBD2 expression alleviated CD4 T cell diabetogenicity following their adoptive transfer into NOD.scid mice. Collectively, our data suggest that MBD2 could be a viable target to develop epigenetic-based therapeutics against T1D in clinical settings.
Collapse
|
3
|
Hanschmann EM, Petry SF, Eitner S, Maresch CC, Lingwal N, Lillig CH, Linn T. Paracrine regulation and improvement of β-cell function by thioredoxin. Redox Biol 2020; 34:101570. [PMID: 32473461 PMCID: PMC7260591 DOI: 10.1016/j.redox.2020.101570] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
The failure of insulin-producing β-cells is the underlying cause of hyperglycemia in diabetes mellitus. β-cell decay has been linked to hypoxia, chronic inflammation, and oxidative stress. Thioredoxin (Trx) proteins are major actors in redox signaling and essential for signal transduction and the cellular stress response. We have analyzed the cytosolic, mitochondrial, and extracellular Trx system proteins in hypoxic and cytokine-induced stress using β-cell culture, isolated pancreatic islets, and pancreatic islet transplantation modelling low oxygen supply. Protein levels of cytosolic Trx1 and Trx reductase (TrxR) 1 significantly decreased, while mitochondrial Trx2 and TrxR2 increased upon hypoxia and reoxygenation. Interestingly, Trx1 was secreted by β-cells during hypoxia. Moreover, murine and human pancreatic islet grafts released Trx1 upon glucose stimulation. Survival of transplanted islets was substantially impaired by the TrxR inhibitor auranofin. Since a release was prominent upon hypoxia, putative paracrine effects of Trx1 on β-cells were examined. In fact, exogenously added recombinant hTrx1 mitigated apoptosis and preserved glucose sensitivity in pancreatic islets subjected to hypoxia and inflammatory stimuli, dependent on its redox activity. Human subjects were studied, demonstrating a transient increase in extracellular Trx1 in serum after glucose challenge. This increase correlated with better pancreatic islet function. Moreover, hTrx1 inhibited the migration of primary murine macrophages. In conclusion, our study offers evidence for paracrine functions of extracellular Trx1 that improve the survival and function of pancreatic β-cells.
Collapse
Affiliation(s)
- Eva-Maria Hanschmann
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, Germany; Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - Susanne Eitner
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, Germany
| | | | - Neelam Lingwal
- Clinical Research Unit, Center of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, Germany.
| | - Thomas Linn
- Clinical Research Unit, Center of Internal Medicine, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
4
|
Burg AR, Tse HM. Redox-Sensitive Innate Immune Pathways During Macrophage Activation in Type 1 Diabetes. Antioxid Redox Signal 2018; 29:1373-1398. [PMID: 29037052 PMCID: PMC6166692 DOI: 10.1089/ars.2017.7243] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE Type 1 diabetes (T1D) is an autoimmune disease resulting in β-cell destruction mediated by islet-infiltrating leukocytes. The role of oxidative stress in human and murine models of T1D is highly significant as these noxious molecules contribute to diabetic complications and β-cell lysis, but their direct impact on dysregulated autoimmune responses is highly understudied. Pro-inflammatory macrophages play a vital role in the initiation and effector phases of T1D by producing free radicals and pro-inflammatory cytokines to facilitate β-cell destruction and to present antigen to autoreactive T cells. Recent Advances: Redox modulation of macrophage functions may play critical roles in autoimmunity. These include enhancing pro-inflammatory innate immune signaling pathways in response to environmental triggers, enforcing an M1 macrophage differentiation program, controlling antigen processing, and altering peptide recognition by oxidative post-translational modification. Therefore, an oxidative environment may act on multiple macrophage functions to orchestrate T1D pathogenesis. CRITICAL ISSUES Mechanisms involved in the initiation of T1D remain unclear, making preventive and early therapeutics difficult to develop. Although many of these advances in the redox regulation of macrophages are in their infancy, they provide insight into how oxidative stress aids in the precipitating event of autoimmune activation. FUTURE DIRECTIONS Future studies should be aimed at mechanistically determining which redox-regulated macrophage functions are pertinent in T1D pathogenesis, as well as at investigating potential targetable therapeutics to halt and/or dampen innate immune activation in T1D.
Collapse
Affiliation(s)
- Ashley R Burg
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham , Birmingham, Alabama
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
5
|
Previte DM, Piganelli JD. Reactive Oxygen Species and Their Implications on CD4 + T Cells in Type 1 Diabetes. Antioxid Redox Signal 2018; 29:1399-1414. [PMID: 28990401 DOI: 10.1089/ars.2017.7357] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous work has indicated that type 1 diabetes (T1D) pathology is highly driven by reactive oxygen species (ROS). One way in which ROS shape the autoimmune response demonstrated in T1D is by promoting CD4+ T cell activation and differentiation. As CD4+ T cells are a significant contributor to pancreatic β cell destruction in T1D, understanding how ROS impact their development, activation, and differentiation is critical. Recent Advances: CD4+ T cells themselves generate ROS via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression and electron transport chain activity. Moreover, T cells can also be exposed to exogenous ROS generated by other immune cells (e.g., macrophages and dendritic cells) and β cells. Genetically modified animals and ROS inhibitors have demonstrated that ROS blockade during activation results in CD4+ T cell hyporesponsiveness and reduced diabetes incidence. Critical Issues and Future Directions: Although the majority of studies with regard to T1D and CD4+ T cells have been done to examine the influence of redox on CD4+ T cell activation, this is not the only circumstance in which a T cell can be impacted by redox. ROS and redox have also been shown to play roles in CD4+ T cell-related tolerogenic mechanisms, including thymic selection and regulatory T cell-mediated suppression. However, the effect of these mechanisms with respect to T1D pathogenesis remains elusive. Therefore, pursuing these avenues may provide valuable insight into the global role of ROS and redox in autoreactive CD4+ T cell formation and function.
Collapse
Affiliation(s)
- Dana M Previte
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| | - Jon D Piganelli
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| |
Collapse
|
6
|
Feduska JM, Tse HM. The proinflammatory effects of macrophage-derived NADPH oxidase function in autoimmune diabetes. Free Radic Biol Med 2018; 125:81-89. [PMID: 29723665 DOI: 10.1016/j.freeradbiomed.2018.04.581] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/22/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease culminating in the destruction of insulin-producing pancreatic β-cells. While ultimately a T cell-mediated disease, macrophages play an indispensable role in disease initiation and progression. Infiltrating macrophages generate an inflammatory environment by releasing NADPH oxidase-derived superoxide and proinflammatory cytokines. The synthesis of reactive oxygen species (ROS) is acknowledged as putative factors contributing to autoimmunity and β-cell damage in T1D. In addition to direct lysis, free radicals collectively participate in β-cell destruction by providing a redox-dependent third signal necessary for islet-reactive CD4 and CD8 T cell maturation and by inducing oxidative post-translational modifications of β-cell epitopes to further exacerbate autoimmune responses. This review will provide an overview of macrophage function and a synergistic cross-talk with redox biology that contributes to autoimmune dysregulation in T1D.
Collapse
Affiliation(s)
- Joseph M Feduska
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182, United States
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182, United States.
| |
Collapse
|
7
|
Danilova IG, Bulavintceva TS, Gette IF, Medvedeva SY, Emelyanov VV, Abidov MT. Partial recovery from alloxan-induced diabetes by sodium phthalhydrazide in rats. Biomed Pharmacother 2017; 95:103-110. [DOI: 10.1016/j.biopha.2017.07.117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 07/24/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
|
8
|
Affiliation(s)
- Xiangwei Xiao
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| |
Collapse
|
9
|
Xiao X, Gittes GK. Concise Review: New Insights Into the Role of Macrophages in β-Cell Proliferation. Stem Cells Transl Med 2015; 4:655-8. [PMID: 25900729 DOI: 10.5966/sctm.2014-0248] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/09/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Diabetes mellitus can potentially be treated with islet transplantation, but additional sources of β cells are necessary to overcome the short supply of donor pancreases. Although controversy still exists, it is generally believed that the postnatal expansion of the β-cell mass is mainly through pre-existing β-cell replication. Thus, understanding the molecular mechanisms underlying the regulation of β-cell proliferation might lead to clinical strategies for increasing β-cell numbers, both in vitro and in vivo. Macrophages have a well-recognized role in the development of insulitis as part of the pathogenesis of type 1 diabetes. However, a potential role for macrophage polarization, triggered by specific environmental stimuli, in promoting β-cell proliferation has only recently been appreciated. In the present review, we discuss several independent studies, using different regeneration models, that demonstrate a substantial inductive role for macrophages in β-cell proliferation. Additional dissection of the involved cell-cell crosstalk through specific signal transduction pathways is expected to improve our understanding of β-cell proliferation and might facilitate the current β-cell replacement therapy. SIGNIFICANCE New independent findings from different β-cell regeneration models, contributed by different research groups, have provided compelling evidence to highlight a previously unappreciated role for macrophages in β-cell proliferation. Additional dissection of the underlying mechanisms and cell-cell crosstalk might shed new light on strategies to increase the functional β-cell mass in vivo and on β-cell replacement therapies.
Collapse
Affiliation(s)
- Xiangwei Xiao
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - George K Gittes
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Padgett LE, Burg AR, Lei W, Tse HM. Loss of NADPH oxidase-derived superoxide skews macrophage phenotypes to delay type 1 diabetes. Diabetes 2015; 64:937-46. [PMID: 25288672 PMCID: PMC4338593 DOI: 10.2337/db14-0929] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Macrophages are early islet-infiltrating cells seen in type 1 diabetes (T1D). While proinflammatory M1 macrophages induce T1D, M2 macrophages have been shown to delay this autoimmune disease in nonobese diabetic (NOD) mice, but the environmental cues that govern macrophage polarization and differentiation remain unresolved. We previously demonstrated the importance of reactive oxygen species (ROS) in T1D, as NOD mice deficient in NADPH oxidase (NOX)-derived superoxide (Ncf1(m1J)) were protected against T1D partly because of blunted Toll-like receptor-dependent macrophage responses. We provide evidence that NOX-derived ROS contribute to macrophage differentiation in T1D. During spontaneous diabetes progression, T1D-resistant NOD.Ncf1(m1J) islet-resident macrophages displayed a dampened M1 and increased M2 phenotype. The transfer of diabetogenic T cells into NOX-deficient NOD.Rag.Ncf1(m1J) recipients resulted in decreased TNF-α(+) and IL-1β(+) islet-infiltrating M1 macrophages and a concomitant enhancement in arginase-1(+) M2 macrophages. Mechanistic analysis of superoxide-deficient bone marrow-derived macrophages revealed a marked diminution in a proinflammatory M1 phenotype due to decreased P-STAT1 (Y701) and interferon regulatory factor 5 compared with NOD mice. We have therefore defined a novel mechanistic link between NOX-derived ROS and macrophage phenotypes, and implicated superoxide as an important factor in macrophage differentiation. Thus, targeting macrophage redox status may represent a promising therapy in halting human T1D.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Department of Microbiology, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Ashley R Burg
- Department of Microbiology, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Weiqi Lei
- Department of Microbiology, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
11
|
Wong FS, Wen L. Type 1 diabetes therapy beyond T cell targeting: monocytes, B cells, and innate lymphocytes. Rev Diabet Stud 2012; 9:289-304. [PMID: 23804267 DOI: 10.1900/rds.2012.9.289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent clinical trials, investigating type 1 diabetes (T1D), have focused mainly on newly diagnosed individuals who have developed diabetes. We need to continue our efforts to understand disease processes and to rationally design interventions that will be safe and specific for disease, but at the same time not induce undesirable immunosuppression. T cells are clearly involved in the pathogenesis of T1D, and have been a major focus for both antigen-specific and non-antigen-specific therapy, but thus far no single strategy has emerged as superior. As T1D is a multifactorial disease, in which multiple cell types are involved, some of these pathogenic and regulatory cell pathways may be important to consider. In this review, we examine evidence for whether monocytes, B cells, and innate lymphocytes, including natural killer cells, may be suitable targets for intervention.
Collapse
Affiliation(s)
- F Susan Wong
- Institute of Molecular and Experimental Medicine, Cardiff School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | | |
Collapse
|
12
|
Lee MS, Kwon HJ, Kim HS. Macrophages from nonobese diabetic mouse have a selective defect in IFN-γ but not IFN-α/β receptor pathway. J Clin Immunol 2012; 32:753-61. [PMID: 22396045 DOI: 10.1007/s10875-012-9682-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 02/28/2012] [Indexed: 10/28/2022]
Abstract
PURPOSE Aberrant regulation of innate immune cells such as macrophages has been implicated in the onset and progression of type 1 diabetes (T1D). Macrophages from nonobese diabetic (NOD) mouse, an animal model of T1D, entail developmental and functional defects that are often associated with hypo-responsiveness to interferon (IFN)-γ. We aimed to uncover a mechanism underlying this phenomenon. METHODS We analyzed the receptor pathway along with the response of macrophages exposed to IFN-γ and the related IFNs such as IFN-α/β. RESULTS We found that NOD macrophages failed to fully respond to IFN-γ but not to IFN-α for the production of inflammatory cytokines (e.g. TNF-α and IL-12). NOD macrophages were also resistant to apoptotic pathway induced by IFN-γ and LPS. Analyses of receptor pathway revealed that STAT1 pathway of intracellular signaling was selectively impaired in NOD macrophages exposed to IFN-γ but not to IFN-α/β. Further, these defects correlated with a low phosphorylation level of JAK2, and were related to impaired up-regulation of surface IFN-γ receptor 2 (IFN-γR2) by IFN-γ. CONCLUSION Taken together, our results suggest that NOD macrophages have a selective defect in IFN-γ but not IFN-α/β receptor pathway. As IFN-γ and IFN-α have been implicated in the development of autoimmunity towards β-cells, such an unanticipated selectivity in IFN responsiveness may provide a new insight into the pathogenesis of T1D.
Collapse
Affiliation(s)
- Myung-Shik Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | |
Collapse
|
13
|
Lee DY, Nam JH, Byun Y. Effect of polyethylene glycol grafted onto islet capsules on prevention of splenocyte and cytokine attacks. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 15:753-66. [PMID: 15255524 DOI: 10.1163/156856204774196144] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the graft rejection of transplanted islets, the host's immune cells recognize the islets as antigens, which then stimulate the immune cells to begin the cytokine secretion and also the proliferation of immune cells. To prevent the recognition of islets by the immune cells, we grafted biocompatible polyethylene glycol (PEG) onto the collagen capsule of islets without incurring any changes in the morphology and function of islets. To evaluate the efficiency of PEG grafting, PEG-grafted islets were cultured with splenocytes consisting mainly of lymphocytes and macrophages. A splenocyte proliferation assessment using a BrdU incorporation assay showed that the PEG-grafted islets did not stimulate the splenocytes. In addition, the viability and microorganisms in islet cells of co-cultured PEG-grafted islets were not altered. However, in the co-culture of free islets (control) splenocytes were stimulated; they mainly secreted TNF-alpha and strongly affected the viability and structure of free islets. Furthermore, when islets were treated with the rat recombinant TNF-alpha for 7 days, the viabilities of PEG-grafted and free islets were significantly damaged, although the viability of PEG-grafted islets was higher than that of free islets by nearly three times. These results demonstrate that PEG grafted on the surface of islets could prevent the recognition of islets by splenocytes, but could not completely protect islets from cytokines.
Collapse
Affiliation(s)
- Dong Yun Lee
- Department of Materials Science and Engineering, Kwangju Institute of Science and Technology, 1 Oryong-dong, Buk-gu, Gwangju 500-712, South Korea
| | | | | |
Collapse
|
14
|
Thayer TC, Delano M, Liu C, Chen J, Padgett LE, Tse HM, Annamali M, Piganelli JD, Moldawer LL, Mathews CE. Superoxide production by macrophages and T cells is critical for the induction of autoreactivity and type 1 diabetes. Diabetes 2011; 60:2144-51. [PMID: 21715554 PMCID: PMC3142064 DOI: 10.2337/db10-1222] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The role of reactive oxygen species (ROS) and their dissipation in type 1 diabetes pathogenesis have garnered considerable controversy. Our recent work has demonstrated the importance of NADPH oxidase (NOX) activity for type 1 diabetes development and modulating T-cell autoreactivity. We previously linked decreased monocyte ROS with diabetes resistance in the alloxan-resistant mouse, and NOD-Ncf1(m1J) mice with a genetic ablation of NOX activity had reduced and delayed type 1 diabetes compared with NOD mice. RESEARCH DESIGN AND METHODS To determine the required cellular sources of ROS that are necessary for type 1 diabetes initiation, we used antibody depletion and adoptive transfer experiments into NOD and NOD-Scid females, respectively. After receiving treatment, female mice were monitored for hyperglycemia and overt diabetes. RESULTS Depletion of macrophages and neutrophils fully protected NOD mice from type 1 diabetes. However, elimination of neutrophils alone showed no significant reduction or delay. Type 1 diabetes induction in NOD-Scid mice by adoptive transfer with NOD-Ncf1(m1J) splenocytes was significantly delayed compared with NOD splenocytes, suggesting macrophage ROS and modulation of effector responses are critical for diabetes. The adaptive immune response was also altered by the absence of NOX activity, as purified T cells from NOD-Ncf1(m1J) mice exhibited delayed transfer kinetics. Cotransfer experiments demonstrated the defect was intrinsic to NOX-deficient CD8(+) T cells. After stimulation, cytotoxic T cells exhibited decreased effector function in the absence of superoxide production. CONCLUSIONS These data demonstrate that the impaired autoreactive response of NOX-deficient NOD-Ncf1(m1J) immune system results from an alteration in the antigen-presenting cell-T-cell axis rather than failure of neutrophils to act as effector cells and that ROS signaling is important for the initiation of β-cell-directed autoimmunity by T cells.
Collapse
Affiliation(s)
- Terri C. Thayer
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Matthew Delano
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Chao Liu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Jing Chen
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Lindsey E. Padgett
- Department of Microbiology, University of Alabama–Birmingham School of Medicine, Birmingham, Alabama
| | - Hubert M. Tse
- Department of Microbiology, University of Alabama–Birmingham School of Medicine, Birmingham, Alabama
| | - Mani Annamali
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Jon D. Piganelli
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lyle L. Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida
- Corresponding author: Clayton E. Mathews,
| |
Collapse
|
15
|
Morran MP, Omenn GS, Pietropaolo M. Immunology and genetics of type 1 diabetes. ACTA ACUST UNITED AC 2009; 75:314-27. [PMID: 18729178 DOI: 10.1002/msj.20052] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes is one of the most well-characterized autoimmune diseases. Type 1 diabetes compromises an individual's insulin production through the autoimmune destruction of pancreatic beta-cells. Although much is understood about the mechanisms of this disease, multiple potential contributing factors are thought to play distinct parts in triggering type 1 diabetes. The immunological diagnosis of type 1 diabetes relies primarily on the detection of autoantibodies against islet antigens in the serum of type 1 diabetes mellitus patients. Genetic analyses of type 1 diabetes have linked human leukocyte antigen, specifically class II alleles, to susceptibility to disease onset. Environmental catalysts include various possible factors, such as viral infections, although the evidence linking infections with type 1 diabetes remains inconclusive. Imbalances within the immune system's system of checks and balances may promote immune activation, while undermining immune regulation. A lack of proper regulation and overactive pathogenic responses provide a framework for the development of autoimmune abnormalities. Type 1 diabetes is a predictable and potentially treatable disease that still requires much research to fully understand and pinpoint the exact triggering events leading to autoimmune activation. In silico research can aid the comprehension of the etiology of complex disease pathways, including Type I diabetes, in order to and help predict the outcome of therapeutic strategies aimed at preserving beta-cell function.
Collapse
Affiliation(s)
- Michael P Morran
- Department of Internal Medicine, Division of Metabolism, Laboratory of Immunogenetics, Brehm Center for Type 1 Diabetes Research and Analysis, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | |
Collapse
|
16
|
Schroeder MM, Belloto RJ, Hudson RA, McInerney MF. Effects of Antioxidants Coenzyme Q10 and Lipoic Acid on Interleukin-1β-Mediated Inhibition of Glucose-Stimulated Insulin Release from Cultured Mouse Pancreatic Islets. Immunopharmacol Immunotoxicol 2008; 27:109-22. [PMID: 15803864 DOI: 10.1081/iph-51755] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
During the development of the autoimmune disease, insulin-dependent diabetes mellitus (IDDM) islet cell death is thought to be mediated in part by oxygen and nitrogen free radicals and interleukin 1beta (IL-1beta), secreted by activated macrophages. Free radicals disrupt the homeostasis of biological systems by damaging major constituent molecules such as lipids, proteins, and DNA. Islet cells are quite susceptible to oxidative damage due to low levels of antioxidant enzymes involved in free radical consumption. If IDDM is associated with an imbalance of oxidative stresses and antioxidant responses in islet cells, then it may be possible to ameliorate disease by supplementating antioxidant defenses. In this study, the antioxidants coenzyme Q10 and lipoic acid were able to block IL-1beta-mediated inhibition of glucose-stimulated insulin secretion from islet cells at 10(-12) M and 10(-9) M, respectively.
Collapse
Affiliation(s)
- Michelle M Schroeder
- Department of Medicinal and Biological Chemistry, College of Pharmacy, University of Toledo, BO 2833, MS# 606, 2801 W. Bancroft St., Toledo, OH 43606-3390, USA
| | | | | | | |
Collapse
|
17
|
O'Sullivan B, Thompson A, Thomas R. NF-kappa B as a therapeutic target in autoimmune disease. Expert Opin Ther Targets 2007; 11:111-22. [PMID: 17227228 DOI: 10.1517/14728222.11.2.111] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
NF-kappaB transmits signals from the cell surface to the nucleus. Signaling through cell surface receptors to activate NF-kappaB and mitogen-activated protein kinases through adaptor molecules is of critical importance to survival and activation of all cells in the body, including those regulating innate and adaptive immunity. As such, NF-kappaB is a key signaling component in autoimmunity and an attractive target for autoimmune disease therapy. However, given its global importance, targeting NF-kappaB tends to be immunosuppressive. In this review, the authors discuss the roles played by NF-kappaB in autoimmunity, drugs which target it, and complexities which need to be addressed to improve the use of NF-kappaB as a target. Finally, the authors highlight some novel approaches that are likely to be important in the next generation of NF-kappaB therapies.
Collapse
Affiliation(s)
- Brendan O'Sullivan
- Centre for Immunology and Cancer Research, University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland, 4102, Australia
| | | | | |
Collapse
|
18
|
Abstract
Type 1 diabetes results from the destruction of insulin-producing pancreatic beta cells by a beta cell-specific autoimmune process. Beta cell autoantigens, macrophages, dendritic cells, B lymphocytes, and T lymphocytes have been shown to be involved in the pathogenesis of autoimmune diabetes. Beta cell autoantigens are thought to be released from beta cells by cellular turnover or damage and are processed and presented to T helper cells by antigen-presenting cells. Macrophages and dendritic cells are the first cell types to infiltrate the pancreatic islets. Naive CD4+ T cells that circulate in the blood and lymphoid organs, including the pancreatic lymph nodes, may recognize major histocompatibility complex and beta cell peptides presented by dendritic cells and macrophages in the islets. These CD4+ T cells can be activated by interleukin (IL)-12 released from macrophages and dendritic cells. While this process takes place, beta cell antigen-specific CD8+ T cells are activated by IL-2 produced by the activated TH1 CD4+ T cells, differentiate into cytotoxic T cells and are recruited into the pancreatic islets. These activated TH1 CD4+ T cells and CD8+ cytotoxic T cells are involved in the destruction of beta cells. In addition, beta cells can also be damaged by granzymes and perforin released from CD8+ cytotoxic T cells and by soluble mediators such as cytokines and reactive oxygen molecules released from activated macrophages in the islets. Thus, activated macrophages, TH1 CD4+ T cells, and beta cell-cytotoxic CD8+ T cells act synergistically to destroy beta cells, resulting in autoimmune type 1 diabetes.
Collapse
Affiliation(s)
- Ji-Won Yoon
- Rosalind Franklin Comprehensive Diabetes Center, Department of Pathology, Chicago Medical School, North Chicago, IL 60064, USA.
| | | |
Collapse
|
19
|
Hessner MJ, Wang X, Meyer L, Geoffrey R, Jia S, Fuller J, Lernmark A, Ghosh S. Involvement of eotaxin, eosinophils, and pancreatic predisposition in development of type 1 diabetes mellitus in the BioBreeding rat. THE JOURNAL OF IMMUNOLOGY 2005; 173:6993-7002. [PMID: 15557196 DOI: 10.4049/jimmunol.173.11.6993] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Allergy and autoimmunity are both examples of deregulated immunity characterized by inflammation and injury of targeted tissues that have until recently been considered disparate disease processes. However, recent findings have implicated mast cells, in coordination with granulocytes and other immune effector cells, in the pathology of these two disorders. The BioBreeding (BB) DRlyp/lyp rat develops an autoimmune insulin-dependent diabetes similar to human type 1 diabetes mellitus (T1DM), whereas the BBDR+/+ rat does not. To better understand immune processes during development of T1DM, gene expression profiling at day (d) 40 (before insulitis) and d65 (before disease onset) was conducted on pancreatic lymph nodes of DRlyp/lyp, DR+/+, and Wistar-Furth (WF) rats. The eosinophil-recruiting chemokine, eotaxin, and the high-affinity IgE receptor (FcepsilonRI) were up-regulated >5-fold in d65 DRlyp/lyp vs d65 DR+/+ pancreatic lymph nodes by microarray (p < 0.05) and quantitative RT-PCR studies (p < 0.05). DR+/+, WF, and d40 DRlyp/lyp animals possessed normal pancreatic histology; however, d65 DRlyp/lyp animals possessed eosinophilic insulitis. Therefore, immunohistochemistry for pancreatic eotaxin expression was conducted, revealing positive staining of d65 DRlyp/lyp islets. Islets of d65 DR+/+ rats also stained positively, consistent with underlying diabetic predisposition in the BB lineage, whereas WF islets did not. Other differentially expressed transcripts included those associated with eosinophils, mast cells, and lymphocytes. These data support an important role for these inflammatory mediators in BB rat T1DM and suggest that the lymphopenia due to the Ian5/(lyp) mutation may result in a deregulation of cells involved in insulitis and beta cell destruction.
Collapse
MESH Headings
- Animals
- Cell Movement/genetics
- Cell Movement/immunology
- Chemokine CCL11
- Chemokines, CC/biosynthesis
- Chemokines, CC/genetics
- Chemokines, CC/physiology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Eosinophils/pathology
- Gene Expression Profiling
- Genetic Predisposition to Disease
- Immunohistochemistry
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Lymph Nodes/chemistry
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Mast Cells/immunology
- Mast Cells/pathology
- Oligonucleotide Array Sequence Analysis
- Pancreas/immunology
- Pancreas/pathology
- Prediabetic State/genetics
- Prediabetic State/immunology
- Prediabetic State/pathology
- Protein Tyrosine Phosphatase, Non-Receptor Type 1
- Protein Tyrosine Phosphatase, Non-Receptor Type 22
- Protein Tyrosine Phosphatases/biosynthesis
- Protein Tyrosine Phosphatases/genetics
- Rats
- Rats, Inbred BB
- Rats, Inbred WF
- Receptors, IgE/biosynthesis
- Receptors, IgE/genetics
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Martin J Hessner
- The Max McGee National Research Center for Juvenile Diabetes, Department of Pediatrics at the Medical College of Wisconsin and the Children's Research Institute of the Children's Hospital of Wisconsin, Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Type 1 diabetes (T1D) results from the destruction of pancreatic beta cells. Genetic factors are believed to be a major component for the development of T1D, but the concordance rate for the development of diabetes in identical twins is only about 40%, suggesting that nongenetic factors play an important role in the expression of the disease. Viruses are one environmental factor that is implicated in the pathogenesis of T1D. To date, 14 different viruses have been reported to be associated with the development of T1D in humans and animal models. Viruses may be involved in the pathogenesis of T1D in at least two distinct ways: by inducing beta cell-specific autoimmunity, with or without infection of the beta cells, [e.g. Kilham rat virus (KRV)] and by cytolytic infection and destruction of the beta cells (e.g. encephalomyocarditis virus in mice). With respect to virus-mediated autoimmunity, retrovirus, reovirus, KRV, bovine viral diarrhoea-mucosal disease virus, mumps virus, rubella virus, cytomegalovirus and Epstein-Barr virus (EBV) are discussed. With respect to the destruction of beta cells by cytolytic infection, encephalomyocarditis virus, mengovirus and Coxsackie B viruses are discussed. In addition, a review of transgenic animal models for virus-induced autoimmune diabetes is included, particularly with regard to lymphocytic choriomeningitis virus, influenza viral proteins and the Epstein-Barr viral receptor. Finally, the prevention of autoimmune diabetes by infection of viruses such as lymphocytic choriomeningitis virus is discussed.
Collapse
Affiliation(s)
- Hee-Sook Jun
- Julia McFarlane Diabetes Research Centre and Department of Microbiology and Infectious Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
21
|
Beshay E, Prud'homme GJ. Inhibitors of phosphodiesterase isoforms III or IV suppress islet-cell nitric oxide production. J Transl Med 2001; 81:1109-17. [PMID: 11502862 DOI: 10.1038/labinvest.3780323] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The general phosphodiesterase (PDE) inhibitor pentoxifylline (PTX), and the PDE type IV inhibitor rolipram (ROL), both increase intracellular cAMP levels and suppress inflammatory cytokine production by T cells and macrophages. We have previously shown that PTX and ROL protect from autoimmune diabetes in nonobese diabetic (NOD) mice. These drugs may mediate some of their anti-inflammatory effects by blocking nitric oxide (NO) production by macrophages. In this study, we investigated the effect of PDE inhibitors in blocking NO production by insulin-secreting NIT-1 insulinoma cells and mouse islet cells in vitro and in vivo. Insulinoma cells and islet cells produced NO when stimulated with a combination of inflammatory cytokines and lipopolysaccharide (LPS). We found that both PTX and ROL markedly suppressed this induced NO production. Islet cells express PDEs III and IV and, accordingly, the PDE III inhibitor cilostamide (CIL) also suppressed NO production, and a combination of ROL and CIL had a synergistic effect. This suppression appeared to be mediated, at least in part, by elevating cAMP level and was mimicked by other cAMP-elevating agents, ie, membrane-permeable cAMP analogs (dibutyryl cAMP and 8-bromo cAMP) and an adenylate cyclase stimulator (forskolin). PDE inhibitors suppressed the expression of inducible nitric oxide synthase (iNOS) mRNA. In vivo treatment with PTX or ROL prevented iNOS protein expression in the islets of NOD mice with cyclophosphamide-accelerated disease. Our findings suggest that PDE inhibitors can protect islets against autoimmunity.
Collapse
Affiliation(s)
- E Beshay
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
22
|
Falcone M, Yeung B, Tucker L, Rodriguez E, Krahl T, Sarvetnick N. IL-4 triggers autoimmune diabetes by increasing self-antigen presentation within the pancreatic Islets. Clin Immunol 2001; 98:190-9. [PMID: 11161975 DOI: 10.1006/clim.2000.4979] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several findings have recently questioned the long held hypothesis that cytokines belonging to the Th2 pathway are protective in T-cell-mediated autoimmunity. Among them, there is our previous report that pancreatic expression of IL-4 activated islet antigen-specific BDC2.5 T cells and rendered them able to trigger insulin-dependent diabetes mellitus in ins-IL-4/BDC2.5 mice (Mueller et al., Immunity, 7, 1997). Here we analyze the mechanisms underlying IL-4-mediated activation of the self-reactive BDC2.5 T cells. IL-4 is mainly known as the Th2-driving cytokine. However, IL-4 is also critical for DC maturation and upregulation of antigen uptake and presentation by macrophages. In our model, we found that pancreatic expression of IL-4 activated self-reactive BDC2.5 T cells by increasing islet antigen presentation by macrophages and dendritic cells. IL-4 could have triggered self-antigen presentation within the pancreatic islets both by driving maturation of DC from a tolerizing to a priming state and by increasing self-antigen uptake by macrophages.
Collapse
Affiliation(s)
- M Falcone
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, 92037, USA
| | | | | | | | | | | |
Collapse
|
23
|
Chung YH, Jun HS, Son M, Bao M, Bae HY, Kang Y, Yoon JW. Cellular and molecular mechanism for Kilham rat virus-induced autoimmune diabetes in DR-BB rats. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:2866-76. [PMID: 10946320 DOI: 10.4049/jimmunol.165.5.2866] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Kilham rat virus (KRV) causes autoimmune diabetes in diabetes-resistant BioBreeding (DR-BB) rats; however, the mechanism by which KRV induces autoimmune diabetes without the direct infection of beta cells is not well understood. We first asked whether molecular mimicry, such as a common epitope between a KRV-specific peptide and a beta cell autoantigen, is involved in the initiation of KRV-induced autoimmune diabetes in DR-BB rats. We found that KRV peptide-specific T cells generated in DR-BB rats infected with recombinant vaccinia virus expressing KRV-specific structural and nonstructural proteins could not induce diabetes, indicating that molecular mimicry is not the mechanism by which KRV induces autoimmune diabetes. Alternatively, we asked whether KRV infection of DR-BB rats could disrupt the finely tuned immune balance and activate autoreactive T cells that are cytotoxic to beta cells, resulting in T cell-mediated autoimmune diabetes. We found that both Th1-like CD45RC+CD4+ and cytotoxic CD8+ T cells were up-regulated, whereas Th2-like CD45RC-CD4+ T cells were down-regulated, and that isolated and activated CD45RC+CD4+ and CD8+ T cells from KRV-infected DR-BB rats induced autoimmune diabetes in young diabetes-prone BioBreeding (DP-BB) rats. We conclude that KRV-induced autoimmune diabetes in DR-BB rats is not due to molecular mimicry, but is due to a breakdown of the finely tuned immune balance of Th1-like CD45RC+CD4+ and Th2-like CD45RC-CD4+ T cells, resulting in the selective activation of beta cell-cytotoxic effector T cells.
Collapse
Affiliation(s)
- Y H Chung
- Julia McFarlane Diabetes Research Center, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
24
|
Shinomiya M, Nadano S, Shinomiya H, Onji M. In situ characterization of dendritic cells occurring in the islets of nonobese diabetic mice during the development of insulitis. Pancreas 2000; 20:290-6. [PMID: 10766456 DOI: 10.1097/00006676-200004000-00011] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Type 1 diabetes mellitus in nonobese diabetic (NOD) mice, a well-known model of human type 1 diabetes, has been considered to be caused by the destruction of insulin-producing beta cells in the islets of the pancreas by self-reactive T cells. Antigen-presenting cells like dendritic cells (DCs) and macrophages are expected to be involved in the processes from their role in generating regulatory or effector T cells. These immunohistochemical studies revealed that CD11c-positive DCs already appeared in the islets of NOD mice as early as 4 weeks old when lymphocytes were not yet infiltrated in the islet, and thus insulitis was not developed. DCs were first observed to locate around swollen parainsular vessels. From age 7 weeks onward to age 13 weeks, more DCs were present in parainsular areas where lymphocytes had also accumulated, and the number of DCs in the islets as well as lymphocytes increased. However, at the end stage of insulitis from age approximately 17 weeks onward, the number of DCs in the islets decreased. In contrast, accumulation of DCs in the para- and periislets was not observed in 7- and 17-week-old ICR female mice that do not develop type 1 diabetes. Double-staining studies using confocal laser scanning microscopy showed that the CD11c-positive DCs coexpress both major histocompatibility (MHC) class II and costimulatory molecules, CD80 and CD86. Electron-microscopy studies further demonstrated that cell bodies and processes of the DCs make close contact with lymphocytes. These results suggest that DCs infiltrated into the pancreatic islets are capable of stimulating T cells by the MHC class II-antigenic peptide complex, together with costimulatory molecules, which eventually lead to the beta-cell destruction in NOD mice.
Collapse
Affiliation(s)
- M Shinomiya
- Third Department of Internal Medicine, Ehime University School of Medicine, Shigenobu, Japan.
| | | | | | | |
Collapse
|
25
|
Bieg S, Simonson W, Ellefsen K, Lernmark A. Rel B is an early marker of autoimmune islet inflammation in the biobreeding (BB) rat. Pancreas 2000; 20:47-54. [PMID: 10630383 DOI: 10.1097/00006676-200001000-00007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Because the development of insulitis and diabetes is predictable in Lyp/Lyp congenic BB rats, we have characterized early islet inflammation in these rats to determine the cell subsets involved in the onset of autoimmune insulitis. Pancreas sections from prediabetic Lyp/Lyp, Lyp/+ and +/+ rats were analyzed by immunohistochemistry. We found W3/25+ cells in the exo- and endocrine tissue from all three genotypes, but intraislet insulitis was never found in Lyp/+ or +/+ rats. The onset of massive, intraislet B- and T-cell infiltration in Lyp/Lyp rats was preceded by Rel B+ cells in and around the islets, followed by ED1+ monocytes/macrophages. Rel B+ cells were more frequent in the parafollicular cortex of pancreatic lymph nodes from Lyp/Lyp than from Lyp/+ and +/+ rats. In the Lyp/Lyp thymus, we found significantly increased expression of IL-12p40 messenger RNA (mRNA; p<0.001), located in the Rel B-protein-rich corticomedullary junction. The NF-KB/Rel B complex specifically transactivates genes involved in antigen presentation in dendritic cells. Rel B+ cells in the islets may therefore mark the onset of autoimmune insulitis and antigen-specific activation of autoreactive T cells in the lymph nodes of diabetes prone Lyp/Lyp BB rats. In the thymus, Rel B+ cells may support the Lyp-dependent development of self-reactive thymocytes by activation of cytokine expression.
Collapse
Affiliation(s)
- S Bieg
- Robert H. Williams Laboratory, Department of Medicine, University of Washington, Seattle, USA.
| | | | | | | |
Collapse
|
26
|
Jun HS, Yoon CS, Zbytnuik L, van Rooijen N, Yoon JW. The role of macrophages in T cell-mediated autoimmune diabetes in nonobese diabetic mice. J Exp Med 1999; 189:347-58. [PMID: 9892617 PMCID: PMC2192977 DOI: 10.1084/jem.189.2.347] [Citation(s) in RCA: 205] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We have shown previously that the inactivation of macrophages in nonobese diabetic (NOD) mice results in the prevention of diabetes; however, the mechanisms involved remain unknown. In this study, we found that T cells in a macrophage-depleted environment lost their ability to differentiate into beta cell-cytotoxic T cells, resulting in the prevention of autoimmune diabetes, but these T cells regained their beta cell-cytotoxic potential when returned to a macrophage-containing environment. To learn why T cells in a macrophage-depleted environment lose their ability to kill beta cells, we examined the islet antigen-specific immune response and T cell activation in macrophage-depleted NOD mice. There was a shift in the immune balance, a decrease in the T helper cell type 1 (Th1) immune response, and an increase in the Th2 immune response, due to the reduced expression of the macrophage-derived cytokine IL-12. As well, there was a deficit in T cell activation, evidenced by significant decreases in the expression of Fas ligand and perforin. The administration of IL-12 substantially reversed the prevention of diabetes in NOD mice conferred by macrophage depletion. We conclude that macrophages play an essential role in the development and activation of beta cell-cytotoxic T cells that cause beta cell destruction, resulting in autoimmune diabetes in NOD mice.
Collapse
Affiliation(s)
- H S Jun
- Julia McFarlane Diabetes Research Centre, Department of Microbiology and Infectious Diseases, Faculty of Medicine, The University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | |
Collapse
|
27
|
Krippeit-Drews P, Kramer C, Welker S, Lang F, Ammon HP, Drews G. Interference of H2O2 with stimulus-secretion coupling in mouse pancreatic beta-cells. J Physiol 1999; 514 ( Pt 2):471-81. [PMID: 9852328 PMCID: PMC2269080 DOI: 10.1111/j.1469-7793.1999.471ae.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
1. We have reported previously that in mouse pancreatic beta-cells H2O2 hyperpolarizes the membrane and increases the ATP-sensitive K+ current recorded in the perforated patch configuration of the patch-clamp technique. The present study was undertaken to elucidate the underlying mechanisms. 2. The intracellular ATP concentration measured by chemoluminescence was reduced by H2O2. The ADP concentration increased in parallel during the first 10 min, resulting in a pronounced decrease in the ATP/ADP ratio. 3. Consistent with these results, glucose-stimulated insulin secretion from isolated islets was inhibited by H2O2. 4. Membrane hyperpolarization measured with intracellular microelectrodes in intact islets and inhibition of insulin secretion were counteracted by tolbutamide, indicating that the channels are still responsive to inhibitors and that the ATP concentration is not too low to trigger exocytosis. However, the sensitivity of the beta-cells to tolbutamide was reduced after treatment with H2O2. 5. H2O2 increased the intracellular Ca2+ activity ([Ca2+]i) in a biphasic manner. A first transient rise in [Ca2+]i due to mobilization of Ca2+ from intracellular stores was followed by a sustained increase, which was at least partly dependent on Ca2+ influx. The first phase seems to reflect Ca2+ mobilization from mitochondria. 6. Our results demonstrate that H2O2 interferes with glucose metabolism, which influences the membrane potential and ATP-sensitive K+ current via the intracellular concentration of ATP. These events finally lead to an inhibition of insulin secretion despite an increase in [Ca2+]i.
Collapse
Affiliation(s)
- P Krippeit-Drews
- Institute of Physiology, Gmelinstrasse 5, University of Tubingen, D-72076 Tubingen, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Carlsson PO, Sandler S, Jansson L. Pancreatic islet blood perfusion in the nonobese diabetic mouse: diabetes-prone female mice exhibit a higher blood flow compared with male mice in the prediabetic phase. Endocrinology 1998; 139:3534-41. [PMID: 9681505 DOI: 10.1210/endo.139.8.6153] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The present study tested the hypothesis that changes in pancreatic islet blood flow correlate with the difference in diabetes incidence between male and female nonobese diabetic (NOD) mice. The blood flows were determined by a microsphere technique. In animals aged 10 and 14 weeks, the islet blood perfusion was 3-fold higher in female NOD mice compared with that in either age-matched male NOD mice or age- and sex-matched control ICR mice. At 5 weeks of age islet blood flow was similar in all groups. No differences between male and female NOD mice in whole pancreatic, duodenal, ileal, or colonic blood flows were observed at any time point. Administration of a bolus dose of aminoguanidine (a blocker of inducible nitric oxide synthase) to 10-week-old animals selectively and markedly decreased islet blood flow in female NOD mice, whereas islet blood flow in ICR mice and male NOD mice remained unaffected. Aminoguanidine did not affect mean arterial blood pressure or whole pancreatic blood flow in any of the groups. Injection of N(G)-methyl-L-arginine, an unspecific inhibitor of both constitutive and inducible nitric oxide synthase, markedly decreased whole pancreatic and islet blood flow to the same level in both male and female NOD mice. These combined findings suggest that diabetes-prone female NOD mice have an increased islet blood flow, which is mediated by an excessive production of nitric oxide formed by inducible nitric oxide synthase. The islet blood hyperperfusion may augment homing to the pancreatic islets of inflammatory cells and soluble factors involved in beta-cell destruction during the development of insulin-dependent diabetes mellitus in this animal model. The presently observed gender difference in the blood flow response could, therefore, at least partially explain why female NOD mice are more prone to develop hyperglycemia than the males.
Collapse
Affiliation(s)
- P O Carlsson
- Department of Medical Cell Biology, Uppsala University, Sweden.
| | | | | |
Collapse
|
29
|
Yoon JW, Jun HS, Santamaria P. Cellular and molecular mechanisms for the initiation and progression of beta cell destruction resulting from the collaboration between macrophages and T cells. Autoimmunity 1998; 27:109-22. [PMID: 9583742 DOI: 10.3109/08916939809008041] [Citation(s) in RCA: 119] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Insulin-dependent diabetes mellitus (IDDM) is caused by the progressive autoimmune destruction of insulin-producing pancreatic beta cells. Although the pathogenesis of autoimmune IDDM has been extensively studied, the precise mechanisms involved in the initiation and progression of beta cell destruction remain unclear. Animal models used in the study of IDDM, such as the BioBreeding (BB) rat and the nonobese diabetic (NOD) mouse, have greatly enhanced our understanding of the pathogenic mechanisms involved in this disease. In these animals, macrophages and/or dendritic cells are the first cell types to infiltrate the pancreatic islets. Macrophages must be involved in the pathogenesis of IDDM early on, since inactivation of macrophages results in the near-complete prevention of insulitis and diabetes in both NOD mice and BB rats. The presentation of beta cell-specific autoantigens by macrophages and/or dendritic cells to CD4+ T helper cells, in association with MHC class II molecules, is considered the initial step in the development of autoimmune IDDM. The activated macrophages secrete IL-12, which stimulates Th1 type CD4+ T cells. The CD4+ T cells secrete IFN-gamma and IL-2. IFN-gamma activates other resting macrophages, which, in turn, release cytokines, such as IL-1beta, TNF-alpha, and free radicals, which are toxic to beta cells. During this process, IL-2 and other cytokines induce the migration of CD8+ peripheral T cells to the inflamed islets, perhaps by inducing the expression of a specific homing receptor. The precytotoxic CD8+ T cells that bear beta cell-specific autoantigen receptors differentiate into cytotoxic effector T cells upon recognition of the beta cell-specific peptide bound to MHC class I molecules in the presence of beta cell-specific CD4+ T helper cells. The cytotoxic CD8+ T cells then effect beta cell damage by releasing perforin and granzyme, and by Fas-mediated apoptosis. In this way, macrophages, CD4+ T cells, and CD8+ T cells synergistically destroy beta cells, resulting in the onset of autoimmune IDDM.
Collapse
Affiliation(s)
- J W Yoon
- Department of Microbiology and Infectious Disease, Julia McFarlane Diabetes Research Centre, Faculty of Medicine, University of Calgary, Alberta, Canada
| | | | | |
Collapse
|
30
|
Chen ZW, Ahren B, Ostenson CG, Cintra A, Bergman T, Möller C, Fuxe K, Mutt V, Jörnvall H, Efendic S. Identification, isolation, and characterization of daintain (allograft inflammatory factor 1), a macrophage polypeptide with effects on insulin secretion and abundantly present in the pancreas of prediabetic BB rats. Proc Natl Acad Sci U S A 1997; 94:13879-84. [PMID: 9391121 PMCID: PMC28401 DOI: 10.1073/pnas.94.25.13879] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A bioactive macrophage factor, the polypeptide daintain/allograft inflammatory factor 1 (AIF1), has been isolated from porcine intestine. It was discovered when searching for intestinal peptides with effects on insulin release, and its purification was monitored by the influence of the peptide fractions on pancreatic glucose-induced insulin secretion. Daintain/AIF1 is a 146-aa residue polypeptide with a mass of 16,603 Da and an acetylated N terminus. An internal 44-residue segment with the sequence pattern -KR-KK-GKR- has a motif typical of peptide hormone precursors, i.e., dibasic sites for potential activation cleavages and at the sequentially last such site, the structure GKR. The latter is a signal for C-terminal amide formation in the processing of peptide hormones. Daintain/AIF1 is immunohistochemically localized to microglial cells in the central nervous system and to dendritic cells and macrophages in several organs. A particularly dense accumulation of daintain/AIF1-immunoreactive macrophages was observed in the insulitis affecting the pancreatic islets of prediabetic BB rats. When injected intravenously in mice, daintain/AIF1 at 75 pmol/kg inhibited glucose (1 g/kg)-stimulated insulin secretion, with a concomitant impairment of the glucose elimination, whereas at higher doses (7.5 and 75 nmol/kg), daintain/AIF1 potentiated glucose-stimulated insulin secretion and enhanced the glucose elimination. Its dual influence on insulin secretion in vivo at different peptide concentrations, and the abundance of macrophages expressing daintain/AIF1 in the pancreatic islets of prediabetic rats, suggest that daintain/AIF1 may have a role in connection with the pathogenesis of insulin-dependent diabetes mellitus.
Collapse
Affiliation(s)
- Z W Chen
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Holstad M, Jansson L, Sandler S. Inhibition of nitric oxide formation by aminoguanidine: an attempt to prevent insulin-dependent diabetes mellitus. GENERAL PHARMACOLOGY 1997; 29:697-700. [PMID: 9347312 DOI: 10.1016/s0306-3623(97)00012-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
1. Insulin-dependent diabetes mellitus is an autoimmune disease leading to pancreatic beta-cell destruction, an event that may, at least partially, be induced by the formation of nitric oxide. 2. Under the influence of cytokines, the enzyme nitric oxide synthase is induced. 3. Blockage of the inducible form of nitric oxide synthase has been found to protect against insulin-dependent diabetes mellitus in some animal models. 4. Aminoguanidine has been found to be a fairly specific inhibitor of cytokine-inducible nitric oxide synthase. 5. Aminoguanidine may reduce the blood flow to the pancreatic islets in vivo and, at higher concentrations, also impair insulin secretion by the beta-cells,--which may make the compound less useful in attempts to prevent insulin-dependent diabetes mellitus.
Collapse
Affiliation(s)
- M Holstad
- Department of Medical Cell Biology, Uppsala University, Sweden
| | | | | |
Collapse
|
32
|
Li XB, Scott FW, Park YH, Yoon JW. Low incidence of autoimmune type I diabetes in BB rats fed a hydrolysed casein-based diet associated with early inhibition of non-macrophage-dependent hyperexpression of MHC class I molecules on beta cells. Diabetologia 1995; 38:1138-47. [PMID: 8690165 DOI: 10.1007/bf00422362] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Diabetes-prone BioBreeding (DPBB) rats were fed a diabetogenic, mainly plant-based rodent diet, Purina Chow 5001, or a diabetes-retardant, hydrolysed casein-based diet. The expression of MHC class I antigens on pancreatic beta cells occurred at around 25 days of age in Purina Chow-fed rats, and progressively increased with the length of time of feeding with the Purina diet. Most of the Purina Chow-fed DPBB rats revealed hyperexpression of MHC class I antigens on their pancreatic beta cells by 50 days of age. Approximately 92% of the hyperexpressed Purina Chow-fed DPBB rats developed severe insulitis and diabetes. In contrast, the majority of hydrolysed casein-fed DPBB rats did not show MHC class I antigen hyperexpression and these rats failed to develop insulitis or diabetes. Purina Chow-fed Wistar-Furth rats and diabetes-resistant BioBreeding (DRBB) rats showed only very weak background staining for MHC class I antigens on their beta cells. When Purina Chow-fed (DPBB rats were treated with silica to inhibit macrophage infiltration into the pancreatic islets, the hyperexpression of MHC class I antigens was seen even more clearly, as beta cells remained intact. MHC class II antigens were not detected on pancreatic beta cells from DPBB, DRBB or Wistar-Furth rats, regardless of their diet. On the basis of these observations, we concluded that hyperexpression of MHC class I antigens on pancreatic beta cells was mainly restricted to Purina Chow-fed DPBB rats and that suppression of non-macrophage-dependent MHC class I antigen hyperexpression on pancreatic beta cells by a hydrolysed caseinbased diet resulted in the prevention of insulitis and diabetes.
Collapse
Affiliation(s)
- X B Li
- Laboratory of Viral and Immunopathogenesis of Diabetes, Julia McFarlane Diabetes Research Centre, University of Calgary Health Science Centre, Alberta, Canada
| | | | | | | |
Collapse
|
33
|
Lernmark A, Klöppel G, Stenger D, Vathanaprida C, Fält K, Landin-Olsson M, Baskin DG, Palmer JP, Gown AM, Petersen JS. Heterogeneity of islet pathology in two infants with recent onset diabetes mellitus. Virchows Arch 1995; 425:631-40. [PMID: 7697219 DOI: 10.1007/bf00199353] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The mechanisms by which the beta cells of pancreatic islets are destroyed in insulin-dependent diabetes mellitus (IDDM) are poorly understood. In this report the pancreatic histo- and immunopathology of two children, both HLA-DR 3/4, DQ 2/8 positive and who both died from cerebral oedema within a day of clinical diagnosis of IDDM, were investigated. Patient 1, a 14-month-old girl, had a 4-week history of polydipsia and polyuria. Patient 2, a 3-year-old boy, had 2 days of illness. Both patients had a similarly severe loss of insulin cells but differed markedly as to the extent of lymphocytic islet infiltration (insulitis). Apart from insulitis, marked islet macrophage infiltration was demonstrated in both patients with the HAM-56 monoclonal antibody. Neither patient showed aberrant expression of HLA class II antigens on insulin-immunoreactive cells, but allele-specific HLA-DQ8 expression was evident on endothelial cells. Glutamic acid decarboxylase immunoreactivity was detected in both insulin- and glucagon-immunoreactive cells. It is concluded that the heterogeneity of islet pathology, especially insulitis, may reflect different dynamics and extent rather than different pathomechanisms of immune destruction of islets in IDDM.
Collapse
Affiliation(s)
- A Lernmark
- Department of Medicine, University of Washington, Seattle 98195
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Josefsen K, Nielsen H, Lorentzen S, Damsbo P, Buschard K. Circulating monocytes are activated in newly diagnosed type 1 diabetes mellitus patients. Clin Exp Immunol 1994; 98:489-93. [PMID: 7994912 PMCID: PMC1534494 DOI: 10.1111/j.1365-2249.1994.tb05517.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Investigations in the BB rat and the non-obese diabetic (NOD) mouse have provided substantial evidence for the involvement of the monocyte/macrophage system in the development of type 1 diabetes mellitus. However, it is not known whether monocytes play the same role in the pathogenesis of human type 1 diabetes. We investigated this problem in a longitudinal study of 29 recent-onset type 1 diabetes mellitus patients. Monocyte chemotaxis, phagocytosis and superoxide production as well as metabolic and haematological parameters were studied immediately after diagnosis and 6 months later. At diagnosis the patients had activated casein and C5a chemotaxis (casein 70 +/- 9 versus 150 +/- 5 (mean +/- s.e.m.), P < 0.001; C5a 137 +/- 10 versus 158 +/- 5, P < 0.05 (activation immobilizes monocytes, reducing the measured values)), and activated superoxide production (3.6 +/- 0.3 versus 3.0 +/- 0.3, P < 0.05). After 6 months casein chemotaxis (115 +/- 16 versus 150 +/- 5, P < 0.05) and Candida phagocytosis (3.3 +/- 0.1 versus 2.8 +/- 0.2, P < 0.001) were still activated. There was no correlation with other clinical or paraclinical parameters. We conclude that the circulating monocytes in newly diagnosed type 1 diabetes patients are activated. It is reasonable to expect that monocytes at the local site of inflammation in pancreas are even further activated. This could play a pathogenic role in beta cell destruction.
Collapse
Affiliation(s)
- K Josefsen
- Bartholin Instituttet, Kommunehospitalet, Copenhagen K, Denmark
| | | | | | | | | |
Collapse
|
35
|
Yderstraede KB, Starklint H, Steinbruchel D, Jørgensen TW, Gotfredsen CF. Fetal rat pancreas transplantation in BB rats: immunohistochemical and functional evaluation. VIRCHOWS ARCHIV. B, CELL PATHOLOGY INCLUDING MOLECULAR PATHOLOGY 1993; 64:13-9. [PMID: 8401812 DOI: 10.1007/bf02915091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Spontaneously diabetic BB/Wor rats received either a syngeneic fetal pancreas transplant or adult islets. In the former, 4-8 fetal pancreases were transplanted, and in the latter, 3-5000 islets. Transplantation was performed by transferring a blood clot containing the pancreases or islets to the renal subcapsular space. Insulin therapy was undertaken postoperatively, except in one experiment with adult islets. Of the fetal pancreas transplanted BB rats, 52% became normoglycaemic, and 21% remained so throughout an observation period of 10 months. Nephrectomy caused a prompt return of diabetes. The histological appearance of the grafts transplanted to the diabetic animals closely resembled that of grafts transplanted to normal rats in a parallel series. For comparison a group of BB rats received a syngeneic transplant of isolated adult islets from WF rats or BBW rats. Following adult islet transplantation, 5 out of 6 animals became hyperglycaemic after a median of 20.5 days when no insulin was given post-transplantation. Four out of 5 animals became hyperglycaemic after a median of 23 days when supportive insulin therapy was administered after the transplantation. The results indicate that recurrent diabetes is not inevitable following syngeneic fetal pancreas transplantation to spontaneously diabetic BB rats. Recurrent diabetes was only occasionally associated with mononuclear cell infiltration. Transplanted tissue was well-preserved and vascularized; mega-islets were a constant finding.
Collapse
Affiliation(s)
- K B Yderstraede
- Department of Medical Endocrinology, Odense University Hospital, Denmark
| | | | | | | | | |
Collapse
|
36
|
Hosszufalusi N, Chan E, Teruya M, Takei S, Granger G, Charles MA. Quantitative phenotypic and functional analyses of islet immune cells before and after diabetes onset in the BB rat. Diabetologia 1993; 36:1146-54. [PMID: 8270129 DOI: 10.1007/bf00401059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Inflammatory cells invading islets are thought to be mediators of islet destruction in spontaneous autoimmune diabetes mellitus. Thus methods were developed to isolate and characterize in situ islet inflammatory cells from 75-95-day-old prediabetic and diabetic BB rats. Islet inflammatory cells were structurally examined using single- and double-colour flow cytometry. Functional studies consisted of cytolytic assays using normal rat islet target cells and in situ islet or spleen effector cells. Structural data reveal natural killer cells to be the major cell population (70%) of total immune cells present in inflamed islets during prediabetes. At diabetes onset, the natural killer cell population remained at a high level (47%), but an increasing population of T cells (40%) was noted also. Analyses of T-cell subsets before and after diabetes onset revealed CD4+ T cells as predominant (50-55% of total T cells) with double-negative (CD4-CD8-) T cells (25-30%) and CD8+ T cells (15-20%) also present in significant quantities. Activated T cells accounted only for a minority of T cells (< 3%). Functional studies indicate that in situ islet-derived cytolytic effector cells are more potent killers (ten-fold) of normal islet target cells than are splenic effector cells. These data suggest that in situ islet inflammatory cells (a) can be quantitatively studied both structurally and functionally; (b) express structural phenotypes differing substantially from splenic mononuclear cell populations; (c) are considerably more cytolytic than splenic effectors; and (d) should prove informative in determining the most significant autoimmune functional events prior to and during islet beta-cell destruction.
Collapse
Affiliation(s)
- N Hosszufalusi
- Diabetes Research Program, University of California, Irvine
| | | | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- A Lernmark
- Department of Endocrinology, Karolinska Institute, Karolinska Hospital, Stockholm, Sweden
| | | | | | | |
Collapse
|
38
|
Wu G, Flynn NE. The activation of the arginine-citrulline cycle in macrophages from the spontaneously diabetic BB rat. Biochem J 1993; 294 ( Pt 1):113-8. [PMID: 8363561 PMCID: PMC1134573 DOI: 10.1042/bj2940113] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The activity of the arginine-citrulline cycle was investigated in macrophages from the spontaneous immunologically mediated diabetic BB rat. Peritoneal macrophages were prepared from male diabetes-prone (BBdp), diabetic (BBd) and age-matched non-diabetes-prone (BBn) rats. Cells were incubated at 37 degrees C for 2 h in Krebs-Henseleit bicarbonate buffer containing 0.5 mM L-arginine, 0.1 mM L-[ureido-14C]citrulline and 5 mM D-glucose to measure the activity of the arginine-citrulline cycle. The uptakes of citrulline and arginine by macrophages were measured during a 5 min incubation period with L-[ureido-14C]citrulline and L-[2,3-3H] arginine respectively. The production of NO3- (the major stable oxidation product of NO) increased (P < 0.01) by 112% and 151% in 75-day-old BBdp and 115-day-old BBd macrophages respectively, compared with age-matched BBn cells. The conversion of [14C]citrulline into [14C]arginine increased (P < 0.01) by 704%, 892% and 904% in 50- and 75-day-old BBdp and 115-day-old BBd macrophages respectively, compared with age-matched BBn cells. The enhanced NO synthesis in BBdp and BBd macrophages was associated with a 25-35% increase in the uptake of L-arginine. However, there were no differences in the uptake of citrulline between BBdp or BBd macrophages and age-matched BBn cells. Our results demonstrate for the first time the activation of the arginine-citrulline cycle in macrophages in an autoimmune condition. The inherent increase in the recycling of L-citrulline to L-arginine in BBdp and BBd macrophages may reflect an innate metabolic disorder in these cells. This increased L-arginine synthesis from L-citrulline may play a role in sustaining a sufficient intracellular L-arginine concentration for prolonged generation of NO in BBdp and BBd macrophages. A role for NO in the autoimmune destruction of pancreatic beta-cells in insulin-dependent diabetes mellitus warrants further investigation.
Collapse
Affiliation(s)
- G Wu
- Department of Animal Science, Texas A&M University, College Station 77843-2471
| | | |
Collapse
|
39
|
Ellerman K, Wrobleski M, Rabinovitch A, Like A. Natural killer cell depletion and diabetes mellitus in the BB/Wor rat (revisited). Diabetologia 1993; 36:596-601. [PMID: 7689498 DOI: 10.1007/bf00404067] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The BB/Wor diabetes-prone rat is an animal model of human insulin-dependent diabetes mellitus. In this model of spontaneous autoimmunity, natural killer cells are candidate cytotoxic effector cells, believed to be the mediators of beta-cell cytolysis in vivo. We therefore studied the effects of an anti-natural killer cell monoclonal antibody on the spontaneous development of diabetes in the BB/Wor rat. The 3.2.3 monoclonal antibody recognizes a molecular present on rat natural killer cells and selectively depletes these cells in vivo. Chronic treatment of diabetic-prone rats with 3.2.3 monoclonal antibody cleared circulating phenotypic natural killer cells, depleted in vitro spleen natural killer cell function, and profoundly reduced intra-islet accumulation of 3.2.3+ cells, but did not prevent or delay the onset of diabetes. These results indicate that natural killer cells are not necessary for the development of spontaneous diabetes in BB/Wor rats.
Collapse
Affiliation(s)
- K Ellerman
- Department of Pathology, University of Massachusetts Medical School, Worcester
| | | | | | | |
Collapse
|
40
|
Tafuri A, Bowers WE, Handler ES, Appel M, Lew R, Greiner D, Mordes JP, Rossini AA. High stimulatory activity of dendritic cells from diabetes-prone BioBreeding/Worcester rats exposed to macrophage-derived factors. J Clin Invest 1993; 91:2040-8. [PMID: 8486773 PMCID: PMC288202 DOI: 10.1172/jci116426] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Dendritic cells (DC) present antigen and initiate T cell-mediated immune responses. To investigate the possible association of autoimmunity with DC function, we compared the accessory activity of splenic DC from Wistar/Furth (WF) and diabetes-prone (DP) BioBreeding (BB) rats. The latter develop autoimmune diabetes and thyroiditis. DC function was quantified in vitro by measuring T cell proliferation in mitogen-stimulated and mixed lymphocyte reactions. When purified without macrophage coculture, WF and DP DC displayed similar levels of accessory activity. In contrast, when purified by a method involving coculture with macrophages, DC from DP rats consistently displayed greater accessory activity. This finding could not be explained by morphological or phenotypic differences between DP and WF DC. In accessory activity assays performed after reciprocal DC cocultures with DP and WF macrophages, DP DC exhibited higher accessory activity irrespective of macrophage donor strain. We also compared the accessory activity of WF and DP DC cultured in the presence of conditioned medium and a mixture of IL-1 and GM-CSF. In all assays, DP DC exhibited higher accessory activity. In studies of (WF x DP) F1 hybrids, the high accessory activity of DP DC was observed to be heritable, and studies of WF and DP radiation chimeras indicated that the effect was an intrinsic property of the DP hematopoietic system. We conclude: (a) splenic DC from DP and WF rats possess similar basal levels of accessory potency; (b) after interaction with macrophages, DC of DP origin are capable of greater stimulatory activity than are WF DC; and (c) the mechanism responsible for this phenomenon involves differential responsiveness of DP and WF DC to macrophage-derived factors such as IL-1 and GM-CSF.
Collapse
Affiliation(s)
- A Tafuri
- Department of Medicine, University of Massachusetts Medical School, Worcester 01655
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Jobe LW, Ubungen R, Goodner CJ, Baskin DG, Braquet P, Lernmark A. Protection from BB rat diabetes by the platelet-activating factor inhibitor BN50730. Autoimmunity 1993; 16:259-66. [PMID: 8025205 DOI: 10.3109/08916939309014644] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The platelet-activating factor inhibitor BN50730, a hetrazepine, was injected intraperitoneally daily from 30 days of age into diabetes-prone BB rats. While 96% (22/23) Tween 80 injected control rats developed diabetes, 0.05 mg/kg BN50730 decreased the frequency to 72% (17/24; n.s.) and 0.5 mg/kg to 56% (14/25; p < 0.01). Mean onset age in controls was 81 +/- 9 days (mean +/- SD), but BN50730 delayed onset to 87 +/- 15 days in the low and 93 +/- 12. days (p < 0.01) in high dose rats. The relative degree of insulitis was reduced in both low (p < 0.01) and high (p < 0.05) dose treated groups. Serum insulin in young prediabetic controls decreased from 84 +/- 34 microU/ml to 38 +/- 20 in the 22 rats developing diabetes (p < 0.001). Serum insulin in BN50730-protected compared to unprotected rats was 114 +/- 49 and 32 +/- 22 (p < 0.001) in the low, and 91 +/- 46 and 21 +/- 15 (p < 0.001) microU/ml in the high dose group, respectively. Increased serum insulin correlated with preserved islet beta cells and decreased insulitis. Treatment did not affect thyroiditis. Thus, platelet-activating factor may be involved in insulitis pathogenesis and platelet-activating factor inhibitors may decrease autoimmune beta cell destruction.
Collapse
Affiliation(s)
- L W Jobe
- University of Washington, School of Medicine, Seattle 98195
| | | | | | | | | | | |
Collapse
|
42
|
Kröncke KD, Rodriguez ML, Kolb H, Kolb-Bachofen V. Cytotoxicity of activated rat macrophages against syngeneic islet cells is arginine-dependent, correlates with citrulline and nitrite concentrations and is identical to lysis by the nitric oxide donor nitroprusside. Diabetologia 1993; 36:17-24. [PMID: 7679656 DOI: 10.1007/bf00399088] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Lysis of rat islet cells by syngeneic activated macrophages in vitro can be completely inhibited by the nitric oxide-synthase-inhibitor NG-methyl-L-arginine. This inhibition can be reversed by an excess of L-arginine. Time-dependent lysis of islet cells by activated macrophages is accompanied by increasing concentrations of nitrite and citrulline in the culture medium both of which are measures of nitric oxide formation derived from L-arginine. Lysis of isolated islet cells and disintegration of isolated whole islets is also obtained within 15 h by culture in the presence of the nitric oxide generating vasodilator sodium nitroprusside. We thus conclude that nitric oxide is extremely toxic for islet cells and that nitric oxide alone and in the absence of other macrophage-generated potentially toxic products can rapidly and completely kill islet cells.
Collapse
Affiliation(s)
- K D Kröncke
- Department of Medicine, Heinrich-Heine-University of Düsseldorf, FRG
| | | | | | | |
Collapse
|
43
|
Guberski DL. Diabetes-Prone and Diabetes-Resistant BB Rats: Animal Models of Spontaneous and Virally Induced Diabetes Mellitus, Lymphocytic Thyroiditis, and Collagen-Induced Arthritis. ILAR J 1993. [DOI: 10.1093/ilar.35.2.29] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
44
|
Burkart V, Koike T, Brenner HH, Kolb H. Oxygen radicals generated by the enzyme xanthine oxidase lyse rat pancreatic islet cells in vitro. Diabetologia 1992; 35:1028-34. [PMID: 1473612 DOI: 10.1007/bf02221677] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The endothelium-associated enzyme xanthine oxidase is known to generate reactive oxygen intermediates which may damage the surrounding tissue. We investigated whether reactive oxygen intermediates released by xanthine oxidase exert a toxic effect on isolated rat islet cells. The xanthine oxidase (25 mU/ml)/hypoxanthine (0.5 mmol/l) system released reactive oxygen intermediates in vitro as detected by luminol in a chemiluminescence analysing system. The addition of nicotinamide inhibited the release of reactive oxygen intermediates in a dose-dependent manner (50% inhibition at 20 mmol/l). Exposure of islet cells to enzyme generated reactive oxygen intermediates caused lysis of 39% of the cells within 15 h. Monitoring the mitochondrial function of islet cells by the conversion of tetrazolium bromide to its formazan product revealed a significant reduction of the respiratory activity down to 51% of that of the controls by 30 min after the initiation of the xanthine oxidase reaction. Mitochondrial dysfunction preceded plasma membrane damage. The addition of nicotinamide, a radical scavenger and inhibitor of the DNA repair enzyme poly(ADP-ribose) synthetase protected the islet cells from lysis and partially preserved their mitochondrial activity in the presence of reactive oxygen intermediates. We conclude that activation of the endothelial enzyme xanthine oxidase, known to be induced by mediators of immune cells or by episodes of ischaemia and reperfusion causes islet cell damage with subsequent cell death in early phases of pancreatic islet cell destruction.
Collapse
Affiliation(s)
- V Burkart
- Diabetes Research Institute, University of Düsseldorf, FRG
| | | | | | | |
Collapse
|
45
|
Affiliation(s)
- A Lernmark
- Department of Medicine, University of Washington, Seattle
| |
Collapse
|
46
|
|
47
|
Papaccio G, Esposito V. Ultrastructural observations on cytotoxic effector cells infiltrating pancreatic islets of low-dose streptozocin treated mice. VIRCHOWS ARCHIV. A, PATHOLOGICAL ANATOMY AND HISTOPATHOLOGY 1992; 420:5-10. [PMID: 1531718 DOI: 10.1007/bf01605977] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The aim of this study was to observe the ultrastructural events, during the onset of diabetes mellitus in the low-dose streptozocin (LDS)-treated mouse model with emphasis on the infiltrating elements. Forty male C57 BL/6J mice were given 40 mg/streptozocin on 5 consecutive days and killed 5, 6, 7, 8, 9, 10, 15, and 18 days after the first injection. Results demonstrated that islet infiltration occurring in LDS-treated mice is characterized by a very early pre-infiltration state in which mononuclear phagocytes in islet capillary vessels were considerably increased in number. A new histopathological time sequence for the early insulitis is described, in which attraction of blood mononuclear phagocytes into the islet capillary lumen is the first step. During the successive stage, occurring on days 6-8 we observed that mononuclear phagocytes migrate through capillary and venule walls into the islet parenchyma, where they differentiate into tissue macrophages. It was only later (step 3) that these macrophages acquired novel properties, typical of their "activated state" and started to phagocytose islet beta-cell debris. These data suggest that during the pre-infiltration and early insulitis the mononuclear phagocyte system plays a key role in the onset of LDS diabetes.
Collapse
Affiliation(s)
- G Papaccio
- Institute of Anatomy, I School of Medicine, Naples, Italy
| | | |
Collapse
|
48
|
Jobe LW, Vertrees S, Wilson CA, Jacobs C, Wilson DL, Picha KS, Baker P, Lernmark A. In vivo effects of interleukin-1 beta on blood leukocytes in BB rats prone or resistant to diabetes. Autoimmunity 1992; 11:233-7. [PMID: 1581467 DOI: 10.3109/08916939209035160] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Previous studies have determined that daily low dose injections of the potent cytokine interleukin-1 beta (IL-1 beta) decreased the frequency of insulin-dependent diabetes mellitus (IDDM) in diabetes-prone (DP) BB rats. In contrast, high dose injections induced an earlier than normal onset. In this study we tested whether the effects of daily human recombinant IL-1 beta injections on leukocyte subsets were associated with its modulation of IDDM onset in BB rats. Prior to the onset of IDDM in DP BB rats, high dose IL-1 beta induced leukocytosis (P less than 0.05), neutrophilia (P less than 0.01), and monocytosis (P less than 0.001). At the onset of IDDM, lymphocyte (P less than 0.01) and neutrophil (P less than 0.001) numbers were increased in high dose treated DP rats but not in rats given saline or low dose IL-1 beta. In 60-day-old diabetes-resistant (DR) BB rats, neurophilia was induced by both low (P less than 0.05) and high (P less than 0.001) dose IL-1 beta without the development of IDDM. At 130 days of age, when the rats were killed, it was discovered that 14/22 (64%) IL-1 beta injected DR rats developed neutralizing IL-1 beta antibodies. Significantly lower neutrophil numbers were observed in high dose DR rats which developed IL-1 beta antibodies compared with those which did not (P = 0.032). Thus, neutrophilia was dissociated from high IL-1 beta acceleration of IDDM onset.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- L W Jobe
- Department of Medicine, University of Washington, Seattle 98195
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Fukazawa H, Baniassadi J, Wall JR. Tissue specific binding of lymphocytes to the thyroid gland of BB/W rats may be an early event in the development of thyroiditis. Autoimmunity 1992; 12:89-93. [PMID: 1617114 DOI: 10.3109/08916939209150314] [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: 12/27/2022]
Abstract
Thyroiditis occurs in about 50% of diabetic Bio Breeding/Worcester (BB/W) rats. In order to investigate the earliest stages of lymphocyte homing to the thyroid gland in the development of experimental autoimmune thyroiditis, we measured the amount of trapping of peripheral blood lymphocytes (PBL) from BB/W rats to the thyroid gland of syngeneic recipient animals. PBL, from donor normal or diabetic BB/W rats, labelled with 51Cr, were injected, i.v., into normal, "potentially diabetic" or diabetic BB/W recipients. After 24 hr the rats were sacrificed and the radioactivity of selected individual organs counted. Results were calculated as % binding of 51Cr-labelled PBL/unit weight of tissue and expressed as a binding index by comparing to binding to recipient blood lymphocytes. A significant binding index was taken as greater than 1.0. PBL from diabetic or normal donor BB/W rats were shown to bind significantly to the thyroid gland of 8 out of 19 "potentially diabetic" or diabetic recipient syngeneic rats, but to none of the normal (non diabetic) recipients tested. Sixty percent of "potentially diabetic" BB/W rats and 50% of diabetic rats at 3-4 months of age showed a significant level of binding of donor lymphocytes from syngeneic diabetic or normal animals to their thyroid gland, while, at 5-6 months the proportion of recipient rats giving positive tests was much less (17%). The source of the donor lymphocytes (diabetic, or normal) did not significantly influence the binding. Lymphocyte binding to pancreas was not significantly greater than to control tissues.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- H Fukazawa
- Thyroid Studies Centre, Montreal General Hospital, Quebec, Canada
| | | | | |
Collapse
|
50
|
Ko IY, Ihm SH, Yoon JW. Studies on autoimmunity for initiation of beta-cell destruction. VIII. Pancreatic beta-cell dependent autoantibody to a 38 kilodalton protein precedes the clinical onset of diabetes in BB rats. Diabetologia 1991; 34:548-54. [PMID: 1936657 DOI: 10.1007/bf00400271] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Autoantibody to a rat islet cell-protein of 38 kilodalton was detectable at around 30 days of age in the sera of diabetes-prone Biobreeding (DP-BB) rats by both immunoprecipitation and differential Western blotting methods. Anti-38 kilodalton islet cell autoantibody was not, however, observed in the sera from 5- to 20-day-old DP-BB rats. Over 90% of DP-BB rats in which the antibody was detected, eventually developed Type 1 (insulin-dependent) diabetes mellitus. The antibody disappeared within 2 weeks after diabetes onset. However, it was preserved in the sera of DP-BB rats which had been treated with silica to prevent insulitis. The anti-38 kilodalton islet cell autoantibody was not detected in sera from control Wistar Furth (WF) rats. The autoantibody also cross-reacted with a rat insulinoma (RINm5F) cell protein of 38 kilodalton, but did not react with protein from mouse fibroblast (L-929 cells), rat pituitary cells (GH3 cells), or normal rat lymphocytes. The production of the autoantibody appears to be pancreatic Beta-cell dependent, since the autoantibody disappears after almost complete depletion of Beta cells, but is consistently present as long as Beta cells remain. Identification of the Beta-cell dependent anti-38 kilodalton islet cell autoantibody, which cross-reacts with a rat insulinoma cell protein of 38 kilodalton and precedes the onset of Type 1 diabetes in BB rats, will be invaluable for study of the molecular nature of a target islet cell autoantigen associated with the induction of autoimmunity in DP-BB rats.
Collapse
Affiliation(s)
- I Y Ko
- Laboratory of Viral and Immunopathogenesis of Diabetes, Julia McFarlane Diabetes Research Centre, Calgary, Alberta, Canada
| | | | | |
Collapse
|