151
|
Baron JL, Reich EP, Visintin I, Janeway CA. The pathogenesis of adoptive murine autoimmune diabetes requires an interaction between alpha 4-integrins and vascular cell adhesion molecule-1. J Clin Invest 1994; 93:1700-8. [PMID: 7512990 PMCID: PMC294222 DOI: 10.1172/jci117153] [Citation(s) in RCA: 87] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
An adoptive transfer model of insulin-dependent diabetes mellitus (IDDM) in the nonobese diabetic mouse was used to examine the roles of alpha 4-integrin, vascular cell adhesion molecule 1 (VCAM-1); and intercellular adhesion molecule 1 (ICAM-1) in the pathogenesis of autoimmune diabetes. Antibodies specific for both alpha 4-integrin and one of its ligands, VCAM-1, were able to delay onset of diabetes and decrease the incidence of the disease in adoptive transfer studies. This blocking of disease was accompanied by a marked decrease in lymphocytic infiltration of the islets of Langerhans. Furthermore, these antibodies preferentially block entrance of CD4 T cells into the tissue. Antibodies specific for ICAM-1 had little effect on the onset or incidence of IDDM. Thus, we conclude that an alpha 4-integrin-VCAM-1 interaction is important in T cell entry into the islets of Langerhans and in the pathogenesis of IDDM. In addition, the cascade of events leading to T cell transit across endothelium may be different for CD4 and CD8 cells, and may differ depending on the endothelium involved. Our results support the more general conclusion that an alpha 4-integrin-VCAM-1 interaction may be crucial in allowing activated effector CD4T cells to leave the blood and enter tissue to clear infection.
Collapse
Affiliation(s)
- J L Baron
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06510
| | | | | | | |
Collapse
|
152
|
Chatenoud L, Thervet E, Primo J, Bach JF. Anti-CD3 antibody induces long-term remission of overt autoimmunity in nonobese diabetic mice. Proc Natl Acad Sci U S A 1994; 91:123-7. [PMID: 8278351 PMCID: PMC42898 DOI: 10.1073/pnas.91.1.123] [Citation(s) in RCA: 480] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Anti-CD3 monoclonal antibodies suppress immune responses by transient T-cell depletion and antigenic modulation of the CD3/T-cell receptor complex. Anti-CD3 treatment of adult nonobese diabetic (NOD) mice, a spontaneous model of T-cell-mediated autoimmune insulin-dependent diabetes mellitus, significantly inhibits the autoimmune process. Short-term low-dose anti-CD3 treatment (5 micrograms/day i.v. for 5 consecutive days) prevented the occurrence of an accelerated form of the disease induced by cyclophosphamide. More unexpectedly, when applied to adult NOD females within 7 days of the onset of full-blown diabetes, the same anti-CD3 regimen induced a complete remission of overt disease (i.e., a return to permanent normoglycemia) in 64-80% of mice. This remission was durable (> 4 months) and was not associated with the disappearance of insulitis (mononuclear cell infiltration of the islets). The immunosuppression was apparently specific for beta-cell-associated antigens, since mice showing anti-CD3-induced remission rejected histoincompatible skin grafts normally, whereas they did not destroy syngeneic islet grafts, unlike control untreated overtly diabetic NOD females. These results open major therapeutic perspectives. They strongly suggest that self-tolerance can be restored in adult mice once autoimmunity is fully established and confirm that this effect can be obtained by transient targeting of the CD3/T-cell receptor without massive T-cell debulking.
Collapse
Affiliation(s)
- L Chatenoud
- Institut National de la Santé et de la Recherche Médicale U 25, Hôpital Necker, Paris, France
| | | | | | | |
Collapse
|
153
|
Jaramillo A, Gill BM, Delovitch TL. Insulin dependent diabetes mellitus in the non-obese diabetic mouse: a disease mediated by T cell anergy? Life Sci 1994; 55:1163-77. [PMID: 7934613 DOI: 10.1016/0024-3205(94)00655-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The non-obese diabetic (NOD) mouse spontaneously develops autoimmune type I insulin-dependent diabetes mellitus (IDDM) with a similar immunopathological profile to the human disease. Development of the disease in both the NOD mouse and in humans is under polygenic control and influenced by many environmental factors. Diabetes results from a specific T cell-mediated destruction of pancreatic insulin-producing islet beta cells. Both CD4 and CD8 T cells as well as macrophages are required for the development of diabetes in NOD mice. An intriguing similarity between murine and human diabetes is a T cell proliferative unresponsiveness (anergy) that may be a susceptibility factor to disease onset. Defective communication between antigen-presenting cells (APC) and T cells, and/or an aberrant production or activity of inflammatory cytokines (e.g. chemokines) in the thymus and periphery (e.g. pancreas) may account for the unresponsiveness of regulatory T cells leading to a loss of immunological tolerance to beta cell autoantigens in NOD mice and in diabetic humans.
Collapse
Affiliation(s)
- A Jaramillo
- Banting and Best Department of Medical Research, University of Toronto, Ontario, Canada
| | | | | |
Collapse
|
154
|
Vardi P. Adjuvant administration modulates the process of beta-cell autoimmunity and prevents IDDM: introduction to human trials. DIABETES/METABOLISM REVIEWS 1993; 9:317-22. [PMID: 7924829 DOI: 10.1002/dmr.5610090412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- P Vardi
- National Center for Childhood Diabetes, Children's Medical Center of Israel, Petah-Tikva
| |
Collapse
|
155
|
Abstract
Although differential cytokine production has been best characterized in CD4+ T cells, it is becoming clear that CD8+ T cells may also be heterogeneous at the level of cytokine production, and that this determines whether they exhibit inflammatory- or suppressor-type properties. Compelling evidence has accumulated in the past few years that cytokines such as interleukin-4, interleukin-10 and transforming growth factor-beta may serve as regulators of cell-mediated immunopathologies by inhibiting the development or effector function of inflammatory T cells that produce cytokines such as interferon-gamma or lymphotoxin.
Collapse
Affiliation(s)
- A O'Garra
- DNAX Research Institute of Molecular and Cellular Biology, Inc., Palo Alto, California 94304-1104
| | | |
Collapse
|
156
|
Singer SM, Tisch R, Yang XD, McDevitt HO. An Abd transgene prevents diabetes in nonobese diabetic mice by inducing regulatory T cells. Proc Natl Acad Sci U S A 1993; 90:9566-70. [PMID: 8415742 PMCID: PMC47610 DOI: 10.1073/pnas.90.20.9566] [Citation(s) in RCA: 86] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Susceptibility to the human autoimmune disease insulin-dependent diabetes mellitus is strongly associated with particular haplotypes of the major histocompatibility complex (MHC). Similarly, in a spontaneous animal model of this disease, the nonobese diabetic (NOD) mouse, the genes of the MHC play an important role in the development of diabetes. We have produced transgenic NOD mice that express the class II MHC molecule I-Ad in addition to the endogenous I-Ag7 molecules in order to study the role of these molecules in the disease process. Although the inflammatory lesions within the islets of Langerhans in the pancreas appear similar in transgenic and nontransgenic animals, transgenic mice develop diabetes with greatly diminished frequency compared to their nontransgenic littermates (10% of transgenic females by 30 weeks of age compared to 45% of nontransgenic females). Furthermore, adoptive transfer experiments show that T cells present in the transgenic mice are able to interfere with the diabetogenic process caused by T cells from nontransgenic mice. Thus, the mechanism by which I-Ad molecules protect mice from diabetes includes selecting in the thymus and/or inducing in the periphery T cells capable of inhibiting diabetes development.
Collapse
Affiliation(s)
- S M Singer
- Department of Microbiology, Stanford University Medical Center, CA 94305
| | | | | | | |
Collapse
|
157
|
Katz JD, Wang B, Haskins K, Benoist C, Mathis D. Following a diabetogenic T cell from genesis through pathogenesis. Cell 1993; 74:1089-100. [PMID: 8402882 DOI: 10.1016/0092-8674(93)90730-e] [Citation(s) in RCA: 564] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Nonobese diabetic (NOD) mice spontaneously develop a disease very similar to type 1 diabetes in humans. We have generated a transgenic mouse strain carrying the rearranged T cell receptor genes from a diabetogenic T cell clone derived from a NOD mouse. Self-reactive T cells expressing the transgene-encoded specificity are not tolerized in these animals, resulting in rampant insulitis and eventually diabetes. Features of the disease process emphasize two so-called check-points, recognized previously in the NOD and human diseases but easily misinterpreted. Although NOD mice are protected from insulitis and diabetes by expression of the E molecule encoded in the major histocompatibility complex, the transgenics are not, permitting us to exclude some possible mechanisms of protection.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Flow Cytometry
- Gene Rearrangement, T-Lymphocyte
- Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Mice
- Mice, Inbred NOD/immunology
- Mice, Transgenic
- Molecular Sequence Data
- Oligodeoxyribonucleotides
- Pancreas/immunology
- Pancreas/pathology
- Polymerase Chain Reaction/methods
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Submandibular Gland/immunology
- Submandibular Gland/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- J D Katz
- Laboratoire de Génétique Moléculaire des Eucaryotes Centre National de la Recherche Scientifique Unité 184 de Biologie Moléculaire l'Institut National de la Santé et de la Recherche Médicale
| | | | | | | | | |
Collapse
|
158
|
|
159
|
Chosich N, Harrison LC. Suppression of diabetes mellitus in the non-obese diabetic (NOD) mouse by an autoreactive (anti-I-Ag7) islet-derived CD4+ T-cell line. Diabetologia 1993; 36:716-21. [PMID: 8405738 DOI: 10.1007/bf00401141] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The non-obese diabetic (NOD) mouse is a spontaneous model of human insulin-dependent diabetes mellitus. Both CD4+ and CD8+ T cells infiltrate the pancreatic islets of NOD mice prior to beta-cell destruction. T-cell lines isolated from the islets of NOD mice are tools for studying the pathogenesis of insulin-dependent diabetes mellitus. During attempts to generate such lines we isolated an autoreactive CD4+ T-cell line, designated C2, from the 'insulitis' lesion of a 20-week-old female non-diabetic NOD/WEHI mouse. Islet T cells were propagated by the addition of interleukin-2 and reexposure every 2 weeks to whole NOD islets and irradiated NOD spleen cells as antigen presenting cells. C2 cells proliferated up to 100-fold upon exposure to NOD antigen presenting cells but did not respond to whole NOD islets or antigen presenting cells from allogeneic mouse strains. Proliferation of C2 cells to NOD antigen presenting cells was blocked by a monoclonal antibody against the unique class II MHC molecule of NOD, I-Ag7. In response to NOD antigen presenting cells, C2 cells secreted interferon-gamma, tumour necrosis factor-alpha and interleukin-6 but no detectable interleukin-2, interleukin-4 or interleukin-10, a pattern of cytokine secretion more characteristic of Th1 CD4 cells. C2 cells displayed significant cytotoxicity in a redirected lysis assay. To explore a possible role for autoreactive T cells in the pathogenesis of autoimmune diabetes, C2 cells were injected i.v. into female NOD mice that had received cyclophosphamide to accelerate development of diabetes.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- N Chosich
- Burnet Clinical Research Unit, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | | |
Collapse
|
160
|
Morrissey PJ, Charrier K, Braddy S, Liggitt D, Watson JD. CD4+ T cells that express high levels of CD45RB induce wasting disease when transferred into congenic severe combined immunodeficient mice. Disease development is prevented by cotransfer of purified CD4+ T cells. J Exp Med 1993; 178:237-44. [PMID: 8100269 PMCID: PMC2191069 DOI: 10.1084/jem.178.1.237] [Citation(s) in RCA: 367] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Purified CD4+ lymph node T cells were sorted into two populations on the basis of their expression of CD45RB (CD45RBhi and CD45RBlo) and injected into congenic severe combined immunodeficient (SCID) mice. After a period of time that was dependent on the number of cells injected, the SCID mice that received CD45RBhi/CD4+ T cells developed a wasting disease that was not seen in SCID mice that received the CD4+/CD45RBlo cells or whole lymph node cells. At death, SCID mice that received the CD4+/CD45RBhi cells had increased spleen and lymph node cellularity compared with normal SCID mice and SCID mice that received the CD4+/CD45RBlo T cells. The spleen and lymph node contained CD4+ cells and neither CD8+ nor surface immunoglobulin M-positive cells, plus a population of cells that did not express any of those markers. At necropsy, the SCID mice that received the CD4+/CD45RBhi cells had significant hyperplasia of the intestinal mucosa with significant lymphoid cell accumulation in the lamina propria. Interestingly, mice that received mixtures of whole lymph node or purified CD4+ cells with CD4+/CD45RBhi cells did not develop weight loss, indicating that the unseparated CD4+ population contained cells that were capable of regulating the reactivity of the CD4+/CD45RBhi cells.
Collapse
Affiliation(s)
- P J Morrissey
- Department of Immunology, Immunex Research and Development Corp., Seattle, Washington 98101
| | | | | | | | | |
Collapse
|
161
|
Abstract
There is no doubt that T cells play a key role in the pathogenesis of autoimmune diseases (AD) both as effector and regulatory cells. Despite spectacular progress in the understanding of natural tolerance to self, owing particularly to transgenic technology, important questions remain open regarding the pathogenesis of AD, the conditions favoring the transition from benign or 'physiological' autoimmunity to deleterious autoimmunity, and the precise effector mechanisms. This review on the cellular basis of T-cell-mediated AD begins with an enumeration of the main arguments in favor of direct T-cell involvement, special emphasis being given to two animal models which have been most extensively investigated: experimental allergic encephalomyelitis, and the nonobese diabetic mouse. The question as to whether pathogenic T cells use a restricted repertoire of V beta genes is examined in the context of these two models. From here we proceed to an evaluation of the mechanisms of onset of AD, discussing both extrinsic and intrinsic factors responsible for the breakdown in T-cell tolerance and reviewing the arguments in favor of suppressor T cells being actively involved in the prevention of autoimmunity. The last two sections are devoted to the effector mechanisms responsible for tissue injury in organ-specific AD and to T-cell-directed therapeutic interventions, respectively. We discuss the two main pathogenic hypotheses based on direct intervention of cytotoxic T cells or indirect involvement of inflammatory cytokines and macrophages, and evaluate the importance of ecotaxis in leading autoreactive T cells to the site of injury. We conclude on a brief and nonexhaustive list of strategies aimed at selectively neutralizing potentially harmful T cells.
Collapse
Affiliation(s)
- C Carnaud
- INSERM U 25, Hôpital Necker, Paris, France
| | | |
Collapse
|
162
|
Abstract
For almost a century self-nonself discrimination has been considered the driving force of the immune system and the dogma of self-tolerance (horror autotoxicus) the essential issue for understanding protective immunity and pathologic autoreactivity. This classical picture has been recently challenged by the discovery that the immune system is influenced by internal activation (amor autocognitus) and autoreactive clones are both present and activated in healthy individuals. Central to the concepts of reactivity and tolerance, in other terms, to physiology and pathology, is the analysis of structures involved in immunorecognition and as they contribute to the outcome of the immune response. The capacity of the immune system to recognize nonself structures and eventually react to them developed during evolution probably before the generation of clonally distributed antigen receptors. The high level of autoreactivity that characterizes the immune system seems to suggest that immunocompetent cells never learned to ignore the self, but rather that, sometime during evolution, they specialized in the function of self-identification, one of the most basic activities of all living cells. Autoimmune diseases would result from a pathologic deviation of a physiologic function, just as many other diseases do.
Collapse
Affiliation(s)
- P del Guercio
- Department of Medicine, University of California at San Diego, La Jolla 92093-0961
| |
Collapse
|
163
|
Abstract
The immune system must not only fight off infections, but also ensure that it does not react against its own body tissues. Since clones of lymphocytes have predetermined reactivities, some will be self-reactive and have the potential to cause damage. They should therefore be neutralized in some way. In a system as complex and important as that governing self-tolerance, many mechanisms must exist to neutralize autoaggressive lymphocytes. They may be classified under two main groups. In one the tolerant state arises from the physical or functional silencing of potentially autoaggressive lymphocytes after antigen encounter. This may involve clonal deletion, clonal abortion or clonal anergy. In the second, regulatory mechanisms of the immune system itself may hold autoreactive lymphocytes in check, for example through the operation of idiotypic network interactions and the action of specialized suppressor cells. Much evidence has accumulated for the physical deletion of autoreactive T cells as they mature in the thymus. The fate of any that escape thymus censorship has been the subject of recent research and is discussed here. Under certain conditions, self-tolerance must also be imposed at the B-cell level to prevent the production of potentially damaging autoantibodies. Although the mechanisms which silence self-reactive lymphocytes are very efficient, self-tolerance can break down, and autoimmunity will thus ensue. The main factors responsible for this are briefly described here.
Collapse
Affiliation(s)
- J F Miller
- Walter and Eliza Hall Institute of Medical Research, Royal Melbourne Hospital, Vic., Australia
| |
Collapse
|
164
|
Honeyman MC, Harrison LC. The immunologic insult in type 1 diabetes. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1993; 14:253-74. [PMID: 8438209 DOI: 10.1007/bf00195977] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- M C Honeyman
- Burnet Clinical Research Unit, Walter and Eliza Hall Institute of Medical Research, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | |
Collapse
|
165
|
Shehadeh NN, Gill RG, Lafferty KJ. Mechanism of self-tolerance to endocrine tissue. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1993; 14:203-20. [PMID: 8438207 DOI: 10.1007/bf00195974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- N N Shehadeh
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Denver 80262
| | | | | |
Collapse
|
166
|
Rashba EJ, Reich EP, Janeway CA, Sherwin RS. Type 1 diabetes mellitus: an imbalance between effector and regulatory T cells? Acta Diabetol 1993; 30:61-9. [PMID: 8219259 DOI: 10.1007/bf00578215] [Citation(s) in RCA: 9] [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
Abundant evidence now exists that autoimmunity plays a critical role in the pathogenesis of type 1 (insulin-dependent) diabetes mellitus. The non-obese diabetic (NOD) mouse is an extensively studied animal model of this T-cell-mediated autoimmune disease. Our laboratory has focused on isolating diabetogenic T cell clones from NOD mice as a means of elucidating the pathogenesis of type 1 diabetes. This experimental approach presupposes that type 1 diabetes in NOD mice results from the action of islet-reactive T cells that are not present in other mouse strains; the diabetogenic T cells would therefore represent "forbidden clones" which exist in NOD mice as a result of a failure of clonal deletion. While the inappropriate presence of diabetogenic T cells probably plays a central role in murine diabetes, it cannot explain all aspects of the disease. Type 1 diabetes is a chronic disorder with a lengthy preclinical stage; if the diabetogenic T cells acted in an unopposed fashion, one might expect to see a much more fulminant clinical course. This observation suggests that regulatory influences are likely to exist in this disease--a possibility supported by recent experimental data. If these regulatory influences could be identified and enhanced, specific immunotherapy for type 1 diabetes could be achieved.
Collapse
Affiliation(s)
- E J Rashba
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06510
| | | | | | | |
Collapse
|
167
|
Boitard C, Timsit J, Larger E, Sempé P, Bach JF. Pathogenesis of IDDM: immune regulation and induction of immune tolerance in the NOD mouse. Autoimmunity 1993; 15 Suppl:12-3. [PMID: 8218842 DOI: 10.3109/08916939309008852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- C Boitard
- INSERUM U25, Hôpital Necker, Paris, France
| | | | | | | | | |
Collapse
|
168
|
Boitard C. The differentiation of the immune system towards anti-islet autoimmunity. Clinical prospects. Diabetologia 1992; 35:1101-12. [PMID: 1478361 DOI: 10.1007/bf00401362] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- C Boitard
- Service d'Immunologie Clinique, INSERM 25, Hôpital Necker, Paris, France
| |
Collapse
|
169
|
Abstract
The demonstration that functionally different T-cell subsets can be defined by the isoforms of the leukocyte-common antigen, CD45, that they express, has prompted studies on the roles of these subsets in autoimmunity. The results have led to the identification of a particular subset of CD4+ T cells that have the ability to inhibit autoimmune disease. Further, it has been shown that diabetes in the B-B rat can be transferred by in vitro activation of T cells by Staphylococcal enterotoxin suggesting that superantigens may play a role in the pathogenesis of this disease. However, in this system too, it appears that a subset of T cells can inhibit the induction of autoaggressive cells. In other experimental autoimmune diseases there is evidence that CD8+ T cells can be protective and that these cells may mediate this protection by the synthesis of transforming growth factor-beta.
Collapse
Affiliation(s)
- D Mason
- MRC Cellular Immunology Unit, Sir William Dunn School of Pathology, Oxford, UK
| | | |
Collapse
|
170
|
Jackman SH, Boyse EA, Goldberg EH. Adoptive transfer of skin-selective autoimmunity induced by Skn alloantigenic disparities. Proc Natl Acad Sci U S A 1992; 89:11041-5. [PMID: 1438311 PMCID: PMC50479 DOI: 10.1073/pnas.89.22.11041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Two unlinked genes of the mouse, Skn-1 and Skn-2, each with alterative alleles, specify alternative cell-surface Skn alloantigens expressed only by epidermal and neural cells. C57BL/6 (B6) and A/J (A) strain mice differ at both Skn loci. Thus lethally irradiated B6 mice restored with (B6 x A)F1 hybrid hematopoietic cells [(B6 x A)/B6 chimeras] reject A strain (Skn-incompatible) skin grafts. Our studies were designed primarily to test the inference that (B6 x A)F1 lymphoid cells, after differentiating in B6 recipients, which lack the Skn alloantigens of A strain mice, may make an Skn-related, skin-selective autoimmune response when returned to their native (B6 x A)F1 habitat. Severe cutaneous lesions did, indeed, ensue after spleen cells of (B6 x A)/B6 chimeras were transferred to (B6 x A)F1 recipients, provided that three conditions were met--namely, (i) priming of the (B6 x A)/B6 chimeric donor by grafting and rejection of Skn-incompatible A strain skin grafts, (ii) stimulation of the recipient's skin as from shaving, at which sites the lesions were mainly located, and (iii) pretreatment of the (B6 x A)F1 recipients with cyclophosphamide or sublethal irradiation. Spleen cells of control female chimeras primed by grafting and rejection of H-Y (Skn-compatible) B6 male skin failed to incite the Skn-typical cutaneous lesions in (B6 x A)F1 recipients, indicating that these lesions were Skn-specific and not a nonspecific consequence of incompatible skin grafting per se. Normally compatible A strain skin grafts, but not Skn-compatible B6 skin grafts, were rejected by cyclophosphamide-treated (B6 x A)F1 recipients of (B6 x A)/B6 spleen cells from Skn-primed chimera donors. Treatment of primed chimeras' spleen cells with antiserum to H-2a (A strain) specifically abolished their capacity to adoptively incite the Skn-related autoimmune syndrome, confirming that the immune cells responsible are of (B6 x A)F1 origin and are not residual B6 derivatives. These findings add weight to the status of Skn systems as agents of tissue-selective histoincompatibility and, perhaps, of clinical disorders with a known or suspected autoimmune basis affecting the skin.
Collapse
Affiliation(s)
- S H Jackman
- Department of Microbiology, Marshall University, School of Medicine, Huntington, WV 25755
| | | | | |
Collapse
|
171
|
Affiliation(s)
- B O Roep
- Department of Immunohaematology and Blood Bank, University Hospital Leiden, The Netherlands
| | | |
Collapse
|
172
|
Strandell E, Sandler S, Boitard C, Eizirik DL. Role of infiltrating T cells for impaired glucose metabolism in pancreatic islets isolated from non-obese diabetic mice. Diabetologia 1992; 35:924-31. [PMID: 1451948 DOI: 10.1007/bf00401420] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Pancreatic islets isolated from non-obese diabetic (NOD) mice, all of which have insulitis, exhibit an impaired glucose metabolism. In order to investigate the role of infiltrating lymphocytes for this altered metabolism, we injected 12- to 13-week-old female NOD mice with monoclonal antibodies directed against either the alpha beta-T cell receptor, CD4+ or CD8+ T cells. Control NOD mice were injected with normal rat IgG or with the vehicle (phosphate buffered saline) alone. Injection of the three different monoclonal antibodies markedly reduced the mononuclear cell infiltration. An intravenous glucose tolerance test showed no differences between the groups. Islet insulin release in response to glucose was similar in all groups. In contrast, islets isolated from the control NOD mice with insulitis showed a high basal (1.7 mmol/l glucose) glucose oxidation rate and a small increase in the glucose oxidation rate in response to a high glucose concentration (16.7 mmol/l glucose). The monoclonal antibodies counteracted the elevated basal glucose oxidation rate of the islets. Parallel studies of stimulated mononuclear cells suggested that the contribution of glucose oxidized by islet-infiltrating lymphocytes could only partially explain the observed alterations in NOD mouse islet metabolism. Culture of islets obtained from NOD mice in the presence of the cytokine interleukin-1 beta induced a similar pattern of glucose metabolism as seen earlier in IgG or phosphate-buffered saline treated control NOD mice. In conclusion, alterations in the glucose oxidation rates seem to be an early sign of disturbance in islets isolated from NOD mice. These early alterations in glucose metabolism can be reversed in vivo by monoclonal antibodies directed against effector lymphocytes.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- E Strandell
- Department of Medical Cell Biology, Uppsala University, Sweden
| | | | | | | |
Collapse
|
173
|
Díaz-Gallo C, Moscovitch-Lopatin M, Strom TB, Kelley VR. An anergic, islet-infiltrating T-cell clone that suppresses murine diabetes secretes a factor that blocks interleukin 2/interleukin 4-dependent proliferation. Proc Natl Acad Sci U S A 1992; 89:8656-60. [PMID: 1528876 PMCID: PMC49979 DOI: 10.1073/pnas.89.18.8656] [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] Open
Abstract
The mechanism of peripheral immunological tolerance has not been fully established. While anergic T cells have been noted in tolerant hosts, the mechanism by which they contribute to the induction and maintenance of tolerance has not been defined. As we previously reported, an accelerated form of diabetogenic autoimmunity in nonobese diabetic mice can be blocked by passive transfer of a CD3+, CD8+, beta-chain variable region 11-positive islet-infiltrating T-cell clone (IS-2.15). In this report we examine the properties of this T-cell clone. We have established that this clone is unresponsive to mitogenic concentrations of anti-T-cell receptor or anti-CD3 monoclonal antibodies and is only weakly responsive to syngeneic islet and spleen cells. Moreover, these T cells secrete an inhibitory factor(s) that irreversibly inhibits interleukin (IL) 2/IL-4-driven proliferation of IL-2/IL-4 indicator T-cell lines. This noncytotoxic factor, which possesses an apparent size of 10-30 kDa, does not interfere with low-affinity IL-2 receptor expression. These data indicate that at least some anergic T cells can play an active role in peripheral tolerance by secreting suppressor factor(s) that regulate IL-2/IL-4-dependent proliferation.
Collapse
Affiliation(s)
- C Díaz-Gallo
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | | | | | | |
Collapse
|
174
|
Clare-Salzler MJ, Brooks J, Chai A, Van Herle K, Anderson C. Prevention of diabetes in nonobese diabetic mice by dendritic cell transfer. J Clin Invest 1992; 90:741-8. [PMID: 1522229 PMCID: PMC329925 DOI: 10.1172/jci115946] [Citation(s) in RCA: 164] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The purpose of this study was to determine the effect of dendritic cell (DC) transfers on the incidence of diabetes in female nonobese diabetic (NOD) mice. Groups of 4-wk-old NOD female mice were given a single foot pad of DCs (70-90% purity) isolated from the draining lymph nodes (LN) of the pancreas (PLN), the cervical LNs, or the axillary/inguinal LNs. In addition, other groups of NOD mice received purified spleen DCs, purified PLN T cells (the major contaminating population in DC preparations), or the injection vehicle PBS. All groups were monitored for diabetes for one year. Significant protection from diabetes was observed in NOD mice receiving greater than 1 x 10(4) PLN DCs in comparison to mice receiving other DCs populations, PLN T cells, or PBS (P less than 0.05). The pancreata of NOD mice that received PLN DCs demonstrated significantly lower levels of lymphocytic infiltration in the islets that age-sex matched nondiabetic female NOD control mice (P less than 0.05). LN cells from nondiabetic NOD mice that received PLN DC protected irradiated female recipients from the adoptive transfer of diabetes to a greater degree than LN cells from age and sex matched nondiabetic female NOD mice that did not receive PLN DC transfers at 36 d (P = 0.014) and at 1 yr (P = 0.0015) after transfer. These data suggest that the PLN DC transfers are able to modulate autoimmunity and limit diabetes expression in the NOD mouse. PLN DCs transfers may regulate autoimmunity by the induction of regulatory cells.
Collapse
|
175
|
Pankewycz OG, Guan JX, Benedict JF. A protective NOD islet-infiltrating CD8+ T cell clone, I.S. 2.15, has in vitro immunosuppressive properties. Eur J Immunol 1992; 22:2017-23. [PMID: 1386312 DOI: 10.1002/eji.1830220810] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Type 1, insulin-dependent diabetes mellitus (IDDM) appears to result from a T cell-dependent destruction of insulin-producing pancreatic beta cells. In non-obese diabetic (NOD) mice and in other rodent models of human IDDM, final expression of disease may be controlled by protective, as well as, destructive T cell influences. Previously, a CD8+ T cell clone, I.S. 2.15, was isolated directly from islets of disease-resistant male NOD mice. Upon transfer to young NOD recipients, the non-cytolytic I.S. 21.5 T cell clone, confers in vivo protection from two forms of accelerated IDDM. The present study demonstrates that I.S. 2.15 T cells induce in vitro immunosuppression. The suppressive effects of I.S. 2.15 T cells are mediated through soluble factor(s) and are independent of T cell activation, cell contact, antigen specificity or the major histocompatibility complex (MHC). By polymerase chain reaction (PCR), I.S. 2.15 T cells contain mRNA species encoding for the potentially immunosuppressive cytokines, interleukin-10 (IL-10) and transforming growth factor-beta (TGF-beta). The T cell suppressive effects engendered by I.S. 2.15 T cells closely mimic those observed with TGF-beta. Moreover, I.S. 2.15-induced immunosuppression correlates with intracellular levels of TGF-beta mRNA. These results establish that immunoregulatory T cells are present within islets in IDDM-resistant NOD mice and may impact on final disease expression through the production of soluble mediator(s).
Collapse
Affiliation(s)
- O G Pankewycz
- Department of Medicine, University of Virginia, Charlottesville 22908
| | | | | |
Collapse
|
176
|
Hutchings P, O'Reilly L, Parish NM, Waldmann H, Cooke A. The use of a non-depleting anti-CD4 monoclonal antibody to re-establish tolerance to beta cells in NOD mice. Eur J Immunol 1992; 22:1913-8. [PMID: 1623929 DOI: 10.1002/eji.1830220735] [Citation(s) in RCA: 94] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The use of immunosuppressive drugs in the management of autoimmunity penalizes a large part of the immune system for the misdemeanors of a small minority of T cells. An ideal form of therapy would be one in which it were possible to render the immune system tolerant of the inciting antigens with minimal effects on other responses. We here show that it is possible to re-establish self tolerance in an animal model of insulin-dependent diabetes mellitus without prior deletion of CD4+ T cells using a short course of therapy with a non-lytic monoclonal antibody to the CD4 adhesion receptor on T cells. This tolerance can be achieved even when diabetogenic cells are already in the pancreas. Primary responses to antigens given after therapy has ceased are normal and secondary responses to antigens seen prior to, but not during, the period of antibody therapy can remain unaffected. This suggests that intervention with selected CD4 antibodies may have significant advantages over and above that provided not only by conventional immunosuppression but also over that provided by a depleting antibody.
Collapse
Affiliation(s)
- P Hutchings
- Department of Pathology, University of Cambridge, GB
| | | | | | | | | |
Collapse
|
177
|
Maki T, Ichikawa T, Blanco R, Porter J. Long-term abrogation of autoimmune diabetes in nonobese diabetic mice by immunotherapy with anti-lymphocyte serum. Proc Natl Acad Sci U S A 1992; 89:3434-8. [PMID: 1565635 PMCID: PMC48882 DOI: 10.1073/pnas.89.8.3434] [Citation(s) in RCA: 91] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
We investigated the therapeutic effect of anti-lymphocyte serum (ALS) on clinically overt diabetes by using a nonobese diabetic (NOD) mouse model of type I diabetes mellitus. ALS given within 14 days of disease onset gradually reversed hyperglycemia with a 76% cumulative incidence of remission. Combined use of anti-CD4 and anti-CD8 monoclonal antibodies, but not anti-CD4 or anti-CD8 antibody alone, was also effective with overall 64% remission. Diabetic NOD mice that failed to respond to ALS treatment accepted subsequent islet isografts for a prolonged period (mostly greater than 100 days), whereas islet isografts in diabetic NOD mice previously treated with normal rabbit serum were all destroyed as acutely as isografts in untreated diabetic NOD mice. These results suggest that persistence of diabetes was due to irreversible beta-cell destruction and that ALS has indeed abrogated autoimmunity. In addition, ALS treatment at the time of islet isografting achieved significant prolongation of graft survival with 8 of 13 mice maintaining euglycemia for greater than 100 days. Although ALS prolonged islet allograft survival in diabetic NOD mice, the degree of prolongation was much less for allografts than for isografts, suggesting that ALS is capable of suppressing autoimmunity more effectively than allograft responses.
Collapse
Affiliation(s)
- T Maki
- Department of Surgery, New England Deaconess Hospital, Boston, MA 02215
| | | | | | | |
Collapse
|
178
|
Itoh M, Mukasa A, Tokunaga Y, Hiramine C, Hojo K. Suppression of efferent limb of testicular autoimmune response by a regulatory CD4+ T cell line in mice. Clin Exp Immunol 1992; 87:455-60. [PMID: 1347494 PMCID: PMC1554346 DOI: 10.1111/j.1365-2249.1992.tb03019.x] [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: 12/01/2022] Open
Abstract
A murine T cell line (designated as C.Ts) as a mediator of suppression of experimental autoimmune orchitis (EAO) was established. The method of establishment of C.Ts cell line was preparing spleen cells from C3H/He mice hyperimmunized with testicular germ cells (TC) and the repeated selection of the lymphocytes in vitro by stimulation with mouse testicular antigens (mTA). The C.Ts cells were Thy1.2+, surface immunoglobulin-, CD3+, CD4+ and CD8-. The cells could suppress the induction of EAO when transferred into actively EAO-sensitized mice only at the pre-clinical stage of the disease (efferent limb of the autoimmune response). The transferred C.Ts cells significantly inhibited both cellular and humoral immune responses to TC in the recipients in an antigen-specific manner. The disease suppression by C.Ts cells was found to depend upon their cell number, and their suppressive activity was markedly augmented by in vitro stimulation with mTA.
Collapse
Affiliation(s)
- M Itoh
- Department of Immunology and Immunopathology, Kagawa Medical School, Japan
| | | | | | | | | |
Collapse
|
179
|
Sheil JM, Shepherd SE, Klimo GF, Paterson Y. Identification of an autologous insulin B chain peptide as a target antigen for H-2Kb-restricted cytotoxic T lymphocytes. J Exp Med 1992; 175:545-52. [PMID: 1370687 PMCID: PMC2119120 DOI: 10.1084/jem.175.2.545] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
We have examined the CD8+ peripheral T cell repertoire of C57BL/6 (H- 2b) mice for cytotoxic T lymphocyte (CTL) reactivities to insulin, using in vitro immunization with a chymotryptic digest of reduced bovine insulin. The results presented in this study demonstrate that potentially autoreactive H-2Kb-restricted cytotoxic T cells specific for an autologous insulin B chain peptide are present in the preimmune splenic T cell repertoire. The immunogenic peptide comprises residues 7- 15 from the insulin B chain and has features in common with naturally processed Kb-restricted peptides identified by others. The minimal peptide sequence recognized by these cytotoxic T cells is 10-15, which is highly conserved in mammalian species and constitutes a self-peptide in mice. The presence of class I major histocompatibility complex- restricted CTLs with potentially autoreactive specificities in preimmune animals raises the possibility of a role for such cells in autoimmune disease states. Possible mechanisms for the in vivo expansion of insulin peptide-specific CTLs are discussed.
Collapse
Affiliation(s)
- J M Sheil
- Department of Microbiology and Immunology, West Virginia University Health Sciences Center, Morgantown 26506
| | | | | | | |
Collapse
|
180
|
MESH Headings
- Adjuvants, Immunologic/therapeutic use
- Animals
- Autoantigens/immunology
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Autoimmune Diseases/pathology
- Autoimmune Diseases/therapy
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Cytokines/therapeutic use
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Disease Susceptibility/immunology
- Female
- Genes, MHC Class I
- Genes, MHC Class II
- Genetic Markers
- Genetic Predisposition to Disease
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Immunosuppressive Agents/therapeutic use
- Macrophages/immunology
- Macrophages/pathology
- Male
- Mice
- Mice, Inbred NOD/genetics
- Mice, Inbred NOD/metabolism
- Mice, Transgenic/immunology
- Rats
- Rats, Inbred BB/genetics
- Rats, Inbred BB/immunology
- Streptozocin
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- H Kikutani
- Institute for Molecular and Cellular Biology, Osaka University, Japan
| | | |
Collapse
|
181
|
Uno M, Miyazaki T, Uehira M, Nishimoto H, Kimoto M, Miyazaki J, Yamamura K. Complete prevention of diabetes in transgenic NOD mice expressing I-E molecules. Immunol Lett 1992; 31:47-52. [PMID: 1548033 DOI: 10.1016/0165-2478(92)90009-d] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Previously, we showed that transgenic expression of the MHC (major histocompatibility complex) class II I-E molecules prevented insulitis in non-obese diabetic (NOD) mice at the age of 19 weeks. To rule out the possibility that the I-E expression merely delays the onset of insulitis, we have further characterized the expression and function of the I-E molecule expressed in transgenic NOD mice and confirmed our previous observations. Northern blot analysis showed that the transgenic E alpha d gene was expressed in a pattern similar to the endogenous E alpha d gene in BALB/c mice. The newly expressed I-E molecules were recognized as an alloantigen by the T lymphocytes of normal NOD mice as shown by mixed lymphocyte reaction (MLR). Transgenic NOD mice were resistant to the treatment by cyclophosphamide, which effectively induces diabetes in normal NOD mice, and did not develop diabetes up to 40 weeks of age. On the basis of these findings, we discuss the role of I-E molecules in the prevention of diabetes in NOD mice.
Collapse
Affiliation(s)
- M Uno
- Institute for Medical Genetics, Kumamoto University Medical School, Japan
| | | | | | | | | | | | | |
Collapse
|
182
|
Boitard C, Villa MC, Becourt C, Gia HP, Huc C, Sempe P, Portier MM, Bach JF. Peripherin: an islet antigen that is cross-reactive with nonobese diabetic mouse class II gene products. Proc Natl Acad Sci U S A 1992; 89:172-6. [PMID: 1729686 PMCID: PMC48198 DOI: 10.1073/pnas.89.1.172] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The nonobese diabetic (NOD) mouse, in which major histocompatibility complex genes may be involved in the susceptibility to diabetes, has been developed as a model of autoimmune diabetes. The NOD mouse expresses I-A-encoded class II major histocompatibility complex antigens, which differ from those of other mouse haplotypes by the presence of a serine at position 57 of the A beta chain. Identifying islet autoantigens may help elucidate the role of class II antigens in the activation of autoreactive T cells and, thus, in the development of diabetes. We have detected autoantibodies directed against a 58-kDa islet cell antigen in NOD mice but not in other strains, including lupus-prone mice. Apart from insulin-secreting cells, the 58-kDa antigen was only found to be expressed by neuroblastoma cells and was identified as peripherin, an intermediate filament protein previously characterized in well-defined neuronal populations. This autoantigen cross-reacted with I-Anod class II antigens, suggesting that it may contribute to defective self-tolerance of islet beta cells in the NOD mouse.
Collapse
Affiliation(s)
- C Boitard
- Institut National de la Santé et de la Recherche Médicale, U 25/Centre National de la Recherche Scientifique A 122, Hôpital Necker, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Abstract
Type I diabetes occurs as a result of T-cell-mediated beta-cell destruction. Several candidate antigens have been described recently, including glutamic acid decarboxylase, heat shock protein 65 and peripherin. Restricted T-cell receptor V beta gene usage in non-obese diabetic mice has been suggested but not yet proven. In addition to major histocompatibility genes, several non-H-2 predisposing genes have been mapped on chromosomes 1, 3 and 11.
Collapse
|
184
|
Zhang ZJ, Davidson L, Eisenbarth G, Weiner HL. Suppression of diabetes in nonobese diabetic mice by oral administration of porcine insulin. Proc Natl Acad Sci U S A 1991; 88:10252-6. [PMID: 1946445 PMCID: PMC52906 DOI: 10.1073/pnas.88.22.10252] [Citation(s) in RCA: 392] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nonobese diabetic (NOD) mice spontaneously develop an autoimmune form of diabetes associated with insulitis. A number of immunomodulatory therapies have been investigated as a treatment for the disease process. Oral administration of the autoantigens myelin basic protein and collagen type II suppresses experimental models of encephalomyelitis and arthritis. We have now found that oral administration of insulin delays the onset and reduces the incidence of diabetes in NOD mice over a 1-year period in animals administered 1 mg of porcine insulin orally twice a week for 5 weeks and then weekly until 1 year of age. As expected, orally administered insulin had no metabolic effect on blood glucose levels. The severity of lymphocytic infiltration of pancreatic islets was also reduced by oral administration of insulin. Furthermore, splenic T cells from animals orally treated with insulin adoptively transfer protection against diabetes, demonstrating that oral insulin administration generates active cellular mechanisms that suppress disease. These results show that oral insulin affects diabetes and the pancreatic cellular inflammatory process in the NOD mouse and raise the possibility that oral administration of insulin or other pancreatic autoantigens may provide a new approach for the treatment of autoimmune diabetes.
Collapse
Affiliation(s)
- Z J Zhang
- Brigham and Women's Hospital, Boston, MA
| | | | | | | |
Collapse
|
185
|
Yagi H, Matsumoto M, Kishimoto Y, Makino S, Harada M. Possible mechanism of the preventive effect of BCG against diabetes mellitus in NOD mouse. II. Suppression of pathogenesis by macrophage transfer from BCG-vaccinated mice. Cell Immunol 1991; 138:142-9. [PMID: 1833073 DOI: 10.1016/0008-8749(91)90139-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Our previous reports showed that a single injection of live BCG, one of the biological response modifiers, prevents insulitis and overt diabetes in NOD mice and that the suppression could be due to the generation of some type of suppressor cells in the BCG-treated mice. Furthermore, a more recent study has revealed that macrophages suppressive against a variety of lymphocyte functions can be induced by BCG, which suggests that these macrophages are involved in the suppression of the pathogenesis. To obtain valid evidence for this speculation, the effects of transfer of macrophage and T-cell fractions on the pathogenesis were examined in the present study. Transfer of macrophage-enriched spleen cell fraction harvested from the BCG-treated females to young females abolished the occurrence of spontaneous diabetes up to the age of 25 to 30 weeks. Also, macrophage transfer prevented the progress of insulitis. In contrast, transfer of a T-cell-enriched fraction did not suppress insulitis and overt diabetes. From these results, it could be concluded that the suppression of the autoimmune pathogenesis of diabetes by BCG is due to the generation of suppressor macrophages.
Collapse
Affiliation(s)
- H Yagi
- Shionogi Research Laboratories, Shionogi & Company, Ltd., Osaka, Japan
| | | | | | | | | |
Collapse
|
186
|
Slattery R. Transgenic approaches to understanding the role of MHC genes in insulin dependent diabetes mellitus. II. The non-obese diabetic (NOD) mouse. BAILLIERE'S CLINICAL ENDOCRINOLOGY AND METABOLISM 1991; 5:449-54. [PMID: 1909863 DOI: 10.1016/s0950-351x(05)80141-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
187
|
Bach JF, Caillat-Zucman S, Garchon HJ, Timsit J, Boitard C. Mechanisms and significance of HLA type I diabetes association. RESEARCH IN IMMUNOLOGY 1991; 142:485-6. [PMID: 1754723 DOI: 10.1016/0923-2494(91)90052-k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- J F Bach
- INSERM U 25, Hôpital Necker, Paris
| | | | | | | | | |
Collapse
|
188
|
Sempé P, Bédossa P, Richard MF, Villà MC, Bach JF, Boitard C. Anti-alpha/beta T cell receptor monoclonal antibody provides an efficient therapy for autoimmune diabetes in nonobese diabetic (NOD) mice. Eur J Immunol 1991; 21:1163-9. [PMID: 1828030 DOI: 10.1002/eji.1830210511] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The nonobese diabetic (NOD) mouse is a relevant model for studying human insulin-dependent diabetes mellitus (IDDM). The selective destruction of insulin-secreting cells in this model is subsequent to an autoimmune reaction directed towards the beta cells inside the islets of Langerhans of the pancreas. Given the key role played by T cells in the development of IDDM, we investigated a model of IDDM prevention in NOD mice by administration of a monoclonal antibody to the alpha/beta dimer of the T cell receptor for antigen. Our data provide evidence that aiming at the T cell receptor protects against both spontaneous and cyclophosphamide-induced diabetes in the NOD mouse. Interestingly, potential clinical application is suggested by the efficient and durable reversal of recent onset diabetes in mice treated with anti-alpha/beta monoclonal antibody within 1 week following the clinical discovery of IDDM.
Collapse
Affiliation(s)
- P Sempé
- INSERM U 25, Clinique Néphrologique, Hôpital Necker, Paris, France
| | | | | | | | | | | |
Collapse
|
189
|
Pankewycz O, Strom TB, Rubin-Kelley VE. Islet-infiltrating T cell clones from non-obese diabetic mice that promote or prevent accelerated onset diabetes. Eur J Immunol 1991; 21:873-9. [PMID: 1902177 DOI: 10.1002/eji.1830210403] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In humans and non-obese diabetic mice (NOD), insulin-dependent diabetes mellitus (IDDM) results from a spontaneous T cell-dependent autoimmune destruction of the insulin-producing pancreatic beta cells. Previous data suggest that a delicate balance between autoaggressive T cells and suppressor-type immune phenomena determine whether expression of autoimmunity is limited to insulitis or progresses to IDDM. To resolve the cellular basis of this intricate network of pathogenic CD4+ and CD8+ T cells and the role of T cells in suppressive immune phenomena. T cell clones were propagated directly from islets of NOD mice at the onset of insulitis. Insofar as insulitis, but not IDDM, is universal in NOD mice, we have screened for the in vivo effects of the islet-infiltrating T cell clones upon expression of IDDM, not insulitis. A CD4+ T cell clone, IS-3S7D, proliferates in response to islet antigen(s) and its transfer into prediabetic NOD mice promotes the rapid onset of IDDM. An interleukin 2 (IL 2)-dependent noncytolytic, V beta 11+ CD8+. T cell clones IS-2.15, prevents an accelerated onset diabetes in two distinct models. The present study, which documents the presence of CD4+ diabetogenic T cell clones and CD8+ T cell clones that dampen autoimmunity, gives tangible evidence that opposing autoimmune processes may determine whether an autoimmune-prone host develops frank disease.
Collapse
Affiliation(s)
- O Pankewycz
- Department of Medicine, Beth Israel Hospital, Boston, MA
| | | | | |
Collapse
|
190
|
Boitard C, Timsit J, Sempé P, Bach JF. Experimental immunoprevention of type I diabetes mellitus. DIABETES/METABOLISM REVIEWS 1991; 7:15-33. [PMID: 1935533 DOI: 10.1002/dmr.5610070105] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- C Boitard
- Service d'Immunologie Clinique, INSERM U25 and CNRS UA 122, Hôpital Necker, Paris, France
| | | | | | | |
Collapse
|
191
|
Prochazka M, Gaskins HR, Leiter EH, Koch-Nolte F, Haag F, Thiele HG. Chromosomal localization, DNA polymorphism, and expression of Rt-6, the mouse homologue of rat T-lymphocyte differentiation marker RT6. Immunogenetics 1991; 33:152-6. [PMID: 1671854 DOI: 10.1007/bf00210829] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
192
|
Shoenfeld Y. Ageing, autoantigen specific-Ts cells, involution of the thymus, autoimmunity and autoimmune diseases. Autoimmunity 1991; 8:251-6. [PMID: 1932511 DOI: 10.3109/08916939108997113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
193
|
Prud'homme GJ, Parfrey NA, Vanier LE. Cyclosporine-induced autoimmunity and immune hyperreactivity. Autoimmunity 1991; 9:345-56. [PMID: 1954315 DOI: 10.3109/08916939108997137] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cyclosporine (CS) is a potent immunosuppressive agent which under some circumstances paradoxically augments DTH responses, aggravates some autoimmune diseases, and induces specific forms of autoimmunity. The enhancement of DTH and other immune responses is closely related to the timing of CS administration relative to immunization. CS inhibits IL-2 production (and several other lymphokines) at a pretranscriptional level, but does not usually prevent the antigen-specific priming of T cells, such that T cells may be poised to respond as soon as CS is withdrawn. Thus, accelerated GVHD and allograft rejection may occur after withdrawal of CS. CS has been shown to aggravate and/or induce relapse in several autoimmune diseases including collagen-induced arthritis, EAE, autoimmune thyroiditis, uveitis in SDA chickens, and an autoimmune form of myocarditis in mice. CS may enhance immune responses by inactivating suppressor cells, by altering Th1/Th2 antagonism (e.g., CS promotes a protective Th1-type response in BALB/c mice infected with Leishmania major), or by promoting T cell activation through a CS-resistant IL-2-independent T cell activation/differentiation pathway. At least three forms of CS-induced autoimmunity have been described: Syngeneic or autologous GVHD which occurs in CS-treated syngeneic or autologous bone marrow transplant recipients after CS is withdrawn in rats, mice, and humans; a systemic autoimmune disease with polyarthritis and glomerulonephritis which occurs in irradiated CBA/N mice treated with CS; and organ-specific autoimmune diseases which occur in mice treated with CS during the neonatal period. The precise mechanisms by which CS induces these autoimmune diseases are not clear, however, CS affects immune tolerance at three levels. CS induces thymic medullary involution with loss of medullary Ia+ cells, and appears to at least partially block the transition from double positive (CD4+CD8+) to single positive (mature type) thymocytes. In syngeneic bone marrow chimeras, CS appears to inhibit the intrathymic deletion of clones with relatively low affinity, but not those with high affinity, to self antigens. CS appears to inhibit the action of suppressor T cells which normally maintain an innate form of resistance to autoimmunity. Finally, CS has been shown to prevent the development of T cell clonal anergy. There is redundancy in immune tolerance mechanisms, i.e., clonal deletion, clonal anergy, and suppressor cells can each maintain tolerance to similar antigens, such that it is likely that CS must cripple more than one tolerance mechanism for autoimmunity to occur.
Collapse
Affiliation(s)
- G J Prud'homme
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
194
|
Abstract
Self-tolerance is generally induced by intrathymic clonal deletion of T cells with reactivity directed to antigens synthesized within the thymus (Kappler et al. 1987, Kisielow et al. 1988). It may also be induced in peripheral T cells when these encounter antigens unique to extra-thymic tissues. Two transgenic models have been particularly useful in the study of peripheral self tolerance: in one model, a known antigen is expressed in a particular extra-thymic site; in the other, the T-cell repertoire is predominantly reactive to this antigen. We, and others, have shown that expression of class I or II MHC molecules in defined extra-thymic sites leads to a state of T-cell tolerance. To account for this, we have proposed two hypotheses which have different implications for autoimmune disease. According to one, tolerance is imposed by deletion or functional silencing of specific high-affinity cytolytic T cells; alternatively, the target cell for tolerance induction may be a regulatory IL-2-producing T-cell, rather than the effector cell itself. To distinguish between these hypotheses it is essential to examine the fate of T cells which have the potential to react to the transgene product. Since the frequency of such T cells is low and there is no dominant clonotype for H-2Kb, which is the class I molecule we used, it was necessary to create double transgenic mice by mating class I transgenic mice with transgenic mice whose T-cell pool was compared of cells reactive to H-2Kb and could be detected by an antibody directed to the TCR. Initial studies showed that such T cells did persist despite the presence of antigen to which they may be reactive. If these double transgenic mice can be shown to be tolerant, they will offer a rich source of tolerant T cells for detailed investigation of their phenotype and fate, and they will be most useful in enabling us to probe the mechanisms responsible for the induction of peripheral self tolerance. Transgenic mouse technology has also been used successfully to unravel the genetic influences which may lead to or prevent autoimmunity. In particular, we have prevented autoimmune diabetes in the nonobese diabetic mouse by introducing a non-NOD MHC class II gene and further work is implicating the failure of intrathymic positive selection of a protective cell as one step in the pathogenesis of diabetes in NOD mice.
Collapse
Affiliation(s)
- J F Miller
- Walter and Eliza Hall Institute of Medical Research, Post Office Royal Melbourne Hospital, Victoria, Australia
| | | | | | | |
Collapse
|
195
|
Lo D, Burkly LC, Flavell RA, Palmiter RD, Brinster RL. Antigen presentation in MHC class II transgenic mice: stimulation versus tolerization. Immunol Rev 1990; 117:121-34. [PMID: 2258189 DOI: 10.1111/j.1600-065x.1990.tb00570.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- D Lo
- Laboratory of Reproductive Physiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia 19104
| | | | | | | | | |
Collapse
|
196
|
Slattery RM, Kjer-Nielsen L, Allison J, Charlton B, Mandel TE, Miller JF. Prevention of diabetes in non-obese diabetic I-Ak transgenic mice. Nature 1990; 345:724-6. [PMID: 1972779 DOI: 10.1038/345724a0] [Citation(s) in RCA: 163] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The non-obese diabetic (NOD) mouse develops insulin-dependent diabetes mellitus (IDDM) with mononuclear cell infiltration of the islets of Langerhans and selective destruction of the insulin-producing beta-cells, as in humans. Most infiltrating cells are T lymphocytes, and most of these carry the CD4 antigen. Adoptive transfer of T cells from diabetic NOD mice into irradiated NOD or athymic nude NOD mice induces diabetes. Susceptibility to IDDM in NOD mice is polygenic, with one gene linked to the major histocompatibility complex class II locus, which in NOD mice expresses a unique I-A molecule but no I-E. Speculation exists as to the role of the I-A molecule in the diabetes susceptibility of NOD mice, especially regarding the significance of specific unique residues. To examine the role of the NOD I-A molecule in IDDM pathogenesis, we made NOD/Lt mice transgenic for I-Ak by microinjecting I-Ak alpha- and beta-genes into fertilized NOD/Lt eggs. Insulitis was markedly reduced and diabetes prevented in NOD/Lt mice expressing I-Ak.
Collapse
Affiliation(s)
- R M Slattery
- Walter and Eliza Hall Institute of Medical Research, Royal Melbourne Hospital, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
197
|
Abstract
DNA sequence analysis of major histocompatibility complex (MHC) class II genes from humans and rodents with type 1 (insulin-dependent) diabetes indicates that a portion of MHC-linked genetic susceptibility in humans is determined by the HLA-DQA1 and -DQB1 loci. In this article John Todd summarizes recent advances in these studies. The conformation of DQ molecules and their levels of expression may influence the efficiency of autoantigen presentation and the degree of pancreatic beta cells destruction during disease development. Certain DAQ1 and DQB1 alleles correlate with decreased susceptibility to disease. The penetrance of class II alleles that are correlated with positive susceptibility may be influenced by environmental factors such as bacterial and viral infections.
Collapse
Affiliation(s)
- J A Todd
- Nuffield Department of Surgery, John Radcliffe Hospital, Headington, Oxford, UK
| |
Collapse
|
198
|
Hutchings PR, Cooke A. The transfer of autoimmune diabetes in NOD mice can be inhibited or accelerated by distinct cell populations present in normal splenocytes taken from young males. J Autoimmun 1990; 3:175-85. [PMID: 1971173 DOI: 10.1016/0896-8411(90)90139-j] [Citation(s) in RCA: 57] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The NOD mouse is characterized by the development of spontaneous autoimmune diabetes which begins with a peri-islet lymphocyte infiltration of the pancreas around 6 weeks of age and progresses to overt diabetes in 50-60% of females from about 12 weeks. Although infiltration occurs around islets in males, the incidence of overt diabetes is much less (about 1%) and suggests that there may be more effective regulatory circuits in these animals. This possibility was examined by using splenocytes from young males to reconstitute irradiated male recipients 6 d before the transfer of diabetogenic spleen cells from spontaneously diabetic females. Those animals which were not reconstituted with male spleen cells developed diabetes 3-5 weeks later, whereas the majority of the reconstituted mice remained normoglycaemic. Characterization of the protective population demonstrated a role for CD4+ T cells. An additional observation was that splenocytes from young normal males also contained a population of non-T cells which could advance the diabetogenic transfer of disease by at least a week.
Collapse
Affiliation(s)
- P R Hutchings
- Department of Immunology, University College, London, UK
| | | |
Collapse
|
199
|
Bach JF, Boitard C, Yasunami R, Dardenne M. Control of diabetes in NOD mice by suppressor cells. J Autoimmun 1990; 3 Suppl 1:97-100. [PMID: 2140263 DOI: 10.1016/s0896-8411(09)90017-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- J F Bach
- INSERM U25, Hôpital Necker, Paris, France
| | | | | | | |
Collapse
|
200
|
Cooke A. An overview on possible mechanisms of destruction of the insulin-producing beta cell. Curr Top Microbiol Immunol 1990; 164:125-42. [PMID: 2127391 DOI: 10.1007/978-3-642-75741-9_7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- A Cooke
- Department of Immunology, ASH, UCMSM, London, UK
| |
Collapse
|