1
|
Lesteberg KE, Araya P, Waugh KA, Chauhan L, Espinosa JM, Beckham JD. Severely ill and high-risk COVID-19 patients exhibit increased peripheral circulation of CD62L+ and perforin+ T cells. Front Immunol 2023; 14:1113932. [PMID: 36817450 PMCID: PMC9932815 DOI: 10.3389/fimmu.2023.1113932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction The emergence of SARS-CoV-2, which causes COVID-19, has led to over 400 million reported cases worldwide. COVID-19 disease ranges from asymptomatic infection to severe disease and may be impacted by individual immune differences. Methods We used multiparameter flow cytometry to compare CD4+ and CD8+ T cell responses in severe (ICU admitted) and non-severe (admitted to observational unit) hospitalized COVID-19 patients. Results We found that patients with severe COVID- 19 had greater frequencies of CD4+ T cells expressing CD62L compared to non-severe patients and greater frequencies of perforin+ CD8+ T cells compared to recovered patients. Furthermore, greater frequencies of CD62L+ CD4+ and CD8+ T cells were seen in severely ill diabetic patients compared to non-severe and non-diabetic patients, and increased CD62L+ CD4+ T cells were also seen in severely ill patients with hypertension. Discussion This is the first report to show that CD62L+ T cells and perforin+ T cells are associated with severe COVID-19 illness and are significantly increased in patients with high-risk pre-existing conditions including older age and diabetes. These data provide a potential biological marker for severe COVID-19.
Collapse
Affiliation(s)
- Kelsey E. Lesteberg
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO, United States
| | - Katherine A. Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lakshmi Chauhan
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, United States
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - J. David Beckham
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Rocky Mountain VA Medical Center, Aurora, CO, United States
| |
Collapse
|
2
|
Akimoto H, Fukuda-Kawaguchi E, Duramad O, Ishii Y, Tanabe K. A Novel Liposome Formulation Carrying Both an Insulin Peptide and a Ligand for Invariant Natural Killer T Cells Induces Accumulation of Regulatory T Cells to Islets in Nonobese Diabetic Mice. J Diabetes Res 2019; 2019:9430473. [PMID: 31781669 PMCID: PMC6855036 DOI: 10.1155/2019/9430473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/03/2019] [Indexed: 12/27/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of pancreatic β cells by autoantigen-reactive diabetogenic cells. Antigen-specific therapies using islet autoantigens for restoring immune tolerance have emerged as promising approaches for the treatment of T1D but have been unsuccessful in humans. Herein, we report that RGI-3100-iB, a novel liposomal formulation carrying both α-galactosylceramide (α-GalCer), which is a representative ligand for invariant natural killer T (iNKT) cells, and insulin B chain 9-23 peptide, which is an epitope for CD4+ T cells, could induce the accumulation of regulatory T cells (Tregs) in islets in a peptide-dependent manner, followed by the remarkable prevention of diabetes onset in nonobese diabetic (NOD) mice. While multiple administrations of a monotherapy using either α-GalCer or insulin B peptide in a liposomal formulation was confirmed to delay/prevent T1D in NOD mice, RGI-3100-iB synergistically enhanced the prevention effect of each monotherapy and alleviated insulitis in NOD mice. Immunopathological analysis showed that Foxp3+ Tregs accumulated in the islets in RGI-3100-iB-treated mice. Cotransfer of diabetogenic T cells and splenocytes of NOD mice treated with RGI-3100-iB, but not liposomal α-GalCer encapsulating an unrelated peptide, to NOD-SCID mice resulted in the prevention of diabetes and elevation of Foxp3 mRNA expression in the islets. These data indicate that the migration of insulin B-peptide-specific Tregs to islet of NOD mice that are involved in the suppression of pathogenic T cells related to diabetes onset and progression could be enhanced by the administration of liposomes containing α-GalCer and insulin B peptide.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/prevention & control
- Disease Models, Animal
- Drug Compounding
- Female
- Forkhead Transcription Factors/metabolism
- Galactosylceramides/administration & dosage
- Hypoglycemic Agents/administration & dosage
- Insulin/administration & dosage
- Islets of Langerhans/drug effects
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Liposomes
- Mice, Inbred NOD
- Mice, SCID
- Natural Killer T-Cells/drug effects
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Peptide Fragments/administration & dosage
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
Collapse
Affiliation(s)
- Hidetoshi Akimoto
- Research Division, REGiMMUNE Corporation, 35-3 Nihonbashi Hakozaki-cho, BRICK GATE 5F, Chuou-Ku, Tokyo 103-0015, Japan
| | - Emi Fukuda-Kawaguchi
- Research Division, REGiMMUNE Corporation, 35-3 Nihonbashi Hakozaki-cho, BRICK GATE 5F, Chuou-Ku, Tokyo 103-0015, Japan
| | - Omar Duramad
- Research Division, REGiMMUNE Inc, 820 Heinz Ave, Berkeley, CA 94710, USA
| | - Yasuyuki Ishii
- Research Division, REGiMMUNE Corporation, 35-3 Nihonbashi Hakozaki-cho, BRICK GATE 5F, Chuou-Ku, Tokyo 103-0015, Japan
| | - Kazunari Tanabe
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-Ku, Tokyo 162-8666, Japan
| |
Collapse
|
3
|
Kuhn C, Rezende RM, da Cunha AP, Valette F, Quintana FJ, Chatenoud L, Weiner HL. Mucosal administration of CD3-specific monoclonal antibody inhibits diabetes in NOD mice and in a preclinical mouse model transgenic for the CD3 epsilon chain. J Autoimmun 2017; 76:115-122. [PMID: 27745778 PMCID: PMC9815832 DOI: 10.1016/j.jaut.2016.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 10/01/2016] [Accepted: 10/03/2016] [Indexed: 01/11/2023]
Abstract
CD3-specific monoclonal antibody (mAb) treats autoimmune disease in animal models and has shown promise in clinical trials of type 1 diabetes. Whereas intravenous administration of CD3-specific mAb acts primarily by transient depletion of activated effector T cells, oral CD3-specific mAb acts primarily by the induction Tregs. We investigated whether oral CD3-specific mAb inhibits disease in non obese diabetic (NOD) mice that spontaneously develop autoimmune diabetes, closely resembling human type 1 diabetes. We found that oral CD3-specific mAb treatment delayed onset and reduced incidence of diabetes in NOD mice, inducing changes in both effector and regulatory T cell compartments. The therapeutic effect was associated with decreased T cell proliferation, decreased IFNγ and IL-17 production, and increased TGF-β and IL-10 production in vitro. In vivo transfer experiments demonstrated that oral CD3-specific mAb decreased diabetogenicity of effector T cells and increased the function of regulatory T cells. Oral OKT3, a monoclonal antibody specific for human CD3 had equivalent effects in transgenic NOD mice expressing the human CD3 epsilon chain which serves as a preclinical model for testing human CD3-specific mAb. These results suggest that oral CD3-specific mAb has the potential for treating autoimmune diabetes in humans.
Collapse
Affiliation(s)
- Chantal Kuhn
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Rafael M. Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Andre Pires da Cunha
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Fabrice Valette
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Université Paris Descartes, Sorbonne Paris Cité, F-75475, Paris, France
| | - Francisco J. Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lucienne Chatenoud
- Université Paris Descartes, Sorbonne Paris Cité, F-75475, Paris, France,INSERM U1151, CNRS UMR 8253, Hôpital Necker-Enfants Malades, Paris, France
| | - Howard L. Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
4
|
Selectin-mediated leukocyte trafficking during the development of autoimmune disease. Autoimmun Rev 2015; 14:984-95. [DOI: 10.1016/j.autrev.2015.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/18/2015] [Indexed: 12/18/2022]
|
5
|
T-lymphocyte recognition of beta cells in type 1 diabetes: clinical perspectives. DIABETES & METABOLISM 2013; 39:459-66. [PMID: 24139825 DOI: 10.1016/j.diabet.2013.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 08/14/2013] [Indexed: 11/23/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by the activation of lymphocytes against pancreatic β cells. Landmarks in the history of T1D were the description of insulitis and of islet cell autoantibodies, and report an association between T1D and a limited number of HLA alleles. Another step was the study of T-lymphocytes, now known to be central to the disease process of T1D whether in mice or men. In humans, T-lymphocytes, and especially CD8⁺ T-cells, are predominant in insulitis. The characterization of antigenic fragments--peptides--recognized by T-cells paves the way towards new assays for predicting T1D and its prevention using antigen- or peptide-specific immunotherapy, while avoiding side effects that may counteract the limited efficacy of immunosuppression and immunomodulation in preserving β-cells from autoimmune destruction in recent-onset T1D patients. The current need for new preclinical models for testing strategies of antigen-specific immune tolerance is also highlighted.
Collapse
|
6
|
Jin Y, Sharma A, Carey C, Hopkins D, Wang X, Robertson DG, Bode B, Anderson SW, Reed JC, Steed RD, Steed L, She JX. The expression of inflammatory genes is upregulated in peripheral blood of patients with type 1 diabetes. Diabetes Care 2013; 36:2794-802. [PMID: 23637351 PMCID: PMC3747909 DOI: 10.2337/dc12-1986] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Our previous gene expression microarray studies identified a number of genes differentially expressed in patients with type 1 diabetes (T1D) and islet autoantibody-positive subjects. This study was designed to validate these gene expression changes in T1D patients and to identify gene expression changes in diabetes complications. RESEARCH DESIGH AND METHODS: We performed high-throughput real-time RT-PCR to validate gene expression changes in peripheral blood mononuclear cells (PBMCs) from a large sample set of 928 T1D patients and 922 control subjects. RESULTS Of the 18 genes analyzed here, eight genes (S100A8, S100A9, MNDA, SELL, TGFB1, PSMB3, CD74, and IL12A) had higher expression and three genes (GNLY, PSMA4, and SMAD7) had lower expression in T1D patients compared with control subjects, indicating that genes involved in inflammation, immune regulation, and antigen processing and presentation are significantly altered in PBMCs from T1D patients. Furthermore, one adhesion molecule (SELL) and three inflammatory genes mainly expressed by myeloid cells (S100A8, S100A9, and MNDA) were significantly higher in T1D patients with complications (odds ratio [OR] 1.3-2.6, adjusted P value = 0.005-10(-8)), especially those patients with neuropathy (OR 4.8-7.9, adjusted P value <0.005). CONCLUSIONS These findings suggest that inflammatory mediators secreted mainly by myeloid cells are implicated in T1D and its complications.
Collapse
Affiliation(s)
- Yulan Jin
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Abstract
Type 1 diabetes (T1D) represents 10 to 15% of all forms of diabetes. Its incidence shows a consistent rise in all countries under survey. Evidence for autoimmunity in human T1D relies on the detection of insulitis, of islet cell antibodies, of activated β-cell-specific T lymphocytes and on the association of T1D with a restricted set of class II major histocompatibility complex (MHC) alleles. However, mechanisms that initiate the failure of immune tolerance to β-cell autoantigens remain elusive in common forms of T1D. T1D commonly develop as a multifactorial disease in which environmental factors concur with a highly multigenic background. The disease is driven by the activation of T-lymphocytes against pancreatic β-cells. Several years elapse between initial triggering of the autoimmune response to β cells, as evidenced by the appearance or islet cell autoantibodies, and the onset of clinical diabetes, defining a prediabetes stage. Active mechanisms hold back autoreactive effector T-cells in prediabetes, in particular a subset of CD4+ T-cells (T(reg)) and other regulatory T-cells, such as invariant NKT cells. There is evidence in experimental models that systemic or local infections can trigger autoimmune reactions to β-cells. However, epidemiological observations that have accumulated over years have failed to identify undisputable environmental factors that trigger T1D. Moreover, multiple environmental factors may intervene in the disease evolution and protective as weel as triggering environmental factors may be involved. Available models also indicate that local signals within the islets are required for full-blown diabetes to develop. Many autoantigens that are expressed by β-cells but also by the other endocrine islet cells and by neurons are recognized by lymphocytes along the development of T1D. The immune image of β-cells is that of native components of the β-cell membrane, as seen by B-lymphocytes, and of fragments of intracellular β-cell proteins in the form of peptides loaded onto class I MHC molecules on the β-cell surface and class I and class II molecules onto professional antigen presenting cells. Given the key role of T lymphocytes in T1D, the cartography of autoantigen-derived peptides that are presented to class I-restricted CD8(+) T-cells and class II-restricted CD4(+) T-cells is of outmost importance and is a necessary step in the development of diagnostic T-cell assays and of immunotherapy of T1D.
Collapse
|
8
|
IL-7 receptor blockade reverses autoimmune diabetes by promoting inhibition of effector/memory T cells. Proc Natl Acad Sci U S A 2012; 109:12668-73. [PMID: 22733744 DOI: 10.1073/pnas.1203692109] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
To protect the organism against autoimmunity, self-reactive effector/memory T cells (T(E/M)) are controlled by cell-intrinsic and -extrinsic regulatory mechanisms. However, how some T(E/M) cells escape regulation and cause autoimmune disease is currently not understood. Here we show that blocking IL-7 receptor-α (IL-7Rα) with monoclonal antibodies in nonobese diabetic (NOD) mice prevented autoimmune diabetes and, importantly, reversed disease in new-onset diabetic mice. Surprisingly, IL-7-deprived diabetogenic T(E/M) cells remained present in the treated animals but showed increased expression of the inhibitory receptor Programmed Death 1 (PD-1) and reduced IFN-γ production. Conversely, IL-7 suppressed PD-1 expression on activated T cells in vitro. Adoptive transfer experiments revealed that T(E/M) cells from anti-IL-7Rα-treated mice had lost their pathogenic potential, indicating that absence of IL-7 signals induces cell-intrinsic tolerance. In addition to this mechanism, IL-7Rα blockade altered the balance of regulatory T cells and T(E/M) cells, hence promoting cell-extrinsic regulation and further increasing the threshold for diabetogenic T-cell activation. Our data demonstrate that IL-7 contributes to the pathogenesis of autoimmune diabetes by enabling T(E/M) cells to remain in a functionally competent state and suggest IL-7Rα blockade as a therapy for established T-cell-dependent autoimmune diseases.
Collapse
|
9
|
Tartar DM, VanMorlan AM, Wan X, Guloglu FB, Jain R, Haymaker CL, Ellis JS, Hoeman CM, Cascio JA, Dhakal M, Oukka M, Zaghouani H. FoxP3+RORgammat+ T helper intermediates display suppressive function against autoimmune diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:3377-85. [PMID: 20181889 PMCID: PMC2843758 DOI: 10.4049/jimmunol.0903324] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recently, traces of double-positive FoxP3(+)RORgammat(+) T cells were identified and viewed as dual programming differentiation intermediates geared toward development into T regulatory or Th17 cells. In this study, we report that FoxP3(+)RORgammat(+) intermediates arise in the NOD mouse T cell repertoire prior to inflammation and can be expanded with tolerogen without further differentiation. Furthermore, FoxP3(+)RORgammat(+) cells express both CD62L and membrane-bound TGFbeta and use the former to traffic to the pancreas and the latter to suppress effector T cells both in vitro and in vivo. The cells perform these functions as FoxP3(+)RORgammat(+) intermediates, despite being able to terminally differentiate into either FoxP3(+)RORgammat(-) T regulatory or FoxP3(-)RORgammat(+) Th17 cells on polarization. These previously unrecognized observations extend plasticity to both differentiation and function and indicate that the intermediates are poised to traffic to sites of inflammation and target diverse pathogenic T cells, likely without prior conditioning by effector T cells, thus broadening efficacy against autoimmunity.
Collapse
MESH Headings
- Animals
- Cell Differentiation/immunology
- Cell Separation
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Flow Cytometry
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred NOD
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Danielle M. Tartar
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Amie M. VanMorlan
- Department of Child Health., University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Xiaoxiao Wan
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - F. Betul Guloglu
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Renu Jain
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Cara L. Haymaker
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Jason S. Ellis
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Christine M. Hoeman
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Jason A. Cascio
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Mermagya Dhakal
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Mohamed Oukka
- Center for Neurologic Diseases Harvard University, Boston, MA, 02115, USA
| | - Habib Zaghouani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Department of Child Health., University of Missouri School of Medicine, Columbia, MO, 65212, USA
| |
Collapse
|
10
|
Gagnerault MC, Lanvin O, Pasquier V, Garcia C, Damotte D, Lucas B, Lepault F. Autoimmunity during Thymectomy-Induced Lymphopenia: Role of Thymus Ablation and Initial Effector T Cell Activation Timing in Nonobese Diabetic Mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:4913-20. [DOI: 10.4049/jimmunol.0901954] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
11
|
You S, Thieblemont N, Alyanakian MA, Bach JF, Chatenoud L. Transforming growth factor-beta and T-cell-mediated immunoregulation in the control of autoimmune diabetes. Immunol Rev 2006; 212:185-202. [PMID: 16903915 DOI: 10.1111/j.0105-2896.2006.00410.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
It is now well-established that CD4+ regulatory T cells are instrumental in controlling immune responses both to self-antigens and to non-self-antigens. However, the precise modalities involved in their differentiation and survival, their mode of action and their antigen specificity are only partially understood. We have been particularly interested in the study of regulatory T cells controlling autoimmune insulin-dependent diabetes. Here, we provide evidence to support the phenotypic and functional diversity of regulatory T cells mediating transferable 'active' or 'dominant' peripheral tolerance in the non-obese diabetic mouse model (NOD). They include natural and adaptive regulatory T cells that are operational both in unmanipulated NOD mice and in animals undergoing treatments aimed at inducing/restoring tolerance to self-beta-cell antigens. At least in our hands, the differential cytokine-dependency appears as a major distinctive feature of regulatory T cells subsets. Among immunoregulatory cytokines, transforming growth factor-beta(TGF-beta) appeared to play a key role. Herein we discuss these results and the working hypothesis they evoke in the context of the present literature, where the role of TGF-beta-dependent T-cell-mediated immunoregulation is still debated.
Collapse
Affiliation(s)
- Sylvaine You
- Université René Descartes Paris 5, Institut National de la Santé et de la Recherche Médicale U580 and Hôpital Necker-Enfants Malades, Paris, France
| | | | | | | | | |
Collapse
|
12
|
Chatenoud L, Bach JF. Resetting the functional capacity of regulatory T cells: a novel immunotherapeutic strategy to promote immune tolerance. Expert Opin Biol Ther 2006; 5 Suppl 1:S73-81. [PMID: 16187942 DOI: 10.1517/14712598.5.1.s73] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Over the last few years, there has been a re-emergence of the concept of suppressor/regulatory T cells among the central players of immune mechanisms controlling a wide variety of immune responses from physiological autoreactivity (i.e., response to self-antigens) to responses to transplants, tumours and infectious antigens. Regulatory T cells are diverse in their phenotypes, antigen specificity, mode of action and immunopathological relevance. This review briefly summarises studies from the authors' group showing that specialised subsets of regulatory T cells are instrumental in the control of autoimmune diseases and more specifically of Type 1 diabetes. In addition, this review will provide evidence supporting the notion that CD3-specific monoclonal antibodies are representatives of a new category of immunotherapeutic agents that possess the unique capacity to promote immunological tolerance (an antigen-specific unresponsiveness in the absence of long-term generalised immunosuppression) through their ability to induce immunoregulatory T cells.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/immunology
- CD3 Complex/immunology
- Clinical Trials as Topic
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Humans
- Immune Tolerance
- Mice
- Mice, Inbred NOD
- Muromonab-CD3/pharmacology
- Muromonab-CD3/therapeutic use
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Lucienne Chatenoud
- Faculté René Descartes Paris 5, INSERM U580, Hôpital Necker, 161 Rue de Sèvres, 75015 Paris, France.
| | | |
Collapse
|
13
|
Li O, Liu JQ, Zhang H, Zheng P, Liu Y, Bai XF. CD62L is required for the priming of encephalitogenic T cells but does not play a major role in the effector phase of experimental autoimmune encephalomyelitis. Scand J Immunol 2006; 64:117-24. [PMID: 16867156 DOI: 10.1111/j.1365-3083.2006.01783.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD62L (l-selectin, mel 14) regulates naïve T cell homing into lymph nodes and the migration of leucocytes to sites of inflammation. The requirement of CD62L in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, has been demonstrated previously. However, it remains controversial as to whether CD62L is required for the induction or the effector phase of EAE. It is also unclear whether other non-T effector cells need CD62L to enter the central nervous system (CNS) parenchyma and exert their damaging effects on myelin. We report that mice with a targeted mutation of CD62L are resistant to Myelin oligodendrocyte glycoprotein peptide-induced EAE. CD62L-deficient mice had no peptide-specific T cell responses in the draining lymph nodes and had lower levels of peptide-specific T cell responses in spleens at a later time point. Adoptive transfer studies showed that CD62L-deficient mice were fully susceptible to adoptive transfer EAE induced by either wildtype or CD62L-deficient T cells. Moreover, CD62L-deficient, F4/80(+) macrophages can be efficiently recruited into the CNS parenchyma. These data suggest that CD62L is required for the induction of encephalitogenic T cells during EAE development, but is not required by T and non-T effector cells to attack the CNS parenchyma.
Collapse
Affiliation(s)
- O Li
- Division of Cancer Immunology, Department of Pathology and Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
14
|
Abstract
The most intensively studied autoimmune disorder, type 1 diabetes mellitus (DM1), has attracted perhaps the greatest interest for gene-based therapeutic and prophylactic interventions. The final clinical manifestation of this immunologically and genetically complex disease, the absence of insulin, is the major starting point for almost all the gene therapy modalities attempted to date. Insulin replacement by transplantation of islets of Langerhans or surrogate beta cells is the obvious choice, but the allogeneic nature of the transplants activates potent antidonor immunoreactivity necessitating gene and cell-based immunosuppressive strategies as an alternative to the toxic pharmacologic immunosuppressives indicated for classic solid organ transplants. Accumulating knowledge of the cellular mechanisms involved in onset, however, have yielded promising tolerance induction prophylactic approaches using genes and cells. Despite the early successes in a number of animal models, the true test of efficacy in humans remains to be demonstrated.
Collapse
Affiliation(s)
- Nick Giannoukakis
- Diabetes Institute, Pediatric Research Section, Children's Hospital of Pittsburgh and University of Pittsburgh, Rangos Research Center, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
15
|
Chatenoud L, Bach JF. Regulatory T cells in the control of autoimmune diabetes: the case of the NOD mouse. Int Rev Immunol 2005; 24:247-67. [PMID: 16036377 DOI: 10.1080/08830180590934994] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Over the last few years, there has been a revival of the concept of suppressor/regulatory T cells being central players in the control of various immune responses, including autoimmune responses and immune response to transplants, tumors, and infectious agents. It appears that regulatory T cells are diverse in their phenotypes, antigen specificity, and modes of action. Here we summarize studies from various groups, including our own, demonstrating that specialized subsets of regulatory T cells are pivotal in the control of autoimmune diabetes as well shown by the compelling evidence accumulated using the non-obese diabetic (NOD) mouse model. We also provide a discussion of the evidence showing that some biological products (such as CD3-specific monoclonal antibodies) are representatives of a new category of immunotherapeutic agents endowed with unique capacities to promote immunological tolerance (an antigen-specific unresponsiveness in the absence of long-term generalized immunosuppression) through their ability to induce immunoregulatory T cells.
Collapse
|
16
|
You S, Belghith M, Cobbold S, Alyanakian MA, Gouarin C, Barriot S, Garcia C, Waldmann H, Bach JF, Chatenoud L. Autoimmune diabetes onset results from qualitative rather than quantitative age-dependent changes in pathogenic T-cells. Diabetes 2005; 54:1415-22. [PMID: 15855328 DOI: 10.2337/diabetes.54.5.1415] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Diabetogenic T-cells can be detected in pre-diabetic nonobese diabetic (NOD) mice after transfer in NOD-SCID recipients. Here we demonstrate that 6-week-old pre-diabetic NOD mice, >2 months before disease onset, already harbor pathogenic T-cells in equal numbers to overtly diabetic animals. The delay in diabetes appearance is explained by the presence of regulatory CD4+ CD25+ T-cells that control diabetogenic effectors and that are, in our hands, transforming growth factor (TGF)-beta-dependent. Our present results suggest, however, that diabetes onset is only partly explained by a decline in this regulatory T-cell activity. Another major factor appears to be the progressive resistance of diabetogenic cells to TGF-beta-dependent mediated inhibition. We propose that progression to overt disease correlates with the pathogenic T-cell's escape from TGF-beta-dependent T-cell-mediated regulation.
Collapse
|
17
|
Lundsgaard D, Holm TL, Hornum L, Markholst H. In vivo control of diabetogenic T-cells by regulatory CD4+CD25+ T-cells expressing Foxp3. Diabetes 2005; 54:1040-7. [PMID: 15793242 DOI: 10.2337/diabetes.54.4.1040] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
To understand the ability of regulatory T-cells to control diabetes development in clinically relevant situations, we established a new model of accelerated diabetes in young DP-BB rats by transferring purified T-cells from DR-BB rats made acutely diabetic. Transfer of 3, 5, 10, or 23 million pure in vitro-activated T-cells accelerated diabetes onset in >90% of the recipients, with the degree of acceleration being dosage dependent. Cotransfer of unfractionated leukocytes from healthy donors prevented diabetes. Full protection was achieved when protective cells were transferred 3-4 days before diabetogenic cells, whereas transfer 2 days before conferred only partial protection. Protection resided in the CD4(+) fraction, as purified CD4(+) T-cells prevented the accelerated diabetes. When CD25(+) cells were depleted from these cells before they were transferred, their ability to prevent diabetes was impaired. In contrast, two million CD4(+)CD25(+) cells (expressing Foxp3) prevented the accelerated diabetes when transferred both before and simultaneously with the diabetogenic T-cells. In addition, 2 million CD4(+)CD25(+) T-cells prevented spontaneous diabetes, even when given to rats age 42 days, whereas 20 million CD4(+)CD25(-) cells (with low Foxp3 expression) were far less effective. We thus demonstrated that CD4(+)CD25(+) cells exhibit powerful regulatory potential in rat diabetes.
Collapse
|
18
|
Yadav D, Judkowski V, Flodstrom-Tullberg M, Sterling L, Redmond WL, Sherman L, Sarvetnick N. B7-2 (CD86) Controls the Priming of Autoreactive CD4 T Cell Response against Pancreatic Islets. THE JOURNAL OF IMMUNOLOGY 2004; 173:3631-9. [PMID: 15356107 DOI: 10.4049/jimmunol.173.6.3631] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The B7-1/2-CD28 system provides the critical signal for the generation of an efficient T cell response. We investigated the role played by B7-2 in influencing pathogenic autoimmunity from islet-reactive CD4 T cells in B7-2 knockout (KO) NOD mice which are protected from type 1 diabetes. B7-2 deficiency caused a profound diminishment in the generation of spontaneously activated CD4 T cells and islet-specific CD4 T cell expansion. B7-2 does not impact the effector phase of the autoimmune response as adoptive transfer of islet Ag-specific BDC2.5 splenocytes stimulated in vitro could easily induce disease in B7-2KO mice. CD4 T cells showed some hallmarks of hyporesponsiveness because TCR/CD28-mediated stimulation led to defective activation and failure to induce disease in NODscid recipients. Furthermore, CD4 T cells exhibited enhanced death in the absence of B7-2. Interestingly, we found that B7-2 is required to achieve normal levels of CD4+CD25+CD62L+ T regulatory cells because a significant reduction of these T regulatory cells was observed in the thymus but not in the peripheral compartments of B7-2KO mice. In addition, our adoptive transfer experiments did not reveal either pathogenic or regulatory potential associated with the B7-2KO splenocytes. Finally, we found that the lack of B7-2 did not induce a compensatory increase in the B7-1 signal on APC in the PLN compartment. Taken together these results clearly indicate that B7-2 plays a critical role in priming islet-reactive CD4 T cells, suggesting a simplified, two-cell model for the impact of this costimulatory molecule in autoimmunity against islets.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, CD/genetics
- Antigens, CD/physiology
- Autoantibodies/biosynthesis
- Autoantigens/immunology
- B7-1 Antigen/biosynthesis
- B7-2 Antigen
- CD28 Antigens/immunology
- CD28 Antigens/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- Cell Division/genetics
- Cell Division/immunology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/prevention & control
- Female
- Interphase/genetics
- Interphase/immunology
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Lymphopenia/genetics
- Lymphopenia/immunology
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Interleukin-2/biosynthesis
- Spleen/metabolism
- Spleen/pathology
- Spleen/transplantation
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- Deepak Yadav
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Elliott JI, Higgins CF. Major histocompatibility complex class I shedding and programmed cell death stimulated through the proinflammatory P2X7 receptor: a candidate susceptibility gene for NOD diabetes. Diabetes 2004; 53:2012-7. [PMID: 15277380 DOI: 10.2337/diabetes.53.8.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
It has been hypothesized that type 1 diabetes is initiated by neonatal physiological pancreatic beta-cell death, indicating that the early stages of this autoimmune response may reflect a dysregulated response to immune "danger" signals. One potential danger signal is ATP, high concentrations of which stimulate the purinergic receptor P2X7 on hematopoietic cells. We compared the sensitivity of lymphocytes from model type 1 diabetic (NOD) and control (C57BL/10) mice to activation of this pathway. Stimulation of the P2X7 receptor of NOD mice resulted in more pronounced shedding of the lymphocyte homing receptor CD62L and in increased programmed cell death. Levels of major histocompatibility complex class I molecules, which have previously been reported to be poorly expressed on NOD lymphocytes, were initially normal, but the molecules were shed preferentially from NOD cells after P2X7 receptor stimulation. Thus, although NOD lymphocytes have been considered resistant to programmed cell death, they are highly sensitive to that stimulated through the P2X7 receptor. Because NOD mice express a low activation threshold allele of the P2X7 receptor and the P2X7 gene maps to a locus associated with disease, P2X7 is a good candidate susceptibility gene for NOD diabetes.
Collapse
Affiliation(s)
- James I Elliott
- Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College, Hammersmith Hospital Campus, London, UK.
| | | |
Collapse
|
20
|
Alyanakian MA, You S, Damotte D, Gouarin C, Esling A, Garcia C, Havouis S, Chatenoud L, Bach JF. Diversity of regulatory CD4+T cells controlling distinct organ-specific autoimmune diseases. Proc Natl Acad Sci U S A 2003; 100:15806-11. [PMID: 14673094 PMCID: PMC307649 DOI: 10.1073/pnas.2636971100] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Depletion of selected regulatory CD4+ T cell subsets induces the spontaneous onset of various immune or autoimmune disorders. It is not clear, however, whether a given subset, notably CD4+CD25+ regulatory T cells, protects from a wide spectrum of immune disorders, or whether specialized subsets of regulatory T cells control each given disease or group of diseases. We report here, using diabetes prone nonobese diabetic (NOD) mice, that depending on the regulatory T cells that are depleted, i.e., CD25+, CD62L+, or CD45RB(low), distinct immune diseases appear after transfer into NOD severe combined immunodeficiency (SCID) recipients. Thus, reconstitution of NOD SCID mice with CD25- T cells induces major gastritis and late-onset diabetes, but no or mild colitis. Reconstitution with CD62L- T cells induces fulminant diabetes with no colitis or gastritis. Reconstitution with CD45RB(high) T cells induces major colitis with wasting disease and no or very moderate gastritis and diabetes. Major differences among the three regulatory T cell subsets are also seen in vitro. The bulk of suppressor cells inhibiting the proliferation of CD4+CD25- T cells in coculture is concentrated within the CD25+ but not the CD62L+ or CD45RB(low) T cell subsets. Similarly, cytokine production patterns are significantly different for each regulatory T cell subset. Collectively, these data point to the diversity and organ selectivity of regulatory T cells controlling distinct autoimmune diseases whatever the underlying mechanisms.
Collapse
Affiliation(s)
- Marie-Alexandra Alyanakian
- Institut National de la Santé et de la Recherche Médicale U580, and Institut Fédératif Necker Enfants Malades, Faculté Necker Enfants Malades, 161 Rue de Sèvres, 75015 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Belghith M, Bluestone JA, Barriot S, Mégret J, Bach JF, Chatenoud L. TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat Med 2003; 9:1202-8. [PMID: 12937416 DOI: 10.1038/nm924] [Citation(s) in RCA: 474] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2003] [Accepted: 08/04/2003] [Indexed: 02/01/2023]
Abstract
CD3-specific antibodies have the unique capacity to restore self-tolerance in established autoimmunity. They induce long-term remission of overt diabetes in nonobese diabetic (NOD) mice and in human type I diabetes. The underlying mechanisms had been unclear until now. Here we report that treatment with CD3epsilon-specific antibodies induces transferable T-cell-mediated tolerance involving CD4+CD25+ cells. However, these CD4+CD25+ T cells are distinct from naturally occurring regulatory T cells that control physiological autoreactivity. CD3-specific antibody treatment induced remission in NOD Cd28-/- mice that were devoid of such regulatory cells. Remission of diabetes was abrogated by coadministration of a neutralizing transforming growth factor (TGF)-beta-specific antibody. The central role of TGF-beta was further suggested by its increased, long-lasting production by CD4+ T cells from tolerant mice. These data explain the intriguing tolerogenic effect of CD3-specific antibodies and position them as the first clinically applicable pharmacological stimulant of TGF-beta-producing regulatory CD4+ T cells.
Collapse
Affiliation(s)
- Mériam Belghith
- INSERM U580, IRNEM, Hôpital Necker, 161 Rue de Sèvres, 75015 Paris, France
| | | | | | | | | | | |
Collapse
|
22
|
Aspord C, Czerkinsky C, Durand A, Stefanutti A, Thivolet C. alpha4 integrins and L-selectin differently orchestrate T-cell activity during diabetes prevention following oral administration of CTB-insulin. J Autoimmun 2002; 19:223-32. [PMID: 12473243 DOI: 10.1006/jaut.2002.0610] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oral administration of insulin conjugated to the B chain of cholera toxin (CTB-insulin) in non-obese diabetic (NOD) mice results in diabetes prevention. We investigated the respective contributions of L-selectin (CD62L) and alpha4-integrin pathways during CTB-driven tolerance. Purified CD62L+CD4+ cells from CTB-insulin fed mice significantly reduced the capacity of diabetogenic T cells to transfer diabetes in syngeneic recipients. In vivo antibody blockade of fed animals during adoptive co-transfer experiments indicated that both CD62L and alpha4-integrins pathways were necessary to develop a protective response after oral tolerance induction. In contrast, when antibodies were given to recipient mice, only CD62L was critical for the protection. In vitro stimulated CD62L+CD4+ cells from the spleen of fed animals secreted lower amounts of IL-4 and IL-10 but comparable levels of TGFbeta than CD62L-cells. A reduced IFN-gamma production between the two cell subsets was specifically observed in CTB-insulin fed mice. Furthermore, antibody treatments induced changes in T-cell migration to the spleen, mesenteric and pancreatic lymph nodes. The protective effect was also associated with migration of regulatory T cells into pancreatic islets. Taken together, our results suggest that L-selectin and alpha4-integrin have distinct but complementary roles in the generation and function of regulatory CD4+ T cells following CTB-insulin administration.
Collapse
|
23
|
Friedline RH, Wong CP, Steeber DA, Tedder TF, Tisch R. L-selectin is not required for T cell-mediated autoimmune diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:2659-66. [PMID: 11884430 DOI: 10.4049/jimmunol.168.6.2659] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Administration of anti-L-selectin (CD62L) mAb to neonatal nonobese diabetic (NOD) mice mediates long term protection against the development of insulitis and overt diabetes. These results suggested that CD62L has a key role in the general function of beta cell-specific T cells. To further examine the role of CD62L in the development of type 1 diabetes, NOD mice lacking CD62L were established. The onset and frequency of overt diabetes were equivalent among CD62L(+/+), CD62L(+/-), and CD62L(-/-) NOD littermates. Furthermore, patterns of T cell activation, migration, and beta cell-specific reactivity were similar in NOD mice of all three genotypes. Adoptive transfer experiments with CD62L(-/-) CD4(+) T cells prepared from BDC2.5 TCR transgenic mice revealed no apparent defects in migration to pancreatic lymph nodes, proliferation in response to beta cell Ag, or induction of diabetes in NOD.scid recipients. In conclusion, CD62L expression is not essential for the development of type 1 diabetes in NOD mice.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Cell Movement/genetics
- Cell Movement/immunology
- Cells, Cultured
- Diabetes Mellitus, Type 1/etiology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Epitopes, T-Lymphocyte/immunology
- Female
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- L-Selectin/biosynthesis
- L-Selectin/genetics
- L-Selectin/physiology
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymphocyte Activation/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Mice, Transgenic
- Pancreas/cytology
- Pancreas/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/transplantation
Collapse
Affiliation(s)
- Randall H Friedline
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
24
|
Martin S, van den Engel NK, Vinke A, Heidenthal E, Schulte B, Kolb H. Dominant role of intercellular adhesion molecule-1 in the pathogenesis of autoimmune diabetes in non-obese diabetic mice. J Autoimmun 2001; 17:109-17. [PMID: 11591119 DOI: 10.1006/jaut.2001.0526] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Intercellular adhesion molecule (ICAM)-1 is involved in forming the immunological synapse. The contribution of ICAM-1 to immune responses is not critical because mice with a disrupted ICAM-1 gene do not have grossly abnormal immune reactivity. Here we report on the surprising finding that diabetes-prone NOD mice with a disrupted ICAM-1 gene (ICAM-1(-/-)) are completely protected from disease development. While 64% of ICAM-1(+/+) and 44% of ICAM-1(+/-) female NOD mice developed overt diabetes until 310 days old, no ICAM-1(-/-) NOD mice became hyperglycaemic. Histological examinations revealed minor infiltration around pancreatic islets of ICAM1(-/-) NOD mice. Administration of cyclophosphamide caused a progression to severe islet destruction in ICAM-1(+/+) NOD mice within 10 days. In contrast, ICAM-1(-/-) mice showed only mild insulitis. Furthermore, ICAM-1(+/+) NOD mice showed an increase of IFN-gamma, interleukin (IL)-12p40 and IL-12p35 pancreatic mRNA levels, leading to an increased ratio of IFN-gamma: IL-4 and IL-12p40: IL-12p35 expression. In contrast, ICAM-1(-/-) NOD mice did not upregulate IFN-gamma or IL-12p40 gene expression but maintained IL-4 and increased IL-12p35 gene expression. These results identify a dominant and non-redundant role of ICAM-1 in the development of autoimmune diabetes.
Collapse
Affiliation(s)
- S Martin
- German Diabetes Centre, German Diabetes Research Institute at the Heinrich Heine University, Düsseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
25
|
Sharif S, Arreaza GA, Zucker P, Mi QS, Sondhi J, Naidenko OV, Kronenberg M, Koezuka Y, Delovitch TL, Gombert JM, Leite-De-Moraes M, Gouarin C, Zhu R, Hameg A, Nakayama T, Taniguchi M, Lepault F, Lehuen A, Bach JF, Herbelin A. Activation of natural killer T cells by alpha-galactosylceramide treatment prevents the onset and recurrence of autoimmune Type 1 diabetes. Nat Med 2001; 7:1057-62. [PMID: 11533711 DOI: 10.1038/nm0901-1057] [Citation(s) in RCA: 469] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Type 1 diabetes (T1D) in non-obese diabetic (NOD) mice may be favored by immune dysregulation leading to the hyporesponsiveness of regulatory T cells and activation of effector T-helper type 1 (Th1) cells. The immunoregulatory activity of natural killer T (NKT) cells is well documented, and both interleukin (IL)-4 and IL-10 secreted by NKT cells have important roles in mediating this activity. NKT cells are less frequent and display deficient IL-4 responses in both NOD mice and individuals at risk for T1D (ref. 8), and this deficiency may lead to T1D (refs. 1,6-9). Thus, given that NKT cells respond to the alpha-galactosylceramide (alpha-GalCer) glycolipid in a CD1d-restricted manner by secretion of Th2 cytokines, we reasoned that activation of NKT cells by alpha-GalCer might prevent the onset and/or recurrence of T1D. Here we show that alpha-GalCer treatment, even when initiated after the onset of insulitis, protects female NOD mice from T1D and prolongs the survival of pancreatic islets transplanted into newly diabetic NOD mice. In addition, when administered after the onset of insulitis, alpha-GalCer and IL-7 displayed synergistic effects, possibly via the ability of IL-7 to render NKT cells fully responsive to alpha-GalCer. Protection from T1D by alpha-GalCer was associated with the suppression of both T- and B-cell autoimmunity to islet beta cells and with a polarized Th2-like response in spleen and pancreas of these mice. These findings raise the possibility that alpha-GalCer treatment might be used therapeutically to prevent the onset and recurrence of human T1D.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, CD1/genetics
- Cyclophosphamide/toxicity
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/prevention & control
- Galactosylceramides/pharmacology
- Interferon-gamma/metabolism
- Interleukin-4/metabolism
- Interleukin-7/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- L-Selectin/metabolism
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, Mutant Strains
- Receptors, Interleukin/drug effects
- Receptors, Interleukin/immunology
- Receptors, Interleukin-10
- Spleen/drug effects
- Spleen/metabolism
Collapse
Affiliation(s)
- S Sharif
- Autoimmunity/Diabetes Group, The John P. Robarts Research Institute, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
The Th1/Th2 concept brought an attractive explanation of the active self tolerance which appears to control the onset of pathogenic autoimmunity. New data coming from various independent horizons indicate that self immunoregulation could also depend to a large extent on non-Th2 cells. Original data derived from the day-3-thymectomy model, selective T-cell lymphocytopenia and nonobese diabetic mice are discussed in an effort to analyze similarities and differences in phenotype (CD25, CD62L and CD45RB) and cytokine pattern (notably interleukin (IL)-4, IL-10 and transforming growth factor (TGF)beta) of regulatory cells involved in these models. The relationship of these cells with Th3, Tr1 and natural killer (NK) T cells are also discussed. The hypothesis is proposed that CD25 CD62L T cells mediate the physiologic regulation of self regulation whereas Th2 and Th3 cells are essentially induced following sensitization against autoantigens.
Collapse
Affiliation(s)
- J F Bach
- INSERM U 25, Hôpital Necker, Paris, France.
| |
Collapse
|
27
|
Mahiou J, Walter U, Lepault F, Godeau F, Bach JF, Chatenoud L. In vivo blockade of the Fas-Fas ligand pathway inhibits cyclophosphamide-induced diabetes in NOD mice. J Autoimmun 2001; 16:431-40. [PMID: 11437491 DOI: 10.1006/jaut.2000.0476] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
There is compelling evidence to show that insulin dependent diabetes ensues from selective apoptosis of pancreatic beta-cells mediated by autoreactive T-lymphocytes. The respective implication in this phenomenon of the various apoptotic pathways driven by Fas, perforin, or tumor necrosis factor is still ill- defined. Here we took advantage of the cyclophosphamide-induced model of accelerated diabetes in NOD mice to explore the physiopathological role of the Fas-Fas Ligand pathway. A single injection of cyclophosphamide (200 mg/kg) to 7-8 week-old prediabetic NOD mice triggered diabetes within 10-15 days in 85-100% of the animals. Cyclophosphamide also induced a significant decrease in spleen T cells, that was most evident by days 6-10 after treatment, and selectively affected the CD3(+)CD62L(+)compartment that includes immunoregulatory T cells. To block the in vivo Fas-Fas ligand (Fas L) interaction we administered a biologically active recombinant fusion protein coupling mouse Fas to the Fc portion of human IgG1 (FAS-Fc). Mice treated with FAS-Fc (10 doses iv of 15 microg) starting on the day of cyclophosphamide injection up to day 22, were fully protected from disease. Unexpectedly this protective effect was not due to blockade of Fas-FasL-mediated beta-cell apoptosis but rather to the inhibition of the cyclophosphamide effect on T cells. Indeed FAS-Fc treatment prevented the drug-induced T cell depletion in general and that of immunoregulatory T cells in particular. Additionally, FAS-Fc administration limited to the phase of beta-cell destruction did not afford any protection.
Collapse
Affiliation(s)
- J Mahiou
- INSERM U 25, Hôpital Necker, 161 rue de Sèvres, Paris, 75015, France
| | | | | | | | | | | |
Collapse
|
28
|
Yamamoto AM, Chernajovsky Y, Lepault F, Podhajcer O, Feldmann M, Bach JF, Chatenoud L. The activity of immunoregulatory T cells mediating active tolerance is potentiated in nonobese diabetic mice by an IL-4-based retroviral gene therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:4973-80. [PMID: 11290776 DOI: 10.4049/jimmunol.166.8.4973] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Splenocytes from nonobese diabetic mice overexpressing murine IL (mIL)-4 upon recombinant retrovirus infection lose their capacity to transfer diabetes to nonobese diabetic-scid recipients. Diabetes appeared in 0-20% of mice injected with mIL-4-transduced cells vs 80-100% of controls injected with beta-galactosidase-transduced cells. Protected mice showed a majority of islets (60%) presenting with noninvasive peri-insulitis at variance with beta-galactosidase controls that exhibited invasive/destructive insulitis. Importantly, in all recipients, the transduced proteins were detected within islet infiltrates. Infiltrating lymphocytes from recipients of mIL-4-transduced cells produced high levels of mIL-4, as assessed by ELISA. In recipients of beta-galactosidase-transduced cells, approximately 60% of TCRalphabeta(+) islet-infiltrating cells expressed beta-galactosidase, as assessed by flow cytometry. The protection from disease transfer is due to a direct effect of mIL-4 gene therapy on immunoregulatory T cells rather than on diabetogenic cells. mIL-4-transduced purified CD62L(-) effector cells or transgenic BDC2.5 diabetogenic T cells still transferred disease efficiently. Conversely, mIL-4 transduction up-regulated the capacity of purified immunoregulatory CD62L(+) cells to inhibit disease transfer. These data open new perspectives for gene therapy in insulin-dependent diabetes using T cells devoid of any intrinsic diabetogenic potential.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Adoptive Transfer
- Animals
- Cell Movement/immunology
- Cells, Cultured
- Clone Cells
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Female
- Genetic Therapy/methods
- Genetic Vectors/administration & dosage
- Genetic Vectors/immunology
- Immune Tolerance/genetics
- Immunity, Active/genetics
- Interleukin-4/administration & dosage
- Interleukin-4/biosynthesis
- Interleukin-4/genetics
- Islets of Langerhans/pathology
- L-Selectin/biosynthesis
- Lymphocyte Transfusion
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Retroviridae/genetics
- Retroviridae/immunology
- Spleen/cytology
- Spleen/immunology
- Spleen/transplantation
- Spleen/virology
- T-Lymphocyte Subsets/enzymology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/transplantation
- T-Lymphocyte Subsets/virology
- Transgenes/immunology
- beta-Galactosidase/administration & dosage
- beta-Galactosidase/biosynthesis
- beta-Galactosidase/genetics
Collapse
Affiliation(s)
- A M Yamamoto
- Institut National de la Santé et de la Recherche Médicale Unité 25, and Centre National de la Recherche Scientifique Unité MR8603, Hôpital Necker, Paris, France
| | | | | | | | | | | | | |
Collapse
|
29
|
Grewal IS, Foellmer HG, Grewal KD, Wang H, Lee WP, Tumas D, Janeway CA, Flavell RA. CD62L is required on effector cells for local interactions in the CNS to cause myelin damage in experimental allergic encephalomyelitis. Immunity 2001; 14:291-302. [PMID: 11290338 DOI: 10.1016/s1074-7613(01)00110-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Adhesion molecules are believed to facilitate infiltration of leukocytes into the CNS of mice with experimental allergic encephalomyelitis (EAE). The role of the adhesion molecule CD62L (L-selectin) in the immunopathology of EAE is not known. To study this, we crossed CD62L-deficient mice with myelin basic protein-specific TCR (MBP-TCR) transgenic mice. CD62L-deficient MBP-TCR transgenic mice failed to develop antigen-induced EAE, and, despite the presence of leukocyte infiltration, damage to myelin in the CNS was not seen. EAE could, however, be induced in CD62L-deficient mice upon adoptive transfer of wild-type macrophages. Our results suggest that CD62L is not required for activation of autoimmune CD4 T cells but is important for the final destructive function of effector cells in the CNS and support a novel mechanism whereby CD62L expressed on effector cells is important in mediating myelin damage.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigen-Presenting Cells/transplantation
- Autoimmunity/genetics
- Autoimmunity/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- Cell Adhesion
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Chemotaxis, Leukocyte
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Deletion
- Immunohistochemistry
- L-Selectin/genetics
- L-Selectin/metabolism
- Lymphocyte Activation
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/transplantation
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myelin Basic Protein/genetics
- Myelin Basic Protein/immunology
- Myelin Basic Protein/metabolism
- Myelin Sheath/metabolism
- Myelin Sheath/pathology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- I S Grewal
- Department of Immunology, Genentech Incorporated, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Dobbs C, Haskins K. Comparison of a T cell clone and of T cells from a TCR transgenic mouse: TCR transgenic T cells specific for self-antigen are atypical. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:2495-504. [PMID: 11160310 DOI: 10.4049/jimmunol.166.4.2495] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It has been widely assumed that T cells from TCR-transgenic (Tg) mice better represent the behavior of T cells from normal mice than do in vitro cultures of T cell clones. We have found that autoreactive T cells arising in the presumably more physiological environment of the BDC-2.5 TCR-Tg mouse, despite being apparently "naive" in surface phenotype, are highly activated functionally and do not resemble CD4(+) T cells from a spontaneously diabetic nonobese diabetic (NOD) mouse or the NOD-derived, diabetogenic CD4(+) T cell clone of origin, BDC-2.5. Our results suggest that autoreactive T cells cloned from the spontaneously diabetic NOD mouse more closely resemble effector T cells arising during the natural disease process.
Collapse
MESH Headings
- Adoptive Transfer
- Aging/genetics
- Aging/immunology
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/genetics
- Autoantigens/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Clone Cells
- Cytokines/biosynthesis
- Cytotoxicity, Immunologic/genetics
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Immunophenotyping
- Injections, Intravenous
- Integrins/biosynthesis
- Integrins/genetics
- Interphase/genetics
- Interphase/immunology
- L-Selectin/immunology
- Leukemia L1210
- Lymphocyte Activation/genetics
- Membrane Glycoproteins/biosynthesis
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Perforin
- Phosphorylation
- Phosphotyrosine/metabolism
- Pore Forming Cytotoxic Proteins
- Prediabetic State/genetics
- Prediabetic State/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Spleen/immunology
- Spleen/metabolism
- Spleen/pathology
- Spleen/transplantation
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Thymus Gland/pathology
- Thymus Gland/transplantation
- Transfection
- Transgenes/immunology
- fas Receptor/genetics
Collapse
Affiliation(s)
- C Dobbs
- Department of Immunology and The Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Denver, CO 80262, USA
| | | |
Collapse
|
31
|
Singh B, Delovitch TL. Immune mechanisms that regulate susceptibility to autoimmune type I diabetes. Clin Rev Allergy Immunol 2000; 19:247-64. [PMID: 11138408 DOI: 10.1385/criai:19:3:247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- B Singh
- Department of Microbiology, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | |
Collapse
|
32
|
Flynn JC, McInerney MF. High density insulin receptor-positive diabetogenic T lymphocytes in nonobese diabetic mice are memory cells. Immunopharmacol Immunotoxicol 2000; 22:387-400. [PMID: 10952038 DOI: 10.3109/08923970009016427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Our previous work examining the importance of insulin receptor (IR) expression on T cells has demonstrated that when T cells from nonobese diabetic mice were sorted into populations expressing a high (IR(High)) and a low (IR(Low)) density of IR, IR(High) T cells rapidly transferred insulitis and diabetes. We have further characterized IR(High) T cells. Both CD4+ and CD8+ cells were detected in the IR(High) T cell population, but IR(High) expression was detected predominantly on CD4+ cells. IRHigh T cells were polyclonal for TCR Vbeta-chain expression. By 3 color flow cytometric analysis, virtually all IR(High) T cells expressed low or negligible levels of CD62L (CD62L(Low)/-) and high levels of CD44 (CD44(High)). The lack of IL-2 receptor and transferrin receptor expression as seen previously, together with the CD62L(Low)/- CD44(High) phenotype suggests that IR(High) T cells are memory cells. However, since only about one quarter of all of the CD62L(Low)/- or CD44(High) T cells were also IR(High), the IR(High) phenotype defines a subpopulation of memory T cells that are aggressively diabetogenic.
Collapse
Affiliation(s)
- J C Flynn
- Department of Medicinal and Biological Chemistry, University of Toledo, College of Pharmacy, OH 43606, USA
| | | |
Collapse
|
33
|
Lepault F, Gagnerault MC. Characterization of peripheral regulatory CD4+ T cells that prevent diabetes onset in nonobese diabetic mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:240-7. [PMID: 10605017 DOI: 10.4049/jimmunol.164.1.240] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The period that precedes onset of insulin-dependent diabetes mellitus corresponds to an active dynamic state in which pathogenic autoreactive T cells are kept from destroying beta cells by regulatory T cells. In prediabetic nonobese diabetic (NOD) mice, CD4+ splenocytes were shown to prevent diabetes transfer in immunodeficient NOD recipients. We now demonstrate that regulatory splenocytes belong to the CD4+ CD62Lhigh T cell subset that comprises a vast majority of naive cells producing low levels of IL-2 and IFN-gamma and no IL-4 and IL-10 upon in vitro stimulation. Consistently, the inhibition of diabetes transfer was not mediated by IL-4 and IL-10. Regulatory cells homed to the pancreas and modified the migration of diabetogenic to the islets, which resulted in a decreased insulitis severity. The efficiency of CD62L+ T cells was dose dependent, independent of sex and disease prevalence. Protection mechanisms did not involve the CD62L molecule, an observation that may relate to the fact that CD4+ CD62Lhigh lymph node cells were less potent than their splenic counterparts. Regulatory T cells were detectable after weaning and persist until disease onset, sustaining the notion that diabetes is a late and abrupt event. Thus, the CD62L molecule appears as a unique marker that can discriminate diabetogenic (previously shown to be CD62L-) from regulatory T cells. The phenotypic and functional characteristics of protective CD4+ CD62L+ cells suggest they are different from Th2-, Tr1-, and NK T-type cells, reported to be implicated in the control of diabetes in NOD mice, and may represent a new immunoregulatory population.
Collapse
Affiliation(s)
- F Lepault
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8603, Université Paris V, Hôpital Necker, Paris, France.
| | | |
Collapse
|
34
|
Savilahti E, Ormälä T, Saukkonen T, Sandini-Pohjavuori U, Kantele JM, Arato A, Ilonen J, Akerblom HK. Jejuna of patients with insulin-dependent diabetes mellitus (IDDM) show signs of immune activation. Clin Exp Immunol 1999; 116:70-7. [PMID: 10209507 PMCID: PMC1905225 DOI: 10.1046/j.1365-2249.1999.00860.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The roles of enteric viruses and food antigens as possible triggers in human insulin-dependent diabetes mellitus and the evidence that mucosal-associated homing receptors are important in both human and experimental diabetes prompted us to undertake an immunohistochemical study of intestinal specimens from patients with IDDM. We studied jejunal morphology and immunohistochemistry in 26 patients with IDDM, 13 of whom had the HLA-DQB1*0201 gene and therefore a higher risk of coeliac disease. The findings were compared with those in specimens from age-matched controls. Villous structure and the density of the intraepithelial lymphocytes were normal in every biopsy specimen. The extent of positivity with anti-DR and -DP antibodies in the villous epithelium was significantly greater in the specimens from patients than in those from controls (P = 0.0002 in both comparisons). The crypts were also more positive: for DR P = 0.0001, and for DP P = 0.002. The densities of T cells, CD4+, CD8+, and T cell receptor alpha/beta+ and gamma/delta+ cells in the epithelium and lamina propria were similar in patients and controls, but the patients had significantly more alpha 4/beta 7 integrin+ cells in the lamina propria (P = 0.006). No difference was seen between HLA-DQB1*0201-positive and -negative patients. These findings reflect a stage of inflammation in the structurally normal intestines of patients with IDDM and suggest secretion of inflammatory Th1-type cytokines in the intestine.
Collapse
Affiliation(s)
- E Savilahti
- Hospital for Children and Adolescents, University of Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Herbelin A, Gombert JM, Lepault F, Bach JF, Chatenoud L. Mature Mainstream TCRαβ+CD4+ Thymocytes Expressing L-Selectin Mediate “Active Tolerance” in the Nonobese Diabetic Mouse. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.5.2620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Pathogenic autoreactive T lymphocytes are mediators of spontaneous insulin-dependent diabetes in nonobese diabetic (NOD) mice. This is demonstrated by their capacity to transfer diabetes into syngeneic immunoincompetent recipients. In addition, especially in prediabetic NOD mice, peripheral CD4+ T lymphocytes were identified that are highly effective, in conventional mixing cotransfer experiments, at preventing disease transfer. The present data demonstrate that mature heat-stable Ag−TCRαβ+CD8− thymocytes from prediabetic NOD mice also express this inhibitory capacity. Selection using an L-selectin (CD62L)-specific Ab showed that TCRαβ+CD4+CD62L+ thymocytes, emerging from the mainstream differentiation pathway, concentrate this ability to regulate autoreactive effectors. Compared with mature TCRαβ+CD8− thymocytes, significantly lower numbers of TCRαβ+CD4+CD62L+ were sufficient to achieve an efficient inhibition of disease transfer into NOD-scid recipients. This protective ability was potentiated following in vitro culture in the presence of IL-7. In contrast, TCRαβ+CD62L− thymocytes, highly enriched in class I-restricted NK T cells, were unable to influence diabetes transfer. Identical results were obtained using thymocytes that have been cultured in vitro for 4 days in the presence of IL-7. These results support the active role in NOD mice of a thymus-derived CD4+ subset that controls peripheral pathogenic autoimmune effectors.
Collapse
Affiliation(s)
- André Herbelin
- *Institut National de la Santé et de la Recherche Médicale, Unité 25, and
| | - Jean-Marc Gombert
- *Institut National de la Santé et de la Recherche Médicale, Unité 25, and
| | - Françoise Lepault
- †Centre National de la Recherche Scientifique, Unité de Recherche Associée 1461, Hôpital Necker, Paris, France
| | - Jean-François Bach
- *Institut National de la Santé et de la Recherche Médicale, Unité 25, and
| | - Lucienne Chatenoud
- *Institut National de la Santé et de la Recherche Médicale, Unité 25, and
| |
Collapse
|
36
|
Michie SA, Sytwu HK, McDevitt JO, Yang XD. The roles of alpha 4-integrins in the development of insulin-dependent diabetes mellitus. Curr Top Microbiol Immunol 1998; 231:65-83. [PMID: 9479861 DOI: 10.1007/978-3-642-71987-5_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lymphocyte/endothelial adhesion followed by transendothelial migration is a key event in the development of organ-specific autoimmunity. Selective interactions of cell surface AM regulate lymphocyte migration under normal as well as pathologic inflammatory conditions. NOD mice are an ideal model for investigating the roles of AM in regulation of lymphocyte migration to target organs in autoimmune diseases such as IDDM. Both in vitro and in vivo studies in NOD mice strongly suggest that the mucosal (alpha 4 beta 7/MAdCAM-1) adhesion system and alpha 4-integrin/VCAM-1 appear to be prominent pathways for insulitis development. In contrast, alpha 4-mediated interactions in NOD inflamed salivary and lacrimal gland and in the inflamed CNS of rodents with EAE seem to be dominated by alpha 4-integrins and VCAM-1. The fact that blocking alpha 4-integrin pathways in NOD mice leads to successful interruption of the diabetogenic process suggests that AM provide a potential therapeutic target for human IDDM. Further studies on IDDM patients will prove helpful for understanding IDDM pathogenesis and in providing a basis for designing AM-based therapeutic approaches.
Collapse
Affiliation(s)
- S A Michie
- Department of Veterans Affairs, Palo Alto Health Care System, CA 94304, USA
| | | | | | | |
Collapse
|