1
|
Zhang M, Jiang W, Lu G, Wang R, Lv Z, Li D. Insight Into Mouse Models of Hyperthyroidism. Front Endocrinol (Lausanne) 2022; 13:929750. [PMID: 35813642 PMCID: PMC9257255 DOI: 10.3389/fendo.2022.929750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/19/2022] [Indexed: 11/26/2022] Open
Abstract
Hyperthyroidism is characterized by an increase in the synthesis and secretion of thyroid hormones in the thyroid gland, and the most common cause of overproduction of thyroid hormones is Graves' disease (GD). Long-term disease models of hyperthyroidism have been established. In general, methods to induce GD include transfection of fibroblasts, injecting plasmids or adenovirus containing thyroid stimulating hormone receptor (TSHR) or TSHR subunit, and exogenous artificial thyroid hormone supplementation. Fortunately, in mouse studies, novel treatments for GD and Graves' orbitopathy (GO) were discovered. It has been reported that prophylactic administration of TSHR A subunit protein in genetically susceptible individuals could induce immune tolerance and provide protection for the future development of GD. Biologically active monoclonal antibody against intracellular adhesion molecule-1 (ICAM-1 mAb) and siRNA targeting TSHR can also be used to treat GD. Moreover, new potential therapeutic targets have been identified in GO mouse models, and these targets could present novel therapeutic approaches. Besides, human placental mesenchymal stem cells (hPMSCs) into the orbit, fucoxanthin and icariin may be new alternative therapies that could be used in addition to the existing drugs, although further research is needed.
Collapse
Affiliation(s)
- Mengyu Zhang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen Jiang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ganghua Lu
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ru Wang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongwei Lv
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Imaging Clinical Medical Center, Tongji University School of Medicine, Shanghai, China
- Clinical Nuclear Medicine Center, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Dan Li, ; Zhongwei Lv,
| | - Dan Li
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Imaging Clinical Medical Center, Tongji University School of Medicine, Shanghai, China
- Clinical Nuclear Medicine Center, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Dan Li, ; Zhongwei Lv,
| |
Collapse
|
2
|
Degen H, Gavvovidis I, Blankenstein T, Uhland K, Ungerer M. Thyrotropin Receptor-Specific Lymphocytes in Adenovirus-TSHR-Immunized Native and Human Leukocyte Antigen-DR3-Transgenic Mice and in Graves' Disease Patient Blood. Thyroid 2021; 31:950-963. [PMID: 33208049 DOI: 10.1089/thy.2020.0338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background: Antigen-specific lymphocytes are increasingly investigated in autoimmune diseases and immune therapies. We sought to identify thyrotropin receptor (TSHR)-specific lymphocytes in mouse models of Graves' disease, including Graves' patient-specific immunotype human leukocyte antigen (HLA)-DR3, and in frozen and thawed Graves' patient blood samples. Methods and Results: Splenic lymphocytes of adenovirus (Ad)-TSHR-immunized BALB/c mice were stimulated with TSHR-specific peptides C, D, or J. Furthermore, CD154-expressing cells were enriched, expanded in vitro, and analyzed for binding of peptide-major histocompatibility complex (MHC) II multimers ("tetramers," immunotype H2-IAd). Only peptides C and J were able to elicit increased expression/secretion of CD154 and interferon-γ, and tetramers which were loaded with peptide C resulted in antigen-specific signals in splenic lymphocytes from Ad-TSHR-immunized mice. Accordingly, TSHR-specific HLA-DR3-MHC class II tetramers loaded with peptide p10 specifically bound to human HLA-DR3-(allele B1*03:01)-transgenic Bl/6 mouse splenic T lymphocytes. In addition, we fine-tuned a protocol to reliably measure thawed human peripheral blood mononuclear cells (PBMCs), which resulted in reliable recovery after freezing and thawing with regard to vitality and B and T cell subpopulation markers including regulatory T cells (CD3, CD4, CD25, FoxP3, CD25high, CD127low). TSHR-specific HLA-DR3-MHC class II tetramers loaded with peptide p10 identified antigen-specific T cells in HLA-DR3-positive Graves' patients' thawed PBMCs. Moreover, stimulation-dependent release of interleukin (IL)-1beta, IL-6, tumor necrosis factor-alpha from thawed PBMCs occurred at the expected levels. Conclusions: Novel MHC II tetramers identified TSHR-specific T lymphocytes in Ad-TSHR-immunized hyperthyroid BALB/c or HLA-DR3-transgenic mice and in thawed human PBMCs from patients with Graves' disease. These assays may contribute to measure both disease severity and effects of novel immune therapies in future animal studies and clinical investigations of Graves' disease.
Collapse
Affiliation(s)
| | - Ioannis Gavvovidis
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch, Germany
- Department of Immunology, Charite - Universitätsmedizin, Berlin, Germany
| | - Thomas Blankenstein
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch, Germany
- Department of Immunology, Charite - Universitätsmedizin, Berlin, Germany
| | | | | |
Collapse
|
3
|
Breaking tolerance in transgenic mice expressing the human TSH receptor A-subunit: thyroiditis, epitope spreading and adjuvant as a 'double edged sword'. PLoS One 2012; 7:e43517. [PMID: 22970131 PMCID: PMC3436763 DOI: 10.1371/journal.pone.0043517] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 07/23/2012] [Indexed: 02/05/2023] Open
Abstract
Transgenic mice with the human thyrotropin-receptor (TSHR) A-subunit targeted to the thyroid are tolerant of the transgene. In transgenics that express low A-subunit levels (Lo-expressors), regulatory T cell (Treg) depletion using anti-CD25 before immunization with adenovirus encoding the A-subunit (A-sub-Ad) breaks tolerance, inducing extensive thyroid lymphocytic infiltration, thyroid damage and antibody spreading to other thyroid proteins. In contrast, no thyroiditis develops in Hi-expressor transgenics or wild-type mice. Our present goal was to determine if thyroiditis could be induced in Hi-expressor transgenics using a more potent immunization protocol: Treg depletion, priming with Complete Freund's Adjuvant (CFA) + A-subunit protein and further Treg depletions before two boosts with A-sub-Ad. As controls, anti-CD25 treated Hi- and Lo-expressors and wild-type mice were primed with CFA+ mouse thyroglobulin (Tg) or CFA alone before A-sub-Ad boosting. Thyroiditis developed after CFA+A-subunit protein or Tg and A-sub-Ad boosting in Lo-expressor transgenics but Hi- expressors (and wild-type mice) were resistant to thyroiditis induction. Importantly, in Lo-expressors, thyroiditis was associated with the development of antibodies to the mouse TSHR downstream of the A-subunit. Unexpectedly, we observed that the effect of bacterial products on the immune system is a “double-edged sword”. On the one hand, priming with CFA (mycobacteria emulsified in oil) plus A-subunit protein broke tolerance to the A-subunit in Hi-expressor transgenics leading to high TSHR antibody levels. On the other hand, prior treatment with CFA in the absence of A-subunit protein inhibited responses to subsequent immunization with A-sub-Ad. Consequently, adjuvant activity arising in vivo after bacterial infections combined with a protein autoantigen can break self-tolerance but in the absence of the autoantigen, adjuvant activity can inhibit the induction of immunity to autoantigens (like the TSHR) displaying strong self-tolerance.
Collapse
|
4
|
Urothelial antigen-specific CD4+ T cells function as direct effector cells and induce bladder autoimmune inflammation independent of CD8+ T cells. Mucosal Immunol 2011; 4:428-37. [PMID: 21270773 PMCID: PMC3118865 DOI: 10.1038/mi.2010.90] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The role of CD4(+) T cells in bladder autoimmune inflammation has not been identified because of the lack of a proper animal model. We investigated CD4(+) T-cell responses to bladder urothelial ovalbumin (OVA), a model self-antigen (Ag), in transgenic URO-OVA mice. The expression of bladder urothelial OVA rendered mice unresponsive to OVA and resulted in quick clearance of Ag-specific CD4(+) T cells. Adoptive transfer of naive OVA-specific CD4(+) T cells led to exogenous T-cell proliferation, activation, and bladder infiltration but no inflammatory induction. In contrast, adoptive transfer of preactivated OVA-specific CD4(+) T cells induced bladder inflammation. Studies further demonstrated that CD4(+) T cells induced bladder inflammation in URO-OVA mice depleted of CD8(+) T cells or deficient in the recombinase activating gene-1 (Rag-1(-/-)). These results indicate that urothelial Ag-specific CD4(+) T cells can function as direct effector cells to induce bladder autoimmune inflammation independent of CD8(+) T cells.
Collapse
|
5
|
Mizutori Y, Nagayama Y, Flower D, Misharin A, Aliesky HA, Rapoport B, McLachlan SM. Role of the transgenic human thyrotropin receptor A-subunit in thyroiditis induced by A-subunit immunization and regulatory T cell depletion. Clin Exp Immunol 2008; 154:305-15. [PMID: 18811696 DOI: 10.1111/j.1365-2249.2008.03769.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Transgenic BALB/c mice that express intrathyroidal human thyroid stimulating hormone receptor (TSHR) A-subunit, unlike wild-type (WT) littermates, develop thyroid lymphocytic infiltration and spreading to other thyroid autoantigens after T regulatory cell (T(reg)) depletion and immunization with human thyrotropin receptor (hTSHR) adenovirus. To determine if this process involves intramolecular epitope spreading, we studied antibody and T cell recognition of TSHR ectodomain peptides (A-Z). In transgenic and WT mice, regardless of T(reg) depletion, TSHR antibodies bound predominantly to N-terminal peptide A and much less to a few downstream peptides. After T(reg) depletion, splenocytes from WT mice responded to peptides C, D and J (all in the A-subunit), but transgenic splenocytes recognized only peptide D. Because CD4(+) T cells are critical for thyroid lymphocytic infiltration, amino acid sequences of these peptides were examined for in silico binding to BALB/c major histocompatibility complex class II (IA-d). High affinity subsequences (inhibitory concentration of 50% < 50 nm) are present in peptides C and D (not J) of the hTSHR and mouse TSHR equivalents. These data probably explain why transgenic splenocytes do not recognize peptide J. Mouse TSHR mRNA levels are comparable in transgenic and WT thyroids, but only transgenics have human A-subunit mRNA. Transgenic mice can present mouse TSHR and human A-subunit-derived peptides. However, WT mice can present only mouse TSHR, and two to four amino acid species differences may preclude recognition by CD4+ T cells activated by hTSHR-adenovirus. Overall, thyroid lymphocytic infiltration in the transgenic mice is unrelated to epitopic spreading but involves human A-subunit peptides for recognition by T cells activated using the hTSHR.
Collapse
Affiliation(s)
- Y Mizutori
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and UCLA School of Medicine, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Muixí L, Alvarez I, Jaraquemada D. Peptides presented in vivo by HLA-DR in thyroid autoimmunity. Adv Immunol 2008; 99:165-209. [PMID: 19117535 DOI: 10.1016/s0065-2776(08)00606-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The association of the major histocompatibility complex (MHC) genes with autoimmune diseases together with the ectopic expression of class II molecules by epithelial cells of the target tissue gives to these molecules a central role in the pathogenesis of the disease, in its regulation and in the persistence of the immune response in situ. HLA-DR molecules expressed by thyroid follicular cells in thyroid autoimmune diseases are compact molecules stably associated with peptides. The nature of these peptides is of vital importance in the understanding of the disease, since these MHC-II-peptide complexes are going to be recognized by both effector and regulatory T cells in situ. In this chapter, we review the current state of the analysis of naturally processed peptides presented by MHC class II molecules in the context of autoimmunity and we discuss our data of natural HLA-DR ligands eluted from Graves' disease affected thyroid glands, from where autoantigen-derived peptides have been identified.
Collapse
Affiliation(s)
- Laia Muixí
- Immunology Unit, Institut de Biotechnologia i Biomedicina, Universitat Autònoma de Barcelona, Campus de Bellaterra, Barcelona, Spain
| | | | | |
Collapse
|
7
|
Abstract
Fifty years after the discovery of thyroid autoimmunity, several animal models of Graves' hyperthyroidism are now available. All are inducible types, and diseases are elicited by injecting living cells (professional or nonprofessional antigen-presenting cells) expressing the recombinant thyrotropin receptor (TSHR) or by DNA vaccination with TSHR cDNA in plasmid or adenovirus vectors. Thus most Graves' models are attributed to the cloning of the TSHR cDNA and involve in vivo expression of the TSHR. These breakthroughs have provided us important insights into our understanding of the pathogenesis of Graves' disease, and also indispensable means to exploring the possibility of development of novel therapeutic modalities. In particular, recent studies have begun to scrutinize the genetic factors contributing to the susceptibility to this ailment, and to delineate the roles for central and peripheral tolerance and also for fine balance between autoreactive effector T cells and regulatory T cells in the pathophysiology of anti-TSHR autoimmunity and Graves' hyperthyroidism. Moreover, preliminary, but novel, therapeutic approaches have also been started to treat experimental hyperthyroidism.
Collapse
Affiliation(s)
- Yuji Nagayama
- Department of Medical Gene Technology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| |
Collapse
|
8
|
Pichurin P, Pham N, David CS, Rapoport B, McLachlan SM. HLA-DR3 transgenic mice immunized with adenovirus encoding the thyrotropin receptor: T cell epitopes and functional analysis of the CD40 Graves' polymorphism. Thyroid 2006; 16:1221-7. [PMID: 17199432 DOI: 10.1089/thy.2006.16.1221] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The major histocompatibility (MHC) molecule HLA-DR3 is a susceptibility gene for Graves' disease (GD) in Caucasians. Mice lacking murine MHC and expressing human HLA-DR3 develop thyrotropin receptor (TSHR) antibodies and sometimes hyperthyroidism after vaccination with TSHR-DNA. MHC molecules present peptides processed from antigens to T cells. Therefore, we used DR3-transgenic mice to investigate recognition of TSHR ectodomain peptides. After immunization with TSHR A-subunit adenovirus (A-subunit-Ad) but not control-adenovirus (Control-Ad), splenocytes from DR3 mice responded to A-subunit protein in culture by producing interferon-gamma (IFN-gamma). When challenged with 29 overlapping TSHR peptides, splenocytes from A-subunit-Ad- or Control-Ad-immunized mice responded to several peptides. However, in splenocytes from A-subunit-Ad but not Control-Ad mice, a peptide containing TSHR residues 142-161 induced significantly more IFN-gamma than the same splenocytes in medium alone. Immunized DR3 mice also permitted testing the TSHR-specific function of the CD40 single nucleotide polymorphism (C vs. T) associated with GD. Of three human DR3 human Epstein-Barr virus lines (EBVL), two had C in both alleles (CC) and one was CT. However, these EBVL presented peptides poorly and there was no difference between CC vs. CT EBVL in peptide presentation to splenocytes from immunized mice. A peptide corresponding to residues 145-163 is one of seven suggested to be important in GD based on HLA-binding affinities, T-epitope algorithms, and human studies. Consequently, as in human GD, TSHR amino acids 142-161 appear to include a major T cell epitope in HLA-DR3 transgenic mice immunized with A-subunit-Ad.
Collapse
Affiliation(s)
- Pavel Pichurin
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and UCLA School of Medicine, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
9
|
Chen CR, Aliesky HA, Guo J, Rapoport B, McLachlan SM. Blockade of costimulation between T cells and antigen-presenting cells: an approach to suppress murine Graves' disease induced using thyrotropin receptor-expressing adenovirus. Thyroid 2006; 16:427-34. [PMID: 16756463 DOI: 10.1089/thy.2006.16.427] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Immune responses require costimulatory interactions between molecules on antigen-presenting cells and T cells: CD40 binding to CD40 ligand and B7 binding to CD28. Graves' hyperthyroidism is induced in BALB/c mice by immunization with thyrotropin receptor (TSHR) A-subunit adenovirus (Ad-A-subunit). We attempted to modulate Ad-A-subunit-induced Graves' disease using adenoviruses expressing costimulation "decoys": CD40-IgG-Fc (CD40-Ig) to block CD40:CD40-ligand interactions and CTLA4-Fc (CTLA4-Ig) to prevent B7:CD28 binding. OUTCOME Unexpectedly, coimmunizing mice with Ad-A-subunit and excess control adenovirus (1:10 Ad-A-subunit:Ad-control) reduced TSHR antibody levels (thyrotropin binding inhibition [TBI]). Furthermore, only 15% of mice developed hyperthyroidism versus 75% using the same Ad-A-subunit dose (10(8) particles) without Ad-control. This effect was related to the dose of control adenovirus but not to the adenovirus insert, the timing or immunization site. Increasing the Ad-subunit dose (10(9) particles) and decreasing the control adenovirus dose (10:1 Ad-A-subunit:Ad-control) induced high TBI levels and 80% of mice were hyperthyroid. Coimmunization with Ad-CD40-Ig (but not Ad-CTLA4-Ig) reduced the incidence of hyperthyroidism to 40%. CONCLUSIONS Using appropriate controls and adenovirus ratios, our data suggest the importance of CD40:CD40-ligand interactions for inducing Graves' hyperthyroidism by Ad-A-subunit. Furthermore, our observations emphasize the potential pitfalls of non-specific inhibition by coimmunization with two adenovirus species.
Collapse
Affiliation(s)
- Chun-Rong Chen
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and UCLA School of Medicine, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
10
|
Pérez-Pérez L, Suárez O, Sánchez-Aguilar D, Toribio J. [Pemphigus vulgaris beginning as the Koebner phenomenon]. ACTAS DERMO-SIFILIOGRAFICAS 2006; 96:681-4. [PMID: 16476320 DOI: 10.1016/s0001-7310(05)73158-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pemphigus vulgaris is an autoimmune disease characterized by acantholysis and intraepidermal bullae formation. The isomorphic Koebner phenomenon is a response to different stimuli, and it has been described in many skin disorders. An association between the two has rarely been described. We present a case of pemphigus vulgaris that appeared over surgical scars. Pemphigus vulgaris over scars is very rare. In our case, the reactivation of the pemphigus antigens in the surgical scar probably caused a generalized autoantibody response, with the formation of bullae on the patient's trunk.
Collapse
Affiliation(s)
- Lidia Pérez-Pérez
- Departamento de Dermatología, Complejo Hospitalario Universitario, Facultad de Medicina, Santiago de Compostela, Spain.
| | | | | | | |
Collapse
|
11
|
Pichurin PN, Chen CR, Chazenbalk GD, Aliesky H, Pham N, Rapoport B, McLachlan SM. Targeted expression of the human thyrotropin receptor A-subunit to the mouse thyroid: insight into overcoming the lack of response to A-subunit adenovirus immunization. THE JOURNAL OF IMMUNOLOGY 2006; 176:668-76. [PMID: 16365463 DOI: 10.4049/jimmunol.176.1.668] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The thyrotropin receptor (TSHR), the major autoantigen in Graves' disease, is posttranslationally modified by intramolecular cleavage to form disulfide-linked A- and B-subunits. Because Graves' hyperthyroidism is preferentially induced in BALB/c mice using adenovirus encoding the free A-subunit rather than full-length human TSHR, the shed A-subunit appears to drive the disease-associated autoimmune response. To further investigate this phenomenon, we generated transgenic mice with the human A-subunit targeted to the thyroid. Founder transgenic mice had normal thyroid function and were backcrossed to BALB/c. The A-subunit mRNA expression was confirmed in thyroid tissue. Unlike wild-type littermates, transgenic mice immunized with low-dose A-subunit adenovirus failed to develop TSHR Abs, hyperthyroidism, or splenocyte responses to TSHR Ag. Conventional immunization with A-subunit protein and adjuvants induced TSHR Abs lacking the characteristics of human autoantibodies. Unresponsiveness was partially overcome using high-dose, full-length human TSHR adenovirus. Although of low titer, these induced Abs recognized the N terminus of the A-subunit, and splenocytes responded to A-subunit peptides. Therefore, "non-self" regions in the B-subunit did not contribute to inducing responses. Indeed, transgenic mice immunized with high-dose A-subunit adenovirus developed TSHR Abs with thyrotropin-binding inhibitory activity, although at lower titers than wild-type littermates, suggesting down-regulation in the transgenic mice. In conclusion, in mice expressing a human A-subunit transgene in the thyroid, non-self human B-subunit epitopes are not necessary to induce responses to the A-subunit. Our findings raise the possibility that autoimmunity to the TSHR in humans may not involve epitopes on a cross-reacting protein, but rather, strong adjuvant signals provided in bystander immune responses.
Collapse
Affiliation(s)
- Pavel N Pichurin
- Autoimmune Disease Unit, Cedars-Sinai Research Institute, and University of California School of Medicine, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Kala M, Chen CR, McLachlan SM, Rapoport B, Aliesky H, Chapman HA. Cathepsin S is not crucial to TSHR processing and presentation in a murine model of Graves' disease. Immunology 2006; 116:532-40. [PMID: 16313367 PMCID: PMC1802445 DOI: 10.1111/j.1365-2567.2005.02255.x] [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: 12/01/2022] Open
Abstract
By regulating invariant (Ii) chain processing and MHC class II peptide loading, the endosomal protease cathepsin S (Cat S) has a potential role in autoimmune susceptibility. Indeed, Cat S null mice are resistant to I-Ab-restricted experimental myasthenia gravis due to inadequate peptide presentation. To explore the role of Cat S in a Graves' disease model, I-Ad-restricted wild-type (WT) and Cat S(-/-) mice were immunized with adenovirus encoding the A subunit of thyroid stimulating hormone receptor (TSHR). TSHR adenovirus immunized mice develop Th1 T cells, TSHR antibodies, and a proportion become overtly hyperthyroid. Although TSHR presentation in vitro was initially impaired in Cat S(-/-) mice, subsequent TSHR presentation in vitro and disease development were similar in both groups but with higher antibody responses in Cat S null mice. WT and Cat S(-/-) mice recognized similar T cell epitopes from a panel of overlapping TSHR peptides. TSHR responses were found to be I-Ad-restricted and Cat S(-/-) I-Ad B cells had marked defects in Ii processing. These data imply that loading of TSHR peptides critical to TSHR antibody responses becomes Ii-independent. Contrasting findings among organ-specific murine autoimmune models imply that potential uses of Cat S inhibitors to ameliorate autoimmunity must be determined empirically.
Collapse
Affiliation(s)
- Mrinalini Kala
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, 93143, USA
| | | | | | | | | | | |
Collapse
|
13
|
Abstract
Graves' hyperthyroidism can be induced in mice or hamsters by novel approaches, namely injecting cells expressing the TSH receptor (TSHR) or vaccination with TSHR-DNA in plasmid or adenoviral vectors. These models provide unique insight into several aspects of Graves' disease: 1) manipulating immunity toward Th1 or Th2 cytokines enhances or suppresses hyperthyroidism in different models, perhaps reflecting human disease heterogeneity; 2) the role of TSHR cleavage and A subunit shedding in immunity leading to thyroid-stimulating antibodies (TSAbs); and 3) epitope spreading away from TSAbs and toward TSH-blocking antibodies in association with increased TSHR antibody titers (as in rare hypothyroid patients). Major developments from the models include the isolation of high-affinity monoclonal TSAbs and analysis of antigen presentation, T cells, and immune tolerance to the TSHR. Studies of inbred mouse strains emphasize the contribution of non-MHC vs. MHC genes, as in humans, supporting the relevance of the models to human disease. Moreover, other findings suggest that the development of Graves' disease is affected by environmental factors, including infectious pathogens, regardless of modifications in the Th1/Th2 balance. Finally, developing immunospecific forms of therapy for Graves' disease will require painstaking dissection of immune recognition and responses to the TSHR.
Collapse
Affiliation(s)
- Sandra M McLachlan
- Autoimmune Disease Unit, Cedars-Sinai Medical Center, University of California Los Angeles School of Medicine, CA 90048, USA.
| | | | | |
Collapse
|
14
|
Chazenbalk GD, Pichurin PN, Guo J, Rapoport B, McLachlan SM. Interactions between the mannose receptor and thyroid autoantigens. Clin Exp Immunol 2005; 139:216-24. [PMID: 15654820 PMCID: PMC1809290 DOI: 10.1111/j.1365-2249.2004.02689.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Thyroid autoantigens require internalization and processing by antigen-presenting cells to induce immune responses. Besides pinocytosis, antigen uptake can be receptor-mediated. The mannose receptor (ManR) has a cysteine rich domain (CR) and eight carbohydrate recognition domains (CRD) that bind glycosylated proteins. The TSH receptor (TSHR), thyroid peroxidase (TPO) and thyroglobulin (Tg) are glycoproteins. To investigate a role for the ManR in thyroid autoimmunity, we tested the interaction between these autoantigens and chimeric ManRs. Plasmids encoding the CR-domain linked to IgG-Fc (CR-Fc) and CDR domains 4-7 linked to IgG-Fc (CDR4-7-Fc) were expressed and purified with Protein A. Enzyme-linked immunosorbent assay (ELISA) plates were coated with human thyroid autoantigens and CR-Fc or CRD4-7-Fc binding detected with peroxidase-conjugated anti-IgG-Fc. CRD4-7-Fc binding was highest for the TSHR, followed by Tg and was minimal for TPO. CR-Fc bound to Tg but not to TSHR or TPO. The interaction between the TSHR and CRD-Fc was calcium-dependent; it was inhibited by mannose (not galactose), and required a glycosylated TSHR A-subunit. Moreover, precomplexing the TSHR A-subunit with CRD-Fc (but not CR-Fc), or adding mannose (but not galactose), decreased in vitro responses of splenocytes from TSHR-immunized mice. Our data indicate that the ManR may participate in autoimmune responses to Tg and the TSHR but not to TPO. Most important, ManR binding of heavily glycosylated TSHR A-subunits suggests a mechanism by which the minute amounts of A-subunit protein shed from the thyroid may be captured by antigen-presenting cells located in the gland or in draining lymph nodes.
Collapse
Affiliation(s)
- G D Chazenbalk
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and UCLA School of Medicine, Los Angeles, CA 90048, USA
| | | | | | | | | |
Collapse
|
15
|
McLachlan SM, Braley-Mullen H, Chen CR, Aliesky H, Pichurin PN, Rapoport B. Dissociation between iodide-induced thyroiditis and antibody-mediated hyperthyroidism in NOD.H-2h4 mice. Endocrinology 2005; 146:294-300. [PMID: 15459116 DOI: 10.1210/en.2004-1126] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NOD.H-2h4 mice are genetically predisposed to thyroid autoimmunity and spontaneously develop thyroglobulin autoantibodies (TgAb) and thyroiditis. Iodide administration enhances TgAb levels and the incidence and severity of thyroiditis. Using these mice, we investigated the interactions between TSH receptor (TSHR) antibodies induced by vaccination and spontaneous or iodide-enhanced thyroid autoimmunity (thyroiditis and TgAb). Mice were immunized with adenovirus expressing the TSHR A-subunit (or control adenovirus). Thyroid antibodies, histology, and serum thyroxine levels were compared in animals on a regular diet or on a high-iodide diet (0.05% NaI-supplemented water). Thyroiditis severity and TgAb levels were enhanced by iodide administration and were independent of the type of adenovirus used for immunization. In contrast, TSHR antibodies, measured by TSH-binding inhibition, thyroid-stimulating activity, and TSH-blocking activity, were induced in the majority of animals immunized with TSHR (but not control) adenovirus and were unaffected by dietary iodide. The NOD.2h4 strain of mice was less susceptible than BALB/c or BALB/k mice to TSHR adenovirus-induced hyperthyroidism. Nevertheless, hyperthyroidism developed in approximately one third of TSHR adenovirus-injected NOD.2h4 mice. This hyperthyroidism was suppressed by a high-iodide diet, probably by a nonimmune mechanism. The fact that inducing an immune response to the TSHR had no effect on thyroiditis raises the possibility that the TSHR may not be the target involved in the variable thyroiditis component in some humans with Graves' disease.
Collapse
Affiliation(s)
- Sandra M McLachlan
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and University of California Los Angeles School of Medicine, Los Angeles, California 90048, USA.
| | | | | | | | | | | |
Collapse
|