1
|
McLachlan SM, Aliesky HA, Rapoport B. A Mouse Thyrotropin Receptor A-Subunit Transgene Expressed in Thyroiditis-Prone Mice May Provide Insight into Why Graves' Disease Only Occurs in Humans. Thyroid 2019; 29:1138-1146. [PMID: 31184281 PMCID: PMC6707033 DOI: 10.1089/thy.2019.0260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background: Graves' disease, caused by autoantibodies that activate the thyrotropin (TSH) receptor (TSHR), has only been reported in humans. Thyroiditis-prone NOD.H2h4 mice develop autoantibodies to thyroglobulin (Tg) and thyroid peroxidase (TPO) but not to the TSHR. Evidence supports the importance of the shed TSHR A-subunit in the initiation and/or amplification of the autoimmune response to the holoreceptor. Cells expressing the gene for the isolated A-subunit secrete A-subunit protein, a surrogate for holoreceptor A-subunit shedding. NOD.H2h4 mice with the human TSHR A-subunit targeted to the thyroid (a "self" antigen in such transgenic (Tgic) animals), unlike their wild-type (wt) siblings, spontaneously develop pathogenic TSHR antibodies to the human-TSH holoreceptor. These autoantibodies do not recognize the endogenous mouse-TSH holoreceptor and do not cause hyperthyroidism. Methods: We have now generated NOD.H2h4 mice with the mouse-TSHR A-subunit transgene targeted to the thyroid. Tgic mice and wt littermates were compared for intrathyroidal expression of the mouse A-subunit. Sera from six-month-old mice were tested for the presence of autoantibodies to Tg and TPO as well as for pathogenic TSHR antibodies (TSH binding inhibition, bioassay for thyroid stimulating antibodies) and nonpathogenic TSHR antibodies (ELISA). Results: Expression of the mouse TSHR A-subunit transgene in the thyroid was confirmed by real-time polymerase chain reaction in the Tgics and had no effect on the spontaneous development of autoantibodies to Tg or TPO. However, unlike the same NOD.H2h4 strain with the human-TSHR A-subunit target to the thyroid, mice expressing intrathyroidal mouse-TSHR A subunit failed to develop either pathogenic or nonpathogenic TSHR antibodies. The mouse TSHR A-subunit differs from the human TSHR A-subunit in terms of its amino acid sequence and has one less glycosylation site than the human TSHR A-subunit. Conclusions: Multiple genetic and environmental factors contribute to the pathogenesis of Graves' disease. The present study suggests that the TSHR A-subunit structure (possibly including posttranslational modification such as glycosylation) may explain, in part, why Graves' disease only develops in humans.
Collapse
Affiliation(s)
- Sandra M. McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, California
- UCLA School of Medicine, University of California, Los Angeles, California
- Address correspondence to: Sandra M. McLachlan, PhD, Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, B-131, Los Angeles, CA 90048
| | - Holly A. Aliesky
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, California
| | - Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, California
- UCLA School of Medicine, University of California, Los Angeles, California
| |
Collapse
|
2
|
McLachlan SM, Aliesky HA, Garcia P, Banuelos B, Rapoport B. Thyroid Hemiagenesis in a Thyroiditis Prone Mouse Strain. Eur Thyroid J 2018; 7:187-192. [PMID: 30283736 PMCID: PMC6140602 DOI: 10.1159/000490700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/04/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Thyroid hemiagenesis, a rare congenital condition detected by ultrasound screening of the neck, is usually not manifested clinically in humans. This condition has been reported in mice with hypothyroidism associated with induced deficiency in paired box 8 and NK2 homeobox 1, sonic hedgehog, or T-box 1. Unexpectedly, we observed thyroid hemiagenesis in NOD.H2h4 mice, an unusual strain that spontaneously develops iodide enhanced thyroid autoimmunity but remains euthyroid. OBJECTIVES AND METHODS First, to compare mice with thyroid hemiagenesis versus bilobed littermates for serum T4, autoantibodies to thyroglobulin (ELISA) and thyroid peroxidase (TPO; flow cytometry with eukaryotic cells expressing mouse TPO), gross anatomy, and thyroid histology; second, to estimate the percentage of mice with thyroid hemiagenesis in the NOD.H2h4 mice we have studied over 6 years. RESULTS Thyroid hemiagenesis was observed in 3 of 1,025 NOD.H2h4 mice (2 females, 1 male; 0.3$). Two instances of hemiagenesis were in wild-type females and one in a transgenic male expressing the human TSHR A-subunit in the thyroid. Two mice had very large unilobed glands, as in some human cases with this condition. Thyroid lymphocytic infiltration, serum T4, and the levels of thyroid autoantibodies were similar in mice with thyroid hemiagenesis and bilobed littermates. CONCLUSIONS Unlike hypothyroidism associated with hemiagenesis in transcription factor knockout mice, hemiagenesis in euthyroid NOD.H2h4 mice occurs spontaneously and is phenotypically similar to that occasionally observed in humans.
Collapse
Affiliation(s)
- Sandra M. McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, California, USA
- UCLA School of Medicine, University of California, Los Angeles, California, USA
- *Sandra M. McLachlan, Cedars-Sinai Medical Center, 8700 Beverly Blvd, B-131, Los Angeles, CA 90048 (USA), E-Mail
| | - Holly A. Aliesky
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, California, USA
| | - Priscilla Garcia
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, California, USA
| | - Bianca Banuelos
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, California, USA
| | - Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, California, USA
- UCLA School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
3
|
McLachlan SM, Aliesky H, Banuelos B, Hee SSQ, Rapoport B. Variable Effects of Dietary Selenium in Mice That Spontaneously Develop a Spectrum of Thyroid Autoantibodies. Endocrinology 2017; 158:3754-3764. [PMID: 28938453 PMCID: PMC5695827 DOI: 10.1210/en.2017-00275] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/18/2017] [Indexed: 12/16/2022]
Abstract
Selenium (Se) is a critical element in thyroid function, and variable dietary Se intake influences immunity. Consequently, dietary Se could influence development of thyroid autoimmunity and provide an adjunct to treat autoimmune thyroid dysfunction. Nonobese diabetic (NOD).H2h4 mice spontaneously develop autoantibodies to thyroglobulin (Tg) and thyroid peroxidase (TPO). This mouse strain expressing a human thyroid-stimulating hormone receptor (TSHR) A-subunit transgene in the thyroid also develops pathogenic TSHR autoantibodies. In this report, we investigated whether dietary Se influences these immune processes. Male and female wild-type and transgenic NOD.H2h4 mice were maintained on normal-, low-, or high-Se (0.1, 0, or 1.0 mg/kg) rodent diets. After 4 months, Se serum levels were extremely low or significantly increased on 0 or 1.0 mg/kg Se, respectively. Varying Se intake affected Tg antibody (TgAb) levels after 2 (but not 4) months; conversely, TPO antibody (TPOAb) levels were altered by dietary Se after 4 (but not 2) months. These data correspond to the earlier development of TgAb than TPOAb in NOD.H2h4 mice. In males, TgAb levels were enhanced by high Se and in females by low Se intake. Se intake had no effect on pathogenic TSHR autoantibodies in TSHR transgenic NOD.H2h4 females. In conclusion, in susceptible NOD.H2h4 mice, we found no evidence that a higher dietary Se intake ameliorates thyroid autoimmunity by reducing autoantibodies to Tg, TPO, or the TSHR. Instead, our finding that low dietary Se potentiates the development of autoantibodies to Tg and TPO in females is consistent with reports in humans of an increased prevalence of autoimmune thyroiditis in low-Se regions.
Collapse
Affiliation(s)
- Sandra M. McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, and University of California, Los Angeles, School of Medicine, Los Angeles, California 90095
| | - Holly Aliesky
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, and University of California, Los Angeles, School of Medicine, Los Angeles, California 90095
| | - Bianca Banuelos
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, and University of California, Los Angeles, School of Medicine, Los Angeles, California 90095
| | - Shane S. Que Hee
- Department of Environmental Health Sciences and Center for Occupational and Environmental Health, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California 90095
| | - Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, and University of California, Los Angeles, School of Medicine, Los Angeles, California 90095
| |
Collapse
|
4
|
McLachlan SM, Aliesky HA, Banuelos B, Lesage S, Collin R, Rapoport B. High-level intrathymic thyrotrophin receptor expression in thyroiditis-prone mice protects against the spontaneous generation of pathogenic thyrotrophin receptor autoantibodies. Clin Exp Immunol 2017; 188:243-253. [PMID: 28099999 PMCID: PMC5383439 DOI: 10.1111/cei.12928] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/03/2017] [Accepted: 01/12/2017] [Indexed: 12/11/2022] Open
Abstract
The thyrotrophin receptor (TSHR) A-subunit is the autoantigen targeted by pathogenic autoantibodies that cause Graves' hyperthyroidism, a common autoimmune disease in humans. Previously, we reported that pathogenic TSHR antibodies develop spontaneously in thyroiditis-susceptible non-obese diabetic (NOD).H2h4 mice bearing a human TSHR A-subunit transgene, which is expressed at low levels in both the thyroid and thymus (Lo-expressor transgene). The present study tested recent evidence that high intrathymic TSHR expression protects against the development of pathogenic TSHR antibodies in humans. By successive back-crossing, we transferred to the NOD.H2h4 background a human TSHR A-subunit transgene expressed at high levels in the thyroid and thymus (Hi-expressor transgene). In the sixth back-cross generation (> 98% NOD.H2h4 genome), only transgenic offspring produced spontaneously immunoglobulin (Ig)G class non-pathogenic human TSHR A-subunit antibodies. In contrast, both transgenic and non-transgenic offspring developed antibodies to thyroglobulin and thyroid peroxidase. However, non-pathogenic human TSHR antibody levels in Hi-expressor offspring were lower than in Lo-expressor transgenic mice. Moreover, pathogenic TSHR antibodies, detected by inhibition of TSH binding to the TSHR, only developed in back-cross offspring bearing the Lo-expressor, but not the Hi-expressor, transgene. High versus low expression human TSHR A-subunit in the NOD.H2h4 thymus was not explained by the transgene locations, namely chromosome 2 (127-147 Mb; Hi-expressor) and chromosome 1 (22.9-39.3 Mb; low expressor). Nevertheless, using thyroiditis-prone NOD.H2h4 mice and two transgenic lines, our data support the association from human studies that low intrathymic TSHR expression is associated with susceptibility to developing pathogenic TSHR antibodies, while high intrathymic TSHR expression is protective.
Collapse
Affiliation(s)
- S. M. McLachlan
- Thyroid Autoimmune Disease UnitCedars‐Sinai Research Institute and UCLA School of Medicine, University of CaliforniaLos AngelesCAUSA
| | - H. A. Aliesky
- Thyroid Autoimmune Disease UnitCedars‐Sinai Research Institute and UCLA School of Medicine, University of CaliforniaLos AngelesCAUSA
| | - B. Banuelos
- Thyroid Autoimmune Disease UnitCedars‐Sinai Research Institute and UCLA School of Medicine, University of CaliforniaLos AngelesCAUSA
| | - S. Lesage
- Department of Immunology‐OncologyMaisonneuve‐Rosemont Hospital, Montréal, Québec, Canada and Département de Microbiologie, Infectiologie et Immunologie, Université de MontréalMontréalQuébecCanada
| | - R. Collin
- Department of Immunology‐OncologyMaisonneuve‐Rosemont Hospital, Montréal, Québec, Canada and Département de Microbiologie, Infectiologie et Immunologie, Université de MontréalMontréalQuébecCanada
| | - B. Rapoport
- Thyroid Autoimmune Disease UnitCedars‐Sinai Research Institute and UCLA School of Medicine, University of CaliforniaLos AngelesCAUSA
| |
Collapse
|
5
|
Rapoport B, Banuelos B, Aliesky HA, Hartwig Trier N, McLachlan SM. Critical Differences between Induced and Spontaneous Mouse Models of Graves' Disease with Implications for Antigen-Specific Immunotherapy in Humans. THE JOURNAL OF IMMUNOLOGY 2016; 197:4560-4568. [PMID: 27913646 DOI: 10.4049/jimmunol.1601393] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/14/2016] [Indexed: 01/20/2023]
Abstract
Graves' hyperthyroidism, a common autoimmune disease caused by pathogenic autoantibodies to the thyrotropin (TSH) receptor (TSHR), can be treated but not cured. This single autoantigenic target makes Graves' disease a prime candidate for Ag-specific immunotherapy. Previously, in an induced mouse model, injecting TSHR A-subunit protein attenuated hyperthyroidism by diverting pathogenic TSHR Abs to a nonfunctional variety. In this study, we explored the possibility of a similar diversion in a mouse model that spontaneously develops pathogenic TSHR autoantibodies, NOD.H2h4 mice with the human (h) TSHR (hTSHR) A-subunit transgene expressed in the thyroid and (shown in this article) the thymus. We hypothesized that such diversion would occur after injection of "inactive" hTSHR A-subunit protein recognized only by nonpathogenic (not pathogenic) TSHR Abs. Surprisingly, rather than attenuating the pre-existing pathogenic TSHR level, in TSHR/NOD.H2h4 mice inactive hTSHR Ag injected without adjuvant enhanced the levels of pathogenic TSH-binding inhibition and thyroid-stimulating Abs, as well as nonpathogenic Abs detected by ELISA. This effect was TSHR specific because spontaneously occurring autoantibodies to thyroglobulin and thyroid peroxidase were unaffected. As controls, nontransgenic NOD.H2h4 mice similarly injected with inactive hTSHR A-subunit protein unexpectedly developed TSHR Abs, but only of the nonpathogenic variety detected by ELISA. Our observations highlight critical differences between induced and spontaneous mouse models of Graves' disease with implications for potential immunotherapy in humans. In hTSHR/NOD.H2h4 mice with ongoing disease, injecting inactive hTSHR A-subunit protein fails to divert the autoantibody response to a nonpathogenic form. Indeed, such therapy is likely to enhance pathogenic Ab production and exacerbate Graves' disease in humans.
Collapse
Affiliation(s)
- Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center and UCLA School of Medicine, Los Angeles, CA 90048; and
| | - Bianca Banuelos
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center and UCLA School of Medicine, Los Angeles, CA 90048; and
| | - Holly A Aliesky
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center and UCLA School of Medicine, Los Angeles, CA 90048; and
| | - Nicole Hartwig Trier
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, DK-2300 Copenhagen S, Denmark
| | - Sandra M McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center and UCLA School of Medicine, Los Angeles, CA 90048; and
| |
Collapse
|
6
|
Rapoport B, McLachlan SM. TSH Receptor Cleavage Into Subunits and Shedding of the A-Subunit; A Molecular and Clinical Perspective. Endocr Rev 2016; 37:114-34. [PMID: 26799472 PMCID: PMC4823380 DOI: 10.1210/er.2015-1098] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/14/2016] [Indexed: 02/07/2023]
Abstract
The TSH receptor (TSHR) on the surface of thyrocytes is unique among the glycoprotein hormone receptors in comprising two subunits: an extracellular A-subunit, and a largely transmembrane and cytosolic B-subunit. Unlike its ligand TSH, whose subunits are encoded by two genes, the TSHR is expressed as a single polypeptide that subsequently undergoes intramolecular cleavage into disulfide-linked subunits. Cleavage is associated with removal of a C-peptide region, a mechanism similar in some respects to insulin cleavage into disulfide linked A- and B-subunits with loss of a C-peptide region. The potential pathophysiological importance of TSHR cleavage into A- and B-subunits is that some A-subunits are shed from the cell surface. Considerable experimental evidence supports the concept that A-subunit shedding in genetically susceptible individuals is a factor contributing to the induction and/or affinity maturation of pathogenic thyroid-stimulating autoantibodies, the direct cause of Graves' disease. The noncleaving gonadotropin receptors are not associated with autoantibodies that induce a "Graves' disease of the gonads." We also review herein current information on the location of the cleavage sites, the enzyme(s) responsible for cleavage, the mechanism by which A-subunits are shed, and the effects of cleavage on receptor signaling.
Collapse
Affiliation(s)
- Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center and UCLA School of Medicine, Los Angeles, California 90048
| | - Sandra M McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center and UCLA School of Medicine, Los Angeles, California 90048
| |
Collapse
|
7
|
Rapoport B, McLachlan SM. Withdrawn: TSH Receptor Cleavage Into Subunits and Shedding of the A-Subunit; A Molecular and Clinical Perspective. Endocr Rev 2016; 2016:23-42. [PMID: 27454362 PMCID: PMC6958993 DOI: 10.1210/er.2015-1098.2016.1.test] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/14/2016] [Indexed: 12/29/2022]
Abstract
The TSH receptor (TSHR) on the surface of thyrocytes is unique among the glycoprotein hormone receptors in comprising two subunits: an extracellular A-subunit, and a largely transmembrane and cytosolic B-subunit. Unlike its ligand TSH, whose subunits are encoded by two genes, the TSHR is expressed as a single polypeptide that subsequently undergoes intramolecular cleavage into disulfide-linked subunits. Cleavage is associated with removal of a C-peptide region, a mechanism similar in some respects to insulin cleavage into disulfide linked A- and B-subunits with lossofaC-peptideregion. The potential pathophysiological importance of TSHR cleavage into A-and B-subunits is that some A-subunits are shed from the cell surface. Considerable experimental evidence supports the concept that A-subunit shedding in genetically susceptible individuals is a factor contributing to the induction and/or affinity maturation of pathogenic thyroid-stimulating autoantibodies, the direct cause of Graves' disease. The noncleaving gonadotropin receptors are not associated with autoantibodies that induce a "Graves' disease of the gonads." We also review herein current information on the location of the cleavage sites, the enzyme(s) responsible for cleavage, the mechanism by which A-subunits are shed, and the effects of cleavage on receptor signaling. (Endocrine Reviews 37: 114-134, 2016).
Collapse
Affiliation(s)
- Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center and UCLA School of Medicine, Los Angeles, California 90048
| | - Sandra M McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center and UCLA School of Medicine, Los Angeles, California 90048
| |
Collapse
|
8
|
Rapoport B, Aliesky HA, Banuelos B, Chen CR, McLachlan SM. A unique mouse strain that develops spontaneous, iodine-accelerated, pathogenic antibodies to the human thyrotrophin receptor. THE JOURNAL OF IMMUNOLOGY 2015; 194:4154-61. [PMID: 25825442 DOI: 10.4049/jimmunol.1500126] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/24/2015] [Indexed: 11/19/2022]
Abstract
Abs that stimulate the thyrotropin receptor (TSHR), the cause of Graves' hyperthyroidism, only develop in humans. TSHR Abs can be induced in mice by immunization, but studying pathogenesis and therapeutic intervention requires a model without immunization. Spontaneous, iodine-accelerated, thyroid autoimmunity develops in NOD.H2(h4) mice associated with thyroglobulin and thyroid-peroxidase, but not TSHR, Abs. We hypothesized that transferring the human TSHR A-subunit to NOD.H2(h4) mice would result in loss of tolerance to this protein. BALB/c human TSHR A-subunit mice were bred to NOD.H2(h4) mice, and transgenic offspring were repeatedly backcrossed to NOD.H2(h4) mice. All offspring developed Abs to thyroglobulin and thyroid-peroxidase. However, only TSHR-transgenic NOD.H2(h4) mice (TSHR/NOD.H2(h4)) developed pathogenic TSHR Abs as detected using clinical Graves' disease assays. As in humans, TSHR/NOD.H2(h4) female mice were more prone than male mice to developing pathogenic TSHR Abs. Fortunately, in view of the confounding effect of excess thyroid hormone on immune responses, spontaneously arising pathogenic human TSHR Abs cross-react poorly with the mouse TSHR and do not cause thyrotoxicosis. In summary, the TSHR/NOD.H2(h4) mouse strain develops spontaneous, iodine-accelerated, pathogenic TSHR Abs in female mice, providing a unique model to investigate disease pathogenesis and test novel TSHR Ag-specific immunotherapies aimed at curing Graves' disease in humans.
Collapse
Affiliation(s)
- Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute/David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90048
| | - Holly A Aliesky
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute/David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90048
| | - Bianca Banuelos
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute/David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90048
| | - Chun-Rong Chen
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute/David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90048
| | - Sandra M McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute/David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90048
| |
Collapse
|
9
|
McLachlan SM, Rapoport B. Breaking tolerance to thyroid antigens: changing concepts in thyroid autoimmunity. Endocr Rev 2014; 35:59-105. [PMID: 24091783 PMCID: PMC3895862 DOI: 10.1210/er.2013-1055] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/24/2013] [Indexed: 02/06/2023]
Abstract
Thyroid autoimmunity involves loss of tolerance to thyroid proteins in genetically susceptible individuals in association with environmental factors. In central tolerance, intrathymic autoantigen presentation deletes immature T cells with high affinity for autoantigen-derived peptides. Regulatory T cells provide an alternative mechanism to silence autoimmune T cells in the periphery. The TSH receptor (TSHR), thyroid peroxidase (TPO), and thyroglobulin (Tg) have unusual properties ("immunogenicity") that contribute to breaking tolerance, including size, abundance, membrane association, glycosylation, and polymorphisms. Insight into loss of tolerance to thyroid proteins comes from spontaneous and induced animal models: 1) intrathymic expression controls self-tolerance to the TSHR, not TPO or Tg; 2) regulatory T cells are not involved in TSHR self-tolerance and instead control the balance between Graves' disease and thyroiditis; 3) breaking TSHR tolerance involves contributions from major histocompatibility complex molecules (humans and induced mouse models), TSHR polymorphism(s) (humans), and alternative splicing (mice); 4) loss of tolerance to Tg before TPO indicates that greater Tg immunogenicity vs TPO dominates central tolerance expectations; 5) tolerance is induced by thyroid autoantigen administration before autoimmunity is established; 6) interferon-α therapy for hepatitis C infection enhances thyroid autoimmunity in patients with intact immunity; Graves' disease developing after T-cell depletion reflects reconstitution autoimmunity; and 7) most environmental factors (including excess iodine) "reveal," but do not induce, thyroid autoimmunity. Micro-organisms likely exert their effects via bystander stimulation. Finally, no single mechanism explains the loss of tolerance to thyroid proteins. The goal of inducing self-tolerance to prevent autoimmune thyroid disease will require accurate prediction of at-risk individuals together with an antigen-specific, not blanket, therapeutic approach.
Collapse
Affiliation(s)
- Sandra M McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, and University of California-Los Angeles School of Medicine, Los Angeles, California 90048
| | | |
Collapse
|
10
|
Wang Y, Wu LP, Fu J, Lv HJ, Guan XY, Xu L, Chen P, Gao CQ, Hou P, Ji MJ, Shi BY. Hyperthyroid monkeys: a nonhuman primate model of experimental Graves' disease. J Endocrinol 2013; 219:183-93. [PMID: 24029729 DOI: 10.1530/joe-13-0279] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Graves' disease (GD) is a common organ-specific autoimmune disease with the prevalence between 0.5 and 2% in women. Several lines of evidence indicate that the shed A-subunit rather than the full-length thyrotropin receptor (TSHR) is the autoantigen that triggers autoimmunity and leads to hyperthyroidism. We have for the first time induced GD in female rhesus monkeys, which exhibit greater similarity to patients with GD than previous rodent models. After final immunization, the monkeys injected with adenovirus expressing the A-subunit of TSHR (A-sub-Ad) showed some characteristics of GD. When compared with controls, all the test monkeys had significantly higher TSHR antibody levels, half of them had increased total thyroxine (T₄) and free T₄, and 50% developed goiter. To better understand the underlying mechanisms, quantitative studies on subpopulations of CD4+T helper cells were carried out. The data indicated that this GD model involved a mixed Th1 and Th2 response. Declined Treg proportions and increased Th17:Treg ratio are also observed. Our rhesus monkey model successfully mimicked GD in humans in many aspects. It would be a useful tool for furthering our understanding of the pathogenesis of GD and would potentially shorten the distance toward the prevention and treatment of this disease in human.
Collapse
Affiliation(s)
- Y Wang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
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: 1.4] [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
|
12
|
McLachlan SM, Aliesky HA, Chen CR, Rapoport B. Role of self-tolerance and chronic stimulation in the long-term persistence of adenovirus-induced thyrotropin receptor antibodies in wild-type and transgenic mice. Thyroid 2012; 22:931-7. [PMID: 22827528 PMCID: PMC3429281 DOI: 10.1089/thy.2012.0008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Graves'-like disease, reflected by thyrotropin receptor (TSHR) antibodies and hyperthyroidism in some mouse strains, can be induced by immunization with adenovirus-expressing DNA for the human TSHR or its A-subunit. The conventional approach involves two or three adenovirus injections at 3-week intervals and euthanasia 10 weeks after the first injection. To investigate TSHR antibody persistence in mice with differing degrees of self-tolerance to the TSHR A-subunit, we studied the effect of delaying euthanasia until 20 weeks after the initial immunization. METHODS Wild-type (WT) mice and transgenic (tg) mice expressing low intrathyroidal levels of the human TSHR A-subunit were immunized with A-subunit-adenovirus on two occasions; a second group of mice was immunized on three occasions. Sera obtained 4, 10, and 20 weeks (euthanasia) after the initial immunization were tested for thyrotropin (TSH) binding inhibition (TBI), antibody binding to TSHR A-subunit protein-coated enzyme-linked immunosorbent assay (ELISA) plates, and thyroid stimulating antibody activity (TSAb; cyclic adenosine monophosphate [cAMP] generation). Serum thyroxine (T4) and thyroid histology were studied at euthanasia. RESULTS THE majority of WT mice retained high TSHR antibody levels measured by TBI or ELISA at euthanasia but only about 50% were TSAb positive. Low-expressor tgs exhibited self-tolerance, with fewer mice positive by TBI or ELISA and antibody levels were lower than in WT littermates. In WT mice, antibody persistence was similar after two or three immunizations; for tgs, only mice immunized three times had detectable TSAb at 20 weeks. Unlike our previous observations of hyperthyroidism in WT mice examined 4 or 10 weeks after immunization, all mice were euthyroid at 20 weeks. CONCLUSIONS Our findings for induced TSHR antibodies in mice, similar to data for human thyroid autoantibodies, indicate that the parameters that contribute to the concentration of the antibody and thereby play a critical role in long-term persistence of TSHR antibodies are the degree of self-tolerance to the TSHR and chronic stimulation.
Collapse
Affiliation(s)
- Sandra M McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, CA 90048, USA.
| | | | | | | |
Collapse
|
13
|
Ye F, Hou P, Wu X, Ma X, Gao L, Wu L, Xu L, Shi B. The significance of immune-related molecule expression profiles in an animal model of Graves' disease. Autoimmunity 2011; 45:143-52. [PMID: 22017306 DOI: 10.3109/08916934.2011.611548] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The thyrotropin receptor (TSHR) A-subunit has been reported to be a critical autoantigen in the generation of thyroid-stimulating antibodies, thereby causing Graves' disease (GD). However, immune mechanisms associated with GD animal models induced by TSHR A-subunit are poorly understood until now. METHODS Female BALB/c mice (n = 23) were randomly divided into two groups, and GD presentation was monitored following injection with either 50 μl phosphate-buffered saline containing 10(9) particles of adenovirus expressing the human TSHR A-subunit (Ad-TSHR289) or the Ad-LacZ control. Expressions of CD40, CD40L, CD80, CD86, CD28, CTLA-4, FOXP3 and IL-17A in various tissues were assessed by quantitative RT-PCR and immunohistochemical assays. RESULTS Compared with control group, mice of the hyperthyroid group showed significant elevation of expression in the thyroid of CD40 and CD86, expression in the heart of CD28, CD40 and CD40L and expression in the liver of CD28, CD40 and CD86. Conversely, there was significantly diminished expression of CTLA-4 in the thymus of mice in the hyperthyroid group. Expression of all genes examined was not significantly different in the spleens of mice from either of the groups and CD40L and FOXP3 expression was not detected in the thyroids of hyperthyroid mice. CONCLUSIONS The expression profile of multiple immune-related molecules differed in mice in the GD group following Ad-TSHR289 immunization, suggesting that these molecules played a potential role in GD pathogenesis.
Collapse
Affiliation(s)
- Feng Ye
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an 710061, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Chen CR, Aliesky HA, Rapoport B, McLachlan SM. An attempt to induce "Graves' disease of the gonads" by immunizing mice with the luteinizing hormone receptor provides insight into breaking tolerance to self-antigens. Thyroid 2011; 21:773-81. [PMID: 21649471 PMCID: PMC3123529 DOI: 10.1089/thy.2010.0460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Gonadotropin receptors, unlike the thyrotropin receptor (TSHR), are not cleaved into disulfide-linked A- and B-subunits, nor do they shed A-subunits. Heavily glycosylated TSHR A-subunits initiate or amplify responses leading to stimulating TSHR-autoantibodies and Graves' hyperthyroidism. METHODS To investigate the possibility that mice immunized with luteinizing hormone receptor (LHR) would develop functional antibodies, we constructed adenoviruses expressing the rat-LH holoreceptor (LHR-Ad) and an LHR A-subunit equivalent (LHR-289-Ad). Female BALB/c mice were immunized with high doses (10(11) particles) of LHR-Ad, LHR-289-Ad, or control (Con)-Ad. Sera were tested using LHR-expressing eukaryotic cells for antibody binding by flow cytometry and for bioactivity by measuring cyclic adenosine monophosphate (cAMP) stimulation. RESULTS Elevated serum binding to LHR cells in some LHR-Ad and LHR-289-Ad immunized mice was not specific for LHR-expressing cells. Moreover, sera lacked bioactivity, consistent with unchanged serum estradiol and ovary histology. The difference between rat and mouse LHR-ectodomains is relatively small (3% at the amino-acid level). In contrast, despite amino-acid identity, immunization of mice with adenovirus expressing membrane-bound mouse thyroid peroxidase (TPO), but not soluble mouse TPO ectodomain, elicited strong TPO-specific antibodies. CONCLUSIONS Our investigations provide insight into antibody responses to self-antigens. First, antibodies are induced to large self-antigens like mouse-TPO when membrane bound. Second, lesser amino acid homology between the immunogen and mouse protein (91% vs. 97% for the human-TSHR and rat-LHR, respectively) favors antibody induction. Finally, from previous studies demonstrating the immunogenicity of the highly glycosylated human TSHR A-subunit versus our present data for the nonimmunogenic less glycosylated rat LHR, we suggest that the extent of glycosylation contributes to breaking self-tolerance.
Collapse
Affiliation(s)
- Chun-Rong Chen
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute and UCLA School of Medicine, Los Angeles, California 90048, USA
| | | | | | | |
Collapse
|
15
|
Hamidi S, Aliesky H, Chen CR, Rapoport B, McLachlan SM. Variable suppression of serum thyroxine in female mice of different inbred strains by triiodothyronine administered in drinking water. Thyroid 2010; 20:1157-62. [PMID: 20860425 PMCID: PMC2947419 DOI: 10.1089/thy.2010.0117] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Recombinant-inbred mouse strains differ in their susceptibility to Graves'-like hyperthyroidism induced by immunization with adenovirus expressing the human thyrotropin (TSH) receptor. Because one genetic component contributing to this susceptibility is altered thyroid sensitivity to TSH receptor agonist stimulation, we wished to quantify thyroid responsiveness to TSH. For such studies, it is necessary to suppress endogenous TSH by administering L-3,5,3′-triiodothyronine (L-T3), with the subsequent decrease in serum thyroxine (T4) reflecting endogenous TSH suppression. Our two objectives were to assess in different inbred strains of mice (i) the extent of serum T4 suppression after L-T3 administration and (ii) the magnitude of serum T4 increase induced by TSH. METHODS Mice were tail-bled to establish baseline-serum T4 before L-T3 administration. We initially employed a protocol of L-T3-supplemented drinking water for 7 days. In subsequent experiments, we injected L-T3 intraperitoneally (i.p.) daily for 3 days. Mice were then injected i.p. with bovine TSH (10 mU) and euthanized 5 hours later. Serum T4 was assayed before L-T3 administration, and before and after TSH injection. In some experiments, serum T3 and estradiol were measured in pooled sera. RESULTS Oral L-T3 (3 or 5 µg/mL) suppressed serum T4 levels by 26%-64% in female BALB/c mice but >95% in males. T4 suppression in female B6 mice ranged from 0% to 90%. In C3H mice, L-T3 at 3 µg/mL was ineffective but 5 µg/mL achieved >80% serum T4 reduction. Unlike inbred mice, in outbred CF1 mice the same protocol was more effective: 83% in females and 100% suppression in males. The degree of T4 suppression was unrelated to baseline T4, T3, or estradiol, but was related to mouse weight and postmortem T3, with greater suppression in larger mice (outbred CF1 animals and inbred males). Among females with serum T4 suppression >80%, the increase in serum T4 after TSH injection was greater for BALB/c and C3H versus B6 mice. Moreover, the T4 increment was higher in female than in male BALB/c. CONCLUSIONS Our data provide important, practical information for future in vivo studies in inbred mice: we recommend that responses to TSH be performed in female animals injected with L-T3 i.p. to suppress baseline T4.
Collapse
Affiliation(s)
- Sepehr Hamidi
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute and UCLA School of Medicine, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | | | | | | | | |
Collapse
|
16
|
Misharin AV, Nagayama Y, Aliesky HA, Rapoport B, McLachlan SM. Studies in mice deficient for the autoimmune regulator (Aire) and transgenic for the thyrotropin receptor reveal a role for Aire in tolerance for thyroid autoantigens. Endocrinology 2009; 150:2948-56. [PMID: 19264867 PMCID: PMC2689795 DOI: 10.1210/en.2008-1690] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 02/20/2009] [Indexed: 01/20/2023]
Abstract
The autoimmune regulator (Aire) mediates central tolerance for many autoantigens, and autoimmunity occurs spontaneously in Aire-deficient humans and mice. Using a mouse model of Graves' disease, we investigated the role of Aire in tolerance to the TSH receptor (TSHR) in Aire-deficient and wild-type mice (hyperthyroid-susceptible BALB/c background). Mice were immunized three times with TSHR A-subunit expressing adenovirus. The lack of Aire did not influence T-cell responses to TSHR protein or TSHR peptides. However, antibody levels were higher in Aire-deficient than wild-type mice after the second (but not the third) immunization. After the third immunization, hyperthyroidism persisted in a higher proportion of Aire-deficient than wild-type mice. Aire-deficient mice were crossed with transgenic strains expressing high or low-intrathyroidal levels of human TSHR A subunits. In the low-expressor transgenics, Aire deficiency had the same effect on the pattern of the TSHR antibody response to immunization as in nontransgenics, although the amplitude of the response was lower in the transgenics. High-expressor A-subunit transgenics were unresponsive to immunization. We examined intrathymic expression of murine TSHR, thyroglobulin, and thyroid peroxidase (TPO), the latter two being the dominant autoantigens in Hashimoto's thyroiditis (particularly TPO). Expression of the TSHR and thyroglobulin were reduced in the absence of Aire. Dramatically, thymic expression of TPO was nearly abolished. In contrast, the human A-subunit transgene, lacking a potential Aire-binding motif, was unaffected. Our findings provide insight into how varying intrathymic autoantigen expression may modulate thyroid autoimmunity and suggest that Aire deficiency may contribute more to developing Hashimoto's thyroiditis than Graves' disease.
Collapse
MESH Headings
- Animals
- Autoantigens/metabolism
- Disease Models, Animal
- Female
- Graves Disease/immunology
- Graves Disease/metabolism
- Graves Disease/pathology
- Hyperthyroidism/immunology
- Hyperthyroidism/metabolism
- Hyperthyroidism/pathology
- Immune Tolerance/immunology
- Immunoglobulins, Thyroid-Stimulating/immunology
- Immunoglobulins, Thyroid-Stimulating/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Receptors, Thyrotropin/genetics
- Receptors, Thyrotropin/immunology
- Receptors, Thyrotropin/metabolism
- T-Lymphocytes, Regulatory/pathology
- Thyroid Gland/immunology
- Thyroid Gland/metabolism
- Thyroid Gland/pathology
- Transcription Factors/genetics
- Transcription Factors/physiology
- AIRE Protein
Collapse
Affiliation(s)
- Alexander V Misharin
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and University of California Los Angeles School of Medicine, Los Angeles, California 90048, USA
| | | | | | | | | |
Collapse
|
17
|
Misharin A, Hewison M, Chen CR, Lagishetty V, Aliesky HA, Mizutori Y, Rapoport B, McLachlan SM. Vitamin D deficiency modulates Graves' hyperthyroidism induced in BALB/c mice by thyrotropin receptor immunization. Endocrinology 2009; 150:1051-60. [PMID: 18927213 PMCID: PMC2646531 DOI: 10.1210/en.2008-1191] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
TSH receptor (TSHR) antibodies and hyperthyroidism are induced by immunizing mice with adenovirus encoding the TSHR or its A-subunit. Depleting regulatory T cells (Treg) exacerbates thyrotoxicosis in susceptible BALB/c mice and induces hyperthyroidism in normally resistant C57BL/6 mice. Vitamin D plays an important role in immunity; high dietary vitamin D intake suppresses (and low intake enhances) adaptive immune responses. Vitamin D-induced immunosuppression may enhance Treg. Therefore, we hypothesized that decreased vitamin D intake would mimic Treg depletion and enhance hyperthyroidism induced by A-subunit adenovirus immunization. BALB/c mice had a reduced ability vs. C57BL/6 mice to generate the active metabolite of vitamin D (1,25-dihydroxyvitamin D3). Vitamin D deficiency induced subtle immune changes in BALB/c (not C57BL/6) mice. Compared with mice fed regular chow, vitamin D-deprived BALB/c mice had fewer splenic B cells and decreased interferon-gamma responses to mitogen and lacked memory T-cell responses to A-subunit protein. However, vitamin D deficiency did not alter TSHR antibody responses measured by ELISA, TSH binding inhibition, or cAMP generation from TSHR-expressing cells. Unexpectedly, compared with vitamin D-sufficient mice, vitamin D-deficient BALB/c mice had lower preimmunization T(4) levels and developed persistent hyperthyroidism. This difference was unrelated to the immunological changes between vitamin D-deficient or -sufficient animals. Previously, we found that different chromosomes or loci confer susceptibility to TSHR antibody induction vs. thyroid function. Our present studies provide evidence that an environmental factor, vitamin D, has only minor effects on induced immunity to the TSHR but directly affects thyroid function in mice.
Collapse
Affiliation(s)
- Alexander Misharin
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and University of California Los Angeles, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
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
|
19
|
Banga JP, Nielsen CH, Gilbert JA, El Fassi D, Hegedus L. Application of new therapies in Graves' disease and thyroid-associated ophthalmopathy: animal models and translation to human clinical trials. Thyroid 2008; 18:973-81. [PMID: 18752425 DOI: 10.1089/thy.2007.0406] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Most current approaches for treating Graves' disease are based essentially upon regimes developed nearly 50 years ago. Moreover, therapeutic approaches for complications such as thyroid-associated ophthalmopathy (TAO) and dermopathy are singularly dependent on conventional approaches of nonspecific immunosuppression. The recent development of an induced model of experimental Graves' disease, although incomplete as it lacks the extrathyroidal manifestations, provided opportunities to investigate immune intervention strategies, including influence upon the autoreactive B and T cell players in the autoimmune process. These major advances are generating new possibilities for therapeutic interventions for patients with Graves' disease and TAO.
Collapse
Affiliation(s)
- J Paul Banga
- Division of Gene and Cell Based Therapy, King's College London School of Medicine, London, United Kingdom.
| | | | | | | | | |
Collapse
|
20
|
McLachlan SM, Nagayama Y, Pichurin PN, Mizutori Y, Chen CR, Misharin A, Aliesky HA, Rapoport B. The link between Graves' disease and Hashimoto's thyroiditis: a role for regulatory T cells. Endocrinology 2007; 148:5724-33. [PMID: 17823263 DOI: 10.1210/en.2007-1024] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hyperthyroidism in Graves' disease is caused by thyroid-stimulating autoantibodies to the TSH receptor (TSHR), whereas hypothyroidism in Hashimoto's thyroiditis is associated with thyroid peroxidase and thyroglobulin autoantibodies. In some Graves' patients, thyroiditis becomes sufficiently extensive to cure the hyperthyroidism with resultant hypothyroidism. Factors determining the balance between these two diseases, the commonest organ-specific autoimmune diseases affecting humans, are unknown. Serendipitous findings in transgenic BALB/c mice, with the human TSHR A-subunit targeted to the thyroid, shed light on this relationship. Of three transgenic lines, two expressed high levels and one expressed low intrathyroidal A-subunit levels (Hi- and Lo-transgenics, respectively). Transgenics and wild-type littermates were depleted of T regulatory cells (Treg) using antibodies to CD25 (CD4(+) T cells) or CD122 (CD8(+) T cells) before TSHR-adenovirus immunization. Regardless of Treg depletion, high-expressor transgenics remained tolerant to A-subunit-adenovirus immunization (no TSHR antibodies and no hyperthyroidism). Tolerance was broken in low-transgenics, although TSHR antibody levels were lower than in wild-type littermates and no mice became hyperthyroid. Treg depletion before immunization did not significantly alter the TSHR antibody response. However, Treg depletion (particularly CD25) induced thyroid lymphocytic infiltrates in Lo-transgenics with transient or permanent hypothyroidism (low T(4), elevated TSH). Neither thyroid lymphocytic infiltration nor hypothyroidism developed in similarly treated wild-type littermates. Remarkably, lymphocytic infiltration was associated with intermolecular spreading of the TSHR antibody response to other self thyroid antigens, murine thyroid peroxidase and thyroglobulin. These data suggest a role for Treg in the natural progression of hyperthyroid Graves' disease to Hashimoto's thyroiditis and hypothyroidism in humans.
Collapse
Affiliation(s)
- Sandra M McLachlan
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
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
|