1
|
Merakchi K, Djerbib S, Soleimani M, Dumont JE, Miot F, De Deken X. Murine Thyroid IL-4 Expression Worsens Hypothyroidism on Iodine Restriction and Mitigates Graves Disease Development. Endocrinology 2022; 163:6650252. [PMID: 35881515 DOI: 10.1210/endocr/bqac107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Indexed: 11/19/2022]
Abstract
Cytokines are known to perturb thyroid function and the role of interleukin-4 (IL-4) in the pathogenesis of Graves disease (GD) remains controversial. In our mouse model overexpressing IL-4 in thyrocytes (Thyr-IL4), we have reported that adult mice preserved normal serum thyroxine despite an iodide uptake defect. In the present work, we evaluated if iodine restriction could uncover the thyroid deficiency in Thyr-IL4 animals as well as the role of pendrin overexpression as a compensatory mechanism. Moreover, using an experimental model of GD we investigated the effect of a local expression of IL-4 on the incidence of hyperthyroidism. Thyr-IL4 mice developed more rapidly elevated serum thyrotropin under low-iodine supply with thyroid enlargement and classical histological modifications. These hallmarks of hypothyroidism were all enhanced in Thyr-IL4 mice with complete pendrin invalidation. Following immunization, a lower proportion of Thyr-IL4 animals developed hyperthyroidism. Surprisingly, immunized Thyr-IL4 animals presented numerous leukocyte infiltrates, associated with increased intrathyroidal expression of IFN-γ. We have demonstrated that thyroid deficiency in Thyr-IL4 mice is partially compensated for by the excessive iodide content of the standard chow and the overexpression of pendrin in these animals. Furthermore, we have shown that the local expression of IL-4 in the thyroid attenuates GD progression, which was associated with enhanced thyroid infiltration by immune cells that could negatively affect thyroid function.
Collapse
Affiliation(s)
- Karima Merakchi
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Sami Djerbib
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Manoocher Soleimani
- Department of Medicine, University of New Mexico, School of Medicine, Albuquerque, New Mexico 87106, USA
| | - Jacques-Emile Dumont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Françoise Miot
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Xavier De Deken
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
2
|
Hotta M, Yoshimura H, Satake A, Tsubokura Y, Ito T, Nomura S. GM-CSF therapy inhibits chronic graft-versus-host disease via expansion of regulatory T cells. Eur J Immunol 2018; 49:179-191. [PMID: 30457669 DOI: 10.1002/eji.201847684] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 10/08/2018] [Accepted: 11/16/2018] [Indexed: 02/02/2023]
Abstract
Regulatory T cells (Tregs) attenuate excessive immune responses, making their expansion beneficial in immune-mediated diseases, including allogeneic bone marrow transplantation associated with graft-versus-host disease (GVHD). In addition to interleukin-2, Tregs require T-cell receptor and costimulatory signals from antigen-presenting cells, such as DCs, for their optimal proliferation. Granulocyte-macrophage colony-stimulating factor (GM-CSF) increases DC number and may promote DC-dependent Treg proliferation. Here, we demonstrate that GM-CSF treatment increases CD4+ CD8- DCs, which are associated with Treg expansion. In a mouse model of chronic GVHD (cGVHD), GM-CSF therapy expanded Tregs, protected against the development of skin GVHD, and regulated both Th1 and Th17 responses in the peripheral lymph nodes, resulting in an attenuation of skin cGVHD. Notably, the expanded Tregs were instrumental to GM-CSF-mediated cGVHD inhibition, which was dependent upon an increased ratio of Tregs to conventional T cells rather than augmentation of suppressive function. These data suggest that GM-CSF induces Treg proliferation by expanding CD4+ CD8- DCs, which in turn regulate alloimmune responses in a cGVHD mouse model. Thus, GM-CSF could be used as a therapeutic DC modulator to induce Treg expansion and to inhibit excessive alloimmune responses in immune-related diseases.
Collapse
Affiliation(s)
- Masaaki Hotta
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Hideaki Yoshimura
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Atsushi Satake
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Yukie Tsubokura
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Tomoki Ito
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| |
Collapse
|
3
|
STAT6 deficiency ameliorates Graves' disease severity by suppressing thyroid epithelial cell hyperplasia. Cell Death Dis 2016; 7:e2506. [PMID: 27906181 PMCID: PMC5260978 DOI: 10.1038/cddis.2016.398] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 10/30/2016] [Accepted: 11/02/2016] [Indexed: 01/02/2023]
Abstract
Signal transducer and activator of transcription 6 (STAT6) is involved in epithelial cell growth. However, little is known regarding the STAT6 phosphorylation status in Graves' disease (GD) and its role in thyroid epithelial cells (TECs). In this study, we found that STAT6 phosphorylation (p-STAT6) was significantly increased in TECs from both GD patients and experimental autoimmune Graves' disease mice and that STAT6 deficiency ameliorated GD symptoms. Autocrine IL-4 signalling in TECs activated the phosphorylation of STAT6 via IL-4 R engagement, and the downstream targets of STAT6 were Bcl-xL and cyclin D1. Thus, the IL-4-STAT6-Bcl-xL/cyclin D1 pathway is crucial for TEC hyperplasia, which aggravates GD. More importantly, in vitro and in vivo experiments demonstrated that STAT6 phosphorylation inhibited by AS1517499 decreased TEC hyperplasia, thereby reducing serum T3 and T4 and ameliorating GD. Thus, our study reveals that in addition to the traditional pathogenesis of GD, in which autoantibody TRAb stimulates thyroid-stimulating hormone receptors and consequently produces T3, T4, TRAb could also trigger TECs producing IL-4, and IL-4 then acts in an autocrine manner to activate p-STAT6 signalling and stimulate unrestricted cell growth, thus aggravating GD. These findings suggest that STAT6 inhibitors could be potent therapeutics for treating GD.
Collapse
|
4
|
Eskalli Z, Achouri Y, Hahn S, Many MC, Craps J, Refetoff S, Liao XH, Dumont JE, Van Sande J, Corvilain B, Miot F, De Deken X. Overexpression of Interleukin-4 in the Thyroid of Transgenic Mice Upregulates the Expression of Duox1 and the Anion Transporter Pendrin. Thyroid 2016; 26:1499-1512. [PMID: 27599561 PMCID: PMC5067804 DOI: 10.1089/thy.2016.0106] [Citation(s) in RCA: 12] [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: 02/07/2023]
Abstract
BACKGROUND The dual oxidases (Duox) are involved in hydrogen peroxide generation, which is essential for thyroid hormone synthesis, and therefore they are markers of thyroid function. During inflammation, cytokines upregulate DUOX gene expression in the airway and the intestine, suggesting a role for these proteins in innate immunity. It was previously demonstrated that interleukin-4 (IL-4) upregulates DUOX gene expression in thyrocytes. Although the role of IL-4 in autoimmune thyroid diseases has been studied extensively, the effects of IL-4 on thyroid physiology remain largely unknown. Therefore, a new animal model was generated to study the impact of IL-4 on thyroid function. METHODS Transgenic (Thyr-IL-4) mice with thyroid-targeted expression of murine IL-4 were generated. Transgene expression was verified at the mRNA and protein level in thyroid tissues and primary cultures. The phenotype of the Thyr-IL-4 animals was characterized by measuring serum thyroxine (T4) and thyrotropin levels and performing thyroid morphometric analysis, immunohistochemistry, whole transcriptome sequencing, quantitative reverse transcription polymerase chain reaction, and ex vivo thyroid function assays. RESULTS Thyrocytes from two Thyr-IL-4 mouse lines (#30 and #52) expressed IL-4, which was secreted into the extracellular space. Although 10-month-old transgenic animals had T4 and thyrotropin serum levels in the normal range, they had altered thyroid follicular structure with enlarged follicles composed of elongated thyrocytes containing numerous endocytic vesicles. These follicles were positive for T4 staining the colloid, indicating their capacity to produce thyroid hormones. RNA profiling of Thyr-IL-4 thyroid samples revealed modulation of multiple genes involved in inflammation, while no major leukocyte infiltration could be detected. Upregulated expression of Duox1, Duoxa1, and the pendrin anion exchanger gene (Slc26a4) was detected. In contrast, the iodide symporter gene Slc5a5 was markedly downregulated resulting in impaired iodide uptake and reduced thyroid hormone levels in transgenic thyroid tissue. Hydrogen peroxide production was increased in Thyr-IL-4 thyroid tissue compared with wild-type animals, but no significant oxidative stress could be detected. CONCLUSIONS This is the first study to show that ectopic expression of IL-4 in thyroid tissue upregulates Duox1/Duoxa1 and Slc26a4 expression in the thyroid. The present data demonstrate that IL-4 could affect thyroid morphology and function, mainly by downregulating Slc5a5 expression, while maintaining a normal euthyroid phenotype.
Collapse
Affiliation(s)
- Zineb Eskalli
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Younes Achouri
- Institut De Duve, Université Catholique de Louvain (UCL), Brussels, Belgium
| | - Stephan Hahn
- Laboratory of Image, Signal processing and Acoustics—Brussels School of Engineering, Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Marie-Christine Many
- Pôle de Morphologie (MORF), Université Catholique de Louvain (UCL), Brussels, Belgium
| | - Julie Craps
- Pôle de Morphologie (MORF), Université Catholique de Louvain (UCL), Brussels, Belgium
| | - Samuel Refetoff
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Xiao-Hui Liao
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Jacques E. Dumont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Jacqueline Van Sande
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Françoise Miot
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Xavier De Deken
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
5
|
Berchner-Pfannschmidt U, Moshkelgosha S, Diaz-Cano S, Edelmann B, Görtz GE, Horstmann M, Noble A, Hansen W, Eckstein A, Banga JP. Comparative Assessment of Female Mouse Model of Graves' Orbitopathy Under Different Environments, Accompanied by Proinflammatory Cytokine and T-Cell Responses to Thyrotropin Hormone Receptor Antigen. Endocrinology 2016; 157:1673-82. [PMID: 26872090 DOI: 10.1210/en.2015-1829] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We recently described a preclinical model of Graves' orbitopathy (GO), induced by genetic immunization of eukaryotic expression plasmid encoding human TSH receptor (TSHR) A-subunit by muscle electroporation in female BALB/c mice. The onset of orbital pathology is characterized by muscle inflammation, adipogenesis, and fibrosis. Animal models of autoimmunity are influenced by their environmental exposures. This follow-up study was undertaken to investigate the development of experimental GO in 2 different locations, run in parallel under comparable housing conditions. Functional antibodies to TSHR were induced in TSHR A-subunit plasmid-immunized animals, and antibodies to IGF-1 receptor α-subunit were also present, whereas control animals were negative in both locations. Splenic T cells from TSHR A-subunit primed animals undergoing GO in both locations showed proliferative responses to purified TSHR antigen and secreted interferon-γ, IL-10, IL-6, and TNF-α cytokines. Histopathological evaluation showed orbital tissue damage in mice undergoing GO, manifest by adipogenesis, fibrosis, and muscle damage with classic signs of myopathy. Although no inflammatory infiltrate was observed in orbital tissue in either location, the appearances were consistent with a "hit-and-run" immune-mediated inflammatory event. A statistically significant increase of cumulative incidence of orbital pathology when compared with control animals was shown for both locations, confirming onset of orbital dysimmune myopathy. Our findings confirm expansion of the model in different environments, accompanied with increased prevalence of T cell-derived proinflammatory cytokines, with relevance for pathogenesis. Wider availability of the model makes it suitable for mechanistic studies into pathogenesis and undertaking of novel therapeutic approaches.
Collapse
Affiliation(s)
- Utta Berchner-Pfannschmidt
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - Sajad Moshkelgosha
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - Salvador Diaz-Cano
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - Bärbel Edelmann
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - Gina-Eva Görtz
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - Mareike Horstmann
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - Alistair Noble
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - Wiebke Hansen
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - Anja Eckstein
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| | - J Paul Banga
- Molecular Ophthalmology (U.B.-P., S.M., G.-E.G., M.H., A.E., J.P.B.), Department of Ophthalmology; Department of Molecular Biology (B.E.); and Institute of Medical Microbiology (W.H.), University Hospital Essen/University of Duisburg-Essen, 45147 Essen, Germany; Faculty of Life Sciences and Medicine (S.M., A.N., J.P.B.), King's College London, London, SE5 9NU United Kingdom; and King's College Hospital NHS Foundation Trust (S.D.-C.), London, SE5 9RS United Kingdom
| |
Collapse
|
6
|
Song RH, Qin Q, Wang X, Yan N, Meng S, Shi XH, He ST, Zhang JA. Differential cytokine expression detected by protein microarray screening in peripheral blood of patients with refractory Graves' disease. Clin Endocrinol (Oxf) 2016; 84:402-7. [PMID: 25817259 DOI: 10.1111/cen.12778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 01/14/2015] [Accepted: 03/23/2015] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The prognosis of Graves' disease (GD) varies among patients. However, the immune pathogenesis of refractory GD is still unknown. The aim of this study was to explore the cytokine expression profile associated with refractory GD. METHODS Preliminary cytokine protein microarray screening was performed to detect differentially expressed cytokines in the plasma of four patients with refractory GD and four patients with stable GD. Some differentially expressed cytokines were then validated in plasma by enzyme-linked immunosorbent assay (ELISA) and in peripheral blood mononuclear cells (PBMCs) by quantitative real-time polymerase chain reaction (qRT-PCR) on another independent set of samples. RESULTS We found that 21 cytokines were differentially expressed between patients with intractable GD and those in remission, including 18 upregulated and 3 downregulated cytokines with a fold change >1·30 and <0·77, respectively. Intractability-related elevation of three cytokines (IL-4, IL-6 and IL-10) was validated by ELISA in plasma on another GD cohort with 30 patients in recurrence and 14 in remission (t-test, P = 0·035, 0·033 and 0·041, respectively). Furthermore, mRNA expression of IL-4, IL-6 and IL-10 in PBMCs, detected by qRT-PCR, was significantly elevated in patients with refractory GD compared with those in remission (P = 0·039, 0·047 and 0·042, respectively). CONCLUSION The severity of GD is associated with the aberrant expression and secretion of several cytokines that may serve as potential biomarkers and predictors for disease prognosis. Targeting these cytokines or their receptors may also lead to a novel therapeutic intervention for GD.
Collapse
Affiliation(s)
- Rong-hua Song
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Qiu Qin
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Xuan Wang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Ni Yan
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Shuai Meng
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Xiao-hong Shi
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Shuang-tao He
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Jin-an Zhang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
7
|
Luo Y, Yoshihara A, Oda K, Ishido Y, Suzuki K. Excessive Cytosolic DNA Fragments as a Potential Trigger of Graves' Disease: An Encrypted Message Sent by Animal Models. Front Endocrinol (Lausanne) 2016; 7:144. [PMID: 27895620 PMCID: PMC5107990 DOI: 10.3389/fendo.2016.00144] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/27/2016] [Indexed: 01/13/2023] Open
Abstract
Graves' hyperthyroidism is caused by autoantibodies directed against the thyroid-stimulating hormone receptor (TSHR) that mimic the action of TSH. The establishment of Graves' hyperthyroidism in experimental animals has proven to be an important approach to dissect the mechanisms of self-tolerance breakdown that lead to the production of thyroid-stimulating TSHR autoantibodies (TSAbs). "Shimojo's model" was the first successful Graves' animal model, wherein immunization with fibroblasts cells expressing TSHR and a major histocompatibility complex (MHC) class II molecule, but not either alone, induced TSAb production in AKR/N (H-2k) mice. This model highlights the importance of coincident MHC class II expression on TSHR-expressing cells in the development of Graves' hyperthyroidism. These data are also in agreement with the observation that Graves' thyrocytes often aberrantly express MHC class II antigens via mechanisms that remain unclear. Our group demonstrated that cytosolic self-genomic DNA fragments derived from sterile injured cells can induce aberrant MHC class II expression and production of multiple inflammatory cytokines and chemokines in thyrocytes in vitro, suggesting that severe cell injury may initiate immune responses in a way that is relevant to thyroid autoimmunity mediated by cytosolic DNA signaling. Furthermore, more recent successful Graves' animal models were primarily established by immunizing mice with TSHR-expressing plasmids or adenovirus. In these models, double-stranded DNA vaccine contents presumably exert similar immune-activating effect in cells at inoculation sites and thus might pave the way toward successful Graves' animal models. This review focuses on evidence suggesting that cell injury-derived self-DNA fragments could act as Graves' disease triggers.
Collapse
Affiliation(s)
- Yuqian Luo
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Aya Yoshihara
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
- Department of Education Planning and Development, Faculty of Medicine, Toho University, Tokyo, Japan
| | - Kenzaburo Oda
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
- Department of Internal Medicine, Division of Diabetes, Metabolism and Endocrinology, Toho University, Tokyo, Japan
| | - Yuko Ishido
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Koichi Suzuki
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
- *Correspondence: Koichi Suzuki,
| |
Collapse
|
8
|
Meta-analysis of the association between vitamin D and autoimmune thyroid disease. Nutrients 2015; 7:2485-98. [PMID: 25854833 PMCID: PMC4425156 DOI: 10.3390/nu7042485] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 02/10/2015] [Accepted: 03/16/2015] [Indexed: 01/13/2023] Open
Abstract
Although emerging evidence suggests that low levels of vitamin D may contribute to the development of autoimmune disease, the relationship between vitamin D reduction and autoimmune thyroid disease (AITD), which includes Graves’ disease (GD) and Hashimoto thyroiditis (HT), is still controversial. The aim was to evaluate the association between vitamin D levels and AITD through systematic literature review. We identified all studies that assessed the association between vitamin D and AITD from PubMed, Embase, CENTRAL, and China National Knowledge Infrastructure (CNKI) databases. We included studies that compared vitamin D levels between AITD cases and controls as well as those that measured the odds of vitamin D deficiency by AITD status. We combined the standardized mean differences (SMD) or the odds ratios (OR) in a random effects model. Twenty case-control studies provided data for a quantitative meta-analysis. Compared to controls, AITD patients had lower levels of 25(OH)D (SMD: −0.99, 95% CI: −1.31, −0.66) and were more likely to be deficient in 25(OH)D (OR 2.99, 95% CI: 1.88, 4.74). Furthermore, subgroup analyses result showed that GD and HT patients also had lower 25(OH)D levels and were more likely to have a 25(OH)D deficiency, suggesting that low levels of serum 25(OH)D was related to AITD.
Collapse
|
9
|
Dastmalchi R, Farazmand A, Noshad S, Mozafari M, Mahmoudi M, Esteghamati A, Amirzargar A. Polymorphism of killer cell immunoglobulin-like receptors (KIR) and their HLA ligands in Graves' disease. Mol Biol Rep 2014; 41:5367-74. [PMID: 24852304 DOI: 10.1007/s11033-014-3408-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 05/11/2014] [Indexed: 12/11/2022]
Abstract
Killer immunoglobulin-like receptors (KIR) play a pivotal role in commencement of both innate and adaptive immunity. Dysregulation of KIRs is associated with an increased risk of autoimmune disorders. This study was designed to assess whether polymorphisms in KIR gene family and their respective HLA class I ligands confer protection or susceptibly to Graves' disease (GD). Eighty patients with confirmed GD (cases) and 176 healthy unrelated subjects (controls) were recruited. Using a polymerase chain reaction sequence-specific primer directed method (PCR-SSP), presence or absence of KIR genes and their HLA ligands were determined. No significant differences were observed between case and control groups regarding individual KIR gene frequencies (p > 0.05 in all cases). The frequency of group A haplotype (the most common KIR haplotype, encompassing 2DL1/2DL3/3DL1/2DS4/2DP1/3DP1/2DL4/3DL2/3DL3), was not different between individuals with and without GD. Moreover, among all other haplotypes (group Bx), no significant differences regarding distribution of centromeric and telomeric gene clusters were identifiable. Inhibitory/activatory gene contents were also comparable between the two groups. Four models of KIR-HLA interaction (inhibition, activation, unrestrained inhibition, and unrestrained activation) were constructed. No combination proved to confer susceptibility to, or offer protection against GD. It seems that the contribution of KIR gene polymorphism to natural killer cell dysfunction and other autoimmune abnormalities observed in GD is limited.
Collapse
Affiliation(s)
- Romina Dastmalchi
- Department of Cellular and Molecular Biology, Kish International Campus, University of Tehran, Kish Island, Iran,
| | | | | | | | | | | | | |
Collapse
|
10
|
Dalan R, Leow MKS. Immune manipulation for Graves' disease: re-exploring an unfulfilled promise with modern translational research. Eur J Intern Med 2012; 23:682-91. [PMID: 22877994 DOI: 10.1016/j.ejim.2012.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 07/11/2012] [Accepted: 07/12/2012] [Indexed: 11/15/2022]
Abstract
Although Graves' disease is the commonest autoimmune thyroid disorder, current therapeutics typically center on the eradication of the antigenic stimulus (i.e. thyroid gland) rather than radically tackling the underlying autoimmune processes. Consequently, it is not a surprising fact that Graves' disease remains essentially a chronic drug-dependent ailment afflicting untold numbers worldwide for decades despite progress in deciphering its autoimmune nature. Addressing the latter is key to a future cure as underscored by appropriate, albeit crude, proof-of-concept scenarios of clinical remissions achieved with hematopoietic stem cell transplantation, immune down-regulation during pregnancy, use of corticosteroids or immunosuppressives, and cytokine biologics in animal models. Ongoing basic and translational research to further elucidate and refine our understanding of the pathogenesis of Graves' disease holds the promise of unraveling novel immune manipulative techniques that will bring the world a step closer to the elusive cure of the underlying autoimmunity amidst skepticisms on the value of the science from the present lack of paralleled advances at the bedside. We review the updated literature and describe the forms of immune manipulation hitherto explored that will offer a route to a future cure, from thionamides, hematopoietic stem cell transplantation to the latest immunomodulatory agents.
Collapse
Affiliation(s)
- Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore.
| | | |
Collapse
|
11
|
El-Malky M, Nabih N, Heder M, Saudy N, El-Mahdy M. Helminth infections: therapeutic potential in autoimmune disorders. Parasite Immunol 2012; 33:589-93. [PMID: 21797885 DOI: 10.1111/j.1365-3024.2011.01324.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Knowledge of immunity enables us to predict that the reactions set in response to infection with helminth would prevent concomitant disease driven by an opposing spectrum of immune events. In another way, the immune response generated to combat the helminth infection could counteract the immunopathological reactions that drive autoimmune diseases. Rodent model systems recapitulate many aspects of human autoimmune diseases and have been enormously useful in defining mechanisms of immunopathology after infection. From this theoretical perspective, many researchers have proved that infection with a variety of helminth can ameliorate disease in murine model systems. Thus, helminth-evoked Th2 events were shown to improve disorders in which Th1 events predominated. This raised the question, 'Can this information be translated into therapies for autoimmune diseases in humans via actual infection, cell delivery or drug intervention?' In this review, we will present some experimental trails to treat autoimmune disorders through establishment of some parasitic infections.
Collapse
Affiliation(s)
- M El-Malky
- Departments of Medical Parasitology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | | | | | | | | |
Collapse
|
12
|
Ganesh BB, Bhattacharya P, Gopisetty A, Prabhakar BS. Role of cytokines in the pathogenesis and suppression of thyroid autoimmunity. J Interferon Cytokine Res 2011; 31:721-31. [PMID: 21823922 DOI: 10.1089/jir.2011.0049] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Autoimmune thyroid diseases (AITD) are one of the most common organ-specific autoimmune disorders, of which Hashimoto's thyroiditis (HT) and Graves' disease (GD) are 2 of the most common clinical expressions. HT is characterized by hypothyroidism that results from the destruction of the thyroid by thyroglobulin-specific T cell-mediated autoimmune response. In contrast, GD is characterized by hyperthyroidism due to excessive production of thyroid hormone induced by thyrotropin receptor-specific stimulatory autoantibodies. Cytokines play a crucial role in modulating immune responses that affect the balance between maintenance of self-tolerance and initiation of autoimmunity. However, the role of cytokines is often confusing and is neither independent nor exclusive of other immune mediators. A regulatory cytokine may either favor induction of tolerance against thyroid autoimmune disease or favor activation and/or exacerbation of autoimmune responses. These apparently contradictory functions of a given cytokine are primarily influenced by the nature of co-signaling delivered by other cytokines. Consequently, a thorough understanding of the role of a particular cytokine in the context of a specific immune response is essential for the development of appropriate strategies to modulate cytokine responses to maintain or restore health. This review provides a summary of recent research pertaining to the role of cytokines in the pathogenesis of AITD with a particular emphasis on the therapeutic applications of cytokine modulation.
Collapse
Affiliation(s)
- Balaji B Ganesh
- Department of Microbiology and Immunology, Research Resources Center, College of Medicine, University of Illinois at Chicago, Illinois, USA
| | | | | | | |
Collapse
|
13
|
Association of an IL-4 gene haplotype with Graves disease in children: experimental study and meta-analysis. Hum Immunol 2011; 72:256-61. [DOI: 10.1016/j.humimm.2010.12.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 12/03/2010] [Accepted: 12/27/2010] [Indexed: 01/26/2023]
|
14
|
Abstract
Apoptosis or programmed cell death plays a central role in regulating not only the development of lymphocytes but also in their homeostasis. A breakdown in apoptosis related signaling mechanisms could result in the development of autoimmune disorders. The past decade has witnessed an explosive increase in knowledge with respect to various apoptotic signaling pathways and their aberrant behavior in autoimmune disorders. Although Fas/FasL mediated signaling appears to be a common paradigm that has emerged from studies in various autoimmune disorders, examples suggesting a role for other cell death pathways have also surfaced. Understanding the definitive role of apoptosis in various autoimmune disorders is likely to define novel targets for future therapeutic intervention.
Collapse
Affiliation(s)
- Kanteti V Prasad
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
15
|
Vasu C, Holterman MJ, Prabhakar BS. Modulation of Dendritic Cell Function and Cytokine Production to Prevent Thyroid Autoimmunity. Autoimmunity 2009; 36:389-96. [PMID: 14669946 DOI: 10.1080/08916930310001603073] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Understanding autoimmune thyroid diseases provides an unique perspective on the role of various components of the immune system in the pathogenesis of organ specific autoimmune diseases, whether the effector mechanism involves autoantibodies or T cells. Hashimoto's thyroiditis (HT) is largely mediated by thyroglobulin specific T cells, while Graves' disease (GD) is mediated by thyrotropin receptor specific autoantibodies. HT is characterized by thyroid destruction mediated by infiltrating or activated resident immune cells through a variety of mechanisms. In contrast GD is characterized by excessive production of thyroid hormone with little or no glandular destruction. Irrespective of the effector mechanism involved, dendritic cells (DCs) are required for the induction of an efficient primary response and thus are the first cells involved in an autoimmune response. DCs also provide the essential link between the innate and the adaptive immune system through co-stimulatory molecules and the production of cytokines and chemokines. Furthermore, inflammatory cytokines also appear to enhance the susceptibility of thyrocytes to apoptosis. In this review, we discuss the role of innate immunity in initiating an adaptive autoimmune response against the thyroid. We will explore the role of different mechanisms involved in breaking self-tolerance to thyroid antigens. Further, we will discuss recent developments in the development of experimental therapeutics against AITD.
Collapse
|
16
|
Abstract
An animal model of Graves' disease (GD) will help us to clearly understand the role of thyroid-stimulating hormone receptor (TSHR)-specific T cells and TSHR-Abs during the development of GD and to develop TSHR-specific immunotherapy. This review focuses on four different recent approaches towards the development of an animal model of GD. These approaches are: (1) Immunization of AKR/N mice with fibroblasts coexpressing syngeneic major histocompatibility complex (MHC) class II and TSHR. (2) Immunization of selected strains of mice with an expression vector containing TSHR cDNA. (3) Immunization of BALB/c mice with syngeneic M12 cells or xenogenic HEK-293 cells expressing full-length or extracellular domain of TSHR (ETSHR). (4) Injection of adenovirus-expressing TSHR into BALB/c mice.
Collapse
Affiliation(s)
- Gattadahalli S Seetharamaiah
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 8600 University Boulevard, Evansville, IN 47712, USA.
| |
Collapse
|
17
|
Dağdelen S, Kong YCM, Banga JP. Toward better models of hyperthyroid Graves' disease. Endocrinol Metab Clin North Am 2009; 38:343-54, viii. [PMID: 19328415 DOI: 10.1016/j.ecl.2009.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Graves' disease affects only humans. Although it is a treatable illness, medical therapy with antithyroid drugs is imperfect, showing high rates of recurrence. Furthermore, the etiology and treatment of the associated ophthalmopathy still represent problematic issues. Animal models could contribute to the solution of such problems by providing a better understanding of the underlying pathogenesis and could be used for evaluating novel therapeutic strategies. This article discusses the pursuit of a better experimental model for hyperthyroid Graves' disease and outlines how this research has clarified the immunology of the disease.
Collapse
Affiliation(s)
- Selçuk Dağdelen
- Department of Diabetes and Endocrinology, King's College London School of Medicine, Denmark Hill Campus, The Rayne Institute, London, UK.
| | | | | |
Collapse
|
18
|
Chu X, Dong C, Lei R, Sun L, Wang Z, Dong Y, Shen M, Wang Y, Wang B, Zhang K, Yang L, Li Y, Yuan W, Wang Y, Song H, Jin L, Xiong M, Huang W. Polymorphisms in the interleukin 3 gene show strong association with susceptibility to Graves' disease in Chinese population. Genes Immun 2009; 10:260-6. [PMID: 19262575 DOI: 10.1038/gene.2009.3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Graves' disease (GD) is a common organ-specific autoimmune disorder, which is multifactorial and develops in genetically susceptible individuals. We had earlier mapped a susceptibility locus for GD to chromosome 5q31-33 in a linkage study. Here we used tag single-nucleotide polymorphisms (SNPs) to search for genetic variants associated with GD, and examined 19 functional candidate genes in this chromosomal region. We identified 192 polymorphisms by re-sequencing the candidate genes, and selected 51 tagSNPs to genotype in a case-control collection of 1118 south Han Chinese subjects (428 cases and 690 controls). Initial analysis suggested that a non-synonymous SNP rs40401 (P27S) of interleukin 3 (IL3) was associated with GD, and further fine-mapping showed that rs40401, or its perfect proxy SNP rs31480 in the 5' flanking region of IL3, fully accounted for the association signal at this locus. We replicated significant association of rs40401 with GD in an independent sample collection of 839 north Han Chinese subjects. A combined analysis revealed strong validation of this association (odds ratio (OR(common))=1.63, combined P (P(comb))=4 x 10(-6) in the Recessive disease model). This study provides convincing evidence that the IL3 gene is a susceptibility locus for GD in the Chinese population.
Collapse
Affiliation(s)
- X Chu
- Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ganesh BB, Cheatem DM, Sheng JR, Vasu C, Prabhakar BS. GM-CSF-induced CD11c+CD8a--dendritic cells facilitate Foxp3+ and IL-10+ regulatory T cell expansion resulting in suppression of autoimmune thyroiditis. Int Immunol 2009; 21:269-82. [PMID: 19174473 DOI: 10.1093/intimm/dxn147] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
GM-CSF plays an essential role in the differentiation of dendritic cells (DCs). Our studies have shown that GM-CSF treatment can induce semi-mature DCs and CD4+CD25+ regulatory T cells (Tregs) and suppress ongoing autoimmunity in mouse models. In this study, we examined the differences in the potential of GM-CSF to exert tolerogenic function on CD8a+ and CD8a- sub-populations of DCs in vivo. We show that GM-CSF modulates CD8a-, but not CD8a+ DCs in vivo, by inhibiting the surface expression of activation markers MHC II and CD80 and production of inflammatory cytokines such as IL-12 and IL-1beta. Self-antigen [mouse thyroglobulin (mTg)] presentation by GM-CSF-exposed CD8a- DCs to T cells from mTg-primed mice induced a profound increase in the frequency of forkhead box P3 (FoxP3)-expressing T cells compared with antigen presentation by GM-CSF-exposed CD8a+ DCs and control CD8a+ and CD8a- DCs. This tolerogenic property of GM-CD8a- DCs was abrogated when IL-12 was added. GM-CSF-exposed CD8a- DCs could also induce secretion of significantly higher amounts of IL-10 by T cells from mTg-primed mice. Importantly, adoptive transfer of CD8a- DCs from GM-CSF-treated SCID mice, but not untreated mice, into wild-type CBA/J mice prevented the development of experimental autoimmune thyroiditis (EAT) in the recipient animals upon immunization with mTg. Collectively, our results show that GM-CSF renders CD8a- DCs tolerogenic, and these DCs induce Foxp3+ and IL-10+ Tregs.
Collapse
Affiliation(s)
- Balaji B Ganesh
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
20
|
Cheatem D, Ganesh BB, Gangi E, Vasu C, Prabhakar BS. Modulation of dendritic cells using granulocyte-macrophage colony-stimulating factor (GM-CSF) delays type 1 diabetes by enhancing CD4+CD25+ regulatory T cell function. Clin Immunol 2009; 131:260-70. [PMID: 19171501 DOI: 10.1016/j.clim.2008.12.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 11/12/2008] [Accepted: 12/01/2008] [Indexed: 11/24/2022]
Abstract
Abnormalities in DC function are implicated in defective immune regulation that leads to type-1 diabetes (T1D) in NOD mice and humans. In this study, we used GM-CSF and Flt3-L to modulate DC function in NOD mice and observed the effects on T1D development. Treatment with either ligand at earlier stages of insulitis suppressed the development of T1D. Unlike Flt3-L, GM-CSF was more effective in suppressing T1D, even when administered at later stages of insulitis. In vitro studies and in vivo adoptive transfer experiments revealed that CD4+CD25+ T cells from GM-CSF-treated mice could suppress effector T cell response and T1D. This suppression is likely mediated through enhanced IL-10 and TGF-beta1 production. Adoptive transfer of GM-CSF exposed DCs to naive mice resulted in an expansion of Foxp3+ T cells and a significant delay in T1D onset. Our results indicate that GM-CSF acted primarily on DCs and caused an expansion of Foxp3+ Tregs which delayed the onset of T1D in NOD mice.
Collapse
Affiliation(s)
- Donald Cheatem
- Department of Microbiology and Immunology (MC790), College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
21
|
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: 0.9] [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
|
22
|
Takahashi H, Amagai M, Nishikawa T, Fujii Y, Kawakami Y, Kuwana M. Novel System Evaluating In Vivo Pathogenicity of Desmoglein 3-Reactive T Cell Clones Using Murine Pemphigus Vulgaris. THE JOURNAL OF IMMUNOLOGY 2008; 181:1526-35. [DOI: 10.4049/jimmunol.181.2.1526] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
23
|
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
|
24
|
Abstract
There is increasing evidence showing that apoptosis plays a role in the development of the autoimmune thyroid diseases-Hashimoto's (lymphocytic) thyroiditis (HT) and Graves' disease (GD). The immune pathogenesis of HT and GD is not yet fully understood, but evidence points toward several steps. A defect in CD4(+)CD25(+) T regulatory cells breaks the immunological tolerance of the host and induces an abnormal production of cytokines, which facilitates the initiation of apoptosis. Though apoptosis appears to play a role in the pathogenesis of both HT and GD, the mechanisms that mediate these processes appear different. The induction of apoptosis in HT results in the destruction of thyrocytes, while apoptosis in the GD leads to damage of thyroid-infiltrating lymphocytes. The differences in the apoptotic mechanisms produce two very different forms of thyroid autoimmune responses, eventually developing into HT and GD, respectively.
Collapse
Affiliation(s)
- Su He Wang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.
| | | |
Collapse
|
25
|
Abstract
Interferon-gamma (IFN-gamma), a prototypic proinflammatory cytokine produced by several different cell types, including the Th1 subset of CD4(+) T cells, plays an important role in inflammation and autoimmune diseases. This review focuses on the varied and often contrasting roles of IFN-gamma in three murine models of autoimmune thyroid disease, experimentally induced autoimmune thyroiditis, the model of iodine-induced spontaneous autoimmune thyroiditis in NOD.H-2h4 mice and several different murine models of Graves' disease.
Collapse
Affiliation(s)
- Yujiang Fang
- Department of Internal Medicine, University of Missouri, Columbia, Missouri, USA
| | | | | |
Collapse
|
26
|
Kaneda T, Honda A, Hakozaki A, Fuse T, Muto A, Yoshida T. An improved Graves' disease model established by using in vivo electroporation exhibited long-term immunity to hyperthyroidism in BALB/c mice. Endocrinology 2007; 148:2335-44. [PMID: 17255207 DOI: 10.1210/en.2006-1077] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In Graves' disease, the overstimulation of the thyroid gland and hyperthyroidism are caused by autoantibodies directed against the TSH receptor (TSHR) that mimics the action of TSH. The establishment of an animal model is an important step to study the pathophysiology of autoimmune hyperthyroidism and for immunological analysis. In this study, we adopted the technique of electroporation (EP) for genetic immunization to achieve considerable enhancement of in vivo human TSHR (hTSHR) expression and efficient induction of hyperthyroidism in mice. In a preliminary study using beta-galactosidase (beta-gal) expression vectors, beta-gal introduced into the muscle by EP showed over 40-fold higher enzymatic activity than that introduced via previous direct gene transfer methods. The sustained hTSHR mRNA expression derived from cDNA transferred by EP was detectable in muscle tissue for at least 2 wk by RT-PCR. Based on these results, we induced hyperthyroidism via two expression vectors inserted with hTSHR or hTSHR289His cDNA. Consequently, 12.0-31.8% BALB/c mice immunized with hTSHR and 79.2-95.7% immunized with hTSHR289His showed high total T(4) levels due to the TSHR-stimulating antibody after three to four times repeated immunization by EP, and thyroid follicles of which were hyperplastic and had highly irregular epithelium. Moreover, TSHR-stimulating antibody surprisingly persisted more than 8 months after the last immunization. These results demonstrate that genetic immunization by in vivo EP is more efficient than previous procedures, and that it is useful for delineating the pathophysiology of Graves' disease.
Collapse
Affiliation(s)
- Toshio Kaneda
- Department of Pathophysiology, Hoshi University, Shinagawa, Tokyo 142-8501, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Sheng JR, Li L, Ganesh BB, Vasu C, Prabhakar BS, Meriggioli MN. Suppression of Experimental Autoimmune Myasthenia Gravis by Granulocyte-Macrophage Colony-Stimulating Factor Is Associated with an Expansion of FoxP3+Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:5296-306. [PMID: 17015715 DOI: 10.4049/jimmunol.177.8.5296] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Dendritic cells (DCs) have the potential to activate or tolerize T cells in an Ag-specific manner. Although the precise mechanism that determines whether DCs exhibit tolerogenic or immunogenic functions has not been precisely elucidated, growing evidence suggests that DC function is largely dependent on differentiation status, which can be manipulated using various growth factors. In this study, we investigated the effects of mobilization of specific DC subsets-using GM-CSF and fms-like tyrosine kinase receptor 3-ligand (Flt3-L)-on the susceptibility to induction of experimental autoimmune myasthenia gravis (EAMG). We administered GM-CSF or Flt3-L to C57BL/6 mice before immunization with acetylcholine receptor (AChR) and observed the effect on the frequency and severity of EAMG development. Compared with AChR-immunized controls, mice treated with Flt3-L before immunization developed EAMG at an accelerated pace initially, but disease frequency and severity was comparable at the end of the observation period. In contrast, GM-CSF administered before immunization exerted a sustained suppressive effect against the induction of EAMG. This suppression was associated with lowered serum autoantibody levels, reduced T cell proliferative responses to AChR, and an expansion in the population of FoxP3+ regulatory T cells. These results highlight the potential of manipulating DCs to expand regulatory T cells for the control of autoimmune diseases such as MG.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/therapy
- Cell Communication/immunology
- Cell Proliferation/drug effects
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Forkhead Transcription Factors
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- Immunization
- Membrane Proteins/administration & dosage
- Membrane Proteins/pharmacology
- Mice
- Mice, Inbred C57BL
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Myasthenia Gravis, Autoimmune, Experimental/prevention & control
- Myasthenia Gravis, Autoimmune, Experimental/therapy
- Receptors, Cholinergic/administration & dosage
- Receptors, Cholinergic/immunology
- T-Lymphocytes, Regulatory/cytology
Collapse
Affiliation(s)
- Jian Rong Sheng
- Department of Neurology and Rehabilitation, University of Illinois, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
28
|
Gilbert JA, Kalled SL, Moorhead J, Hess DM, Rennert P, Li Z, Khan MZ, Banga JP. Treatment of autoimmune hyperthyroidism in a murine model of Graves' disease with tumor necrosis factor-family ligand inhibitors suggests a key role for B cell activating factor in disease pathology. Endocrinology 2006; 147:4561-8. [PMID: 16794009 DOI: 10.1210/en.2006-0507] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Hyperthyroid Graves' disease is a common autoimmune disorder mediated by agonistic antibodies to the TSH receptor, termed thyroid stimulating antibodies (TSAbs). Recently members of the TNF superfamily, B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL), have been identified along with their receptors, B cell maturation antigen and transmembrane activator and calcium-modulator and cyclophilin ligand interactor, and the BAFF-specific receptor. BAFF is a fundamental B cell survival/maturation factor, and both BAFF and APRIL have been implicated in antibody production. We investigated the effect of interfering with BAFF- and APRIL-mediated signals in an induced model of Graves' disease by blockade of these factors using soluble decoy receptors. In a therapeutic setting in mice with established hyperthyroidism, we show that blockade of BAFF or BAFF+APRIL with BAFF-specific receptor-Fc and B cell maturation antigen-Fc, respectively, leads to significant reductions in the induced hyperthyroidism. This was supported by a parallel pattern of declining TSAbs in the responding animals. Histopathological analysis of splenic sections from treated animals revealed marked reductions in the B cell follicle regions, but staining with anti-CD138 revealed the persistence of plasma cells. Thus, the reductions in TSAbs in the treated animals were not related to overall plasma cell numbers in the secondary lymphoid organs. Our results are the first to demonstrate attenuation of established hyperthyroidism by therapeutic intervention aimed at autoreactive B cells and indicate that both BAFF and APRIL appear to play important roles in the development and survival of the autoantibody producing cells in this model.
Collapse
Affiliation(s)
- Jacqueline A Gilbert
- Division of Gene and Cell-Based Therapy, King's College London School of Medicine, Bessemer Road, London SE5 9PJ, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Land KJ, Gudapati P, Kaplan MH, Seetharamaiah GS. Differential requirement of signal transducer and activator of transcription-4 (Stat4) and Stat6 in a thyrotropin receptor-289-adenovirus-induced model of Graves' hyperthyroidism. Endocrinology 2006; 147:111-9. [PMID: 16195404 DOI: 10.1210/en.2005-1031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T helper type 1 (Th1) and Th2 cells have critical roles in the development of cell-mediated and humoral immune responses, respectively. This division of function predicts that Th1 cells mediate inflammatory diseases and Th2 cells promote antibody (Ab)-mediated autoimmunity. Our previous studies using HEK-293 cells expressing the extracellular domain of the TSH receptor (TSHR) showed that Stat4-/- mice, which lack Th1 cells, are susceptible, whereas Stat6-/- mice, which lack Th2 cells, are resistant to the induction of Graves' hyperthyroidism. To investigate the role of Stat4 and Stat6 genes in other murine models of hyperthyroidism, we injected wild-type BALB/c, Stat4-/-, and Stat6-/- mice with an adenovirus expressing amino acid residues 1-289 of TSHR (TSHR-289-ad or 289-ad). The viral system induces a much stronger immune response with much more rapid onset of disease. Our results showed that 56% of wild-type, 75% of Stat4-/-, and 39% of Stat6-/- mice developed hyperthyroidism. Hyperthyroid mice exhibited thyroid stimulatory Abs. The Stat4-/- mice developed a higher incidence and greater severity of hyperthyroidism compared with wild-type and Stat6-/- mice. BALB/c and Stat4-/- mice showed significantly higher TSHR Abs of the IgG1 subclass and IL-4 compared with Stat6-/- mice. In contrast, Stat6-/- mice had predominantly the IgG2a subclass of TSHR Ab and produced significantly higher amounts of IFN-gamma than BALB/c and Stat4-/- mice. All hyperthyroid mice showed enlarged thyroid glands with hyperactivity. These results suggest that in the TSHR-289-ad model, the Th2 cells are more efficient in mediating disease, but in the absence of Th2 cells, Th1 cells may still initiate a reduced incidence of Graves' hyperthyroidism.
Collapse
Affiliation(s)
- Kimberly J Land
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Evansville, Indiana 47712, USA
| | | | | | | |
Collapse
|
30
|
Aust G, Krohn K, Morgenthaler NG, Schröder S, Schütz A, Edelmann J, Brylla E. Graves' disease and Hashimoto's thyroiditis in monozygotic twins: case study as well as transcriptomic and immunohistological analysis of thyroid tissues. Eur J Endocrinol 2006; 154:13-20. [PMID: 16381986 DOI: 10.1530/eje.1.02063] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To report on the rare simultaneous occurrence of Graves' disease (GD) and Hashimoto's thyroiditis (HT) in monozygotic twins. DESIGN We compared the pattern of thyroid tissue-derived cDNAs to gain insight into previous and ongoing immune destruction and reconstruction processes using microarrays. The results were confirmed by immunohistology and real-time PCR. RESULTS Destruction of thyroid tissue in HT reduced levels of thyrocyte-related cDNAs and cDNAs encoding extracellular matrix components, but increased levels of proteases involved in extracellular matrix degradation compared with GD. Lymphocytic infiltrates forming ectopic follicles replaced the thyroid tissue almost completely in HT. Thus, lymphocyte-related cDNA levels were higher in HT than in GD. The same was true for many chemokines and their receptors, which not only enable migration towards the thyroid but also maintain the lymphocytic infiltrate. HT also showed increased levels of cDNAs encoding molecules related to apoptosis than did GD. Surprisingly, the Th1- and Th2-specific cytokine profiles suggested for HT and GD respectively could not be confirmed. cDNAs encoding factors and receptors involved in angiogenesis were increased in GD compared with HT. CONCLUSIONS Comparison of gene expression reflects the cellular differences between the two types of autoimmune thyroid disease in twins with identical genetic and similar environmental background.
Collapse
Affiliation(s)
- G Aust
- Research Laboratories, Center of Surgery, University of Leipzig, Germany.
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The most common cause of hyperthyroidism is Graves' disease, which represents a typical example of an organ-specific autoimmune condition. The exact triggers for the disease remain unknown, but are likely to involve a complex interaction between multiple environmental factors in a genetically predisposed individual. The main feature of the condition is the presence of thyroid-stimulating antibodies, which activate the thyroid- stimulating hormone receptor, resulting in hyperthyroidism. These antibodies may also be involved in the extrathyroidal complications of the disease. The recent generation of thyroid-stimulating antibodies in animal models and the isolation of monoclonal thyroid-stimulating antibodies from a patient with Graves' disease should allow the detailed study of thyroid-stimulating antibodies-thyroid-stimulating hormone receptor interactions. This will help to shed more light on disease pathogenesis and may offer new treatment strategies in difficult cases, particularly in patients with extrathyroidal complications.
Collapse
Affiliation(s)
- Ramzi A Ajjan
- a Academic Unit of Molecular and Vascular Medicine, The LIGHT Laboratories, University of Leeds, Leeds LS2 9JT UK
| | - Anthony P Weetman
- b Division of Clinical Sciences, University of Sheffield, Northern General Hospital, Sheffield S5 7AU UK.
| |
Collapse
|
32
|
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
|
33
|
Abstract
Graves' disease is a common organ-specific autoimmune disease characterized by overstimulation of the thyroid gland with agonistic anti-thyrotropin (TSH) receptor autoantibodies, which leads to hyperthyroidism and diffuse hyperplasia of the thyroid gland. Several groups including us have recently established several animal models of Graves' hyperthyroidism using novel immunization approaches, such as in vivo expression of the TSH receptor by injecting syngeneic living cells co-expressing the TSH receptor, the major histocompatibility complex (MHC) class II antigen and a costimulatory molecule, or genetic immunization using plasmid or adenovirus vectors coding the TSH receptor. This breakthrough has made it possible for us to study the pathogenesis of Graves' disease in more detail and has provided important insights into our understanding of disease pathogenesis. The important new findings that have emerged include: (i) the shed A subunit being the major autoantigen for TSAb, (ii) the significant role played by dendritic cells (DCs) as professional antigen-presenting cells in initiating disease development, (iii) contribution of MHC and particularly non-MHC genetic backgrounds in disease susceptibility, and (iv) influence of some particular infectious pathogens on disease development. However, the data regarding Th1/Th2 balance of TSH receptor-specific immune response or the association of Graves' hyperthyroidism with intrathyroidal lymphocytic infiltration are rather inconsistent. Future studies with these models will hopefully lead to better understanding of disease pathogenesis and help develop novel strategies for treatment and ultimately prevention of Graves' disease in humans.
Collapse
Affiliation(s)
- Yuji Nagayama
- Department of Medical Gene Technology, Atomic Bomb Disease Institute, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki
| |
Collapse
|
34
|
Aust G, Kamprad M, Lamesch P, Schmücking E. CXCR6 within T-helper (Th) and T-cytotoxic (Tc) type 1 lymphocytes in Graves' disease (GD). Eur J Endocrinol 2005; 152:635-43. [PMID: 15817921 DOI: 10.1530/eje.1.01892] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE In Graves' disease (GD), stimulating anti-TSH receptor antibodies are responsible for hyperthyroidism. T-helper 2 (Th2) cells were expected to be involved in the underlying immune mechanism, although this is still controversial. The aim of this study was to examine the expression of CXCR6, a chemokine receptor that marks functionally specialized T-cells within the Th1 and T-cytotoxic 1 (Tc1) cell pool, to gain new insights into the running immune processes. METHODS CXCR6 expression was examined on peripheral blood lymphocytes (PBLs) and thyroid-derived lymphocytes (TLs) of GD patients in flow cytometry. CXCR6 cDNA was quantified in thyroid tissues affected by GD (n = 16), Hashimoto's thyroiditis (HT; n = 2) and thyroid autonomy (TA; n = 11) using real-time reverse transcriptase PCR. RESULTS The percentages of peripheral CXCR6(+) PBLs did not differ between GD and normal subjects. CXCR6 was expressed by small subsets of circulating T-cells and natural killer (NK) cells. CXCR6(+) cells were enriched in thyroid-derived T-cells compared with peripheral CD4(+) and CD8(+) T-cells in GD. The increase was evident within the Th1 (CD4(+) interferon-gamma(+) (IFN-gamma(+))) and Tc1 (CD8(+)IFN-gamma(+)) subpopulation and CD8(+) granzyme A(+) T-cells (cytotoxic effector type). Thyroid-derived fibro-blasts and thyrocytes were CXCR6(-). There was no significant difference between the CXCR6 mRNA levels in GD compared with HT and normal TA tissues. The lowest CXCR6 mRNA levels were obtained from thyroid nodules from TA patients and GD patients with low thyroid peroxidase autoantibody levels. CONCLUSIONS CXCR6 was overexpressed in Th1 and Tc1 TLs compared with PBLs in GD. CXCR6 could be a marker for lymphocytes that have migrated into the thyroid and assist in the thyroid, independently of the bias of the underlying disease.
Collapse
MESH Headings
- Adolescent
- Adult
- DNA, Complementary/analysis
- Female
- Flow Cytometry
- Graves Disease/immunology
- Humans
- Killer Cells, Natural/chemistry
- Male
- Middle Aged
- Polymerase Chain Reaction
- RNA, Messenger/analysis
- Receptors, CXCR6
- Receptors, Chemokine
- Receptors, Cytokine/analysis
- Receptors, Cytokine/genetics
- Receptors, G-Protein-Coupled/analysis
- Receptors, G-Protein-Coupled/genetics
- Receptors, Virus/analysis
- Receptors, Virus/genetics
- T-Lymphocytes/chemistry
- T-Lymphocytes, Cytotoxic/chemistry
- Th1 Cells/chemistry
- Thyroid Gland/chemistry
Collapse
Affiliation(s)
- G Aust
- Institute of Anatomy, University of Leipzig, Germany.
| | | | | | | |
Collapse
|
35
|
Nagayama Y, Saitoh O, McLachlan SM, Rapoport B, Kano H, Kumazawa Y. TSH receptor-adenovirus-induced Graves' hyperthyroidism is attenuated in both interferon-gamma and interleukin-4 knockout mice; implications for the Th1/Th2 paradigm. Clin Exp Immunol 2005; 138:417-22. [PMID: 15544617 PMCID: PMC1809247 DOI: 10.1111/j.1365-2249.2004.02641.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The role of the Th1/Th2 balance in the pathogenesis of murine Graves' hyperthyroidism is controversial. In BALB/c mice injected with adenovirus expressing TSH receptor (TSHR-adeno model), we found that suppression of TSHR-specific Th1 immune responses by exogenous interleukin-4 (IL-4), alpha-galactosylceramide or helminth (Schistosoma mansoni) infection was associated with inhibition of hyperthyroidism, indicating the critical role for Th1 cytokines. In contrast, BALB/c IL-4 knockout (KO), but not interferon-gamma (IFN-gamma) KO mice failed to develop Graves' hyperthyroidism when injected with TSHR-expressing M12 B lymphoma cells (TSHR-M12 model), suggesting the importance of Th2 cytokine IL-4. To reconcile differences in these two models, we used IL-4 KO and IFN-gamma KO BALB/c mice in the TSHR-adeno model. Unlike wild-type (wt) BALB/c mice in which 60% developed hyperthyroidism, only 13 and 7% of IL-4 KO and IFN-gamma KO mice, respectively, became hyperthyroid. Thyroid stimulating antibodies were positive in most hyperthyroid mice. TSHR antibody titres determined by TSH binding inhibition and ELISA were comparable in all three groups. IgG1 and IgG2a TSHR antibody titres were similar in IFN-gamma KO and wt mice, whereas IgG1 TSHR antibody titres and TSHR-specific splenocyte IFN-gamma secretion were lower in IL-4 KO than in IFN-gamma KO and wt mice, respectively. Our results clearly implicate both IFN-gamma and IL-4 in development of hyperthyroidism in the TSHR-adeno model. These data, together with the previous report, also indicate different cytokine requirements in these two Graves' models, with IFN-gamma being more important in the TSHR-adeno than the TSHR-M12 model. Moreover, our previous and present observations indicate a difference in the role of exogenous versus endogenous IL-4 in TSHR-adenovirus induced Graves' hyperthyroidism.
Collapse
Affiliation(s)
- Y Nagayama
- Department of Medical Gene Technology, Atomic Bomb Disease Institute, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| | | | | | | | | | | |
Collapse
|
36
|
Chen CR, Aliesky H, Pichurin PN, Nagayama Y, McLachlan SM, Rapoport B. Susceptibility rather than resistance to hyperthyroidism is dominant in a thyrotropin receptor adenovirus-induced animal model of Graves' disease as revealed by BALB/c-C57BL/6 hybrid mice. Endocrinology 2004; 145:4927-33. [PMID: 15284197 DOI: 10.1210/en.2004-0716] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We investigated why TSH receptor (TSHR) adenovirus immunization induces hyperthyroidism more commonly in BALB/c than in C57BL/6 mice. Recent modifications of the adenovirus model suggested that using adenovirus expressing the TSHR A subunit (A-subunit-Ad), rather than the full-length TSHR, and injecting fewer viral particles would increase the frequency of hyperthyroidism in C57BL/6 mice. This hypothesis was not fulfilled; 65% of BALB/c but only 5% of C57BL/6 mice developed hyperthyroidism. TSH binding inhibitory antibody titers were similar in each strain. Functional TSHR antibody measurements provided a better indication for this strain difference. Whereas thyroid-stimulating antibody activity was higher in C57BL/6 than BALB/c mice, TSH blocking antibody activity was more potent in hyperthyroid-resistant C57BL/6 mice. F(1) hybrids (BALB/c x C57BL/6) responded to A-subunit-Ad immunization with hyperthyroidism and TSHR antibody profiles similar to those of the hyperthyroid-susceptible parental BALB/c strain. In contrast, ELISA of TSHR antibodies revealed that the IgG subclass distribution in the F(1) mice resembled the disease-resistant C57BL/6 parental strain. Because the IgG subclass distribution is dependent on the T helper 1/T helper 2 cytokine balance, this paradigm can likely be excluded as an explanation for susceptibility to hyperthyroidism. In summary, our data for BALB/c, C57BL/6, and F(1) strains suggest that BALB/c mice carry a dominant gene(s) for susceptibility to induction of a thyroid-stimulating antibody/TSH blocking antibody balance that results in hyperthyroidism. Study of this genetic influence will provide useful information on potential candidate genes in human Graves' disease.
Collapse
Affiliation(s)
- Chun-Rong Chen
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite B-131, Los Angeles, California 90048, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Recent research in autoimmune thyroid disease (AITD) has largely focused on delineation of the autoantigens and their epitopes, but there is now renewed interest in the immunoregulatory properties of T cells, an understanding of which may explain the emergence of AITD in experimental settings. T cell recognition of autoantigens has shown considerable intra- and interindividual heterogeneity, and a mixed pattern of cytokine production indicates that both the Th1 and Th2 limbs of the helper T cell response are involved in all types of AITD. It is now clear that secretion of chemokines and cytokines within the thyroid accounts for the accumulation and expansion of the intrathyroidal lymphocyte pool, and that the thyroid cells themselves contribute to this secretion. The thyroid cells also produce a number of proinflammatory molecules which will tend to exacerbate the autoimmune process. Thyroid cell destruction in autoimmune hypothyroidism is dependent on T cell-mediated cytotoxicity with the likely additional effect of death receptor-mediated apoptosis.
Collapse
Affiliation(s)
- A P Weetman
- Clinical Sciences Centre, University of Sheffield, Northern General Hospital, UK.
| |
Collapse
|
38
|
Nagayama Y, Watanabe K, Niwa M, McLachlan SM, Rapoport B. Schistosoma mansoniand α-Galactosylceramide: Prophylactic Effect of Th1 Immune Suppression in a Mouse Model of Graves’ Hyperthyroidism. THE JOURNAL OF IMMUNOLOGY 2004; 173:2167-73. [PMID: 15265954 DOI: 10.4049/jimmunol.173.3.2167] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Graves' hyperthyroidism, an organ-specific autoimmune disease mediated by stimulatory thyrotropin receptor (TSHR) autoantibodies, has been considered a Th2-dominant disease. However, recent data with mouse Graves' models are conflicting. For example, we recently demonstrated that injection of BALB/c mice with adenovirus coding the TSHR induced Graves' hyperthyroidism characterized by mixed Th1 and Th2 immune responses against the TSHR, and that transient coexpression of the Th2 cytokine IL-4 by adenovirus skewed Ag-specific immune response toward Th2 and suppressed disease induction. To gain further insight into the relationship between immune polarization and Graves' disease, we evaluated the effect of Th2 immune polarization by helminth Schistosoma mansoni infection and alpha-galactosylceramide (alpha-GalCer), both known to bias the systemic immune response to Th2, on Graves' disease. S. mansoni infection first induced mixed Th1 and Th2 immune responses to soluble worm Ags, followed by a Th2 response to soluble egg Ags. Prior infection with S. mansoni suppressed the Th1-type anti-TSHR immune response, as demonstrated by impaired Ag-specific IFN-gamma secretion of splenocytes and decreased titers of IgG2a subclass anti-TSHR Abs, and also prevented disease development. Similarly, alpha-GalCer suppressed Ag-specific splenocyte secretion of IFN-gamma and prevented disease induction. However, once the anti-TSHR immune response was fully induced, S. mansoni or alpha-GalCer was ineffective in curing disease. These data support the Th1 theory in Graves' disease and indicate that suppression of the Th1-type immune response at the time of Ag priming may be crucial for inhibiting the pathogenic anti-TSHR immune response.
Collapse
Affiliation(s)
- Yuji Nagayama
- Department of Medical Gene Technology, Atomic Bomb Disease Institute, Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto, Nagasaki, Japan.
| | | | | | | | | |
Collapse
|
39
|
Barrett K, Liakata E, Rao PV, Watson PF, Weetman AP, Lymberi P, Banga JP, Carayanniotis G. Induction of hyperthyroidism in mice by intradermal immunization with DNA encoding the thyrotropin receptor. Clin Exp Immunol 2004; 136:413-22. [PMID: 15147342 PMCID: PMC1809053 DOI: 10.1111/j.1365-2249.2004.02483.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Intramuscular injection with plasmid DNA encoding the human thyrotropin receptor (TSHR) has been known to elicit symptoms of Graves' disease (GD) in outbred but not inbred mice. In this study, we have examined, firstly, whether intradermal (i.d.) injection of TSHR DNA can induce hyperthyroidism in BALB/c mice and, secondly, whether coinjection of TSHR- and cytokine-producing plasmids can influence the outcome of disease. Animals were i.d. challenged at 0, 3 and 6 weeks with TSHR DNA and the immune response was assessed at the end of the 8th or 10th week. In two experiments, a total of 10 (67%) of 15 mice developed TSHR-specific antibodies as assessed by flow cytometry. Of these, 4 (27%) mice had elevated thyroxine (TT4) levels and goitrous thyroids with activated follicular epithelial cells but no evidence of lymphocytic infiltration. At 10 weeks, thyroid-stimulating antibodies (TSAb) were detected in two out of the four hyperthyroid animals. Interestingly, in mice that received a coinjection of TSHR- and IL-2- or IL-4-producing plasmids, there was no production of TSAbs and no evidence of hyperthyroidism. On the other hand, coinjection of DNA plasmids encoding TSHR and IL-12 did not significantly enhance GD development since two out of seven animals became thyrotoxic, but had no goitre. These results demonstrate that i.d. delivery of human TSHR DNA can break tolerance and elicit GD in inbred mice. The data do not support the notion that TSAb production is Th2-dependent in murine GD but they also suggest that codelivery of TSHR and Th1-promoting IL-12 genes may not be sufficient to enhance disease incidence and/or severity in this model.
Collapse
Affiliation(s)
- K Barrett
- Division of Endocrinology, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Graves' disease (GD) is a very common autoimmune disorder of the thyroid in which stimulatory antibodies bind to the thyrotropin receptor and activate glandular function, resulting in hyperthyroidism. In addition, some patients with GD develop localized manifestations including ophthalmopathy (GO) and dermopathy. Since the cloning of the receptor cDNA, significant progress has been made in understanding the structure-function relationship of the receptor, which has been discussed in a number of earlier reviews. In this paper, we have focused our discussion on studies related to the molecular mechanisms of the disease pathogenesis and the development of animal models for GD. It has become apparent that multiple factors contribute to the etiology of GD, including host genetic as well as environmental factors. Studies in experimental animals indicate that GD is a slowly progressing disease that involves activation and recruitment of thyrotropin receptor-specific T and B cells. This activation eventually results in the production of stimulatory antibodies that can cause hyperthyroidism. Similarly, significant new insights have been gained in our understanding of GO that occurs in a subset of patients with GD. As in GD, both environmental and genetic factors play important roles in the development of GO. Although a number of putative ocular autoantigens have been identified, their role in the pathogenesis of GO awaits confirmation. Extensive analyses of orbital tissues obtained from patients with GO have provided a clearer understanding of the roles of T and B cells, cytokines and chemokines, and various ocular tissues including ocular muscles and fibroblasts. Equally impressive is the progress made in understanding why connective tissues of the orbit and the skin in GO are singled out for activation and undergo extensive remodeling. Results to date indicate that fibroblasts can act as sentinel cells and initiate lymphocyte recruitment and tissue remodeling. Moreover, these fibroblasts can be readily activated by Ig in the sera of patients with GD, suggesting a central role for them in the pathogenesis. Collectively, recent studies have led to a better understanding of the pathogenesis of GD and GO and have opened up potential new avenues for developing novel treatments for GD and GO.
Collapse
Affiliation(s)
- Bellur S Prabhakar
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612-7344, USA.
| | | | | |
Collapse
|
41
|
Pichurin P, Chen CR, Pichurina O, David C, Rapoport B, McLachlan SM. Thyrotropin receptor-DNA vaccination of transgenic mice expressing HLA-DR3 or HLA-DQ6b. Thyroid 2003; 13:911-7. [PMID: 14611699 DOI: 10.1089/105072503322511300] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Graves' disease in Caucasians is associated with the major histocompatibility (MHC) antigen HLA-DR3. One approach to studying the role of susceptibility genes involves the use of mice that lack murine MHC and instead express human HLA antigens. Although Graves' disease does not arise spontaneously in animals, thyrotropin receptor (TSHR) antibodies can be induced in mice by vaccination with TSHR-DNA in a plasmid. In the present study, we characterized TSHR antibodies and thyroiditis developing in HLA-DR3 transgenic mice vaccinated with TSHR-DNA. As controls, we used mice transgenic for HLA-DQ6b, an MHC antigen rarely associated with Graves' disease. We observed that approximately 30% of DR3-, but none of DQ6b-transgenic mice, developed TSHR antibodies detectable by enzyme-linked immunosorbent assay (ELISA). The cysteine-rich amino terminal peptide was the dominant linear antibody epitope in DR3 mice, as in other strains vaccinated with TSHR-DNA. Sera from some vaccinated DR3 mice were positive on flow cytometry using intact cells expressing the TSHR, demonstrating recognition of the native TSHR on the cell surface. Although none of the these mice had thyroid stimulating antibodies or were hyperthyroid, a few developed lymphocytic infiltration of the thyroid. These data, together with information for other mouse strains, demonstrate that MHC (human and murine) and non-MHC genes contribute to the outcome of TSHR-DNA vaccination and indicate the potential value of DR3 transgenic mice for dissecting immune responses to the TSHR.
Collapse
Affiliation(s)
- Pavel Pichurin
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and UCLA School of Medicine, Los Angeles, California 90048, USA
| | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Vasu C, Dogan RNE, Holterman MJ, Prabhakar BS. Selective induction of dendritic cells using granulocyte macrophage-colony stimulating factor, but not fms-like tyrosine kinase receptor 3-ligand, activates thyroglobulin-specific CD4+/CD25+ T cells and suppresses experimental autoimmune thyroiditis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5511-22. [PMID: 12759428 DOI: 10.4049/jimmunol.170.11.5511] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fms-like tyrosine kinase receptor 3-ligand (Flt3-L) and GM-CSF cause expansion of different subsets of dendritic cells and skew the immune response toward predominantly Th1 and Th2 type, respectively. In the present study, we investigated their effects on experimental autoimmune thyroiditis in CBA/J mice. Relative to mouse thyroglobulin (mTg) immunized controls, mTg-immunized mice treated with Flt3-L showed more severe thyroiditis characterized by enhanced lymphocytic infiltration of the thyroid, and IFN-gamma and IL-2 production. In contrast, mice treated with GM-CSF, either before or after immunization with mTg, showed suppressed T cell response to mTg and failed to develop thyroiditis. Lymphocytes from these mice, upon activation with mTg in vitro, produced higher levels of IL-4 and IL-10. Additionally, GM-CSF-treated mice showed an increase in the frequency of CD4(+)/CD25(+) T cells, which suppressed the mTg-specific T cell response. Neutralization of IL-10, but not IL-4, or depletion of CD4(+)/CD25(+) cells resulted in increased mTg-specific in vitro T cell proliferation suggesting that IL-10 produced by the Ag-specific CD4(+)/CD25(+) regulatory T cells might be critical for disease suppression. These results indicate that skewing immune response toward Th2, through selective activation of dendritic cells using GM-CSF, may have therapeutic potential in Th1 dominant autoimmune diseases including Hashimoto's thyroiditis.
Collapse
|