Skip to main content
Top
Published in: Endocrine 2/2019

01-11-2019 | Adenovirus | Review

A transgenic mouse that spontaneously develops pathogenic TSH receptor antibodies will facilitate study of antigen-specific immunotherapy for human Graves’ disease

Authors: Sandra M. McLachlan, Basil Rapoport

Published in: Endocrine | Issue 2/2019

Login to get access

Abstract

Graves’ hyperthyroidism can be treated but not cured. Antigen-specific immunotherapy would accomplish this goal, for which purpose an animal model is an invaluable tool. Two types of animal models are available. First, pathogenic TSHR antibodies (TSHRAb) can be induced by injecting mice with fibroblasts co-expressing the human TSHR (hTSHR) and MHC class II, or in mammals using plasmid or adenovirus vectors encoding the hTSHR or its A-subunit. Second, a mouse model that spontaneously develops pathogenic TSHRAb resembling those in human disease was recently described. This outcome was accomplished by transgenic intrathyroidal expression of the hTSHR A-subunit in NOD.H2h4 mice that are genetically predisposed to develop thyroiditis but, without the transgene, do not generate TSHRAb. Recently, novel approaches to antigen-specific immunotherapy have been tested, primarily in the induced model, by injecting TSHR A-subunit protein or cyclic TSHR peptides. T-cell tolerance has also been induced in “humanized” HLA-DR3 mice by injecting synthetic peptides predicted in silico to mimic naturally processed TSHR T-cell epitopes. Indeed, a phase 1 study based on the latter approach has been conducted in humans. In the spontaneous model (hTSHR/NOD.H2h mice), injection of soluble or nanoparticle-bearing hTSHR A-subunits had the unwanted effect of exacerbating pathogenic TSHRAb levels. A promising avenue for tolerance induction, successful in other conditions and yet to be tested with the TSHR, involves encapsulating the antigen. In conclusion, these studies provide insight into the potential outcome of immunotherapeutic approaches and emphasize the importance of a spontaneous model to test future novel, antigen-specific immunotherapies for Graves’ disease.
Literature
2.
3.
go back to reference O. Torring, T. Watt, G. Sjolin, K. Bystrom, M. Abraham-Nordling, J. Calissendorff, P.K. Cramon, H. Filipsson Nystrom, B. Hallengren, M. Holmberg, S. Khamisi, M. Lantz, G. Wallin, Impaired quality of life after radioiodine therapy compared to antithyroid drugs or surgical treatment for Graves’ hyperthyroidism: a long-term follow-up with the thyroid-related patient-reported outcome questionnaire and 36-item short form health status survey. Thyroid 29(3), 322–331 (2019). https://doi.org/10.1089/thy.2018.0315 CrossRefPubMed O. Torring, T. Watt, G. Sjolin, K. Bystrom, M. Abraham-Nordling, J. Calissendorff, P.K. Cramon, H. Filipsson Nystrom, B. Hallengren, M. Holmberg, S. Khamisi, M. Lantz, G. Wallin, Impaired quality of life after radioiodine therapy compared to antithyroid drugs or surgical treatment for Graves’ hyperthyroidism: a long-term follow-up with the thyroid-related patient-reported outcome questionnaire and 36-item short form health status survey. Thyroid 29(3), 322–331 (2019). https://​doi.​org/​10.​1089/​thy.​2018.​0315 CrossRefPubMed
4.
go back to reference B. Rapoport, S.M. McLachlan, The thyrotropin receptor in Graves’ disease. Thyroid 17(10), 911–922 (2007)PubMed B. Rapoport, S.M. McLachlan, The thyrotropin receptor in Graves’ disease. Thyroid 17(10), 911–922 (2007)PubMed
7.
go back to reference H. Aliesky, C.L. Courtney, B. Rapoport, S.M. McLachlan, Thyroid autoantibodies are rare in nonhuman great apes and hypothyroidism cannot be attributed to thyroid autoimmunity. Endocrinology 154(12), 4896–4907 (2013)PubMedPubMedCentral H. Aliesky, C.L. Courtney, B. Rapoport, S.M. McLachlan, Thyroid autoantibodies are rare in nonhuman great apes and hypothyroidism cannot be attributed to thyroid autoimmunity. Endocrinology 154(12), 4896–4907 (2013)PubMedPubMedCentral
8.
go back to reference M. Parmentier, F. Libert, C. Maenhaut, A. Lefort, C. Gerard, J. Perret, J. Van Sande, J.E. Dumont, G. Vassart, Molecular cloning of the thyrotropin receptor. Science 246, 1620–1622 (1989)PubMed M. Parmentier, F. Libert, C. Maenhaut, A. Lefort, C. Gerard, J. Perret, J. Van Sande, J.E. Dumont, G. Vassart, Molecular cloning of the thyrotropin receptor. Science 246, 1620–1622 (1989)PubMed
9.
go back to reference Y. Nagayama, K.D. Kaufman, P. Seto, B. Rapoport, Molecular cloning, sequence and functional expression of the cDNA for the human thyrotropin receptor. Biochem. Biophys. Res. Commun. 165, 1184–1190 (1989)PubMed Y. Nagayama, K.D. Kaufman, P. Seto, B. Rapoport, Molecular cloning, sequence and functional expression of the cDNA for the human thyrotropin receptor. Biochem. Biophys. Res. Commun. 165, 1184–1190 (1989)PubMed
10.
go back to reference F. Libert, A. Lefort, C. Gerard, M. Parmentier, J. Perret, M. Ludgate, J.E. Dumont, G. Vassart, Cloning, sequencing and expression of the human thyrotropin (TSH) receptor: Evidence for binding of autoantibodies. Biochem. Biophys. Res. Commun. 165, 1250–1255 (1989)PubMed F. Libert, A. Lefort, C. Gerard, M. Parmentier, J. Perret, M. Ludgate, J.E. Dumont, G. Vassart, Cloning, sequencing and expression of the human thyrotropin (TSH) receptor: Evidence for binding of autoantibodies. Biochem. Biophys. Res. Commun. 165, 1250–1255 (1989)PubMed
11.
go back to reference P.R. Buckland, C.R. Rickards, R.D. Howells, E. Davies Jones, B. Rees Smith, Photo-affinity labelling of the thyrotropin receptor. FEBS Lett. 145(2), 245–249 (1982) P.R. Buckland, C.R. Rickards, R.D. Howells, E. Davies Jones, B. Rees Smith, Photo-affinity labelling of the thyrotropin receptor. FEBS Lett. 145(2), 245–249 (1982)
12.
go back to reference J. Furmaniak, F.A. Hashim, P.R. Buckland, V.B. Petersen, K. Beever, R.D. Howells, B. Rees Smith, Photoaffinity labelling of the TSH receptor on FRTL5 cells. FEBS Lett. 215, 316–322 (1987)PubMed J. Furmaniak, F.A. Hashim, P.R. Buckland, V.B. Petersen, K. Beever, R.D. Howells, B. Rees Smith, Photoaffinity labelling of the TSH receptor on FRTL5 cells. FEBS Lett. 215, 316–322 (1987)PubMed
13.
go back to reference B. Rapoport, S.M. McLachlan, TSH receptor cleavage into subunits and shedding of the A-subunit; a molecular and clinical perspective. Endocr. Rev. 37, 114–134 (2016)PubMedPubMedCentral B. Rapoport, S.M. McLachlan, TSH receptor cleavage into subunits and shedding of the A-subunit; a molecular and clinical perspective. Endocr. Rev. 37, 114–134 (2016)PubMedPubMedCentral
14.
go back to reference A.V. Misharin, Y. Nagayama, H. Aliesky, Y. Mizutori, B. Rapoport, S.M. McLachlan, Attenuation of induced hyperthyroidism in mice by pretreatment with thyrotropin receptor protein: deviation of thyroid-stimulating antibody to non-functional antibodies. Endocrinology 150(8), 3944–3952 (2009)PubMedPubMedCentral A.V. Misharin, Y. Nagayama, H. Aliesky, Y. Mizutori, B. Rapoport, S.M. McLachlan, Attenuation of induced hyperthyroidism in mice by pretreatment with thyrotropin receptor protein: deviation of thyroid-stimulating antibody to non-functional antibodies. Endocrinology 150(8), 3944–3952 (2009)PubMedPubMedCentral
16.
go back to reference S.M. McLachlan, Y. Nagayama, B. Rapoport, Insight into Graves’ hyperthyroidism from animal models. Endocr. Rev. 26(6), 800–832 (2005)PubMed S.M. McLachlan, Y. Nagayama, B. Rapoport, Insight into Graves’ hyperthyroidism from animal models. Endocr. Rev. 26(6), 800–832 (2005)PubMed
17.
go back to reference T. Hanafusa, R. Pujol-Borrell, L. Chiovato, R.C.G. Russell, D. Doniach, G.F. Bottazzo, M. Feldmann, Aberrant expression of HLA-DR antigen on thyrocytes in Graves’ disease: relevance for autoimmunity. Lancet ii, 1111–1115 (1983) T. Hanafusa, R. Pujol-Borrell, L. Chiovato, R.C.G. Russell, D. Doniach, G.F. Bottazzo, M. Feldmann, Aberrant expression of HLA-DR antigen on thyrocytes in Graves’ disease: relevance for autoimmunity. Lancet ii, 1111–1115 (1983)
18.
go back to reference G.F. Bottazzo, R. Pujol-Borrell, T. Hanafusa, M. Feldmann, Role of aberrant HLA-DR expression and antigen presentation in induction of endocrine autoimmunity. Lancet 2, 1115–1119 (1983)PubMed G.F. Bottazzo, R. Pujol-Borrell, T. Hanafusa, M. Feldmann, Role of aberrant HLA-DR expression and antigen presentation in induction of endocrine autoimmunity. Lancet 2, 1115–1119 (1983)PubMed
19.
go back to reference N. Shimojo, Y. Kohno, K.-I. Yamaguchi, S.-I. Kikuoka, A. Hoshioka, H. Niimi, A. Hirai, Y. Tamura, Y. Saito, L.D. Kohn, K. Tahara, Induction of Graves-like disease in mice by immunization with fibroblasts transfected with the thyrotropin repector and a class II molecule. Proc. Natl Acad. Sci. USA 93, 11074–11079 (1996)PubMed N. Shimojo, Y. Kohno, K.-I. Yamaguchi, S.-I. Kikuoka, A. Hoshioka, H. Niimi, A. Hirai, Y. Tamura, Y. Saito, L.D. Kohn, K. Tahara, Induction of Graves-like disease in mice by immunization with fibroblasts transfected with the thyrotropin repector and a class II molecule. Proc. Natl Acad. Sci. USA 93, 11074–11079 (1996)PubMed
20.
go back to reference T. Ando, M. Imaizumi, P. Graves, P. Unger, T.F. Davies, Induction of thyroid-stimulating hormone receptor autoimmunity in hamsters. Endocrinol 144(2), 671–680 (2003) T. Ando, M. Imaizumi, P. Graves, P. Unger, T.F. Davies, Induction of thyroid-stimulating hormone receptor autoimmunity in hamsters. Endocrinol 144(2), 671–680 (2003)
21.
go back to reference S. Kaithamana, J. Fan, Y. Osuga, S.G. Liang, B.S. Prabhakar, Induction of experimental autoimmune Graves’ disease in BALB/c mice. J. Immunol. 163(9), 5157–5164 (1999)PubMed S. Kaithamana, J. Fan, Y. Osuga, S.G. Liang, B.S. Prabhakar, Induction of experimental autoimmune Graves’ disease in BALB/c mice. J. Immunol. 163(9), 5157–5164 (1999)PubMed
22.
go back to reference S. Costagliola, P. Rodien, M.-C. Many, M. Ludgate, G. Vassart, Genetic immunization against the human thyrotropin receptor causes thyroiditis and allows production of monoclonal antibodies recognizing the native receptor. J. Immunol. 160, 1458–1465 (1998)PubMed S. Costagliola, P. Rodien, M.-C. Many, M. Ludgate, G. Vassart, Genetic immunization against the human thyrotropin receptor causes thyroiditis and allows production of monoclonal antibodies recognizing the native receptor. J. Immunol. 160, 1458–1465 (1998)PubMed
23.
go back to reference S.X. Zhao, S. Tsui, A. Cheung, R.S. Douglas, T.J. Smith, J.P. Banga, Orbital fibrosis in a mouse model of Graves’ disease induced by genetic immunization of thyrotropin receptor cDNA. J. Endocrinol. 210(3), 369–377 (2011)PubMedPubMedCentral S.X. Zhao, S. Tsui, A. Cheung, R.S. Douglas, T.J. Smith, J.P. Banga, Orbital fibrosis in a mouse model of Graves’ disease induced by genetic immunization of thyrotropin receptor cDNA. J. Endocrinol. 210(3), 369–377 (2011)PubMedPubMedCentral
24.
go back to reference T. Kaneda, A. Honda, A. Hakozaki, T. Fuse, A. Muto, T. Yoshida, An improved Graves’ disease model established by using in vivo electroporation exhibited long-term immunity to hyperthyroidism in BALB/c mice. Endocrinology 148(5), 2335–2344 (2007)PubMed T. Kaneda, A. Honda, A. Hakozaki, T. Fuse, A. Muto, T. Yoshida, An improved Graves’ disease model established by using in vivo electroporation exhibited long-term immunity to hyperthyroidism in BALB/c mice. Endocrinology 148(5), 2335–2344 (2007)PubMed
25.
go back to reference Y. Nagayama, M. Kita-Furuyama, T. Ando, K. Nakao, H. Mizuguchi, T. Hayakawa, K. Eguchi, M. Niwa, A novel murine model of Graves’ hyperthyroidism with intramuscular injection of adenovirus expressing the thyrotropin receptor. J. Immunol. 168(6), 2789–2794 (2002)PubMed Y. Nagayama, M. Kita-Furuyama, T. Ando, K. Nakao, H. Mizuguchi, T. Hayakawa, K. Eguchi, M. Niwa, A novel murine model of Graves’ hyperthyroidism with intramuscular injection of adenovirus expressing the thyrotropin receptor. J. Immunol. 168(6), 2789–2794 (2002)PubMed
26.
go back to reference C.-R. Chen, P. Pichurin, Y. Nagayama, F. Latrofa, B. Rapoport, S.M. McLachlan, The thyrotropin receptor autoantigen in Graves’ disease is the culprit as well as the victim. J. Clin. Investig. 111(12), 1897–1904 (2003)PubMed C.-R. Chen, P. Pichurin, Y. Nagayama, F. Latrofa, B. Rapoport, S.M. McLachlan, The thyrotropin receptor autoantigen in Graves’ disease is the culprit as well as the victim. J. Clin. Investig. 111(12), 1897–1904 (2003)PubMed
27.
go back to reference S.M. McLachlan, B. Rapoport, Breaking tolerance to thyroid antigens: changing concepts in thyroid autoimmunity. Endocr. Rev. 35(1), 59–105 (2014)PubMed S.M. McLachlan, B. Rapoport, Breaking tolerance to thyroid antigens: changing concepts in thyroid autoimmunity. Endocr. Rev. 35(1), 59–105 (2014)PubMed
28.
go back to reference Y. Wang, L.P. Wu, J. Fu, H.J. Lv, X.Y. Guan, L. Xu, P. Chen, C.Q. Gao, P. Hou, M.J. Ji, B.Y. Shi, Hyperthyroid monkeys: a nonhuman primate model of experimental Graves’ disease. J. Endocrinol. 219(3), 183–193 (2013)PubMed Y. Wang, L.P. Wu, J. Fu, H.J. Lv, X.Y. Guan, L. Xu, P. Chen, C.Q. Gao, P. Hou, M.J. Ji, B.Y. Shi, Hyperthyroid monkeys: a nonhuman primate model of experimental Graves’ disease. J. Endocrinol. 219(3), 183–193 (2013)PubMed
29.
go back to reference G.D. Chazenbalk, P. Pichurin, C.R. Chen, F. Latrofa, A.P. Johnstone, S.M. McLachlan, B. Rapoport, Thyroid-stimulating autoantibodies in Graves disease preferentially recognize the free A subunit, not the thyrotropin holoreceptor. J. Clin. Invest. 110(2), 209–217 (2002)PubMedPubMedCentral G.D. Chazenbalk, P. Pichurin, C.R. Chen, F. Latrofa, A.P. Johnstone, S.M. McLachlan, B. Rapoport, Thyroid-stimulating autoantibodies in Graves disease preferentially recognize the free A subunit, not the thyrotropin holoreceptor. J. Clin. Invest. 110(2), 209–217 (2002)PubMedPubMedCentral
30.
go back to reference Y. Mizutori, C.R. Chen, F. Latrofa, S.M. McLachlan, B. Rapoport, Evidence that shed TSH receptor A-subunits drive affinity maturation of autoantibodies causing Graves’ disease. J. Clin. Endocrinol. Metab. 94(3), 927–935 (2009)PubMed Y. Mizutori, C.R. Chen, F. Latrofa, S.M. McLachlan, B. Rapoport, Evidence that shed TSH receptor A-subunits drive affinity maturation of autoantibodies causing Graves’ disease. J. Clin. Endocrinol. Metab. 94(3), 927–935 (2009)PubMed
31.
go back to reference S.M. McLachlan, K. Alpi, B. Rapoport, Hypothesis and review: does Graves’ disease develop in non-human great apes? Thyroid 21(12), 1359–1366 (2011)PubMedPubMedCentral S.M. McLachlan, K. Alpi, B. Rapoport, Hypothesis and review: does Graves’ disease develop in non-human great apes? Thyroid 21(12), 1359–1366 (2011)PubMedPubMedCentral
33.
go back to reference B. Rapoport, H.A. Aliesky, B. Banuelos, C.R. Chen, S.M. McLachlan, A unique mouse strain that develops spontaneous, iodine-accelerated, pathogenic antibodies to the human thyrotrophin receptor. J. Immunol. 194(9), 4154–4161 (2015)PubMedPubMedCentral B. Rapoport, H.A. Aliesky, B. Banuelos, C.R. Chen, S.M. McLachlan, A unique mouse strain that develops spontaneous, iodine-accelerated, pathogenic antibodies to the human thyrotrophin receptor. J. Immunol. 194(9), 4154–4161 (2015)PubMedPubMedCentral
34.
go back to reference S.M. McLachlan, Y. Nagayama, P.N. Pichurin, Y. Mizutori, C.R. Chen, A. Misharin, H.A. Aliesky, B. Rapoport, The link between Graves’ disease and Hashimoto’s thyroiditis: a role for regulatory T cells. Endocrinology 148(12), 5724–5733 (2007)PubMed S.M. McLachlan, Y. Nagayama, P.N. Pichurin, Y. Mizutori, C.R. Chen, A. Misharin, H.A. Aliesky, B. Rapoport, The link between Graves’ disease and Hashimoto’s thyroiditis: a role for regulatory T cells. Endocrinology 148(12), 5724–5733 (2007)PubMed
35.
go back to reference L. Rasooly, C.L. Burek, N.R. Rose, Iodine-induced autoimmune thyroiditis in NOD-H2h4 mice. Clin. Immunol. Immunopathol. 81, 287–292 (1996)PubMed L. Rasooly, C.L. Burek, N.R. Rose, Iodine-induced autoimmune thyroiditis in NOD-H2h4 mice. Clin. Immunol. Immunopathol. 81, 287–292 (1996)PubMed
36.
go back to reference H. Braley-Mullen, G.C. Sharp, B. Medling, H. Tang, Spontaneous autoimmune thyroiditis in NOD.H-2h4 mice. J. Autoimmun. 12(3), 157–165 (1999)PubMed H. Braley-Mullen, G.C. Sharp, B. Medling, H. Tang, Spontaneous autoimmune thyroiditis in NOD.H-2h4 mice. J. Autoimmun. 12(3), 157–165 (1999)PubMed
37.
go back to reference P.R. Hutchings, S. Verma, J.M. Phillips, S.Z. Harach, S. Howlett, A. Cooke, Both CD4(+) T cells and CD8(+) T cells are required for iodine accelerated thyroiditis in NOD mice. Cell Immunol. 192(2), 113–121 (1999)PubMed P.R. Hutchings, S. Verma, J.M. Phillips, S.Z. Harach, S. Howlett, A. Cooke, Both CD4(+) T cells and CD8(+) T cells are required for iodine accelerated thyroiditis in NOD mice. Cell Immunol. 192(2), 113–121 (1999)PubMed
38.
go back to reference C.R. Chen, S. Hamidi, H. Braley-Mullen, Y. Nagayama, C. Bresee, H.A. Aliesky, B. Rapoport, S.M. McLachlan, Antibodies to thyroid peroxidase arise spontaneously with age in NOD.H-2h4 mice and appear after thyroglobulin antibodies. Endocrinology 151(9), 4583–4593 (2010)PubMedPubMedCentral C.R. Chen, S. Hamidi, H. Braley-Mullen, Y. Nagayama, C. Bresee, H.A. Aliesky, B. Rapoport, S.M. McLachlan, Antibodies to thyroid peroxidase arise spontaneously with age in NOD.H-2h4 mice and appear after thyroglobulin antibodies. Endocrinology 151(9), 4583–4593 (2010)PubMedPubMedCentral
39.
go back to reference G.D. Chazenbalk, J.C. Jaume, S.M. McLachlan, B. Rapoport, Engineering the human thyrotropin receptor ectodomain from a non-secreted form to a secreted, highly immunoreactive glycoprotein that neutralizes autoantibodies in Graves’ patients’ sera. J. Biol. Chem. 272, 18959–18965 (1997)PubMed G.D. Chazenbalk, J.C. Jaume, S.M. McLachlan, B. Rapoport, Engineering the human thyrotropin receptor ectodomain from a non-secreted form to a secreted, highly immunoreactive glycoprotein that neutralizes autoantibodies in Graves’ patients’ sera. J. Biol. Chem. 272, 18959–18965 (1997)PubMed
41.
go back to reference C.R. Chen, H. Aliesky, P.N. Pichurin, Y. Nagayama, S.M. McLachlan, B. Rapoport, 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 145, 4927–4933 (2004)PubMed C.R. Chen, H. Aliesky, P.N. Pichurin, Y. Nagayama, S.M. McLachlan, B. Rapoport, 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 145, 4927–4933 (2004)PubMed
42.
go back to reference B. Rapoport, R.W. Williams, C.R. Chen, S.M. McLachlan, Immunoglobulin heavy chain variable region genes contribute to the induction of thyroid stimulating antibodies in recombinant inbred mice. Genes Immun. 11(3), 254–263 (2010)PubMedPubMedCentral B. Rapoport, R.W. Williams, C.R. Chen, S.M. McLachlan, Immunoglobulin heavy chain variable region genes contribute to the induction of thyroid stimulating antibodies in recombinant inbred mice. Genes Immun. 11(3), 254–263 (2010)PubMedPubMedCentral
43.
go back to reference S.M. McLachlan, H. Braley-Mullen, C.R. Chen, H. Aliesky, P.N. Pichurin, B. Rapoport, Dissociation between iodide-induced thyroiditis and antibody-mediated hyperthyroidism in NOD.H-2h4 mice. Endocrinology 146, 294–300 (2005)PubMed S.M. McLachlan, H. Braley-Mullen, C.R. Chen, H. Aliesky, P.N. Pichurin, B. Rapoport, Dissociation between iodide-induced thyroiditis and antibody-mediated hyperthyroidism in NOD.H-2h4 mice. Endocrinology 146, 294–300 (2005)PubMed
44.
go back to reference M. Dedecjus, M. Stasiolek, J. Brzezinski, K. Selmaj, A. Lewinski, Thyroid hormones influence human dendritic cells’ phenotype, function, and subsets distribution. Thyroid 21(5), 533–540 (2011)PubMed M. Dedecjus, M. Stasiolek, J. Brzezinski, K. Selmaj, A. Lewinski, Thyroid hormones influence human dendritic cells’ phenotype, function, and subsets distribution. Thyroid 21(5), 533–540 (2011)PubMed
45.
go back to reference C. Mao, S. Wang, Y. Xiao, J. Xu, Q. Jiang, M. Jin, X. Jiang, H. Guo, G. Ning, Y. Zhang, Impairment of regulatory capacity of CD4+CD25+ regulatory T cells mediated by dendritic cell polarization and hyperthyroidism in Graves’ disease. J. Immunol. 186(8), 4734–4743 (2011)PubMed C. Mao, S. Wang, Y. Xiao, J. Xu, Q. Jiang, M. Jin, X. Jiang, H. Guo, G. Ning, Y. Zhang, Impairment of regulatory capacity of CD4+CD25+ regulatory T cells mediated by dendritic cell polarization and hyperthyroidism in Graves’ disease. J. Immunol. 186(8), 4734–4743 (2011)PubMed
47.
go back to reference H.J. Lee, C.W. Li, S.S. Hammerstad, M. Stefan, Y. Tomer, Immunogenetics of autoimmune thyroid diseases: a comprehensive review. J. Autoimmun. 64, 82–90 (2015)PubMedPubMedCentral H.J. Lee, C.W. Li, S.S. Hammerstad, M. Stefan, Y. Tomer, Immunogenetics of autoimmune thyroid diseases: a comprehensive review. J. Autoimmun. 64, 82–90 (2015)PubMedPubMedCentral
48.
go back to reference S.M. McLachlan, H. Aliesky, B. Banuelos, J. Magana, R.W. Williams, B. Rapoport, Immunoglobulin heavy chain variable region and major histocompatibility region genes are linked to induced graves’ disease in females from two very large families of recombinant inbred mice. Endocrinology 155(10), 4094–4103 (2014)PubMedPubMedCentral S.M. McLachlan, H. Aliesky, B. Banuelos, J. Magana, R.W. Williams, B. Rapoport, Immunoglobulin heavy chain variable region and major histocompatibility region genes are linked to induced graves’ disease in females from two very large families of recombinant inbred mice. Endocrinology 155(10), 4094–4103 (2014)PubMedPubMedCentral
49.
go back to reference F. Latrofa, G.D. Chazenbalk, P. Pichurin, C.-R. Chen, S.M. McLachlan, B. Rapoport, Characterization of TSH receptor autoantibodies affinity-enriched to near purity from the serum of Graves’ patients. Thyroid 13(7), 734–734 (2003) F. Latrofa, G.D. Chazenbalk, P. Pichurin, C.-R. Chen, S.M. McLachlan, B. Rapoport, Characterization of TSH receptor autoantibodies affinity-enriched to near purity from the serum of Graves’ patients. Thyroid 13(7), 734–734 (2003)
50.
go back to reference A.P. Weetman, M.E. Yateman, P.A. Ealey, C.M. Black, C.B. Reimer, R.C. Williams Jr. B. Shine, N.J. Marshall, Thyroid-stimulating antibody activity between different immunoglobulin G subclasses. J. Clin. Invest. 86, 723–727 (1990)PubMedPubMedCentral A.P. Weetman, M.E. Yateman, P.A. Ealey, C.M. Black, C.B. Reimer, R.C. Williams Jr. B. Shine, N.J. Marshall, Thyroid-stimulating antibody activity between different immunoglobulin G subclasses. J. Clin. Invest. 86, 723–727 (1990)PubMedPubMedCentral
52.
go back to reference Y. Nagayama, O. Saitoh, S.M. McLachlan, B. Rapoport, H. Kano, Y. Kumazawa, TSHR receptor-adenovirus-induced Graves’ hyperthyroidism is attenuated in both interferon-g and interleukin-4 knockout mice: Implications for the Th1/Th2 paradigm. Clin. Exp. Immunol. 138, 417–422 (2004)PubMedPubMedCentral Y. Nagayama, O. Saitoh, S.M. McLachlan, B. Rapoport, H. Kano, Y. Kumazawa, TSHR receptor-adenovirus-induced Graves’ hyperthyroidism is attenuated in both interferon-g and interleukin-4 knockout mice: Implications for the Th1/Th2 paradigm. Clin. Exp. Immunol. 138, 417–422 (2004)PubMedPubMedCentral
53.
go back to reference O. Saitoh, Y. Mizutori, N. Takamura, H. Yamasaki, A. Kita, H. Kuwahara, Y. Nagayama, Adenovirus-mediated gene delivery of interleukin-10, but not transforming growth factor beta, ameliorates the induction of Graves’ hyperthyroidism in BALB/c mice. Clin. Exp. Immunol. 141(3), 405–411 (2005)PubMedPubMedCentral O. Saitoh, Y. Mizutori, N. Takamura, H. Yamasaki, A. Kita, H. Kuwahara, Y. Nagayama, Adenovirus-mediated gene delivery of interleukin-10, but not transforming growth factor beta, ameliorates the induction of Graves’ hyperthyroidism in BALB/c mice. Clin. Exp. Immunol. 141(3), 405–411 (2005)PubMedPubMedCentral
54.
go back to reference J.W. Kappler, N. Roehm, P. Marrack, T cell tolerance by clonal elimination in the thymus. Cell 49(2), 273–280 (1987)PubMed J.W. Kappler, N. Roehm, P. Marrack, T cell tolerance by clonal elimination in the thymus. Cell 49(2), 273–280 (1987)PubMed
55.
go back to reference M. Stefan, C. Wei, A. Lombardi, C.W. Li, E.S. Concepcion, W.B. Inabnet III, R. Owen, W. Zhang, Y. Tomer, Genetic-epigenetic dysregulation of thymic TSH receptor gene expression triggers thyroid autoimmunity. Proc. Natl Acad. Sci. USA 111(34), 12562–12567 (2014)PubMed M. Stefan, C. Wei, A. Lombardi, C.W. Li, E.S. Concepcion, W.B. Inabnet III, R. Owen, W. Zhang, Y. Tomer, Genetic-epigenetic dysregulation of thymic TSH receptor gene expression triggers thyroid autoimmunity. Proc. Natl Acad. Sci. USA 111(34), 12562–12567 (2014)PubMed
56.
go back to reference R. Colobran, M.P. Armengol, R. Faner, M. Gartner, L.O. Tykocinski, A. Lucas, M. Ruiz, M. Juan, B. Kyewski, R. Pujol-Borrell, Association of an SNP with intrathymic transcription of TSHR and Graves’ disease: a role for defective thymic tolerance. Hum. Mol. Genet. 20(17), 3415–3423 (2011)PubMed R. Colobran, M.P. Armengol, R. Faner, M. Gartner, L.O. Tykocinski, A. Lucas, M. Ruiz, M. Juan, B. Kyewski, R. Pujol-Borrell, Association of an SNP with intrathymic transcription of TSHR and Graves’ disease: a role for defective thymic tolerance. Hum. Mol. Genet. 20(17), 3415–3423 (2011)PubMed
57.
go back to reference M. Nakahara, N. Mitsutake, H. Sakamoto, C.R. Chen, B. Rapoport, S.M. McLachlan, Y. Nagayama, Enhanced response to mouse thyroid-stimulating hormone (TSH) receptor immunization in TSH receptor-knockout mice. Endocrinology 151(8), 4047–4054 (2010)PubMed M. Nakahara, N. Mitsutake, H. Sakamoto, C.R. Chen, B. Rapoport, S.M. McLachlan, Y. Nagayama, Enhanced response to mouse thyroid-stimulating hormone (TSH) receptor immunization in TSH receptor-knockout mice. Endocrinology 151(8), 4047–4054 (2010)PubMed
58.
go back to reference A. Schluter, M. Horstmann, S. Diaz-Cano, S. Plohn, K. Stahr, S. Mattheis, M. Oeverhaus, S. Lang, U. Flogel, U. Berchner-Pfannschmidt, A. Eckstein, J.P. Banga, Genetic immunization with mouse thyrotrophin hormone receptor plasmid breaks self-tolerance for a murine model of autoimmune thyroid disease and Graves’ orbitopathy. Clin. Exp. Immunol. 191(3), 255–267 (2018). https://doi.org/10.1111/cei.13075 CrossRefPubMed A. Schluter, M. Horstmann, S. Diaz-Cano, S. Plohn, K. Stahr, S. Mattheis, M. Oeverhaus, S. Lang, U. Flogel, U. Berchner-Pfannschmidt, A. Eckstein, J.P. Banga, Genetic immunization with mouse thyrotrophin hormone receptor plasmid breaks self-tolerance for a murine model of autoimmune thyroid disease and Graves’ orbitopathy. Clin. Exp. Immunol. 191(3), 255–267 (2018). https://​doi.​org/​10.​1111/​cei.​13075 CrossRefPubMed
59.
go back to reference A.V. Misharin, Y. Nagayama, H.A. Aliesky, B. Rapoport, S.M. McLachlan, 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 150(6), 2948–2956 (2009)PubMedPubMedCentral A.V. Misharin, Y. Nagayama, H.A. Aliesky, B. Rapoport, S.M. McLachlan, 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 150(6), 2948–2956 (2009)PubMedPubMedCentral
60.
go back to reference S.M. McLachlan, H.A. Aliesky, B. Banuelos, S. Lesage, R. Collin, B. Rapoport, High-level intrathymic thyrotrophin receptor expression in thyroiditis-prone mice protects against the spontaneous generation of pathogenic thyrotrophin receptor autoantibodies. Clin. Exp. Immunol. 188(2), 243–253 (2017)PubMedPubMedCentral S.M. McLachlan, H.A. Aliesky, B. Banuelos, S. Lesage, R. Collin, B. Rapoport, High-level intrathymic thyrotrophin receptor expression in thyroiditis-prone mice protects against the spontaneous generation of pathogenic thyrotrophin receptor autoantibodies. Clin. Exp. Immunol. 188(2), 243–253 (2017)PubMedPubMedCentral
61.
go back to reference C.-R. Chen, P. Pichurin, G.D. Chazenbalk, H. Aliesky, Y. Nagayama, S.M. McLachlan, B. Rapoport, Low-dose immunization with adenovirus expressing the thyroid-stimulating hormone receptor A-subunit deviates the antibody response toward that of autoantibodies in human Graves’ disease. Endocrinology 145(1), 228–233 (2004)PubMed C.-R. Chen, P. Pichurin, G.D. Chazenbalk, H. Aliesky, Y. Nagayama, S.M. McLachlan, B. Rapoport, Low-dose immunization with adenovirus expressing the thyroid-stimulating hormone receptor A-subunit deviates the antibody response toward that of autoantibodies in human Graves’ disease. Endocrinology 145(1), 228–233 (2004)PubMed
63.
go back to reference S. Moshkelgosha, G. Masetti, U. Berchner-Pfannschmidt, H.L. Verhasselt, M. Horstmann, S. Diaz-Cano, A. Noble, B. Edelman, D. Covelli, S. Plummer, J.R. Marchesi, M. Ludgate, F. Biscarini, A. Eckstein, J.P. Banga, Gut microbiome in BALB/c and C57BL/6J mice undergoing experimental thyroid autoimmunity associate with differences in immunological responses and thyroid function. Horm. Metab. Res. 50(12), 932–941 (2018). https://doi.org/10.1055/a-0653-3766 CrossRefPubMed S. Moshkelgosha, G. Masetti, U. Berchner-Pfannschmidt, H.L. Verhasselt, M. Horstmann, S. Diaz-Cano, A. Noble, B. Edelman, D. Covelli, S. Plummer, J.R. Marchesi, M. Ludgate, F. Biscarini, A. Eckstein, J.P. Banga, Gut microbiome in BALB/c and C57BL/6J mice undergoing experimental thyroid autoimmunity associate with differences in immunological responses and thyroid function. Horm. Metab. Res. 50(12), 932–941 (2018). https://​doi.​org/​10.​1055/​a-0653-3766 CrossRefPubMed
64.
65.
go back to reference N. Manji, J.D. Carr-Smith, K. Boelaert, A. Allahabadia, M. Armitage, V.K. Chatterjee, J.H. Lazarus, S.H. Pearce, B. Vaidya, S.C. Gough, J.A. Franklyn, Influences of age, gender, smoking, and family history on autoimmune thyroid disease phenotype. J. Clin. Endocrinol. Metab. 91(12), 4873–4880 (2006). https://doi.org/10.1210/jc.2006-1402 CrossRefPubMed N. Manji, J.D. Carr-Smith, K. Boelaert, A. Allahabadia, M. Armitage, V.K. Chatterjee, J.H. Lazarus, S.H. Pearce, B. Vaidya, S.C. Gough, J.A. Franklyn, Influences of age, gender, smoking, and family history on autoimmune thyroid disease phenotype. J. Clin. Endocrinol. Metab. 91(12), 4873–4880 (2006). https://​doi.​org/​10.​1210/​jc.​2006-1402 CrossRefPubMed
66.
go back to reference J.C. Jaume, B. Rapoport, S.M. McLachlan, Lack of female bias in a mouse model of autoimmune hyperthyroidism (Graves’ disease). Autoimmunity 29(4), 269–272 (1999)PubMed J.C. Jaume, B. Rapoport, S.M. McLachlan, Lack of female bias in a mouse model of autoimmune hyperthyroidism (Graves’ disease). Autoimmunity 29(4), 269–272 (1999)PubMed
67.
go back to reference B. Rapoport, H.A. Aliesky, C.R. Chen, S.M. McLachlan, Evidence that TSH receptor A-subunit multimers, not monomers, drive antibody affinity maturation in Graves’ disease. J. Clin. Endocrinol. Metab. 100(6), E871–E875 (2015)PubMedPubMedCentral B. Rapoport, H.A. Aliesky, C.R. Chen, S.M. McLachlan, Evidence that TSH receptor A-subunit multimers, not monomers, drive antibody affinity maturation in Graves’ disease. J. Clin. Endocrinol. Metab. 100(6), E871–E875 (2015)PubMedPubMedCentral
68.
go back to reference C.R. Chen, P.A. Hubbard, L.M. Salazar, S.M. McLachlan, R. Murali, B. Rapoport, Crystal structure of a TSH receptor monoclonal antibody: insight into Graves’ disease pathogenesis. Mol. Endocrinol. 29(1), 99–107 (2015)PubMed C.R. Chen, P.A. Hubbard, L.M. Salazar, S.M. McLachlan, R. Murali, B. Rapoport, Crystal structure of a TSH receptor monoclonal antibody: insight into Graves’ disease pathogenesis. Mol. Endocrinol. 29(1), 99–107 (2015)PubMed
69.
go back to reference J. Sanders, D.Y. Chirgadze, P. Sanders, S. Baker, A. Sullivan, A. Bhardwaja, J. Bolton, M. Reeve, N. Nakatake, M. Evans, T. Richards, M. Powell, R.N. Miguel, T.L. Blundell, J. Furmaniak, B.R. Smith, Crystal structure of the TSH receptor in complex with a thyroid-stimulating autoantibody. Thyroid 17(5), 395–410 (2007)PubMed J. Sanders, D.Y. Chirgadze, P. Sanders, S. Baker, A. Sullivan, A. Bhardwaja, J. Bolton, M. Reeve, N. Nakatake, M. Evans, T. Richards, M. Powell, R.N. Miguel, T.L. Blundell, J. Furmaniak, B.R. Smith, Crystal structure of the TSH receptor in complex with a thyroid-stimulating autoantibody. Thyroid 17(5), 395–410 (2007)PubMed
70.
go back to reference J.A. Gilbert, S.L. Kalled, J. Moorhead, D.M. Hess, P. Rennert, Z. Li, M.Z. Khan, J.P. Banga, 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 147(10), 4561–4568 (2006)PubMed J.A. Gilbert, S.L. Kalled, J. Moorhead, D.M. Hess, P. Rennert, Z. Li, M.Z. Khan, J.P. Banga, 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 147(10), 4561–4568 (2006)PubMed
72.
go back to reference L. Wu, L. Xun, J. Yang, L. Xu, Z. Tian, S. Gao, Y. Zhang, P. Hou, B. Shi, Induction of murine neonatal tolerance against Graves’ disease using recombinant adenovirus expressing the TSH receptor A-subunit. Endocrinology 152(3), 1165–1171 (2011)PubMed L. Wu, L. Xun, J. Yang, L. Xu, Z. Tian, S. Gao, Y. Zhang, P. Hou, B. Shi, Induction of murine neonatal tolerance against Graves’ disease using recombinant adenovirus expressing the TSH receptor A-subunit. Endocrinology 152(3), 1165–1171 (2011)PubMed
76.
go back to reference B. Rapoport, B. Banuelos, H.A. Aliesky, N. Hartwig Trier, S.M. McLachlan, Critical differences between induced and spontaneous mouse models of Graves’ disease with implications for antigen-specific immunotherapy in humans. J. Immunol. 197, 4560–4568 (2016)PubMedPubMedCentral B. Rapoport, B. Banuelos, H.A. Aliesky, N. Hartwig Trier, S.M. McLachlan, Critical differences between induced and spontaneous mouse models of Graves’ disease with implications for antigen-specific immunotherapy in humans. J. Immunol. 197, 4560–4568 (2016)PubMedPubMedCentral
77.
go back to reference A.M. McGregor, M.M. Petersen, R. Capiferri, D.C. Evered, B.R. Smith, R. Hall, Effects of radioiodine on thyrotrophin binding inhibiting immunoglobulins in Graves’ disease. Clin. Endocrinol. 11(4), 437–444 (1979) A.M. McGregor, M.M. Petersen, R. Capiferri, D.C. Evered, B.R. Smith, R. Hall, Effects of radioiodine on thyrotrophin binding inhibiting immunoglobulins in Graves’ disease. Clin. Endocrinol. 11(4), 437–444 (1979)
79.
go back to reference A. Yeste, M.C. Takenaka, I.D. Mascanfroni, M. Nadeau, J.E. Kenison, B. Patel, A.M. Tukpah, J.A. Babon, M. DeNicola, S.C. Kent, D. Pozo, F.J. Quintana, Tolerogenic nanoparticles inhibit T cell-mediated autoimmunity through SOCS2. Sci. Signal. 9(433), ra61 (2016)PubMed A. Yeste, M.C. Takenaka, I.D. Mascanfroni, M. Nadeau, J.E. Kenison, B. Patel, A.M. Tukpah, J.A. Babon, M. DeNicola, S.C. Kent, D. Pozo, F.J. Quintana, Tolerogenic nanoparticles inhibit T cell-mediated autoimmunity through SOCS2. Sci. Signal. 9(433), ra61 (2016)PubMed
82.
go back to reference S.H.S. Pearce, C. Dayan, D.C. Wraith, K. Barrell, N. Olive, L. Jansson, T. Walker-Smith, C. Carnegie, K.F. Martin, K. Boelaert, J. Gilbert, C.E. Higham, I. Muller, R.D. Murray, P. Perros, S. Razvi, B. Vaidya, F. Wernig, G.J. Kahaly, Antigen-specific immunotherapy with thyrotropin receptor peptides in Graves’ hyperthyroidism: a phase I study. Thyroid (2019). https://doi.org/10.1089/thy.2019.0036 PubMedPubMedCentral S.H.S. Pearce, C. Dayan, D.C. Wraith, K. Barrell, N. Olive, L. Jansson, T. Walker-Smith, C. Carnegie, K.F. Martin, K. Boelaert, J. Gilbert, C.E. Higham, I. Muller, R.D. Murray, P. Perros, S. Razvi, B. Vaidya, F. Wernig, G.J. Kahaly, Antigen-specific immunotherapy with thyrotropin receptor peptides in Graves’ hyperthyroidism: a phase I study. Thyroid (2019). https://​doi.​org/​10.​1089/​thy.​2019.​0036 PubMedPubMedCentral
83.
go back to reference A.J. Coles, M. Wing, S. Smith, F. Coraddu, S. Greer, C. Taylor, A. Weetman, G. Hale, V.K. Chatterjee, H. Waldmann, A. Compston, Pulsed monoclonal antibody treatment and autoimmune thyroid disease in multiple sclerosis. Lancet 354(9191), 1691–1695 (1999)PubMed A.J. Coles, M. Wing, S. Smith, F. Coraddu, S. Greer, C. Taylor, A. Weetman, G. Hale, V.K. Chatterjee, H. Waldmann, A. Compston, Pulsed monoclonal antibody treatment and autoimmune thyroid disease in multiple sclerosis. Lancet 354(9191), 1691–1695 (1999)PubMed
84.
go back to reference C.B. Smarr, W.T. Yap, T.P. Neef, R.M. Pearson, Z.N. Hunter, I. Ifergan, D.R. Getts, P.J. Bryce, L.D. Shea, S.D. Miller, Biodegradable antigen-associated PLG nanoparticles tolerize Th2-mediated allergic airway inflammation pre- and postsensitization. Proc. Natl Acad. Sci. USA 113(18), 5059–5064 (2016). https://doi.org/10.1073/pnas.1505782113 CrossRefPubMed C.B. Smarr, W.T. Yap, T.P. Neef, R.M. Pearson, Z.N. Hunter, I. Ifergan, D.R. Getts, P.J. Bryce, L.D. Shea, S.D. Miller, Biodegradable antigen-associated PLG nanoparticles tolerize Th2-mediated allergic airway inflammation pre- and postsensitization. Proc. Natl Acad. Sci. USA 113(18), 5059–5064 (2016). https://​doi.​org/​10.​1073/​pnas.​1505782113 CrossRefPubMed
Metadata
Title
A transgenic mouse that spontaneously develops pathogenic TSH receptor antibodies will facilitate study of antigen-specific immunotherapy for human Graves’ disease
Authors
Sandra M. McLachlan
Basil Rapoport
Publication date
01-11-2019
Publisher
Springer US
Keyword
Adenovirus
Published in
Endocrine / Issue 2/2019
Print ISSN: 1355-008X
Electronic ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-019-02083-9

Other articles of this Issue 2/2019

Endocrine 2/2019 Go to the issue