Horm Metab Res 2015; 47(10): 727-734
DOI: 10.1055/s-0035-1559633
Review

Graves’ Disease Mechanisms: The Role of Stimulating, Blocking, and Cleavage Region TSH Receptor Antibodies

S. A. Morshed
1   Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, James J. Peters VA Medical Center, New York, USA
,
T. F. Davies
1   Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, James J. Peters VA Medical Center, New York, USA
› Author Affiliations

Abstract

The immunologic processes involved in Graves’ disease (GD) have one unique characteristic – the autoantibodies to the TSH receptor (TSHR) – which have both linear and conformational epitopes. Three types of TSHR antibodies (stimulating, blocking, and cleavage) with different functional capabilities have been described in GD patients, which induce different signaling effects varying from thyroid cell proliferation to thyroid cell death. The establishment of animal models of GD by TSHR antibody transfer or by immunization with TSHR antigen has confirmed its pathogenic role and, therefore, GD is the result of a breakdown in TSHR tolerance. Here we review some of the characteristics of TSHR antibodies with a special emphasis on new developments in our understanding of what were previously called “neutral” antibodies and which we now characterize as autoantibodies to the “cleavage” region of the TSHR ectodomain.



Publication History

Received: 13 April 2015

Accepted: 10 July 2015

Article published online:
11 September 2015

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Bahn RS, Dutton CM, Natt N, Joba W, Spitzweg C, Heufelder AE. Thyrotropin receptor expression in Graves’ orbital adipose/connective tissues: potential autoantigen in Graves’ ophthalmopathy. J Clin Endocrinol Metab 1998; 83: 998-1002
  • 2 Chazenbalk GD, Pichurin P, Chen CR, Latrofa F, Johnstone AP, McLachlan SM, Rapoport B. Thyroid-stimulating autoantibodies in Graves disease preferentially recognize the free A subunit, not the thyrotropin holoreceptor. J Clin Invest 2002; 110: 209-217
  • 3 Kajita Y, Rickards CR, Buckland PR, Howells RD, Rees SB. Analysis of thyrotropin receptors by photoaffinity labelling. Orientation of receptor subunits in the cell membrane. Biochem J 1985; 227: 413-420
  • 4 Loosfelt H, Pichon C, Jolivet A, Misrahi M, Caillou B, Jamous M, Vannier B, Milgrom E. Two-subunit structure of the human thyrotropin receptor. Proc Natl Acad Sci USA 1992; 89: 3765-3769
  • 5 Vlase H, Davies TF. Insights into the molecular mechanisms of the autoimmune thyroid diseases.In: Endocrine and Organ Specific Autoimmunity. Eisenbarth GS (ed.). CA: R. G. Landes Co; 1999: 98–132
  • 6 Bagriacik EU, Klein JR. The thyrotropin (thyroid-stimulating hormone) receptor is expressed on murine dendritic cells and on a subset of CD45RBhigh lymph node T cells: functional role for thyroid-stimulating hormone during immune activation. J Immunol 2000; 164: 6158-6165
  • 7 Meffre E, Wardemann H. B-cell tolerance checkpoints in health and autoimmunity. Curr Opin Immunol 2008; 20: 632-638
  • 8 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: 4561-4568
  • 9 Lai Kwan LQ, King Hung KO, Zheng BJ, Lu L. Local BAFF gene silencing suppresses Th17-cell generation and ameliorates autoimmune arthritis. Proc Natl Acad Sci USA 2008; 105: 14993-14998
  • 10 Salvi M, Vannucchi G, Campi I, Rossi S, Bonara P, Sbrozzi F, Guastella C, Avignone S, Pirola G, Ratiglia R, Beck-Peccoz P. Efficacy of rituximab treatment for thyroid-associated ophthalmopathy as a result of intraorbital B-cell depletion in one patient unresponsive to steroid immunosuppression. Eur J Endocrinol 2006; 154: 511-517
  • 11 Adams DD, Purves HD. Abnormal responses in the assay of thyrotropin. Proc Univ Otago Med School 1956; 34: 11-12
  • 12 Ando T, Davies TF. Monoclonal antibodies to the thyrotropin receptor. Clin Dev Immunol 2005; 12: 137-143
  • 13 Ando T, Latif R, Pritsker A, Moran T, Nagayama Y, Davies TF. A monoclonal thyroid-stimulating antibody. J Clin Invest 2002; 110: 1667-1674
  • 14 Ando T, Latif R, Davies TF. Antibody-induced modulation of TSH receptor post-translational processing. J Endocrinol 2007; 195: 179-186
  • 15 Ando T, Latif R, Daniel S, Eguchi K, Davies TF. Dissecting linear and conformational epitopes on the native thyrotropin receptor. Endocrinology 2004; 145: 5185-5193
  • 16 Mizutori Y, Chen CR, McLachlan SM, Rapoport B. The thyrotropin receptor hinge region is not simply a scaffold for the leucine-rich domain but contributes to ligand binding and signal transduction. Mol Endocrinol 2008; 22: 1171-1182
  • 17 Mueller S, Jaeschke H, Gunther R, Paschke R. The hinge region: an important receptor component for GPHR function. Trends Endocrinol Metab 2010; 21: 111-122
  • 18 Morshed SA, Ma R, Latif R, Davies TF. How one TSH receptor antibody induces thyrocyte proliferation while another induces apoptosis. J Autoimmun 2013; 47: 17-24
  • 19 Morshed SA, Latif R, Davies TF. Delineating the autoimmune mechanisms in Graves’ disease. Immunol Res 2012; 54: 191-203
  • 20 Morshed SA, Ando T, Latif R, Davies TF. Neutral antibodies to the TSH receptor are present in Graves’ disease and regulate selective signaling cascades. Endocrinology 2010; 151: 5537-5549
  • 21 Costagliola S, Many MC, Denef JF, Pohlenz J, Refetoff S, Vassart G. Genetic immunization of outbred mice with thyrotropin receptor cDNA provides a model of Graves’ disease. J Clin Invest 2000; 105: 803-811
  • 22 Sanders J, Allen F, Jeffreys J, Bolton J, Richards T, Depraetere H, Nakatake N, Evans M, Kiddie A, Premawardhana LD, Chirgadze DY, Miguel RN, Blundell TL, Furmaniak J, Smith BR. Characteristics of a monoclonal antibody to the thyrotropin receptor that acts as a powerful thyroid-stimulating autoantibody antagonist. Thyroid 2005; 15: 672-682
  • 23 Muehlberg T, Gilbert JA, Rao PV, McGregor AM, Banga JP. Dynamics of thyroid-stimulating and -blocking antibodies to the thyrotropin receptor in a murine model of Graves’ disease. Endocrinology 2004; 145: 1539-1545
  • 24 Sanders J, Allen F, Jeffreys J, Bolton J, Richards T, Depraetere H, Nakatake N, Evans M, Kiddie A, Premawardhana LD, Chirgadze DY, Miguel RN, Blundell TL, Furmaniak J, Smith BR. Characteristics of a monoclonal antibody to the thyrotropin receptor that acts as a powerful thyroid-stimulating autoantibody antagonist. Thyroid 2005; 15: 672-682
  • 25 Sanders J, Jeffreys J, Depraetere H, Evans M, Richards T, Kiddie A, Brereton K, Premawardhana LD, Chirgadze DY, Nunez MR, Blundell TL, Furmaniak J, Rees SB. Characteristics of a human monoclonal autoantibody to the thyrotropin receptor: sequence structure and function. Thyroid 2004; 14: 560-570
  • 26 Nagy EV, Burch HB, Mahoney K, Lukes YG, Morris III JC, Burman KD. Graves’ IgG recognizes linear epitopes in the human thyrotropin receptor. Biochem Biophys Res Commun 1992; 188: 28-33
  • 27 Sanders J, Chirgadze DY, Sanders P, Baker S, Sullivan A, Bhardwaja A, Bolton J, Reeve M, Nakatake N, Evans M, Richards T, Powell M, Miguel RN, Blundell TL, Furmaniak J, Smith BR. Crystal Structure of the TSH Receptor in Complex with a Thyroid-Stimulating Autoantibody. Thyroid 2007; 17: 395-410
  • 28 Latif R, Teixeira A, Michalek K, Ali MR, Schlesinger M, Baliram R, Morshed SA, Davies TF. Antibody protection reveals extended epitopes on the human TSH receptor. PLoS One 2012; 7: e44669
  • 29 Morshed SA, Latif R, Davies TF. Characterization of thyrotropin receptor antibody-induced signaling cascades. Endocrinology 2009; 150: 519-529
  • 30 Sanders P, Young S, Sanders J, Kabelis K, Baker S, Sullivan A, Evans M, Clark J, Wilmot J, Hu X, Roberts E, Powell M, Nunez Miguel R, Furmaniak J, Rees Smith B. Crystal structure of the TSH receptor (TSHR) bound to a blocking-type TSHR autoantibody. J Mol Endocrinol 2011; 46: 81-99
  • 31 Evans M, Sanders J, Tagami T, Sanders P, Young S, Roberts E, Wilmot J, Hu X, Kabelis K, Clark J, Holl S, Richards T, Collyer A, Furmaniak J, Smith BR. Monoclonal autoantibodies to the TSH receptor, one with stimulating activity and one with blocking activity, obtained from the same blood sample. Clin Endocrinol (Oxf) 2010; 73: 404-412
  • 32 Davies TF, Ando T, Lin RY, Tomer Y, Latif R. Thyrotropin receptor-associated diseases: from adenomata to Graves disease. J Clin Invest 2005; 115: 1972-1983
  • 33 Vives-Pi M, Rodriguez-Fernandez S, Pujol-Autonell I. How apoptotic beta-cells direct immune response to tolerance or to autoimmune diabetes: a review. Apoptosis 2015; 20: 263-272
  • 34 Mao J, Yuan H, Xie W, Wu D. Guanine nucleotide exchange factor GEF115 specifically mediates activation of Rho and serum response factor by the G protein alpha subunit Galpha13. Proc Natl Acad Sci USA 1998; 95: 12973-12976
  • 35 Back CM, Stohr S, Schafer EA, Biebermann H, Boekhoff I, Breit A, Gudermann T, Buch TR. TSH induces metallothionein 1 in thyrocytes via Gq/11- and PKC-dependent signaling. J Mol Endocrinol 2013; 51: 79-90
  • 36 Zaballos MA, Garcia B, Santisteban P. Gbetagamma dimers released in response to thyrotropin activate phosphoinositide 3-kinase and regulate gene expression in thyroid cells. Mol Endocrinol 2008; 22: 1183-1199
  • 37 Ye RD. Regulation of nuclear factor kappaB activation by G-protein-coupled receptors. J Leukoc Biol 2001; 70: 839-848
  • 38 Aaronson DS, Horvath CM. A road map for those who don't know JAK-STAT. Science 2002; 296: 1653-1655
  • 39 Stassi G, De MR. Autoimmune thyroid disease: new models of cell death in autoimmunity. Nat Rev Immunol 2002; 2: 195-204
  • 40 Wang SH, Baker JR. The role of apoptosis in thyroid autoimmunity. Thyroid 2007; 17: 975-979
  • 41 Mao C, Wang S, Xiao Y, Xu J, Jiang Q, Jin M, Jiang X, Guo H, Ning G, Zhang Y. Impairment of regulatory capacity of CD4+CD25+ regulatory T cells mediated by dendritic cell polarization and hyperthyroidism in Graves’ disease. J Immunol 2011; 186: 4734-4743
  • 42 Bossowski A, Czarnocka B, Bardadin K, Stasiak-Barmuta A, Urban M, Dadan J, Ratomski K, Bossowska A. Identification of apoptotic proteins in thyroid gland from patients with Graves’ disease and Hashimoto’s thyroiditis. Autoimmunity 2008; 41: 163-173
  • 43 Bossowski A, Czarnocka B, Bardadin K, Urban M, Niedziela M, Dadan J. Expression of Bcl-2 family proteins in thyrocytes from young patients with immune and nonimmune thyroid diseases. Horm Res 2008; 70: 155-164
  • 44 Newby PR, Roberts-Davies EL, Brand OJ, Heward JM, Franklyn JA, Gough SCL, Simmonds MJ. Tag SNP screening of the PDCD1 gene for association with Graves' disease. Clinical Endocrinology 2007; 67: 125-128
  • 45 Nagata S, Hanayama R, Kawane K. Autoimmunity and the clearance of dead cells. Cell 2010; 140: 619-630
  • 46 Marchi S, Giorgi C, Suski JM, Agnoletto C, Bononi A, Bonora M, De Marchi E, Missiroli S, Patergnani S, Poletti F, Rimessi A, Duszynski J, Wieckowski MR, Pinton P. Mitochondria-ros crosstalk in the control of cell death and aging. J Signal Transduct 2012; 2012: 329635
  • 47 Mano T, Shinohara R, Iwase K, Kotake M, Hamada M, Uchimuro K, Hayakawa N, Hayashi R, Nakai A, Ishizuki Y, Nagasaka A. Changes in free radical scavengers and lipid peroxide in thyroid glands of various thyroid disorders. Horm Metab Res 1997; 29: 351-354
  • 48 Gimenez-Barcons M, Colobran R, Gomez-Pau A, Marin-Sanchez A, Casteras A, Obiols G, Abella R, Fernandez-Doblas J, Tonacchera M, Lucas-Martin A, Pujol-Borrell R. Graves’ Disease TSHR-Stimulating Antibodies (TSAbs) Induce the Activation of Immature Thymocytes: A Clue to the Riddle of TSAbs Generation?. J Immunol 2015; 194: 4199-4206
  • 49 Kamath C, Young S, Kabelis K, Sanders J, Adlan MA, Furmaniak J, Rees Smith B, Premawardhana LD. Thyrotrophin receptor antibody characteristics in a woman with long-standing Hashimoto’s who developed Graves’ disease and pretibial myxoedema. Clin Endocrinol (Oxf) 2012; 77: 465-470
  • 50 Takasu N, Yamada T, Sato A, Nakagawa M, Komiya I, Nagasawa Y, Asawa T. Graves’ disease following hypothyroidism due to Hashimoto’s disease: studies of eight cases. Clin Endocrinol (Oxf) 1990; 33: 687-698