Skip to main content
Erschienen in: Autoimmunity Highlights 1/2013

01.04.2013 | Review Article

Blocking type TSH receptor antibodies

verfasst von: Jadwiga Furmaniak, Jane Sanders, Bernard Rees Smith

Erschienen in: Autoimmunity Highlights | Ausgabe 1/2013

Einloggen, um Zugang zu erhalten

Abstract

TSH receptor (TSHR) autoantibodies (TRAbs) play a key role in the pathogenesis of Graves’ disease. In the majority of patients, TRAbs stimulate thyroid hormone synthesis via activation of the TSHR (stimulating TRAbs, TSHR agonists). In some patients, TRAbs bind to the receptor but do not cause activation (blocking TRAbs, TSHR antagonists). Isolation of human TSHR monoclonal antibodies (MAbs) with either stimulating (M22 and K1-18) or blocking activities (5C9 and K1-70) has been a major advance in studies on the TSHR. The binding characteristics of the blocking MAbs, their interaction with the TSHR and their effect on TSHR constitutive activity are summarised in this review. In addition, the binding arrangement in the crystal structures of the TSHR in complex with the blocking MAb K1-70 and with the stimulating MAb M22 (2.55 Å and 1.9 Å resolution, respectively) are compared. The stimulating effect of M22 and the inhibiting effect of K1-70 on thyroid hormone secretion in vivo is discussed. Furthermore the ability of K1-70 to inhibit the thyroid stimulating activity of M22 in vivo is shown. Human MAbs which act as TSHR antagonists are potentially important new therapeutics. For example, in Graves’ disease, K1-70 may well be effective in controlling hyperthyroidism and the eye signs caused by stimulating TRAb. In addition, hyperthyroidism caused by autonomous TSH secretion should be treatable by K1-70, and 5C9 has the potential to control hyperthyroidism associated with TSHR activating mutations. Furthermore, K1-70 has potential applications in thyroid imaging as well as targeted drug delivery to TSHR expressing tissues.
Literatur
2.
3.
Zurück zum Zitat Rees Smith B, Bolton J, Young S, Collyer A, Weeden A, Bradbury J, Weightman D, Perros P, Sanders J, Furmaniak J (2004) A new assay for thyrotropin receptor autoantibodies. Thyroid 14:830–835. doi:10.1089/thy.2004.14.830CrossRef Rees Smith B, Bolton J, Young S, Collyer A, Weeden A, Bradbury J, Weightman D, Perros P, Sanders J, Furmaniak J (2004) A new assay for thyrotropin receptor autoantibodies. Thyroid 14:830–835. doi:10.​1089/​thy.​2004.​14.​830CrossRef
5.
Zurück zum Zitat McLachlan SM, Rapoport B (1996) Monoclonal, human autoantibodies to the TSH receptor—the holy grail and why are we looking for it? J Clin Endocrinol Metab 81:3152–3154PubMedCrossRef McLachlan SM, Rapoport B (1996) Monoclonal, human autoantibodies to the TSH receptor—the holy grail and why are we looking for it? J Clin Endocrinol Metab 81:3152–3154PubMedCrossRef
9.
10.
Zurück zum Zitat Costagliola S, Bonomi M, Morganthaler NG, Van Durme J, Panneels V, Refetoff S, Vassart G (2004) Delineation of the discontinuous-conformational epitope of a monoclonal antibody displaying full in vitro and in vivo thyrotropin activity. Mol Endocrinol 18:3020–3034. doi:10.1210/me.2004-0231PubMedCrossRef Costagliola S, Bonomi M, Morganthaler NG, Van Durme J, Panneels V, Refetoff S, Vassart G (2004) Delineation of the discontinuous-conformational epitope of a monoclonal antibody displaying full in vitro and in vivo thyrotropin activity. Mol Endocrinol 18:3020–3034. doi:10.​1210/​me.​2004-0231PubMedCrossRef
11.
Zurück zum Zitat Gilbert J, Gianoukakis A, Salehi S, Moorhead J, Rao P, Kahn MZ, McGregor A, Smith T, Banga JP (2006) Monoclonal pathogenic antibodies to the thyroid stimulating hormone receptor in Graves’ disease with potent thyroid-stimulating activity but differential blocking activity activate multiple signalling pathways. J Immunol 176:5084–5092PubMedCrossRef Gilbert J, Gianoukakis A, Salehi S, Moorhead J, Rao P, Kahn MZ, McGregor A, Smith T, Banga JP (2006) Monoclonal pathogenic antibodies to the thyroid stimulating hormone receptor in Graves’ disease with potent thyroid-stimulating activity but differential blocking activity activate multiple signalling pathways. J Immunol 176:5084–5092PubMedCrossRef
12.
Zurück zum Zitat Sanders J, Allen F, Jeffreys J, Bolton J, Richards T, Depraetere H, Nakatake N, Evans M, Kiddie A, Premawardhana LDKE, Chirgadze DY, Miguel RN, Blundell TL, Furmaniak J, Rees Smith B (2005) Characteristics of a monoclonal antibody to the thyrotropin receptor that acts as a powerful thyroid-stimulating autoantibody antagonist. Thyroid 15:672–682. doi:10.1089/thy.2005.15.672PubMedCrossRef Sanders J, Allen F, Jeffreys J, Bolton J, Richards T, Depraetere H, Nakatake N, Evans M, Kiddie A, Premawardhana LDKE, Chirgadze DY, Miguel RN, Blundell TL, Furmaniak J, Rees Smith B (2005) Characteristics of a monoclonal antibody to the thyrotropin receptor that acts as a powerful thyroid-stimulating autoantibody antagonist. Thyroid 15:672–682. doi:10.​1089/​thy.​2005.​15.​672PubMedCrossRef
13.
Zurück zum Zitat Shepherd PS, Da Costa CR, Cridland JC, Gilmore KS, Johnstone AP (1999) Identification of an important thyrotrophin binding site on the human thyrotrophin receptor using monoclonal antibodies. Mol Cell Endocrinol 149:197–206PubMedCrossRef Shepherd PS, Da Costa CR, Cridland JC, Gilmore KS, Johnstone AP (1999) Identification of an important thyrotrophin binding site on the human thyrotrophin receptor using monoclonal antibodies. Mol Cell Endocrinol 149:197–206PubMedCrossRef
16.
Zurück zum Zitat Davies TF, Bobovnikova Y, Weiss M, Vlase H, Moran T, Graves PN (1998) Development and characterization of monoclonal antibodies specific for the murine thyrotropin receptor. Thyroid 8:693–701PubMedCrossRef Davies TF, Bobovnikova Y, Weiss M, Vlase H, Moran T, Graves PN (1998) Development and characterization of monoclonal antibodies specific for the murine thyrotropin receptor. Thyroid 8:693–701PubMedCrossRef
19.
Zurück zum Zitat Oda Y, Sanders J, Roberts S, Maruyama M, Kato R, Perez M, Petersen VB, Wedlock N, Furmaniak J, Rees Smith B (1998) Binding characteristics of antibodies to the TSH receptor. J Mol Endocrinol 20:233–244. doi:10.1677/jme.0.0200233PubMedCrossRef Oda Y, Sanders J, Roberts S, Maruyama M, Kato R, Perez M, Petersen VB, Wedlock N, Furmaniak J, Rees Smith B (1998) Binding characteristics of antibodies to the TSH receptor. J Mol Endocrinol 20:233–244. doi:10.​1677/​jme.​0.​0200233PubMedCrossRef
20.
Zurück zum Zitat Oda Y, Sanders J, Evans M, Kiddie A, Munkley A, James C, Richards T, Wills J, Furmaniak J, Rees Smith B (2000) Epitope analysis of the human thyrotropin (TSH) receptor using monoclonal antibodies. Thyroid 10:1051–1059. doi:10.1089/thy.2000.10.1051PubMedCrossRef Oda Y, Sanders J, Evans M, Kiddie A, Munkley A, James C, Richards T, Wills J, Furmaniak J, Rees Smith B (2000) Epitope analysis of the human thyrotropin (TSH) receptor using monoclonal antibodies. Thyroid 10:1051–1059. doi:10.​1089/​thy.​2000.​10.​1051PubMedCrossRef
23.
Zurück zum Zitat Sanders J, Jeffreys J, Depraetere H, Evans M, Richards T, Kiddie A, Brereton K, Premawardhana LD, Chirgadze DY, Núñez Miguel R, Blundell TL, Furmaniak J, Rees Smith B (2004) Characteristics of a human monoclonal autoantibody to the thyrotropin receptor: sequence structure and function. Thyroid 14:560–570. doi:10.1089/1050725041692918PubMedCrossRef Sanders J, Jeffreys J, Depraetere H, Evans M, Richards T, Kiddie A, Brereton K, Premawardhana LD, Chirgadze DY, Núñez Miguel R, Blundell TL, Furmaniak J, Rees Smith B (2004) Characteristics of a human monoclonal autoantibody to the thyrotropin receptor: sequence structure and function. Thyroid 14:560–570. doi:10.​1089/​1050725041692918​PubMedCrossRef
24.
Zurück zum Zitat Sanders J, Chirgadze DY, Sanders P, Baker S, Sullivan A, Bhardwaja A, Bolton J, Reeve M, Nakatake N, Evans M, Richards T, Powell M, Núñez Miguel R, Blundell TL, Furmaniak J, Rees Smith B (2007) Crystal structure of the TSH receptor in complex with a thyroid-stimulating autoantibody. Thyroid 17:395–410. doi:10.1089/thy.2007.0034PubMedCrossRef Sanders J, Chirgadze DY, Sanders P, Baker S, Sullivan A, Bhardwaja A, Bolton J, Reeve M, Nakatake N, Evans M, Richards T, Powell M, Núñez Miguel R, Blundell TL, Furmaniak J, Rees Smith B (2007) Crystal structure of the TSH receptor in complex with a thyroid-stimulating autoantibody. Thyroid 17:395–410. doi:10.​1089/​thy.​2007.​0034PubMedCrossRef
25.
Zurück zum Zitat Sanders J, Evans M, Betterle C, Sanders P, Bhardwaja A, Young S, Roberts E, Wilmot J, Richards T, Kiddie A, Small K, Platt H, Summerhayes S, Harris R, Reeve M, Coco G, Zanchetta R, Chen S, Furmaniak J, Rees Smith B (2008) A human monoclonal autoantibody to the thyrotropin receptor with thyroid-stimulating blocking activity. Thyroid 18:735–746. doi:10.1089/thy.2007.0327PubMedCrossRef Sanders J, Evans M, Betterle C, Sanders P, Bhardwaja A, Young S, Roberts E, Wilmot J, Richards T, Kiddie A, Small K, Platt H, Summerhayes S, Harris R, Reeve M, Coco G, Zanchetta R, Chen S, Furmaniak J, Rees Smith B (2008) A human monoclonal autoantibody to the thyrotropin receptor with thyroid-stimulating blocking activity. Thyroid 18:735–746. doi:10.​1089/​thy.​2007.​0327PubMedCrossRef
27.
Zurück zum Zitat 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, Rees Smith B (2010) Monoclonal autoantibodies to the TSH receptor, one with stimulating activity and one with blocking activity, obtained from the same blood sample. Clin Endocrinol 73:404–412. doi:10.1111/j.1365-2265.2010.03831.xCrossRef 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, Rees Smith B (2010) Monoclonal autoantibodies to the TSH receptor, one with stimulating activity and one with blocking activity, obtained from the same blood sample. Clin Endocrinol 73:404–412. doi:10.​1111/​j.​1365-2265.​2010.​03831.​xCrossRef
28.
Zurück zum Zitat Sanders P, Young S, Sanders J, Kabelis K, Baker S, Sullivan A, Evans M, Clark J, Wilmot J, Hu X, Roberts E, Powell M, Núñez Miguel R, Furmaniak J, Rees Smith B (2011) Crystal structure of the TSH receptor (TSHR) bound to a blocking-type TSHR autoantibody. J Mol Endocrinol 46:81–99. doi:10.1530/JME-10-0127PubMed Sanders P, Young S, Sanders J, Kabelis K, Baker S, Sullivan A, Evans M, Clark J, Wilmot J, Hu X, Roberts E, Powell M, Núñez Miguel R, Furmaniak J, Rees Smith B (2011) Crystal structure of the TSH receptor (TSHR) bound to a blocking-type TSHR autoantibody. J Mol Endocrinol 46:81–99. doi:10.​1530/​JME-10-0127PubMed
30.
31.
32.
Zurück zum Zitat Zhang M, Phuong K, Tong T, Fremont V, Chen J, Narayan P, Puett D, Weintraub BD, Szkudlinski MW (2000) The extracellular domain suppresses constitutive activity of the transmembrane domain of the human TSH receptor: implications for hormone-receptor interaction and antagonist design. Endocrinology 141:3514–3517. doi:10.1210/en.141.9.3514PubMedCrossRef Zhang M, Phuong K, Tong T, Fremont V, Chen J, Narayan P, Puett D, Weintraub BD, Szkudlinski MW (2000) The extracellular domain suppresses constitutive activity of the transmembrane domain of the human TSH receptor: implications for hormone-receptor interaction and antagonist design. Endocrinology 141:3514–3517. doi:10.​1210/​en.​141.​9.​3514PubMedCrossRef
33.
Zurück zum Zitat Vlaeminck-Guillem VK, Ho SC, Rodien P, Vassart G, Costagliola S (2002) Activation of the cAMP pathway by the TSH receptor involves switching of the ectodomain from a tethered inverse agonist to an agonist. Mol Endocrinol 16:736–746. doi:10.1210/me.16.4.736PubMedCrossRef Vlaeminck-Guillem VK, Ho SC, Rodien P, Vassart G, Costagliola S (2002) Activation of the cAMP pathway by the TSH receptor involves switching of the ectodomain from a tethered inverse agonist to an agonist. Mol Endocrinol 16:736–746. doi:10.​1210/​me.​16.​4.​736PubMedCrossRef
34.
Zurück zum Zitat Sanders J, Oda Y, Roberts S, Maruyama M, Furmaniak J, Rees Smith B (1997) Understanding the thyrotropin receptor function-structure relationship. Baillière’s Clin Endocr Metab 11:451–479. doi:10.1016/S0950-351X(97)80693-3CrossRef Sanders J, Oda Y, Roberts S, Maruyama M, Furmaniak J, Rees Smith B (1997) Understanding the thyrotropin receptor function-structure relationship. Baillière’s Clin Endocr Metab 11:451–479. doi:10.​1016/​S0950-351X(97)80693-3CrossRef
36.
Zurück zum Zitat Kopp P, Muirhead S, Jourdain N, Gu W-X, Jameson JL, Rodd C (1997) Congenital hyperthyroidism caused by solitary toxic adenoma harboring a novel somatic mutation (serine281 to isoleucine) in the extracellular domain of the thyrotropin receptor. J Clin Investig 100:1634–1639. doi:10.1172/JCI119687PubMedPubMedCentralCrossRef Kopp P, Muirhead S, Jourdain N, Gu W-X, Jameson JL, Rodd C (1997) Congenital hyperthyroidism caused by solitary toxic adenoma harboring a novel somatic mutation (serine281 to isoleucine) in the extracellular domain of the thyrotropin receptor. J Clin Investig 100:1634–1639. doi:10.​1172/​JCI119687PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Parma J, Van Sande J, Swillens S, Tonacchera M, Dumont J, Vassart G (1995) Somatic mutations causing constitutive activity of the thyrotropin receptor are the major cause of hyperfunctioning thyroid adenomas: identification of additional mutations activating both the cyclic adenosine 3′,5′-monophosphate and inositol phosphate-Ca2 + cascades. Mol Endocrinol 9:725–733. doi:10.1210/me.9.6.725PubMed Parma J, Van Sande J, Swillens S, Tonacchera M, Dumont J, Vassart G (1995) Somatic mutations causing constitutive activity of the thyrotropin receptor are the major cause of hyperfunctioning thyroid adenomas: identification of additional mutations activating both the cyclic adenosine 3′,5′-monophosphate and inositol phosphate-Ca2 + cascades. Mol Endocrinol 9:725–733. doi:10.​1210/​me.​9.​6.​725PubMed
38.
Zurück zum Zitat Parma J, Duprez L, Van Sande J, Cochaux P, Gervy C, Mockel J, Dumont J, Vassart G (1993) Somatic mutations in the thyrotropin receptor gene cause hyperfunctioning thyroid adenomas. Nature 365:649–651. doi:10.1038/365649a0PubMedCrossRef Parma J, Duprez L, Van Sande J, Cochaux P, Gervy C, Mockel J, Dumont J, Vassart G (1993) Somatic mutations in the thyrotropin receptor gene cause hyperfunctioning thyroid adenomas. Nature 365:649–651. doi:10.​1038/​365649a0PubMedCrossRef
39.
Zurück zum Zitat Sanders J, Bolton J, Sanders P, Jeffreys J, Nakatake N, Richards T, Evans M, Kiddie A, Summerhayes S, Roberts E, Núñez Miguel R, Furmaniak J, Rees Smith B (2006) Effects of TSH receptor mutations on binding and biological activity of monoclonal antibodies and TSH. Thyroid 16:1195–1206. doi:10.1089/thy.2006.16.1195PubMedCrossRef Sanders J, Bolton J, Sanders P, Jeffreys J, Nakatake N, Richards T, Evans M, Kiddie A, Summerhayes S, Roberts E, Núñez Miguel R, Furmaniak J, Rees Smith B (2006) Effects of TSH receptor mutations on binding and biological activity of monoclonal antibodies and TSH. Thyroid 16:1195–1206. doi:10.​1089/​thy.​2006.​16.​1195PubMedCrossRef
40.
Zurück zum Zitat Kosugi S, Ban T, Akamizu T, Kohn LD (1991) Site-directed mutagenesis of a portion of the extracellular domain of the rat thyrotropin receptor important in autoimmune thyroid disease and nonhomologous with gonadotropin receptors: relationship of functional and immunogenic domains. J Biol Chem 266:19413–19418PubMed Kosugi S, Ban T, Akamizu T, Kohn LD (1991) Site-directed mutagenesis of a portion of the extracellular domain of the rat thyrotropin receptor important in autoimmune thyroid disease and nonhomologous with gonadotropin receptors: relationship of functional and immunogenic domains. J Biol Chem 266:19413–19418PubMed
41.
Zurück zum Zitat Mueller S, Kleinau G, Jaeschke H, Paschke R, Krause G (2008) Extended hormone binding site of the human thyroid stimulating hormone receptor: distinctive acidic residues in the hinge region are involved in bovine thyroid stimulating hormone binding and receptor activation. J Biol Chem 283:18048–18055. doi:10.1074/jbc.M800449200PubMedCrossRef Mueller S, Kleinau G, Jaeschke H, Paschke R, Krause G (2008) Extended hormone binding site of the human thyroid stimulating hormone receptor: distinctive acidic residues in the hinge region are involved in bovine thyroid stimulating hormone binding and receptor activation. J Biol Chem 283:18048–18055. doi:10.​1074/​jbc.​M800449200PubMedCrossRef
42.
Zurück zum Zitat Mueller S, Kleinau G, Szkudlinski MW, Jaeschke H, Krause G, Paschke R (2009) The superagonistic activity of bovine thyroid-stimulating hormone (TSH) and the human TR1401 TSH analog is determined by specific amino acids in the hinge region of the human TSH receptor. J Biol Chem 284:16317–16324. doi:10.1074/jbc.M109.005710PubMedPubMedCentralCrossRef Mueller S, Kleinau G, Szkudlinski MW, Jaeschke H, Krause G, Paschke R (2009) The superagonistic activity of bovine thyroid-stimulating hormone (TSH) and the human TR1401 TSH analog is determined by specific amino acids in the hinge region of the human TSH receptor. J Biol Chem 284:16317–16324. doi:10.​1074/​jbc.​M109.​005710PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Sanders J, Núñez Miguel R, Bolton J, Bhardwaja A, Sanders P, Nakatake N, Evans M, Furmaniak J, Rees Smith B (2007) Molecular interactions between the TSH receptor and a thyroid-stimulating monoclonal autoantibody. Thyroid 17:699–706. doi:10.1089/thy.2007.0041PubMedCrossRef Sanders J, Núñez Miguel R, Bolton J, Bhardwaja A, Sanders P, Nakatake N, Evans M, Furmaniak J, Rees Smith B (2007) Molecular interactions between the TSH receptor and a thyroid-stimulating monoclonal autoantibody. Thyroid 17:699–706. doi:10.​1089/​thy.​2007.​0041PubMedCrossRef
45.
Zurück zum Zitat Chen C-R, Tanaka K, Chazenbalk GD, McLachlan SM, Rapoport B (2001) A full biological response to autoantibodies in Graves’ disease requires a disulfide-bond loop in the thyrotropin N-terminus homologous to a laminin EGF-like domain. J Biol Chem 276:14767–14772. doi:10.1074/jbc.M008001200PubMedCrossRef Chen C-R, Tanaka K, Chazenbalk GD, McLachlan SM, Rapoport B (2001) A full biological response to autoantibodies in Graves’ disease requires a disulfide-bond loop in the thyrotropin N-terminus homologous to a laminin EGF-like domain. J Biol Chem 276:14767–14772. doi:10.​1074/​jbc.​M008001200PubMedCrossRef
46.
Zurück zum Zitat Chazenbalk GD, Latrofa F, McLachlan SM, Rapoport B (2004) Thyroid stimulation does not require antibodies with identical epitopes but does involve recognition of a critical conformation at the N terminus of the thyrotropin receptor A-subunit. J Clin Endocrinol Metab 89:1788–1793. doi:10.1210/jc.2003-031554PubMedCrossRef Chazenbalk GD, Latrofa F, McLachlan SM, Rapoport B (2004) Thyroid stimulation does not require antibodies with identical epitopes but does involve recognition of a critical conformation at the N terminus of the thyrotropin receptor A-subunit. J Clin Endocrinol Metab 89:1788–1793. doi:10.​1210/​jc.​2003-031554PubMedCrossRef
47.
Zurück zum Zitat Hamidi S, Chen C-R, McLachlan SM, Rapoport B (2011) Insight into thyroid-stimulating autoantibody interaction with the thyrotropin receptor N-terminus based on mutagenesis and re-evaluation of ambiguity in this region of the receptor crystal structure. Thyroid 21:1013–1020. doi:10.1089/thy.2011.0147PubMedPubMedCentralCrossRef Hamidi S, Chen C-R, McLachlan SM, Rapoport B (2011) Insight into thyroid-stimulating autoantibody interaction with the thyrotropin receptor N-terminus based on mutagenesis and re-evaluation of ambiguity in this region of the receptor crystal structure. Thyroid 21:1013–1020. doi:10.​1089/​thy.​2011.​0147PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Núñez Miguel R, Sanders J, Jeffreys J, Depraetere H, Evans M, Richards T, Blundell TL, Rees Smith B, Furmaniak J (2004) Analysis of the thyrotropin receptor-thyrotropin interaction by comparative modeling. Thyroid 14:991–1011. doi:10.1089/thy.2004.14.991PubMedCrossRef Núñez Miguel R, Sanders J, Jeffreys J, Depraetere H, Evans M, Richards T, Blundell TL, Rees Smith B, Furmaniak J (2004) Analysis of the thyrotropin receptor-thyrotropin interaction by comparative modeling. Thyroid 14:991–1011. doi:10.​1089/​thy.​2004.​14.​991PubMedCrossRef
52.
Zurück zum Zitat Núñez Miguel R, Sanders J, Chirgadze DY, Blundell TL, Furmaniak J, Rees Smith B (2008) FSH and TSH binding to their respective receptors: similarities, differences and implication for glycoprotein hormone specificity. J Mol Endocrinol 41:145–164. doi:10.1677/JME-08-0040PubMedCrossRef Núñez Miguel R, Sanders J, Chirgadze DY, Blundell TL, Furmaniak J, Rees Smith B (2008) FSH and TSH binding to their respective receptors: similarities, differences and implication for glycoprotein hormone specificity. J Mol Endocrinol 41:145–164. doi:10.​1677/​JME-08-0040PubMedCrossRef
53.
Zurück zum Zitat Núñez Miguel R, Sanders J, Chirgadze DY, Furmaniak J, Rees Smith B (2009) Thyroid stimulating autoantibody M22 mimics TSH binding to the TSH receptor leucine rich domain: a comparative structural study of protein–protein interactions. J Mol Endocrinol 42:381–395. doi:10.1677/JME-08-0152PubMedCrossRef Núñez Miguel R, Sanders J, Chirgadze DY, Furmaniak J, Rees Smith B (2009) Thyroid stimulating autoantibody M22 mimics TSH binding to the TSH receptor leucine rich domain: a comparative structural study of protein–protein interactions. J Mol Endocrinol 42:381–395. doi:10.​1677/​JME-08-0152PubMedCrossRef
54.
Zurück zum Zitat Horimoto M, Petersen VB, Pegg CAS, Fukuma N, Wabayashi N, Kiso Y, Furmaniak J, Rees Smith B (1993) Production and characterization of a human monoclonal thyroid peroxidase autoantibody. Autoimmunity 14:1–7. doi:10.3109/08916939309077350CrossRef Horimoto M, Petersen VB, Pegg CAS, Fukuma N, Wabayashi N, Kiso Y, Furmaniak J, Rees Smith B (1993) Production and characterization of a human monoclonal thyroid peroxidase autoantibody. Autoimmunity 14:1–7. doi:10.​3109/​0891693930907735​0CrossRef
55.
Zurück zum Zitat Costagliola S, Rodien P, Many MC, Ludgate M, Vassart G (1998) Genetic immunisation against the human thyrotropin receptor causes thyroiditis and allows production of monoclonal antibodies recognising the native receptor. J Immunol 160:1458–1465PubMed Costagliola S, Rodien P, Many MC, Ludgate M, Vassart G (1998) Genetic immunisation against the human thyrotropin receptor causes thyroiditis and allows production of monoclonal antibodies recognising the native receptor. J Immunol 160:1458–1465PubMed
56.
Zurück zum Zitat Sequeira M, Jasani B, Fuhrer D, Wheeler M, Ludgate M (2002) Demonstration of reduced in vivo surface expression of activating mutant thyrotropin receptors in thyroid sections. Eur J Endocrinol 146:163–171. doi:10.1530/eje.0.1460163PubMedCrossRef Sequeira M, Jasani B, Fuhrer D, Wheeler M, Ludgate M (2002) Demonstration of reduced in vivo surface expression of activating mutant thyrotropin receptors in thyroid sections. Eur J Endocrinol 146:163–171. doi:10.​1530/​eje.​0.​1460163PubMedCrossRef
57.
Zurück zum Zitat Boschi A, Daumerie Ch, Spiritus M, Beguin C, Senou M, Yuksel D, Duplicy M, Costagliola S, Ludgate M, Many MC (2005) Quantification of cells expressing the thyrotropin receptor in extraocular muscles in thyroid associated orbitopathy. Br J Ophthalmol 89:724–729. doi:10.1136/bjo.2004.050807PubMedPubMedCentralCrossRef Boschi A, Daumerie Ch, Spiritus M, Beguin C, Senou M, Yuksel D, Duplicy M, Costagliola S, Ludgate M, Many MC (2005) Quantification of cells expressing the thyrotropin receptor in extraocular muscles in thyroid associated orbitopathy. Br J Ophthalmol 89:724–729. doi:10.​1136/​bjo.​2004.​050807PubMedPubMedCentralCrossRef
68.
70.
Zurück zum Zitat Rapoport B, Alsabeh R, Aftergood D, McLachlan SM (2000) Elephantiasic pretibial myedema: insight into and a hypothesis regarding the pathogenesis of the extrathyroidal manifestations of Graves’ disease. Thyroid 10:685–692. doi:10.1089/10507250050137761PubMedCrossRef Rapoport B, Alsabeh R, Aftergood D, McLachlan SM (2000) Elephantiasic pretibial myedema: insight into and a hypothesis regarding the pathogenesis of the extrathyroidal manifestations of Graves’ disease. Thyroid 10:685–692. doi:10.​1089/​1050725005013776​1PubMedCrossRef
71.
76.
Zurück zum Zitat Hayakawa N, Premawardhana LDKE, Powell M, Masuda M, Arnold C, Sanders J, Evans M, Chen S, Jaume JC, Baekkeskov S, Rees Smith B, Furmaniak J (2002) Isolation and characterization of human monoclonal autoantibodies to glutamic acid decarboxylase. Autoimmunity 35:343–355. doi:10.1080/0891693021000003206PubMedCrossRef Hayakawa N, Premawardhana LDKE, Powell M, Masuda M, Arnold C, Sanders J, Evans M, Chen S, Jaume JC, Baekkeskov S, Rees Smith B, Furmaniak J (2002) Isolation and characterization of human monoclonal autoantibodies to glutamic acid decarboxylase. Autoimmunity 35:343–355. doi:10.​1080/​0891693021000003​206PubMedCrossRef
77.
Zurück zum Zitat Sanders J, Evans M, Betterle C, Sanders P, Bhardwaja A, Young S, Roberts E, Wilmot J, Richards T, Kiddie A, Small K, Platt H, Summerhayes S, Harris R, Reeve M, Coco G, Zanchetta R, Chen S, Furmaniak J, Rees Smith B (2008) A human monoclonal autoantibody to the thyrotropin receptor with thyroid-stimulating blocking activity. Thyroid 18(7):735–746. doi:10.1089/thy.2007.0327PubMedCrossRef Sanders J, Evans M, Betterle C, Sanders P, Bhardwaja A, Young S, Roberts E, Wilmot J, Richards T, Kiddie A, Small K, Platt H, Summerhayes S, Harris R, Reeve M, Coco G, Zanchetta R, Chen S, Furmaniak J, Rees Smith B (2008) A human monoclonal autoantibody to the thyrotropin receptor with thyroid-stimulating blocking activity. Thyroid 18(7):735–746. doi:10.​1089/​thy.​2007.​0327PubMedCrossRef
78.
Zurück zum Zitat Furmaniak J, Sanders J, Young S, Kabelis K, Sanders P, Evans M et al (2012) In vivo effects of a human thyroid timulating monoclonal autoantibody (M22) and a human thyroid blocking autoantibody (K1-70). Autoimmun Highlights (in press) doi:10.1007/s13317-011-0025-9 Furmaniak J, Sanders J, Young S, Kabelis K, Sanders P, Evans M et al (2012) In vivo effects of a human thyroid timulating monoclonal autoantibody (M22) and a human thyroid blocking autoantibody (K1-70). Autoimmun Highlights (in press) doi:10.​1007/​s13317-011-0025-9
Metadaten
Titel
Blocking type TSH receptor antibodies
verfasst von
Jadwiga Furmaniak
Jane Sanders
Bernard Rees Smith
Publikationsdatum
01.04.2013
Verlag
Springer International Publishing
Erschienen in
Autoimmunity Highlights / Ausgabe 1/2013
Print ISSN: 2038-0305
Elektronische ISSN: 2038-3274
DOI
https://doi.org/10.1007/s13317-012-0028-1

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.