Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Nature ; 609(7928): 846-853, 2022 09.
Article in English | MEDLINE | ID: mdl-35940205

ABSTRACT

Thyroid hormones are vital in metabolism, growth and development1. Thyroid hormone synthesis is controlled by thyrotropin (TSH), which acts at the thyrotropin receptor (TSHR)2. In patients with Graves' disease, autoantibodies that activate the TSHR pathologically increase thyroid hormone activity3. How autoantibodies mimic thyrotropin function remains unclear. Here we determined cryo-electron microscopy structures of active and inactive TSHR. In inactive TSHR, the extracellular domain lies close to the membrane bilayer. Thyrotropin selects an upright orientation of the extracellular domain owing to steric clashes between a conserved hormone glycan and the membrane bilayer. An activating autoantibody from a patient with Graves' disease selects a similar upright orientation of the extracellular domain. Reorientation of the extracellular domain transduces a conformational change in the seven-transmembrane-segment domain via a conserved hinge domain, a tethered peptide agonist and a phospholipid that binds within the seven-transmembrane-segment domain. Rotation of the TSHR extracellular domain relative to the membrane bilayer is sufficient for receptor activation, revealing a shared mechanism for other glycoprotein hormone receptors that may also extend to other G-protein-coupled receptors with large extracellular domains.


Subject(s)
Cryoelectron Microscopy , Immunoglobulins, Thyroid-Stimulating , Receptors, Thyrotropin , Thyrotropin , Cell Membrane/metabolism , Graves Disease/immunology , Graves Disease/metabolism , Humans , Immunoglobulins, Thyroid-Stimulating/chemistry , Immunoglobulins, Thyroid-Stimulating/immunology , Immunoglobulins, Thyroid-Stimulating/pharmacology , Immunoglobulins, Thyroid-Stimulating/ultrastructure , Phospholipids/metabolism , Protein Domains , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/ultrastructure , Receptors, Thyrotropin/agonists , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/ultrastructure , Rotation , Thyrotropin/chemistry , Thyrotropin/metabolism , Thyrotropin/pharmacology
2.
Med Hypotheses ; 127: 23-25, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31088643

ABSTRACT

The theory that antibody (Ab) directed against the TSH receptor (TSHR) (TSHRAb) is the causal factor of Graves' disease seems unlikely. Corticosteroids have not had a curative effect on the hyperthyroidism of Graves' disease despite their effectiveness for other autoimmune diseases. Two kinds of TSHRAb, thyroid-stimulating Ab (TSAb) and thyroid-blocking Ab (TBAb), are known as causal factors of hyperthyroidism and hypothyroidism, respectively. Previously, we reported that TSAb may be thyroid stimulating animal IgG-like hormone and TBAb may be the precursor of TSAb. In this paper we suggested that TBAb (precursor) converts to TSAb (active form) via the action of the protease, colloid antigen 2 (CA2). We speculate that the conversion of TBAb to TSAb is controlled by two factors: the protease and an anti-protease Ab. When anti-protease Ab levels are high, the patient exhibits hypothyroidism due to the increase in TBAb levels caused by neutralization of the protease. When anti-protease Ab levels are negative, the patient's hypothyroidism disappeared by the negative serum TBAb due to increased protease. An immunoglobulin G (IgG) with enzyme activity is known as an abzyme, which may be an undeveloped form. IgG with hormone activity may be likewise called an abhormone, which could also be an undeveloped form. The tumor marker CEA is a known member of the IgG supergene family. Many ancestral versions of proteins may have been produced as an IgG form. Possible participation of colloid antigen 2 and abhormone for the etiology of Graves' disease is suggested.


Subject(s)
Antibodies/chemistry , Antigens/chemistry , Graves Disease/etiology , Immunoglobulin G/chemistry , Immunoglobulins, Thyroid-Stimulating/chemistry , Thyrotropin/chemistry , Acetylcholine/chemistry , Animals , Autoantibodies/blood , Carcinoembryonic Antigen/analysis , Humans , Hyperthyroidism/complications , Hypothyroidism/complications , Models, Biological , Receptors, Thyrotropin/chemistry , Swine , Thyroid Gland/pathology , Thyroxine/chemistry
3.
Clin Exp Immunol ; 170(2): 115-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23039881

ABSTRACT

Hyperthyroidism, defined by overproduction of thyroid hormones, has a 2-3% prevalence in the population. The most common form of hyperthyroidism is Graves' disease. A diagnostic biomarker for Graves' disease is the presence of immunoglobulins which bind to, and stimulate, the thyroid stimulating hormone receptor (TSHR), a G-protein coupled receptor (GPCR). We hypothesized that the ectopically expressed TSHR gene in a thyroid stimulating immunoglobulin (TSI) assay could be engineered to increase the accumulation of the GPCR pathway second messenger, cyclic AMP (cAMP), the molecule measured in the assay as a marker for pathway activation. An ectopically expressing TSHR-mutant guanine nucleotide-binding protein, (GNAS) Chinese hamster ovary (CHO) cell clone was constructed using standard molecular biology techniques. After incubation of the new clone with sera containing various levels of TSI, GPCR pathway activation was then quantified by measuring cAMP accumulation in the clone. The clone, together with a NaCl-free cell assay buffer containing 5% polyethylene glycol (PEG)6000, was tested against 56 Graves' patients, 27 toxic thyroid nodule patients and 119 normal patients. Using receiver operating characteristic analysis, when comparing normal with Graves' sera, the assay yielded a sensitivity of 93%, a specificity of 99% and an efficiency of 98%. Total complex precision (within-run, across runs and across days), presented as a percentage coefficient of variation, was found to be 7·8, 8·7 and 7·6% for low, medium and high TSI responding serum, respectively. We conclude that the performance of the new TSI assay provides sensitive detection of TSI, allowing for accurate, early detection of Graves' disease.


Subject(s)
Biological Assay/methods , GTP-Binding Proteins/chemistry , Immunoglobulins, Thyroid-Stimulating/blood , Immunoglobulins, Thyroid-Stimulating/chemistry , Receptors, G-Protein-Coupled/chemistry , Receptors, Thyrotropin/chemistry , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cyclic AMP/genetics , Cyclic AMP/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Graves Disease/blood , Graves Disease/genetics , Graves Disease/metabolism , Humans , Hyperthyroidism/blood , Hyperthyroidism/genetics , Hyperthyroidism/metabolism , Immunoglobulins, Thyroid-Stimulating/metabolism , Polyethylene Glycols/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Sensitivity and Specificity , Thyroid Gland/metabolism
4.
Thyroid ; 21(9): 1013-20, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21834684

ABSTRACT

BACKGROUND: Thyroid-stimulating autoantibodies (TSAb) bind to the thyrotropin receptor (TSHR) extracellular domain, or ectodomain (ECD), comprising a leucine-rich repeat domain (LRD) linked by a hinge region to the transmembrane domain (TMD). The LRD (residues 22-260; signal peptide 1-21) contains two disulfide-bonded loops at its N-terminus. In the crystal structure of the isolated LRD complexed with human TSAb monoclonal antibody (mAb) M22, N-terminal disulfide loop 1 (residues 22-30) could not be determined because of crystal disorder. Nevertheless, present crystal structure data are interpreted to exclude a role for the LRD N-terminal disulfide loops in the TSAb epitope(s), contradicting prior functional evidence of a role for these loops in TSAb function. MATERIALS AND METHODS: To re-examine this issue we studied two cell types expressing the TSHR with the extreme N-terminal loop 1 (residues 22-30) deleted: the TSHR ECD lacking the TMD and tethered to the plasma membrane by a glycosyl-phosphatidylinositol (GPI) anchor, and the TSH holoreceptor containing the TMD. Because TSAb including M22 "see" the holoreceptor poorly relative to the TSHR ECD-GPI, we used the latter to examine the effect of deleting residues 22-30 on M22 binding by flow cytometry and the holoreceptor to test the effect of this deletion on the functional response to M22. RESULTS: Deletion of TSHR N-terminal loop 1 (residues 22-30) reduced the number of TSHR-ECD-GPI recognized by M22 relative to two TSHR mAb with epitopes far downstream of the LRD N-terminal loops. Relative to control mAb 2C11, M22 recognized only 60.4% of cell surface receptors (p = 0.02). In contrast to M22 binding to TSHR-ECD-GPI, in functional studies with the TSH holoreceptor, M22 stimulation of cAMP generation was unaltered by the loop 1 deletion. CONCLUSIONS: Our data support the concept that TSAb interact with the cysteine-rich N-terminus of the TSHR. Comparison of crystal structures of the same TSHR LRD in complex with TSAb M22 or blocking antibody K1-70 helps reconcile contradictory viewpoints. A difference between M22 interaction with the identical TSHR N-terminus expressed on the TSHR-ECD-GPI and holoreceptor suggests that crystallization of the TSHR LRD-M22 complex may not provide a complete understanding of the functional TSAb epitope(s) in Graves' disease.


Subject(s)
Antibodies, Monoclonal/chemistry , Epitopes , Immunoglobulins, Thyroid-Stimulating/chemistry , Mutagenesis, Site-Directed , Receptors, Thyrotropin/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Binding Sites, Antibody , CHO Cells , Cricetinae , Cricetulus , Crystallography , Epitope Mapping , Flow Cytometry , Immunoglobulins, Thyroid-Stimulating/metabolism , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Structure-Activity Relationship , Transfection
5.
J Mol Endocrinol ; 42(5): 381-95, 2009 May.
Article in English | MEDLINE | ID: mdl-19221175

ABSTRACT

The TSH receptor (TSHR) ligands M22 (a thyroid stimulating human monoclonal antibody) and TSH, bind to the concave surface of the leucine rich repeats domain (LRD) of the TSHR and here, we show that M22 mimics closely the binding of TSH. We compared interactions produced by M22 with the TSHR in the M22-TSHR crystal structure (2.55 A resolution) and produced by TSH with the TSHR in a TSH-TSHR comparative model. The crystal structure of the TSHR and a comparative model of TSH based on the crystal structure of FSH were used as components to build the TSH-TSHR model. This model was built based on the FSH-FSH receptor structure (2.9 A) and then the structure of the TSHR in the model was replaced by the TSHR crystal structure. The analysis shows that M22 light chain mimics the TSHbeta chain in its interaction with TSHR LRD, while M22 heavy chain mimics the interactions of the TSHalpha chain. The M22-TSHR complex contains a greater number of hydrogen bonds and salt bridges and fewer hydrophobic interactions than the TSH-TSHR complex, consistent with a higher M22 binding affinity. Furthermore, the surface area formed by TSHR residues N208, Q235, R255, and N256 has been identified as a candidate target region for small molecules which might selectively block binding of autoantibodies to the TSHR.


Subject(s)
Autoantibodies/chemistry , Autoantibodies/metabolism , Immunoglobulins, Thyroid-Stimulating/chemistry , Immunoglobulins, Thyroid-Stimulating/metabolism , Proteins/chemistry , Proteins/metabolism , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/metabolism , Thyrotropin/metabolism , Amino Acids , Humans , Leucine-Rich Repeat Proteins , Ligands , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Surface Properties , Thyrotropin/chemistry
6.
J Mol Endocrinol ; 41(3): 145-64, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18606720

ABSTRACT

The crystal structures of the leucine-rich repeat domain (LRD) of the FSH receptor (FSHR) in complex with FSH and the TSH receptor (TSHR) LRD in complex with the thyroid-stimulating autoantibody (M22) provide opportunities to assess the molecular basis of the specificity of glycoprotein hormone-receptor binding. A comparative model of the TSH-TSHR complex was built using the two solved crystal structures and verified using studies on receptor affinity and activation. Analysis of the FSH-FSHR and TSH-TSHR complexes allowed identification of receptor residues that may be important in hormone-binding specificity. These residues are in leucine-rich repeats at the two ends of the FSHR and the TSHR LRD structures but not in their central repeats. Interactions in the interfaces are consistent with a higher FSH-binding affinity for the FSHR compared with the binding affinity of TSH for the TSHR. The higher binding affinity of porcine (p)TSH and bovine (b)TSH for human (h)TSHR compared with hTSH appears not to be dependent on interactions with the TSHR LRD as none of the residues that differ among hTSH, pTSH or bTSH interact with the LRD. This suggests that TSHs are likely to interact with other parts of the receptors in addition to the LRD with these non-LRD interactions being responsible for affinity differences. Analysis of interactions in the FSH-FSHR and TSH-TSHR complexes suggests that the alpha-chains of both hormones tend to be involved in the receptor activation process while the beta-chains are more involved in defining binding specificity.


Subject(s)
Follicle Stimulating Hormone/metabolism , Immunoglobulins, Thyroid-Stimulating/metabolism , Receptors, FSH/metabolism , Receptors, Thyrotropin/metabolism , Amino Acids , Animals , Cattle , Follicle Stimulating Hormone/chemistry , Humans , Immunoglobulins, Thyroid-Stimulating/chemistry , Models, Molecular , Protein Structure, Secondary , Receptors, FSH/chemistry , Receptors, Thyrotropin/chemistry , Swine
7.
Thyroid ; 17(5): 395-410, 2007 May.
Article in English | MEDLINE | ID: mdl-17542669

ABSTRACT

OBJECTIVE: To analyze interactions between the thyroid-stimulating hormone receptor (TSHR) and a thyroid-stimulating human monoclonal autoantibody (M22) at the molecular level. DESIGN: A complex of part of the TSHR extracellular domain (amino acids 1-260; TSHR260) bound to M22 Fab was prepared and purified. Crystals suitable for X-ray diffraction analysis were obtained and the structure solved at 2.55 A resolution. MAIN OUTCOME: TSHR260 comprises of a curved helical tube and M22 Fab clasps its concave surface at 90 degrees to the tube length axis. The interface buried in the complex is large (2,500 A(2)) and an extensive network of ionic, polar, and hydrophobic bonding is involved in the interaction. There is virtually no movement in the atoms of M22 residues on the binding interface compared to unbound M22 consistent with "lock and key" binding. Mutation of residues showing strong interactions in the structure influenced M22 activity, indicating that the binding detail observed in the complex reflects interactions of M22 with intact, functionally active TSHR. The receptor-binding arrangements of the autoantibody are very similar to those reported for follicle-stimulating hormone (FSH) binding to the FSH receptor (amino acids 1-268) and consequently to those of TSH itself. CONCLUSIONS: It is remarkable that the thyroid-stimulating autoantibody shows almost identical receptor-binding features to TSH although the structures and origins of these two ligands are very different. Furthermore, our structure of the TSHR and its complex with M22 provide foundations for developing new strategies to understand and control both glycoprotein hormone receptor activation and the autoimmune response to the TSHR.


Subject(s)
Immunoglobulins, Thyroid-Stimulating/chemistry , Receptors, Thyrotropin/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallization , Follicle Stimulating Hormone/chemistry , Humans , Immunoglobulin Fab Fragments/chemistry , Molecular Sequence Data , Mutation , Receptors, FSH/chemistry , Receptors, Thyrotropin/genetics , X-Ray Diffraction
8.
J Immunol Methods ; 315(1-2): 11-8, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-16920148

ABSTRACT

Accurate characterization of the antigen binding region of antibodies is of great value in many fields of research, assay development and clinical diagnostics. Up to now, there is an unmet clinical need to use antibodies as diagnostic markers for the prediction of both prognosis and therapeutic response. To this end, comprehensive but differentiated immunoassays need to be generated. We have developed a peptide microarray for the diagnosis and epitope mapping of anti-thyrotropin receptor antibodies. The primary sequence of the human thyrotropin receptor (hTSHR) was represented by a library of 251 synthetic peptides. The peptides were site-specifically immobilized in a two-step procedure first by coupling of biotinylated peptides to hydrazide-modified streptavidin and then utilizing a subsequent chemoselective reaction between the hydrazide linkers of the streptavidin and an aldehyde coated glass surface. The technology was used to map the epitopes of seven commercially available murine monoclonal antibodies specific for the human TSH receptor (mTSHRAb). A previously unknown epitope recognized by mTSHRAb 4C1 was identified at amino acids (AA) 379 through 384 and the epitope recognized by mTSHRAb A9 was also localized (AA 214-222). Previously identified epitopes recognized by mTSHRAbs 2C11 (AA 349-360), 28 (AA 34-39), 49 (AA 289-297), A7 (AA 406-411) and A10 (AA 34-39) were confirmed. The peptide microarray exhibited excellent performance in single and multiplex antibody analysis and high specificity. This technology may have potential as a multi-determinate in vitro diagnostic assay for the differential analysis of a heterogeneity of antibodies involved in the pathogenesis of autoimmune diseases.


Subject(s)
Epitope Mapping/methods , Immunoglobulins, Thyroid-Stimulating/genetics , Protein Array Analysis/methods , Receptors, Thyrotropin/immunology , Aldehydes/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Humans , Immunoglobulins, Thyroid-Stimulating/chemistry , Immunoglobulins, Thyroid-Stimulating/immunology , Mice , Models, Molecular , Molecular Sequence Data , Molecular Structure
9.
J Immunol Methods ; 296(1-2): 159-70, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15680160

ABSTRACT

Stability of single-chain Fvs (scFvs) can be improved by mutagenesis followed by phage display selection where the unstable variants are first inactivated by, for example, denaturing treatment. Here we describe a modified strategy for the selection of stabilized antibody fragments by phage display, based on denaturation under reducing conditions. This strategy was applied to an anti-thyroid-stimulating hormone (TSH) scFv fragment which refolded remarkably during the selection if denaturation was carried out in conventionally used non-reducing conditions. Refolding was, however, efficiently prevented by combining denaturation with reduction of the intra-domain disulfide bridges, which created favourable conditions for selection of clones with improved stability. Using this strategy, scFv mutants with 8-9 degrees C improved thermal stability and 0.8-0.9 M improved stability for guanidinium chloride were found after 4-5 enrichment cycles. The most stable mutants selected contained either Lys(H)66Arg or Asn(H)52aSer mutations, which are known to stabilize other scFvs. Periplasmic expression level of the mutants was also improved.


Subject(s)
Immunoglobulin Variable Region/genetics , Immunoglobulins, Thyroid-Stimulating/genetics , Peptide Library , Animals , Disulfides/chemistry , Guanidine/chemistry , Hot Temperature , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Immunoglobulins, Thyroid-Stimulating/chemistry , Immunoglobulins, Thyroid-Stimulating/immunology , Mice , Mutagenesis/genetics , Oxidation-Reduction , Point Mutation/genetics , Protein Denaturation/genetics , Protein Folding , Thermodynamics
10.
J Endocrinol Invest ; 26(11): 1076-80, 2003 Nov.
Article in English | MEDLINE | ID: mdl-15008244

ABSTRACT

Anti-TSH receptor antibodies (TRAbs) have been known to be involved in Graves' disease and primary hypothyroidism. We previously isolated and reconstituted immunoglobulin (Ig) genes of Epstein-Barr virus-transformed B cell clones producing monoclonal TRAbs obtained from Graves' patients. In the present study, we performed a similar experiment using a B cell clone, 32A-5, derived from a patient with primary hypothyroidism. The variable region genes of Ig heavy (H) and light (L) chains were isolated and sequenced from the 32A-5 clone. A significant number of somatic mutations were found in variable regions of H and L chain gene segments. Each pair of H and L chain cDNAs was ligated into an expression vector for IgG1 production and stably introduced into myeloma cells. The transfectants were injected ip into BALB/c mice to yield ample volume of the antibody for following applications. Interactions of recombinant 32A-5 with Graves' sera with varying thyroid-stimulating antibody (TSAb) activities were studied. The recombinant antibody tended to suppress TSAb activities in 10 of 15 Graves' sera, in which four were significantly inhibited. In summary, this is the first study to analyze human monoclonal TSH-stimulation blocking antibodies (TSBAb) at the molecular level. Use of human recombinant monoclonal TSBAb may be an analytical tool for molecular-basis etiology and an alternative therapeutic path for Graves' disease.


Subject(s)
Antibodies, Monoclonal/blood , Hypothyroidism/immunology , Immunoglobulins, Thyroid-Stimulating/blood , Myxedema/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Base Sequence , Cricetinae , Graves Disease/blood , Graves Disease/immunology , Humans , Hypothyroidism/blood , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/immunology , Immunoglobulins, Thyroid-Stimulating/chemistry , Immunoglobulins, Thyroid-Stimulating/pharmacology , Lymphocytes/chemistry , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Myxedema/blood , Recombinant Proteins , Sequence Analysis, DNA , Thyrotropin/immunology
12.
Thyroid ; 5(6): 449-54, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8808094

ABSTRACT

Thyroid stimulating (TS) activity (cAMP production in thyroid cells) and TSH binding inhibition (TBI) activity (determined by TSH receptor assay) in fragments released from TSAb-IgG by protease digestion were examined. The unbound fraction (UF) and the bound fraction (BF) were separated using a protein A-Sepharose column after papain hydrolysis (more hydrolysis at pH 5.0 than pH 7.5) of TSAb-IgG. When both fractions were gel filtrated on a Sephadex G-100 column, the TS and TBI activity were found in both Fab fraction (Mr 50 kDa) and the retarded fraction (between Mr 50 and 20 kDa) in the UF, and also in the first fraction (undigested IgG, Mr 160 kDa), the second fraction (Fc with tracer amounts of Fab, Mr 50 kDa), and the retarded fraction (between Mr 50 and 20 kDa) of the BF. The biological activity in the second fraction was suggested as being derived from Fab, because the activity bound to the anti-F(ab')2 column but did not bind to the anti-Fc column. Anti-Tg and anti-TPO activities were found in Fab, but were not found in the retarded fraction that consisted of Mr 20-30 kDa. In pepsin hydrolysis the UF from the protein A column consisted of both F(ab')2 (Mr 100 kDa) and pF'c (CH3) (Mr 25 kDa), and the BF consisted of only the undigested IgG. The biological activities were found in both the F(ab')2 fraction and the retarded fraction (between Mr 100 and 25 kDa). Anti-Tg and anti-TPO activities were found in F(ab')2, but no activity was observed in the Mr 25-kDa fraction. The present study showed that the biological activity of TSAb is distributed in not only the Fab or F(ab')2 fragment, but also in thyroactive smaller components (TSC) (Mr 20-30 kDa) without antigen-binding activity such as anti-Tg and anti-TPO. We suggest that TSC may be released from the Fab fragment region of TSAb-IgG by protease hydrolysis.


Subject(s)
Immunoglobulins, Thyroid-Stimulating/chemistry , Immunoglobulins, Thyroid-Stimulating/pharmacology , Animals , Chromatography, Affinity , Chromatography, Gel , Endopeptidases , Graves Disease/metabolism , Humans , Hydrolysis , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/pharmacology , Iodide Peroxidase/metabolism , Long-Acting Thyroid Stimulator/chemistry , Long-Acting Thyroid Stimulator/pharmacology , Papain/chemistry , Pepsin A/chemistry , Sepharose , Staphylococcal Protein A/metabolism , Swine , Thyroglobulin/metabolism , Thyrotropin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL