Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Int J Biol Macromol ; 107(Pt B): 2484-2491, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29079441

ABSTRACT

This work is devoted to the ascertainment of serological cross-reactivity between OmpF porin from Yersinia pseudotuberculosis (YpOmpF) and human thyroid-stimulating hormone receptor (hTSHR). Extracts containing hTSHR were isolated from surgical thyroid tissue of patients with clinical and diagnostic signs of diffuse toxic goiter. Monoclonal antibodies to hTSHR (mAbs) were shown to interact both with antigens in thyroid tissue extracts and with YpOmpF. Models of spatial structures of trimer and monomer complexes of YpOmpF with antibodies to hTSHR were also constructed. According to the results of molecular modeling, YpOmpF, being in monomeric form, can, like hTSHR, interact freely with the mAbs. But when the porin trimer is formed the hydrophobic region that comprises in the porin-antibody interaction zone is closed. This circumstance as well as other spatial rearrangement of amino acid residues that determine the efficiency of binding prevents the interaction between YpOmpF trimer and monoclonal antibody to receptor. These in vitro and in silico results confirmed the existence of the phenomenon of molecular mimicry. Thus, an autoimmune disease of the thyroid gland (Graves' disease) that sometimes arises after suffering pseudotuberculosis may be the consequence of the structural and antigenic similarity between YpOmpF and hTSHR.


Subject(s)
Computer Simulation , Cross Reactions/immunology , Porins/immunology , Receptors, Thyrotropin/immunology , Yersinia pseudotuberculosis/metabolism , Amino Acid Sequence , Animals , Female , Humans , Mice, Inbred BALB C , Models, Molecular , Protein Binding , Protein Structure, Secondary , Rabbits , Receptors, Thyrotropin/isolation & purification , Thyroid Gland/metabolism , Tissue Extracts
2.
Rev. lab. clín ; 6(3): 132-134, jul.-sept. 2013.
Article in Spanish | IBECS | ID: ibc-115470

ABSTRACT

El hipotiroidismo materno clínico y el subclínico tienen consecuencias graves tanto para la madre como para el feto. Debido a la compleja fisiología de la función tiroidea durante el embarazo, debería realizarse la evaluación hormonal según valores de referencia para cada trimestre de gestación y para cada zona y con las técnicas propias de cada laboratorio. Si no se dispone en el laboratorio de estos valores de referencia trimestrales propios, se recomiendan los siguientes valores de referencia de tirotropina: primer trimestre < 2,5 mUI/L; segundo y tercer trimestres < 3,0 mUI/L(AU)


Overt and subclinical maternal hypothyroidism is known to have serious adverse effects for both mother and fetus. Given the complex physiology of thyroid function during pregnancy, hormone assessment should be performed according to reference values for each gestational trimester and generated locally in each reference laboratory. If trimester-specific references intervals are not available in the laboratory, the following reference ranges of tirotropine are recommended: first trimester < 2,5 mUI/L; second and third trimesters < 3,0 mUI/L(AU)


Subject(s)
Humans , Female , Receptors, Thyrotropin/isolation & purification , Thyrotropin , Thyrotropin-Releasing Hormone , Receptors, Thyrotropin-Releasing Hormone/analysis , Pregnancy/blood , Reference Values , Congenital Hypothyroidism/blood , Hypothyroidism/blood , Congenital Hypothyroidism/complications , Congenital Hypothyroidism/diagnosis , Hypothyroidism/complications , Hypothyroidism/diagnosis
3.
J Immunol ; 181(6): 4397-405, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18768899

ABSTRACT

Thyroid-stimulating hormone receptor (TSHR) plays a central role in regulating thyroid function and is targeted by IgGs in Graves' disease (GD-IgG). Whether TSHR is involved in the pathogenesis of thyroid-associated ophthalmopathy (TAO), the orbital manifestation of GD, remains uncertain. TSHR signaling overlaps with that of insulin-like grow factor 1 receptor (IGF-1R). GD-IgG can activate fibroblasts derived from donors with GD to synthesize T cell chemoattractants and hyaluronan, actions mediated through IGF-1R. In this study, we compare levels of IGF-1R and TSHR on the surfaces of TAO and control orbital fibroblasts and thyrocytes and explore the physical and functional relationship between the two receptors. TSHR levels are 11-fold higher on thyrocytes than on TAO or control fibroblasts. In contrast, IGF-1R levels are 3-fold higher on TAO vs control fibroblasts. In pull-down studies using fibroblasts, thyrocytes, and thyroid tissue, Abs directed specifically against either IGF-1Rbeta or TSHR bring both proteins out of solution. Moreover, IGF-1Rbeta and TSHR colocalize to the perinuclear and cytoplasmic compartments in fibroblasts and thyrocytes by confocal microscopy. Examination of orbital tissue from patients with TAO reveals similar colocalization to cell membranes. Treatment of primary thyrocytes with recombinant human TSH results in rapid ERK phosphorylation which can be blocked by an IGF-1R-blocking mAb. Our findings suggest that IGF-1R might mediate some TSH-provoked signaling. Furthermore, they indicate that TSHR levels on orbital fibroblasts are considerably lower than those on thyrocytes and that this receptor associates with IGF-1R in situ and together may comprise a functional complex in thyroid and orbital tissue.


Subject(s)
Autoantigens/physiology , Graves Disease/immunology , Graves Disease/metabolism , Receptor, IGF Type 1/physiology , Receptors, Thyrotropin/physiology , Cell Proliferation , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Graves Disease/pathology , Humans , Insulin-Like Growth Factor I/physiology , Orbit/immunology , Orbit/metabolism , Orbit/pathology , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/isolation & purification , Receptors, Thyrotropin/isolation & purification , Thyroid Gland/immunology , Thyroid Gland/metabolism , Thyroid Gland/pathology
4.
Endocrinology ; 143(4): 1182-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11897670

ABSTRACT

TSH receptor (TSHR) DNA vaccination induces high TSHR antibody levels in BALB/c mice housed in a conventional facility. However, under pathogen-free conditions, we observed a Th1 cellular response to TSHR antigen characterized by interferon-gamma (IFN gamma) production. In the present study we investigated the effect on TSHR DNA vaccination of diverting the cytokine milieu away from Th1 using 1) IFN gamma knockout BALB/c mice, and 2) wild-type mice covaccinated with DNA for the TSHR and for IFN gamma/receptor-Fc protein that prevents IFN gamma from binding to its receptor. Neither approach enhanced TSHR antibody levels, although splenocyte IFN gamma production in response to TSHR antigen was absent (IFN gamma knockouts) or reduced (IFN gamma receptor-Fc). Moreover, production of IL-2, another Th1 cytokine, but not Th2 cytokines, indicated that neither strategy overcame the Th1 bias of im DNA vaccination. Importantly, splenocyte production of IFN gamma and IL-2 provides a sensitive detection system for TSHR-specific T cells. Unexpectedly, higher TSHR antibody levels developed in rare mice. High titer animals had TSHR-specific responses of both Th2 and Th1 types, whereas low titer animals had Th1-restricted TSHR responses. The heterogeneity of responses induced by TSHR DNA vaccination in mice may provide insight into the titers and IgG subclasses of spontaneous autoantibodies in humans.


Subject(s)
Interferon-gamma/genetics , Interferon-gamma/physiology , Th1 Cells/metabolism , Animals , Cytokines/biosynthesis , DNA/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoglobulin G/immunology , Immunoglobulins, Thyroid-Stimulating , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Interferon/immunology , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/isolation & purification , Receptors, Thyrotropin/metabolism , Recombinant Fusion Proteins/immunology , Thyrotropin/metabolism , Vaccination , Interferon gamma Receptor
5.
Protein Expr Purif ; 23(3): 369-73, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11722172

ABSTRACT

For the first time soluble, full-length, recombinant, human thyroid-stimulating hormone (TSH) receptor (TSHR) has been expressed in a prokaryotic system. The full-length TSHR cDNA, obtained from normal human thyroid, was cloned into a pQE-9 vector, sequenced, and confirmed to be identical to the published sequence, to be full length, and to be in frame. Expression of the receptor was as a fusion protein with a hexahistidine tail at the amino terminal, in an Escherichia coli expression system. Approximately 2.5 mg of protein per liter of bacterial culture was recovered from the cell homogenate, after a single passage through a nickel-nitrilotriacetic acid resin column. An estimated 60% increase in purity of a band of expected size, 87 kDa, was observed upon gel electrophoresis and staining with Coomassie blue, after the single purification step. Immunoreactivity of the 87-kDa protein with Graves' sera was confirmed by Western blotting.


Subject(s)
Escherichia coli/genetics , Receptors, Thyrotropin/genetics , Amino Acid Sequence , Autoantibodies/immunology , Blotting, Western , Chromatography, Affinity , Gene Expression , Graves Disease/immunology , Histidine/metabolism , Humans , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/isolation & purification , Receptors, Thyrotropin/metabolism , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Solubility
6.
J Immunol ; 164(12): 6158-65, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843665

ABSTRACT

Thyroid-stimulating hormone (TSH), a central neuroendocrine mediator of the hypothalamus-pituitary-thyroid axis, has been shown to affect various aspects of immunological development and function. To gain a better understanding of TSH involvement within the mammalian immune system, the expression and distribution of the TSH receptor (TSHr) has been studied by immunoprecipitation and by flow cytometric analyses. Using highly enriched populations of B cells, T cells, and dendritic cells, trace amounts of TSHr were precipitated from B cells and T cells, whereas high levels of TSHr were precipitated from the dendritic cell fraction. Flow cytometric analyses of TSHr expression on splenic and lymph node T cells revealed a major difference between those tissues in that only 2-3% of splenic T cells were TSHr+, whereas 10-20% of CD4+8- and CD4-8+ lymph node T cells expressed the TSHr, which was exclusively associated with CD45RB(high) cells and was not expressed during or after activation. The TSHr was not present on cells of the immune system during fetal or neonatal life. However, recombinant TSHbeta was found to significantly enhance the phagocytic activity of dendritic cells from adult animals and to selectively augment IL-1beta and IL-12 cytokine responses of dendritic cells following phagocytic activation. These findings identify a novel immune-endocrine bridge associated with professional APCs and naive T cells.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Leukocyte Common Antigens/biosynthesis , Lymph Nodes/metabolism , Lymphocyte Activation , Receptors, Thyrotropin/biosynthesis , T-Lymphocyte Subsets/metabolism , Thyrotropin/physiology , Animals , Cell Differentiation/immunology , Cells, Cultured , Cytokines/metabolism , Immunologic Memory , Interleukin-1/metabolism , Interleukin-12/metabolism , Interphase/immunology , Lymph Nodes/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Phagocytosis/drug effects , Precipitin Tests , Receptors, Thyrotropin/isolation & purification , Spleen/cytology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , Thyrotropin/pharmacology
7.
Clin Exp Immunol ; 118(2): 205-12, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10540180

ABSTRACT

Graves' disease (GD) is characterized by the presence of autoantibodies against the TSH-receptor (TSH-R) which are pathogenic and, upon binding to the receptor, trigger intracellular signal transduction. The autoantibodies are oligoclonal and as they are responsible for disease activity, their characterization would lead to a better understanding of the development of GD. Attempts to isolate anti-TSH-R antibodies from patients have proved to be difficult due to the exceedingly low serum levels due to rarity of these B cells, together with difficulties in obtaining purified TSH-R capable of interacting with patients autoantibodies. We employed phage antibody display technology and performed selection with a previously characterized semisynthetic antibody library on the purified extracellular ectodomain of the TSH-R. We report the isolation of six different anti-TSH-R monoclonal phage antibodies (moPhabs) from this library. All the moPhabs recognized TSH-R and its recombinant fragments by Western blotting, but failed to recognize the native TSH-R by flow cytometry. Consequently, the moPhabs did not lead to TSH-R activation. As these were the first moPhabs to TSH-R, they were analysed in terms of nucleotide and amino acid sequence and epitope specificity on the receptor. The moPhabs used immunoglobulin VH1 and VH3 germ line genes, all associated with Vlambda3 genes. Interestingly, the CDR3 regions of all moPhabs were remarkably similar, though not identical. In light of the common CDR3 usage, the epitopes recognized on TSH-R appeared to be restricted to amino acids residues 405-411 and 357-364. In summary, our results show that semisynthetic libraries may be limited in isolating human monoclonal antibodies that resemble pathogenic antithyrotropin receptor autoantibodies present in patients with GD. It is likely that until preparations of purified TSH-R that can be recognized by patients autoantibodies become available, similar to the recently described glycosylphosphatidylinositol (GPI) anchored TSH-R ectodomain, monoclonal antibodies from phage antibody display to TSH-R will be limited for isolating the rare, pathogenic antibodies of GD.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Autoantibodies/isolation & purification , Graves Disease/immunology , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/isolation & purification , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Autoantibodies/genetics , Autoantibodies/metabolism , B-Lymphocytes/metabolism , Bacteriophages/genetics , Bacteriophages/immunology , Binding Sites, Antibody , CHO Cells , Cricetinae , Epitope Mapping , Humans , Immunoglobulins, Thyroid-Stimulating , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/isolation & purification , Protein Binding , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Sequence Analysis, DNA
8.
J Clin Endocrinol Metab ; 84(10): 3797-802, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10523032

ABSTRACT

Detergent-solubilized porcine TSH receptor (TSHR) has been labeled with 125I using a monoclonal antibody to the C-terminal domain of the receptor. The ability of sera containing TSHR autoantibody to immunoprecipitate the labeled receptor was then investigated. Sera negative for TSHR autoantibody (as judged by assays based on inhibition of labeled TSH binding to detergent-solubilized porcine TSHR) immunoprecipitated about 4% of the labeled receptor, whereas sera with high levels of receptor autoantibody immunoprecipitated more than 25% of the labeled receptor. The ability to immunoprecipitate labeled TSHR correlated well with ability of the sera to inhibit labeled TSH binding to the receptor (r = 0.92; n = 63), and this is consistent with TSHR autoantibodies in these samples being directed principally to a region of the receptor closely related to the TSH binding site. Preincubation of labeled TSHR with unlabeled TSH before reaction with test sera inhibited the immunoprecipitation reaction, providing further evidence for a close relationship between the TSHR autoantibody binding site(s) and the TSH binding site. This was the case whether the sera had TSH agonist (i.e., thyroid stimulating) or TSH antagonist (i.e., blocking) activities, thus, providing no clear evidence for different regions of the TSHR being involved in forming the binding site(s) for TSHR autoantibodies with stimulating and with blocking activities. The ability of TSHR autoantibodies to stimulate cyclic AMP production in isolated porcine thyroid cells was compared with their ability to immunoprecipitate labeled porcine TSHR. A significant correlation was observed (r = 0.58; n = 50; P < 0.001) and the correlation was improved when stimulation of cyclic AMP production was compared with inhibition of labeled TSH binding to porcine TSHR (r = 0.76). Overall, our results indicate that TSHR autoantibodies bind principally to a region on the TSHR closely related to the TSH binding site, and this seems to be the case whether the autoantibodies act as TSH agonists or antagonists.


Subject(s)
Autoantibodies/immunology , Receptors, Thyrotropin/immunology , Animals , Antibodies, Monoclonal/immunology , Blotting, Western , CHO Cells , Cricetinae , Graves Disease/blood , Humans , Iodine Radioisotopes , Precipitin Tests , Receptors, Thyrotropin/blood , Receptors, Thyrotropin/isolation & purification , Receptors, Thyrotropin/metabolism , Recombinant Proteins , Reference Values , Swine , Thyroiditis, Autoimmune/blood , Thyrotropin/metabolism
9.
J Clin Endocrinol Metab ; 84(4): 1391-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10199784

ABSTRACT

We produced substantial amount of the extracellular domain of the human TSH receptor (TSHRE) that has a tag of six histidines at C-terminus as a soluble form in the human cell line HeLa using a vaccinia virus system. By sequential nickel-chelating and lentil lectin column chromatography, TSHRE was purified to about 70% purity, with the recovery of around 0.1-0.2 mg TSHRE/L culture (5 x 10(8) cells/liter culture). The purified TSHRE interacted with TSH as well as Graves' autoantibodies to TSHR. However, the affinity of TSHRE for TSH was much lower than that of intact TSHR. The IC50 value for inhibition of TSH-dependent cAMP synthesis by TSHRE was about 10(-8) mol/L. Most importantly, the purified TSHRE inhibited the binding of the IgG of Graves' patients to thyroid membrane. About 1 microg/mL (2 x 10(-8) mol/L) TSHRE neutralized most of the autoantibody activity of patients' sera tested in the TSH binding inhibitory immunoglobulin (TBII) assay. Moreover, this protein neutralized thyroid stimulatory antibody-induced cAMP synthesis with an IC50 of 1 x 10(-9) mol/L and completely at 0.5-1 microg/mL (1-2 x 10(-8) mol/L). In the simple enzyme-linked immunosorbent assay, the TSHRE immobilized on the wells coated with nickel showed significantly higher binding with the IgGs from Graves' patients than in those from normal individuals. This autoantibody-reactive TSHRE will be useful for further studies on the diagnosis, pathogenesis, and the development of therapy of Graves' disease.


Subject(s)
Autoantibodies/immunology , Receptors, Thyrotropin/isolation & purification , Enzyme-Linked Immunosorbent Assay , Graves Disease/immunology , HeLa Cells , Humans , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/metabolism , Recombinant Proteins/isolation & purification , Thyrotropin/metabolism , Vaccinia virus/genetics
10.
J Clin Endocrinol Metab ; 84(2): 702-10, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10022441

ABSTRACT

We used the secreted TSH receptor (TSHR) ectodomain variant TSHR-289 (truncated at amino acid residue 289 with a 6-histidine tail) to investigate properties of TSHR autoantibodies in Graves' disease. Sequential concanavalin A and Ni-chelate chromatography extracted milligram quantities of TSHR-289 (approximately 20-40% purity) from the culture medium. Nanogram quantities of this material neutralized the TSH binding inhibitory activity in all 15 Graves' sera studied. We generated a mouse monoclonal antibody (mAb), 3BD10, to partially purified TSHR-289. Screening of a TSHR complementary DNA fragment expression library localized the 3BD10 epitope to 27 amino acids at the N-terminus of the TSHR, a cysteine-rich segment predicted to be highly conformational. 3BD10 preferentially recognized native, as opposed to reduced and denatured, TSHR-289, but did not interact with the TSH holoreceptor on the cell surface. Moreover, mAb 3BD10 could extract from culture medium TSHR-289 nonreactive with autoantibodies, but not the lesser amount (approximately 25%) of TSHR-289 molecules capable of neutralizing autoantibodies. Although the active form of TSHR-289 in culture medium was stable at ambient temperature, stability was reduced at 37 C, explaining the mixture of active and inactive molecules in medium harvested from cell cultures. In conclusion, studies involving a TSHR ectodomain variant indicate the exquisite conformational requirements of TSHR autoantibodies. Even under "native" conditions, only a minority of molecules in highly potent TSHR-289 preparations neutralize patients' autoantibodies. Therefore, Graves' disease is likely to be caused by even lower concentrations of autoantibodies than previously thought. Finally, reciprocally exclusive binding to TSHR-289 by human autoantibodies and a mouse mAb with a defined epitope suggests that the extreme N-terminus of the TSHR is important for autoantibody recognition.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens/chemistry , Autoantibodies/blood , Peptide Fragments/immunology , Protein Conformation , Receptors, Thyrotropin/immunology , Animals , Antibody Specificity , Antigens/immunology , Chromatography/methods , Culture Media, Conditioned , Culture Techniques , Drug Stability , Epitopes/immunology , Humans , Immunosorbent Techniques , Mice , Mice, Inbred BALB C , Protein Sorting Signals/immunology , Receptors, Thyrotropin/isolation & purification , Temperature
11.
J Endocrinol ; 160(2): 239-45, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9924193

ABSTRACT

We report a method for the purification and radioactive labeling of human TSH receptor (TSHR). The method is based on the construction of a fusion TSHR (TSHR-Xa-BIO) which consists of the N-terminal 725 amino acids of human TSHR linked to the 4-amino acid Xa protease cleavage site and the 87-amino acid C-terminal domain of the biotin carboxyl carrier protein subunit of Escherichia coli acetyl-CoA carboxylase (the C-terminal domain directs the efficient posttranslational biotinylation of the protein). TSHR-Xa-BIO was produced in HeLa cells using recombinant vaccinia virus. The expressed protein was fully functional and was biotinylated with an efficiency of about 90%. Streptavidin-agarose-immobilized TSHR-Xa-BIO was labeled with 125I using the chloramine T oxidation procedure and specifically eluted from the solid phase after cleavage with protease Xa. Isolated native radiochemically pure 125I-labeled TSHR specifically interacted with pathological autoantibodies in the sera of patients with Graves' disease, and thus could be useful for the detection of these autoantibodies by immunoprecipitation analysis.


Subject(s)
Autoantibodies/blood , Autoantigens/isolation & purification , Graves Disease/diagnosis , Receptors, Thyrotropin/isolation & purification , Biotinylation , Gene Expression , Genetic Vectors , HeLa Cells , Humans , Iodine Radioisotopes , Isotope Labeling , Precipitin Tests , Receptors, Thyrotropin/genetics , Vaccinia virus/genetics
12.
Thyroid ; 5(2): 101-5, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7647568

ABSTRACT

Attempts to purify the thyroid-stimulating hormone receptor (TSHR) have been complicated by its susceptibility to proteolysis and its low level of expression in thyrocytes and many transfected cells. Controversy exists over its size and structure. Multiple, single-polypeptide forms of the TSHR (230, 187, and 95-100 kDa) have been recently identified in immunoblots of crude plasma membranes prepared from COS-7 cells transfected with rat or human cDNA, but the relationship of these receptor species to the TSHR in human thyroid tissue has been heretofore unknown. We have developed a technique for immunoprecipitation of the TSHR which employed IgG purified from a polyclonal rabbit antiserum to TSHR residues 352-366. We have used immunoprecipitation to isolate the previously characterized 95-100 kDa TSH-holoreceptor, 187 kDa intermediate, and 230 kDa precursor forms of the TSHR from plasma membrane prepared from transfected COS-7 cells and human thyroid tissue. The presence of all three forms was not altered by the addition of reducing agent to the sample buffer, demonstrating the single polypeptide structure of the TSHR. This is, to our knowledge, the first report of the purification from transfected COS-7 cells of these TSHR species identified previously only in immunoblots of crude plasma membrane, and the first report of the identification by any means of these TSHR forms in human thyroid tissue. The isolation of TSHR from human thyroid tissue requires confirmation by direct means, but promises to make the receptor available in a soluble form for studies of its structure and function.


Subject(s)
Receptors, Thyrotropin/isolation & purification , Thyroid Gland/chemistry , Cell Line , Cell Membrane/chemistry , Humans , Immunoblotting , Isomerism , Molecular Weight , Precipitin Tests , Transfection
13.
J Biol Chem ; 270(4): 1543-9, 1995 Jan 27.
Article in English | MEDLINE | ID: mdl-7829482

ABSTRACT

Conventional baculovirus vectors that utilize the very late polyhedrin promoter have not proved successful for expressing a thyrotropin (TSH) receptor capable of ligand and Graves' disease autoantibody binding comparable to the receptor produced in mammalian cells. Because of the clinical importance of high level expression of this protein, we reassessed the baculovirus system using a new transfer vector (pAcMP3) containing the late basic protein promoter, which functions earlier than the classical polyhedrin promoter. Maximal synthesis of the [35S]methionine-labeled TSH receptor extracellular domain, affinity-purified using a 6-histidine tag, occurred earlier (1 day after insect cell infection) than with a vector (pVL1393) containing the polyhedrin promoter. The pAcMP3-derived TSH receptor extracellular domain was larger (approximately 68 kDa) than the pVL1393-derived protein (approximately 63 kDa). Only the 68-kDa product was secreted, albeit in trace amounts detectable only by precursor labeling. Enzymatic deglycosylation reduced both 68- and 63-kDa cellular proteins to approximately 54 kDa, indicating that the pAcMP3 vector generated a protein with greater carbohydrate content. However, despite its greater degree of glycosylation, most of the 68-kDa protein remained within the cell, almost entirely in the particulate fraction. Remarkably, the trace amounts of 68-kDa receptor protein affinity-purified from the soluble cytosolic fraction of infected insect cells completely neutralized TSH receptor autoantibodies in patients' sera and partly inhibited TSH binding. In conclusion, a baculovirus vector with a promoter active earlier than the conventional polyhedrin promoter generates a more glycosylated and functional TSH receptor extracellular domain protein, albeit at low levels. These data carry important implications for the expression by baculovirus vectors of functional, highly glycosylated proteins.


Subject(s)
Promoter Regions, Genetic , Receptors, Thyrotropin/biosynthesis , Transfection , Animals , Baculoviridae , Base Sequence , CHO Cells , Cricetinae , DNA Primers , Genetic Vectors , Glycosylation , Humans , Kinetics , Methionine/metabolism , Molecular Sequence Data , Mutagenesis, Insertional , Occlusion Body Matrix Proteins , Polymerase Chain Reaction , Receptors, Thyrotropin/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Restriction Mapping , Sequence Deletion , Spodoptera , Time Factors , Viral Proteins/genetics , Viral Structural Proteins
14.
Cell Immunol ; 158(2): 329-41, 1994 Oct 15.
Article in English | MEDLINE | ID: mdl-7923386

ABSTRACT

The development of autoimmunity was investigated after repeated immunizations with human thyrotropin receptor (hTSH-R) of five congenic strains of female and male mice. After each immunization, free T3 levels and antibodies to hTSH-R and to six peptides of the hTSH-R were assayed. Our results showed that H-2s and H-2q female mice developed features of autoimmunity such as antibody responses to hTSH-R and to hTSH-R peptides, transient variations in the levels of free T3 thyroid hormone, and lymphocytic infiltrations in their thyroid glands. Concerning the antibody responses to hTSH-R peptides, we found that peptide P1 (352-366) contained a major B cell epitope. Furthermore, strain-specific B cell epitope was exemplified by peptide 92 (12-30) and two male- and female-specific B cell epitopes were located in peptides 91 (32-46) and 93 (316-330), respectively. These features appeared rather related to hyperthyroidism.


Subject(s)
Autoantibodies/biosynthesis , Receptors, Thyrotropin/immunology , Amino Acid Sequence , Animals , Binding, Competitive/immunology , Female , Humans , Major Histocompatibility Complex/immunology , Male , Mice , Mice, Inbred Strains , Molecular Sequence Data , Peptide Fragments/immunology , Receptors, Thyrotropin/isolation & purification , Thyroglobulin/immunology , Thyroid Gland/pathology , Triiodothyronine/blood
15.
Eur J Biochem ; 222(2): 711-9, 1994 Jun 01.
Article in English | MEDLINE | ID: mdl-8020509

ABSTRACT

The complementary DNA for human thyroid-stimulating hormone (TSH) receptor encodes a single protein with a deduced molecular mass of 84.5 kDa. This protein is cleaved during its maturation in the human thyroid since the receptor protein has been shown to be composed of two subunits (alpha subunit of approximately 53 kDa and beta subunit of approximately 38 kDa) held together by disulfide bridges [Loosfelt, H., Pichon, C., Jolivet, A., Misrahi, M., Caillou, B., Jamous, M., Vannier, B. & Miligrom, E. (1992) Proc. Natl Acad. Sci. USA 89, 3765-3769]. A similar processing occurs in an L cell line permanently expressing the human TSH receptor. The processing is however incomplete, resulting in a permanent accumulation of a 95-kDa high-mannose precursor which is present only in trace amounts in the thyroid. Pulse-chase experiments show the successive appearance in the L cells of two precursors: initially the approximately 95-kDa high-mannose glycoprotein followed by a approximately 120-kDa species containing mature oligosaccharides. This latter precursor is then processed into the alpha and beta subunits. In primary cultures of human thyrocytes precursors of similar size are detected. Spodoptera frugiperda insect cells (Sf9 and Sf21) infected with a recombinant baculovirus encoding the human TSH receptor synthesize a monomeric protein of about 90 kDa soluble only in denaturing conditions. Comparison with the product of in vitro transcription-translation experiments (approximately 80 kDa), suggests that it may be incompletely or improperly glycosylated. The TSH receptor expressed in these cells is unable to bind the hormone. Immunoelectron microscopy studies show that in human thyrocytes most of the receptor is present on the cell surface; in L cells the receptor is detected on the cell surface, as well as in the endoplasmic reticulum and in the Golgi apparatus (this intracellular pool of receptor molecules probably corresponding to the high-mannose precursor); in insect cells nearly all the receptor molecules are trapped in the endoplasmic reticulum. These differences in receptor distribution are concordant with the differences observed for receptor processing.


Subject(s)
Eukaryotic Cells/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Receptors, Thyrotropin/biosynthesis , Thyroid Gland/metabolism , Animals , Autoradiography , Baculoviridae/genetics , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cysteine/metabolism , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Humans , Immunoblotting , L Cells , Methionine/metabolism , Mice , Microscopy, Immunoelectron , Moths , Receptors, Thyrotropin/isolation & purification , Receptors, Thyrotropin/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sulfur Radioisotopes/metabolism , Transfection
16.
Endocrinology ; 134(2): 549-54, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8299554

ABSTRACT

We produced large quantities of the extracellular domain of the human TSH receptor (ETSHR) using the baculovirus expression system. Insect cells containing the ETSHR protein were sequentially extracted using lysis, nuclease, and high salt buffers to enrich for recombinant protein. The ETSHR protein was purified to homogeneity on a C4 reverse phase semipreparative column using HPLC. The recombinant protein was identified as ETSHR by immunoreactivity with antibodies prepared against TSHR-derived synthetic peptides. The identity of the ETSHR was further confirmed by amino acid compositional analyses, which agreed with the amino acid composition predicted from reported cDNA sequence analyses. Protein sequence analyses confirmed that the first 26 amino acids of the N-terminal region and the C-terminal amino acid were identical to the predicted amino acid sequence. The purified ETSHR was refolded in the presence of 1.5 M guanidine-HCl and 1 mM each of cystine and cysteine. [125I] TSH bound to the refolded ETSHR in vitro in a dose-dependent manner and was specifically blocked by unlabeled TSH, but not by LH or FSH. It was notable that a membrane requirement was not essential for TSH to bind to ETSHR.


Subject(s)
Receptors, Thyrotropin/metabolism , Thyrotropin/metabolism , Amino Acid Sequence , Animals , Antibodies , Baculoviridae , Binding Sites , Binding, Competitive , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Cysteine/pharmacology , Cystine/pharmacology , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Follicle Stimulating Hormone/pharmacology , Guanidine , Guanidines/pharmacology , Humans , Immunoblotting , Luteinizing Hormone/pharmacology , Molecular Sequence Data , Molecular Weight , Protein Folding , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Thyrotropin/pharmacology , Transfection
17.
J Mol Endocrinol ; 10(2): 127-42, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8484862

ABSTRACT

Since the cloning of the TSH receptor (TSH-R), the target autoantigen of Graves' disease, the receptor has been expressed in a variety of eukaryotic cells to obtain a functional molecule. Despite this success, the levels of receptor expression have been marginally higher than the extremely low levels found in thyroid cells, preventing any progress on the purification of the molecule. In this study, the large extracellular region of the TSH-R, without the membrane spanning segments, has been expressed in insect cells using recombinant baculovirus to generate substantial quantities of the receptor protein. A monoclonal antibody previously generated to a bacterial TSH-R fusion protein was used to characterize and monitor the expression of the truncated receptor in insect cells. Two polypeptides of 63 and 49 kDa were recognized as the components of the truncated recombinant receptor. The 63 kDa protein was shown to be the glycosylated form of the smaller, 49 kDa, component. Expression in different insect cell lines showed that an increase in expression of approximately tenfold was apparent in High Five cells when compared with Sf21 cells. Very small quantities of the truncated receptor were secreted by the three insect cell lines examined, with the majority of the molecule being retained within the cells. Immunoaffinity purification of milligram quantities of the truncated receptor was achieved using the monoclonal antibody. The availability of the purified TSH-R has allowed the establishment of an enzyme-linked immunosorbent assay to measure autoantibodies in the sera of patients with Graves' disease. Although the truncated receptor interacts with autoantibodies, our results show that it does not bind TSH and differs in this respect from other glycoprotein hormone receptors.


Subject(s)
Graves Disease/metabolism , Receptors, Thyrotropin/metabolism , Animals , Antibodies, Monoclonal/immunology , Autoantibodies/blood , Baculoviridae/genetics , Base Sequence , Cell Line , Chromatography, Affinity , DNA , Enzyme-Linked Immunosorbent Assay , Glycosylation , Graves Disease/immunology , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Moths , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/isolation & purification , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
18.
Biochem Biophys Res Commun ; 186(2): 1074-80, 1992 Jul 31.
Article in English | MEDLINE | ID: mdl-1497642

ABSTRACT

An antiserum raised against an alignment of amino acid-(32-56), termed TSHRP-1, in the extracellular domain of human thyrotropin (TSH) receptor was used to identify the TSH receptor-like substance in plasma of Graves' disease. The dilution curve of plasma TSHRP-1-like immunoreactivity was observed in a manner parallel to the standard synthetic peptide curve in radioimmunoassay, and its molecular weight estimated approximately 60 kDa. The amounts of TSHRP-1-like immunoreactivity were significantly higher in Graves' plasma than those in plasma of normal and hypothyroid patients due to Hashimoto's thyroiditis. The present results indicate that human peripheral blood possesses a soluble form of the extracellular domain of TSH receptor which may contribute to the pathophysiology of Graves' disease.


Subject(s)
Graves Disease/blood , Receptors, Thyrotropin/analysis , Biomarkers/blood , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Graves Disease/immunology , Humans , Immune Sera , Immunoglobulin G , Kinetics , Molecular Weight , Peptides/chemical synthesis , Peptides/immunology , Radioimmunoassay/methods , Receptors, Thyrotropin/isolation & purification , Receptors, Thyrotropin/metabolism
19.
Mol Endocrinol ; 5(11): 1607-12, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1779967

ABSTRACT

The most widely held model for the human TSH receptor is of holoreceptor of 80 kDa with two subunits of approximately 50 and 30 kDa linked by disulfide bridges, with the former subunit containing the major hormone-binding site. We reexamined this model by covalently cross-linking radiolabeled TSH to the recombinant human TSH receptor stably expressed in Chinese hamster ovary (CHO) cells. When cross-linking was performed after the preparation of CHO membranes, analysis of hormone-receptor complexes under reducing and nonreducing conditions provided results supporting the two-subunit TSH receptor model. In contrast, however, cross-linking of TSH to the TSH receptor in intact CHO cells before membrane preparation revealed, even under reducing conditions, an approximately 100-kDa receptor as well as an approximately 54-kDa hormone-binding subunit. The approximately 100-kDa holoreceptor size is consistent with the size of the TSH receptor, as predicted from its derived amino acid sequence. The proportions of the approximately 100-kDa TSH receptor and the 54-kDa fragment varied in different experiments, suggesting the occurrence of proteolytic cleavage. Cross-linking of radiolabeled TSH to intact cells expressing a mutant TSH receptor (TSHR-D1) lacking amino acids 317-366 localized the proteolytic cleavage site to just up-stream of amino acid residue 317. In summary, the present data obtained by cross-linking TSH to recombinant human TSH receptors in intact cells provides evidence that the receptor exists in vivo as an approximately 100-kDa glycoprotein with a single polypeptide chain with intramolecular disulfide bridges.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Receptors, Thyrotropin/metabolism , Thyrotropin/metabolism , Animals , Autoradiography , CHO Cells , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cricetinae , Cross-Linking Reagents , Electrophoresis, Polyacrylamide Gel , Humans , Iodine Radioisotopes , Models, Structural , Protein Conformation , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/isolation & purification , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
20.
Proc Natl Acad Sci U S A ; 88(5): 1991-5, 1991 Mar 01.
Article in English | MEDLINE | ID: mdl-2000404

ABSTRACT

Gangliosides were extracted from purified human and porcine thyrotropin (TSH) receptors (TSH-R) and were detected by probing with an 125I-labeled sialic acid-specific lectin, Limax flavus agglutinin. Gangliosides copurified with human and porcine TSH-R migrated between monosialoganglioside GM1 and disialoganglioside GD1a. Ceramide glycanase digestion of the purified human TSH-R-associated glycolipid confirmed its ganglioside nature. It was resistant to Vibrio cholerae sialidase, which digests all gangliosides except GM1, but was sensitive to Arthrobacter ureafaciens sialidase, which digests all gangliosides including GM1. These findings indicate that the human TSH-R contains ganglioside that belongs to the galactosyl(beta 1----3)-N-acetylgalactosaminyl (beta 1----4)-[N-acetylneuraminyl(alpha 2----3)]galactosyl(beta 1----4) glucosyl(beta 1----1)ceramide (GM1) family. Its intimate association with receptor protein implies a key role for ganglioside in the structure and function of the TSH-R.


Subject(s)
Gangliosides/analysis , Receptors, Thyrotropin/chemistry , Adrenal Glands/metabolism , Amino Acid Sequence , Animals , Carbohydrate Sequence , Cell Membrane/metabolism , Dogs , Gangliosides/isolation & purification , Goiter, Nodular/metabolism , Graves Disease/metabolism , Humans , Molecular Sequence Data , Oligosaccharides/chemistry , Oligosaccharides/isolation & purification , Receptors, Thyrotropin/isolation & purification , Receptors, Thyrotropin/metabolism , Sequence Homology, Nucleic Acid , Swine , Thyroid Gland/metabolism , Thyrotropin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL