Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Clin Exp Immunol ; 139(2): 216-24, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15654820

RESUMO

Thyroid autoantigens require internalization and processing by antigen-presenting cells to induce immune responses. Besides pinocytosis, antigen uptake can be receptor-mediated. The mannose receptor (ManR) has a cysteine rich domain (CR) and eight carbohydrate recognition domains (CRD) that bind glycosylated proteins. The TSH receptor (TSHR), thyroid peroxidase (TPO) and thyroglobulin (Tg) are glycoproteins. To investigate a role for the ManR in thyroid autoimmunity, we tested the interaction between these autoantigens and chimeric ManRs. Plasmids encoding the CR-domain linked to IgG-Fc (CR-Fc) and CDR domains 4-7 linked to IgG-Fc (CDR4-7-Fc) were expressed and purified with Protein A. Enzyme-linked immunosorbent assay (ELISA) plates were coated with human thyroid autoantigens and CR-Fc or CRD4-7-Fc binding detected with peroxidase-conjugated anti-IgG-Fc. CRD4-7-Fc binding was highest for the TSHR, followed by Tg and was minimal for TPO. CR-Fc bound to Tg but not to TSHR or TPO. The interaction between the TSHR and CRD-Fc was calcium-dependent; it was inhibited by mannose (not galactose), and required a glycosylated TSHR A-subunit. Moreover, precomplexing the TSHR A-subunit with CRD-Fc (but not CR-Fc), or adding mannose (but not galactose), decreased in vitro responses of splenocytes from TSHR-immunized mice. Our data indicate that the ManR may participate in autoimmune responses to Tg and the TSHR but not to TPO. Most important, ManR binding of heavily glycosylated TSHR A-subunits suggests a mechanism by which the minute amounts of A-subunit protein shed from the thyroid may be captured by antigen-presenting cells located in the gland or in draining lymph nodes.


Assuntos
Autoantígenos/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo , Receptores de Superfície Celular/metabolismo , Linfócitos T/imunologia , Hormônios Tireóideos/imunologia , Tireoidite Autoimune/imunologia , Animais , Apresentação de Antígeno , Carboidratos/análise , Linhagem Celular , Feminino , Humanos , Iodeto Peroxidase/química , Iodeto Peroxidase/imunologia , Receptor de Manose , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica , Receptores da Tireotropina/química , Receptores da Tireotropina/imunologia , Tireoglobulina/química , Tireoglobulina/imunologia
2.
J Endocrinol Invest ; 25(8): 696-701, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12240901

RESUMO

Constitutively activating germline mutations of the TSH receptor (TSH-R) are considered the cause of hereditary non-autoimmune hyperthyroidism. In this study, 10 members (8 affected and 2 unaffected) of an Italian family with hereditary non-autoimmune hyperthyroidism were investigated for the presence of mutations in the TSH-R gene. The clinical features of the disease were also analyzed. PCR-amplified fragments of the TSH-R gene were obtained from genomic DNA extracted from peripheral blood leukocytes of each family member and analyzed by direct nucleotide sequencing and restriction analysis. An identical germline TSH-R mutation was detected in all the patients with hyperthyroidism but in none of the unaffected family members. The mutation was heterozygotic and determined the substitution of valine for methionine (codon 463; ATG-->GTG) in the second transmembrane domain of the TSH-R. When expressed in chinese hamster ovary (CHO) cells, the Val463 mutant TSH-R induced constitutive activation of the TSH receptor. Analysis of the clinical features of our family and those of other families with hereditary non-autoimmune hyperthyroidism, including one with the same Val463 mutation, revealed wide variability in the phenotypical expression of the disease. Our findings indicate that an activating germline mutation in the TSH-R gene plays a key role in hereditary non-autoimmune hyperthyroidism although the onset of clinical manifestations and the evolution of the disease seem to depend heavily on other factors, thus far unidentified. The absence of a clear correlation between mutant genotypes and phenotypic expression of the disease currently limits the prognostic value of genetic testing in families with hereditary non-autoimmune hyperthyroidism.


Assuntos
Mutação em Linhagem Germinativa , Hipertireoidismo/genética , Receptores da Tireotropina/genética , Adulto , Animais , Células CHO , Cricetinae , Feminino , Mutação em Linhagem Germinativa/genética , Mutação em Linhagem Germinativa/fisiologia , Humanos , Masculino , Metionina , Pessoa de Meia-Idade , Linhagem , Fenótipo , Valina
3.
Thyroid ; 12(2): 115-20, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11916280

RESUMO

Recently, many exceptions have been reported that undermine the concept that the epitopes for thyroid-stimulating autoantibodies (TSAb) and thyrotropin-blocking autoantibodies (TBAb) lie within the N-terminal and C-terminal portions of the thyrotropin receptor (TSHR) ectodomain, respectively. Here we have used a new approach to examine the issue by attempting to neutralize autoantibodies with the purified, N-terminal 289 amino acids of the TSHR ectodomain (TSHR-289), essentially the A subunit. Immunoglobulin G (IgG) with TSAb activity from 10 patients with Graves' disease was preincubated with or without purified active or inactive TSHR-289. Active, but not inactive, TSHR-289 completely neutralized TSAb activity in all sera. We then tested the ability of active TSHR-289 to neutralize TBAb activity in two rare hypothyroid patients with pure TBAb activity lacking agonist activity. IgG from both patients was extremely potent in the TBAb assay. Unlike with TSAb, preincubation of the IgG with a large excess of active TSHR-289 (20 microg/mL) revealed a remarkable divergence. TBAb activity from one patient was totally neutralized whereas in the other patient TBAb activity was totally unaffected. In conclusion, using a new approach, we confirm that the major portion of the TSAb epitope in the 418 amino acid ectodomain lies upstream of residue 289 (within the A subunit). In contrast, TBAb that cause hyperthyroidism can be more heterogeneous, with epitopes that lie largely upstream (A subunit) or downstream (B subunit) of residue 289.


Assuntos
Autoanticorpos/imunologia , Hipotireoidismo/imunologia , Receptores da Tireotropina/imunologia , Autoanticorpos/análise , Autoanticorpos/farmacologia , Epitopos , Feminino , Doença de Graves/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulinas Estimuladoras da Glândula Tireoide/análise , Imunoglobulinas Estimuladoras da Glândula Tireoide/imunologia , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , Fragmentos de Peptídeos/imunologia , Receptores da Tireotropina/genética , Glândula Tireoide/efeitos dos fármacos , Tireotropina/antagonistas & inibidores , Tireotropina/imunologia
4.
Endocrinology ; 142(8): 3530-6, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11459799

RESUMO

Two approaches have been developed to induce TSH receptor antibodies in mice with properties resembling those in Graves' disease, the Shimojo model of injecting live fibroblasts coexpressing the TSH receptor and major histocompatibility complex antigen Class II, and TSH receptor-DNA vaccination. Thyroid-stimulating antibodies appear to occur less commonly after DNA vaccination, but there has been no direct comparison of these models. We performed a three-way comparison of 1) AKR/N and 2) BALB/c mice vaccinated with TSH receptor-DNA and 3) AKR/N mice injected with fibroblasts expressing the TSH receptor and the major histocompatibility complex antigen class II of AKR/N mice. TSH receptor-DNA vaccinated mice had low or undetectable levels of TSH receptor antibodies determined by ELISA or flow cytometry. Nonspecific binding precluded comparisons with sera from Shimojo mice by these assays. TSH binding inhibition and thyroid-stimulating antibody were undetectable in TSH receptor-DNA vaccinated mice. In Shimojo mice, TSH binding inhibition was positive in approximately 60%, and thyroid-stimulating antibodies were positive in hyperthyroid animals. Unlike the negative antibody data, splenocytes from TSH receptor-vaccinated (but not Shimojo) mice proliferated and produced the Th1 cytokine interferon-gamma in response to TSH receptor antigen. In conclusion, DNA vaccination is less effective at inducing TSH receptor antibodies than the Shimojo approach, but it permits the future characterization of TSH receptor-specific T cells generated without adjuvant.


Assuntos
Interferon gama/biossíntese , Receptores da Tireotropina/genética , Receptores da Tireotropina/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Vacinação , Vacinas de DNA/imunologia , Animais , Anticorpos/análise , Formação de Anticorpos/fisiologia , Antígenos/farmacologia , Divisão Celular/efeitos dos fármacos , Humanos , Interleucina-4/biossíntese , Linfócitos/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Baço/metabolismo , Glândula Tireoide/citologia
5.
Thyroid ; 11(2): 111-4, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11288979

RESUMO

Cleavage of thyrotropin receptors (TSHR) on the cell surface into disulfide-linked A and B subunits involves deletion of an intervening region that corresponds approximately to a 50 amino acid "insertion" in the TSHR relative to the noncleaving luteinizing hormone/choriogonadotropin receptor (LH/CGR). The location of this insertion is imprecise because of the relatively low homology between the two receptors in this region. We tested the hypothesis that the TSHR 50 amino acid insertion was further downstream than we previously concluded, a possibility that would relocate the crucial LH/CGR glycan at N291 relative to the position of the TSHR insertion, and that would mitigate against the 50 amino acid insertion playing a role in TSHR intramolecular cleavage. Thus, we transferred the LH/CGR glycan at amino acid 291 from downstream (N367) to upstream of the 50 amino acid insertion (N317) in the TSHR, leaving this insertion intact. TSHR cleavage persisted. Moreover, deletion of amino acid residues 320-366 in addition to the upstream N291 substitution (ALN317-319NET) also did not prevent cleavage. On the other hand, deletion of three contiguous downstream residues (GQE367-369) in the TSHR 50 amino acid insertion abolished receptor cleavage into subunits. In summary, the present data are consistent with our previous location of the TSHR 50 amino acid insertion and, therefore, do not undermine evidence for the involvement of this insertion in TSHR cleavage. In addition, the data regarding TSHR residues GQE367-369 (far downstream of cleavage site 1) support the controversial possibility of a secondary cleavage site downstream of the insertion.


Assuntos
Elementos de DNA Transponíveis , Receptores da Tireotropina/genética , Sequência de Aminoácidos/genética , Animais , Células CHO , Cricetinae , Reagentes de Ligações Cruzadas/farmacologia , Deleção de Genes , Dados de Sequência Molecular , Mutação/genética
6.
Eur J Biochem ; 268(8): 2261-9, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11298743

RESUMO

To gain insight into the thyrotropin hormone (TSH) receptor (TSHR) cleavage, we sought to convert the noncleaving luteinizing hormone (LH) receptor (LHR) into a cleaved, two-subunit molecule. For this purpose, we generated a series of LHR mutants and chimeric LH-TSH receptors. Cleavage of mature, ligand binding receptors on the cell surface was determined by covalent 125I-labeled hCG crosslinking to intact, stably transfected mammalian cells. We first targeted a cluster of three N-linked glycans in the LHR (N295, N303, N317) in a region corresponding to the primary TSHR cleavage site, which has only one N-linked glycan. Elimination by mutagenesis of the most strategic N-linked glycan (LHR-N317Q) generated only a trace amount of LHR cleavage. Removal of the other N-linked glycans had no additive effect. A much greater degree of cleavage ( approximately 50%) was evident in a chimeric LH-TSHR in which the juxtamembrane segment of the LHR (domain E; amino acids 317-367) was replaced with the corresponding domain of the TSHR (residues 363-418). Similarly cleaving LHR were created using a much smaller component within this region, namely LHR-NET317-319 replaced with TSHR-GQE367-369, or by substitution of the same three amino-acid residues with AAA (LHR-NET317-319AAA). In summary, our data alter current concepts regarding TSHR cleavage by suggesting limited (not absent) amino-acid specificity in a region important for TSHR cleavage (GQE367-369). The data also support the concept of a separate and distinct downstream cleavage site 2 in the TSHR.


Assuntos
Receptores do LH/metabolismo , Receptores da Tireotropina/metabolismo , Sequência de Aminoácidos , Animais , Ligação Competitiva , Células CHO , Códon , Cricetinae , Reagentes de Ligações Cruzadas , DNA Complementar/metabolismo , Densitometria , Citometria de Fluxo , Humanos , Ligantes , Dados de Sequência Molecular , Mutagênese , Mutação , Plasmídeos/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Homologia de Sequência de Aminoácidos , Transfecção
7.
J Biol Chem ; 276(18): 14767-72, 2001 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-11278376

RESUMO

We observed amino acid homology between the cysteine-rich N terminus of the thyrotropin receptor (TSHR) ectodomain and epidermal growth factor-like repeats in the laminin gamma1 chain. Thyroid-stimulating autoantibodies (TSAb), the cause of Graves' disease, interact with this region of the TSHR in a manner critically dependent on antigen conformation. We studied the role of the cluster of four cysteine (Cys) residues in this region of the TSHR on the functional response to TSAb in Graves' patients' sera. As a benchmark we also studied TSH binding and action. Removal in various permutations of the four cysteines at TSHR positions 24, 29, 31, and 41 (signal peptide residues are 1-21) revealed Cys(41) to be the key residue for receptor expression. Forced pairing of Cys(41) with any one of the three upstream Cys residues was necessary for trafficking to the cell surface of a TSHR with high affinity TSH binding similar to the wild-type receptor. However, for a full biological response to TSAb, forced pairing of Cys(41) with Cys(29) or with Cys(31), but not with Cys(24), retained functional activity comparable with the wild-type TSHR. These data suggest that an N-terminal disulfide-bonded loop between Cys(41) and Cys(29) or its close neighbor Cys(31) comprises, in part, the highly conformational epitope for TSAb at the critical N terminus of the TSHR. Amino acid homology, as well as cysteine pairing similar to the laminin gamma1 chain epidermal growth factor-like repeat 11, suggests conformational similarity between the two molecules and raises the possibility of molecular mimicry in the pathogenesis of Graves' disease.


Assuntos
Autoanticorpos/imunologia , Dissulfetos/imunologia , Fator de Crescimento Epidérmico/química , Doença de Graves/imunologia , Laminina/química , Receptores da Tireotropina/química , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
8.
J Clin Endocrinol Metab ; 86(3): 1287-93, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238522

RESUMO

A secreted recombinant TSH receptor (TSHR) ectodomain variant (TSHR-289) neutralizes TSHR autoantibodies in Graves' disease, but is heterogeneous in containing both immunologically active and inactive molecules and is also unstable. We have now purified each form of TSHR-289 using sequential affinity chromatography with a mouse mAb (3BD10) specific for the inactive form, and a mAb to C-terminal His residues that recognizes both forms. The immunological difference between active and inactive TSHR-289 was unrelated to primary amino acid sequence or carbohydrate content and was, therefore, attributable to its folded state. The epitopes for Graves' autoantibodies and 3BD10 overlap, and both are destroyed by denaturation. Therefore, reciprocal binding by autoantibodies and 3BD10 to conformational determinants involving the same TSHR segment suggests a prion-like shift between two folded states of the molecule. Despite purification, immunologically active TSHR-289 remained labile, as determined by loss of autoantibody, and gain of 3BD10, recognition. However, using chemical chaperones we have, for the first time, been able to stabilize purified TSHR antigen in immunologically intact form. In summary, purification of immunologically active and stable antigen in milligram quantities provides a powerful tool for future diagnostic and therapeutic studies in Graves' disease.


Assuntos
Autoanticorpos/imunologia , Doença de Graves/imunologia , Conformação Proteica , Receptores da Tireotropina/química , Receptores da Tireotropina/imunologia , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Células CHO , Carboidratos/análise , Cromatografia de Afinidade , Cricetinae , Estabilidade de Medicamentos , Epitopos/química , Epitopos/imunologia , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide , Metilaminas/farmacologia , Camundongos , Prolina/farmacologia , Receptores da Tireotropina/genética , Proteínas Recombinantes/química , Relação Estrutura-Atividade , Tireotropina/metabolismo
9.
Endocrinology ; 141(10): 3573-7, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11014210

RESUMO

Some TSH receptors (TSHR) on the cell surface cleave into A and B subunits. Cleavage at upstream Site 1 is followed by the proteolytic excision of an intervening C peptide region terminating at a downstream Site 2. Although present evidence suggests that Site 1 lies between amino acid residues 303 and 317, the mechanism and exact amino acid(s) involved in cleavage are unknown. Previous amino acid substitutions at Site 1 failed to abrogate cleavage. We, therefore, performed deletion mutations within this region. Cleavage of cell surface TSHR, detected by 125I-TSH cross-linking to intact cells, was not prevented by deletion of four individual segments within the Site 1 cleavage region (delta305-308, delta309-312, delta313-316, delta317-320). However, deletion of the entire region (delta305-320) reduced the extent of cleavage and shifted the cleavage site upstream of the glycan at amino acid residue N302. Elimination of this glycan (N302Q substitution) reversed the effect of deleting amino acid residues 305-320 on TSHR cleavage, suggesting that reduced cleavage at the new, upstream cleavage site was caused by steric hindrance by the glycan at N302. In summary, deletion, as opposed to mutagenesis, of the TSHR cleavage Site 1 region produces a spatial shift in TSHR cleavage Site 1 from downstream to upstream of the glycan at N302. These observations provide strong evidence that TSHR cleavage at this site does not occur at a particular amino acid motif and suggests that cleavage involves a "molecular ruler" mechanism involving cleavage at a fixed distance from a protease attachment site.


Assuntos
Deleção de Genes , Receptores da Tireotropina/genética , Motivos de Aminoácidos , Sequência de Aminoácidos/genética , Animais , Células CHO , Cricetinae , Dados de Sequência Molecular , Mutação/genética , Polissacarídeos/genética
10.
Thyroid ; 9(11): 1057-61, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10595452

RESUMO

There is, at present, no assay in clinical use for the direct assay of autoantibody binding to the thyrotropin receptor (TSHR). We now describe a direct thyrotropin receptor autoantibody binding assay (DTAb) using a secreted form of the TSHR ectodomain (TSHR-289) without the need for antigen purification. The assay compensates for the low TSHR autoantibody concentration in serum by capturing a relatively large amount of patient immunoglobulin G (IgG) on high-capacity beads, a reversal of standard methods that typically first immobilize antigen. TSHR-289 captured by Graves' IgG was detected in a colorimetric reaction using a biotinylated murine monoclonal antibody to the poly-histidine tail engineered into the antigen. By this approach, sera from 11 normal individuals provided a mean optical density (OD) value of 0.20 +/- 0.08 SD (range 0.06-0.33). Of 38 sera from unselected patients with a history of Graves' disease (untreated and treated), 29 (76%) generated OD values > 0.37 (2 SD above the mean for the normal sera), the highest being OD 1.38. Surprisingly, 3 of 13 (23%) sera from TPO autoantibody-positive patients with Hashimoto's thyroiditis also provided values > 2 SD above the normal sera. The extent of direct autoantibody binding to the TSHR correlated closely with the thyrotropin binding inhibition (TBI) values (r = 0.881; p < 0.001). One serum was clearly positive in only the direct binding assay and another in only the TBI assay. The data obtained with the direct binding assay correlated less well with the thyroid-stimulating antibody (TSAb) assay (r = 0.582; p < 0.001). In summary, we describe a new direct DTAb assay that correlates more closely with the TBI than with the TSI assays. Future studies in a large series of clinically defined patients will be needed to evaluate the clinical utility of the DTAb assay.


Assuntos
Autoanticorpos/sangue , Receptores da Tireotropina/imunologia , Humanos
11.
J Biol Chem ; 274(48): 33979-84, 1999 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-10567361

RESUMO

We studied cell surface thyrotropin receptor (TSHR) by biotinylating proteins on the surface of metabolically labeled, intact cells. In addition to TSHR cleaved into A and B subunits, mature single-chain receptors with complex carbohydrate were also present on the cell surface. A low A/B subunit ratio indicated partial shedding of extracellular A subunits from transmembrane B subunits. TSHR cleavage at upstream site 1 (within amino acid residues 305-316) would generate a B subunit of 51-52 kDa. However, only smaller B subunits (40-46 kDa) were detected, corresponding to N termini from residues approximately 370 (site 2) extending downstream to the region of B subunit insertion into the plasma membrane. The intervening C peptide region between sites 1 and 2 could not be purified from TSHR epitope-tagged (c-myc) within this region. However, the small proportion of B subunits recovered with a c-myc antibody were larger (45-52 kDa) than the majority of B subunits recovered with a C-terminal antibody. In conclusion, our study provides the first characterization of cell surface TSHR including their A and B subunits. Single-chain, mature TSHR do exist on the cell surface. The C peptide lost during intramolecular cleavage disintegrates rapidly following cleavage at upstream site 1 of the single-chain TSHR into A and B subunits. N-terminal disintegration of the B subunit pauses at site 2, but then progresses downstream to the vicinity of the plasma membrane, revealing a novel mechanism for A subunit shedding.


Assuntos
Membrana Celular/metabolismo , Receptores da Tireotropina/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Sítios de Ligação , Células CHO , Cricetinae , Meios de Cultivo Condicionados/química , Expressão Gênica , Glicosilação , Humanos , Hidrólise , Camundongos , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/isolamento & purificação , Fragmentos de Peptídeos/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores da Tireotropina/química , Receptores da Tireotropina/genética
12.
Endocrinology ; 140(10): 4516-20, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10499506

RESUMO

We examined the relationship between TSH receptor (TSHR) cleavage into two subunits and ligand-independent, constitutive activity characteristic of this receptor. Because of homology to the thrombin receptor-tethered ligand, we focused initially on a region in the vicinity of the second, downstream cleavage site of the TSHR ectodomain. We introduced into the wild-type TSHR three mutations. One mutation, TSHR(GQE(367-369)NET) prevents cleavage at site 2. The other two mutations, ELK(369-371)T-Y (TSHR-E1a2) and NPQE(372-375)SAIF (TSHR-E1b), introduce major changes into the potential tethered ligand. Basal, steady state intracellular cAMP levels in cloned, stably transfected Chinese hamster ovary cells were expressed as a function of the number of receptors (cAMP/receptor). None of these three mutations decreased ligand-independent constitutive activity, thereby excluding the tethered ligand hypothesis as well as a requirement for cleavage at site 2 in this process. Turning to the more upstream site 1 in the TSHR ectodomain, we examined a receptor (TSHR-delta50AA) with deletion of a unique 50-amino acid insertion (residues 317-366) that appears to be involved in cleavage at this site. Constitutive cAMP production was similar to that of the wild-type TSHR. Finally, we studied a TSHR mutant that cleaves at neither site 1 (deletion of residues 317-366) nor site 2 (GQE(367-369)NET substitution) and, therefore, does not cleave into A and B subunits. Again, the basal, constitutive level of cAMP production was similar to that of the wild-type TSHR. In summary, contrary to the prevailing hypothesis based on several lines of evidence, TSHR cleavage into subunits is not associated with constitutive, ligand-independent activity.


Assuntos
Fragmentos de Peptídeos/fisiologia , Receptores da Tireotropina/química , Receptores da Tireotropina/fisiologia , Sequência de Aminoácidos/genética , Animais , Células CHO , Cricetinae , AMP Cíclico/biossíntese , Membranas Intracelulares/metabolismo , Dados de Sequência Molecular , Mutação/fisiologia , Receptores da Tireotropina/genética , Tireotropina/farmacologia
13.
Mol Cell Endocrinol ; 150(1-2): 113-9, 1999 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-10411305

RESUMO

The TSH receptor (TSHR) sheds its A subunit, particularly when cells are cultured in serum-poor medium. This shed A subunit is reported to be smaller than the cell-associated receptor because of the loss of glycan without change in its polypeptide core. Contrary to previous deductions, we now find that the 'small' shed A subunit has lost a glycan moiety because of the proteolytic clipping of a small C-terminal fragment containing an Asn-linked glycan. Moreover, this lost peptide fragment contains cysteine residues likely involved in A subunit linkage to the membrane-associated B subunit. Progressive lowering of the serum concentration in culture medium accentuates the process. Therefore, 'small' A subunit shedding does not appear related to a physiological mechanism involving disulfide bond reduction. On the other hand, we detected, for the first time, shedding of a lesser amount of normal-sized, in addition to small, A subunits, especially by cells cultured in standard serum concentrations.


Assuntos
Receptores da Tireotropina/química , Receptores da Tireotropina/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Meios de Cultura Livres de Soro , Humanos , Dados de Sequência Molecular , Conformação Proteica , Receptores da Tireotropina/genética , Transdução de Sinais
14.
J Clin Endocrinol Metab ; 84(2): 702-10, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10022441

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos/química , Autoanticorpos/sangue , Fragmentos de Peptídeos/imunologia , Conformação Proteica , Receptores da Tireotropina/imunologia , Animais , Especificidade de Anticorpos , Antígenos/imunologia , Cromatografia/métodos , Meios de Cultivo Condicionados , Técnicas de Cultura , Estabilidade de Medicamentos , Epitopos/imunologia , Humanos , Técnicas de Imunoadsorção , Camundongos , Camundongos Endogâmicos BALB C , Sinais Direcionadores de Proteínas/imunologia , Receptores da Tireotropina/isolamento & purificação , Temperatura
15.
J Biol Chem ; 274(4): 2093-6, 1999 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-9890969

RESUMO

Among the glycoprotein hormone receptors, only the thyrotropin receptor (TSHR) cleaves (at two sites) into disulfide-linked A and B subunits. A 50-amino acid insertion unique to the TSHR ectodomain (residues 317-366) plays no role in ligand binding or signal transduction, but its deletion abrogates cleavage at Site 1, closely upstream of the insertion. We sought to define the region within the 50-amino acid tract involved in TSHR cleavage at Site 1. Mutation of small segments within this region previously failed to prevent cleavage at Site 1. We, therefore, divided the 50-amino acid insertion into quartiles and deleted each one individually (TSHR residues 317-327, 328-338, 339-350, and 351-362). As determined by covalent cross-linking of 125I-TSH to intact cells expressing the mutant receptors, none of these deletions prevented TSHR cleavage at Site 1. Neither did larger deletions of quartiles 1 + 2, 2 + 3, and 3 + 4. However, qualitative differences in the extent of receptor cleavage suggested that quartiles 1 and 4 were playing a greater role in cleavage at Site 1 than were the middle two quartiles. In support of this hypothesis, deletion of these two discontinuous segments almost completely eliminated TSHR cleavage at Site 1. In conclusion, intramolecular cleavage at Site 1 requires the presence of the N-terminal and C-terminal quartiles of the 50-amino acid insertion unique to the TSHR. Taken together with previous observations, our data suggest that this tract may provide a discontinuous binding site for a protease that clips the TSHR at Site 1.


Assuntos
Aminoácidos/química , Receptores da Tireotropina/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Hidrólise , Dados de Sequência Molecular , Mutação , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Deleção de Sequência
17.
J Biol Chem ; 273(4): 1959-63, 1998 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-9442031

RESUMO

Thyrotropin (TSH) receptor (TSHR) A and B subunits are formed by intramolecular cleavage of the single chain receptor at two separate sites. The region involved in cleavage at Site 2 has been identified, but previous mutagenesis studies failed to identify Site 1. We now report fortuitous observations on the effect of trypsin on the TSHR that localizes a small region harboring Site 1. Thus, as detected by immunoblotting and by 125I-TSH cross-linking to TSHR expressed on the surface of intact CHO cells, trypsin clipped a small polypeptide fragment bearing a glycan moiety from the C terminus of the A subunit. Based on the TSHR primary structure, this small fragment (1-2 kDa) contains Asn-302. This information, together with estimation of the size of the deglycosylated A subunit relative to a series of C-terminal truncated TSHR ectodomain variants, places cleavage Site 1 in the vicinity of, or closely upstream to, residue 317. Remarkably, mutagenesis of every amino acid residue between residues 298-316 (present study) and 317-362 (previous data) did not prevent cleavage at Site 1. However, cleavage at this site was abrogated by deletion of a 50-amino acid segment (residues 317-366) unique to the TSHR in the glycoprotein hormone receptor family. In summary, these data provide novel insight into TSHR intramolecular cleavage. Cleavage at Site 1 does not depend on a specific amino acid motif and differs from cleavage at Site 2 by involvement of a mechanism requiring the presence of the enigmatic TSHR 50-amino acid "insertion."


Assuntos
Receptores da Tireotropina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Células CHO , Cricetinae , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidase/metabolismo , Dados de Sequência Molecular , Peso Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Tripsina/metabolismo
18.
J Biol Chem ; 272(45): 28296-300, 1997 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-9353284

RESUMO

The thyrotropin receptor (TSHR) exists in two forms (single polypeptide and two subunits), whereas the lutropin/chorionic gonadotropin receptor (LH/CGR) is a single chain. Recent data suggest that the TSHR cleaves at two sites. We mutagenized selected chimeric TSH-LH/CGR to localize the cleavage sites in the TSHR. All 23 receptors mutated in the estimated vicinity of the upstream site cleaved into two subunits as determined by 125I-TSH cross-linking to intact cells. In contrast, in a series of mutations homologous to the noncleaving LH/CGR, the downstream TSHR cleavage site localized to three amino acids (GQE367-369). Remarkably, group substitution of these residues, but not substitution of individual residues, abolished cleavage. Moreover, the mutation that prevented cleavage (GQE367-369NET) transposed a motif (NET291-293) that is glycosylated in the LH/CGR. TSHR cleavage or noncleavage after substitution of GQE367-369 with other triplets (AAA, NQE, and NQT) was consistent with a role for N-linked glycosylation at this site. In summary, our data (i) support the concept that the TSHR cleaves at two sites, (ii) relate TSHR residues GQE367-369 to cleavage at the second, downstream site, and (iii) suggest that cleavage or noncleavage at site two is related to N-linked glycosylation. These findings provide new insight into the evolutionary divergence of two closely related receptors.


Assuntos
Receptores do LH/química , Receptores do LH/metabolismo , Receptores da Tireotropina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Bovinos , Glutamina/metabolismo , Glicina/metabolismo , Glicosilação , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Receptores do LH/genética , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
19.
J Biol Chem ; 272(30): 18959-65, 1997 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-9228077

RESUMO

Previous attempts to generate autoantibody-reactive, secreted thyrotropin receptor (TSHR) ectodomain in mammalian cells have failed because of retention within the cell of material with immature carbohydrate. We have overcome this difficulty by performing progressive carboxyl-terminal truncations of the human TSHR ectodomain (418 amino acid residues including signal peptide). Three ectodomain variants (TSHR-261, TSHR-289, and TSHR-309) were truncated at residues 261, 289, and 309, respectively. Unlike the full ectodomain, ectodomain variants were secreted with an efficiency inversely proportional to their size. Secreted ectodomain variants contained approximately 20 kDa of complex carbohydrate. TSHR-261 was chosen for further study because it was secreted very efficiently and neutralized autoantibodies in Graves' patients' sera. This ectodomain variant was partially purified using sequential lectin and nickel-chelate chromatography, permitting the first direct visualization and quantitation of the mammalian TSHR. Most important, very small (nanogram) quantities of this material neutralized 70-100% of TSHR autoantibody activity in all 18 Graves' sera studied. In summary, carboxyl-terminal truncation of the human TSHR ectodomain generates a secreted protein with complex carbohydrate that neutralizes autoantibodies in Graves' patients' sera. Antigenically active TSHR will be valuable for future studies on the diagnosis, pathogenesis, and immunotherapy of Graves' disease.


Assuntos
Autoanticorpos/imunologia , Glicoproteínas/imunologia , Doença de Graves/imunologia , Receptores da Tireotropina/imunologia , Adsorção , Animais , Células CHO , Cricetinae , Meios de Cultura , Glicoproteínas/genética , Glicoproteínas/metabolismo , Doença de Graves/fisiopatologia , Humanos , Lectinas/metabolismo , Modelos Moleculares , Testes de Neutralização , Fragmentos de Peptídeos/imunologia , Engenharia de Proteínas , Receptores da Tireotropina/genética
20.
J Clin Endocrinol Metab ; 82(7): 2129-34, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9215283

RESUMO

Seven years after the molecular cloning of the human TSH receptor (TSHR), the porcine TSHR remains in general use in the TSH binding inhibition (TBI) assay for autoantibodies to the TSHR. We compared porcine and recombinant human TSHR in two types of TBI assays: one using intact Chinese hamster ovary cells expressing the recombinant human TSHR on their surface, and the other using soluble receptors extracted from these cells with detergent. In the intact cell TBI assay, monolayers expressing large numbers of TSHR were less effective than cells expressing few receptors. These findings are consistent with the very low concentration of TSHR autoantibodies in serum. Binding of [125I]human TSH was about 5-fold lower than that of [125I]bovine TSH to the intact cells. Nevertheless, TBI values with the two ligands were similar for most sera. However, a few sera produced greater inhibition of human than of bovine TSH binding. In the solubilized human TSHR TBI assay, in contrast to the intact cell TBI assay, cells expressing very large number of TSHR were an excellent source for detergent extraction of soluble human TSHR, but only if the cells were extracted while still on the dish and not after scraping. A 10-cm diameter dish of cells provided TSHR for 100-200 replicate determinations when substituted for solubilized porcine TSHR in a commercial TBI kit. TBI values in serum from 30 individuals with suspected Graves' disease correlated closely when tested with solubilized human and porcine TSHR (r = 0.954; P < 0.001). However, 2 sera that were negative with the porcine TSHR were positive with the human TSHR. TBI and thyroid-stimulating activity in these sera correlated weakly regardless of whether the TBI used human or porcine TSHR. These findings open the way to a practical TBI assay using recombinant human TSHR.


Assuntos
Autoanticorpos/análise , Receptores da Tireotropina/imunologia , Tireotropina/metabolismo , Animais , Ligação Competitiva , Células CHO , Bovinos , Cricetinae , Humanos , Métodos , Receptores da Tireotropina/metabolismo , Proteínas Recombinantes/metabolismo , Suínos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...