Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; : fj201800211R, 2018 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-29799790

RESUMEN

Thyroid function is controlled by thyroid-stimulating hormone (TSH), which binds to its G protein-coupled receptor [thyroid-stimulating hormone receptor (TSHR)] on thyrocytes. TSHR can potentially couple to all G protein families, but it mainly activates the Gs- and Gq/11-mediated signaling cascades. To date, there is a knowledge gap concerning the role of the individual G protein cascades in thyroid pathophysiology. Here, we demonstrate that the thyrocyte-specific deletion of Gs-protein α subunit (Gαs) in adult mice [tamoxifen-inducible Gs protein α subunit deficient (iTGαsKO) mice] rapidly impairs thyrocyte function and leads to hypothyroidism. Consequently, iTGαsKO mice show reduced food intake and activity. However, body weight and the amount of white adipose tissue were decreased only in male iTGαsKO mice. Unexpectedly, hyperplastic follicles and papillary thyroid cancer-like tumor lesions with increased proliferation and slightly increased phospho-ERK1/2 staining were found in iTGαsKO mice at an older age. These tumors developed from nonrecombined thyrocytes still expressing Gαs in the presence of highly elevated serum TSH. In summary, we report that partial thyrocyte-specific Gαs deletion leads to hypothyroidism but also to tumor development in thyrocytes with remaining Gαs expression. Thus, these mice are a novel model to elucidate the pathophysiological consequences of hypothyroidism and TSHR/Gs/cAMP-mediated tumorigenesis.-Patyra, K., Jaeschke, H., Löf, C., Jännäri, M., Ruohonen, S. T., Undeutsch, H., Khalil, M., Kero, A., Poutanen, M., Toppari, J., Chen, M., Weinstein, L. S., Paschke, R., Kero, J. Partial thyrocyte-specific Gαs deficiency leads to rapid-onset hypothyroidism, hyperplasia, and papillary thyroid carcinoma-like lesions in mice.

2.
J Biol Chem ; 291(27): 14095-14108, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27129207

RESUMEN

The thyroid stimulating hormone receptor (TSHR) is a G protein-coupled receptor (GPCR) with a characteristic large extracellular domain (ECD). TSHR activation is initiated by binding of the hormone ligand TSH to the ECD. How the extracellular binding event triggers the conformational changes in the transmembrane domain (TMD) necessary for intracellular G protein activation is poorly understood. To gain insight in this process, the knowledge on the relative positioning of ECD and TMD and the conformation of the linker region at the interface of ECD and TMD are of particular importance. To generate a structural model for the TSHR we applied an integrated structural biology approach combining computational techniques with experimental data. Chemical cross-linking followed by mass spectrometry yielded 17 unique distance restraints within the ECD of the TSHR, its ligand TSH, and the hormone-receptor complex. These structural restraints generally confirm the expected binding mode of TSH to the ECD as well as the general fold of the domains and were used to guide homology modeling of the ECD. Functional characterization of TSHR mutants confirms the previously suggested close proximity of Ser-281 and Ile-486 within the TSHR. Rigidifying this contact permanently with a disulfide bridge disrupts ligand-induced receptor activation and indicates that rearrangement of the ECD/extracellular loop 1 (ECL1) interface is a critical step in receptor activation. The experimentally verified contact of Ser-281 (ECD) and Ile-486 (TMD) was subsequently utilized in docking homology models of the ECD and the TMD to create a full-length model of a glycoprotein hormone receptor.


Asunto(s)
Receptores de Tirotropina/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Glicosilación , Humanos , Espectrometría de Masas , Modelos Moleculares , Mutación , Proteolisis , Receptores de Tirotropina/química , Receptores de Tirotropina/genética , Resonancia por Plasmón de Superficie
3.
J Biol Chem ; 286(25): 22622-31, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21525003

RESUMEN

The extracellular region of the thyrotropin receptor (TSHR) can be subdivided into the leucine-rich repeat domain (LRRD) and the hinge region. Both the LRRD and the hinge region interact with thyrotropin (TSH) or autoantibodies. Structural data for the TSHR LRRD were previously determined by crystallization (amino acids Glu(30)-Thr(257), 10 repeats), but the structure of the hinge region is still undefined. Of note, the amino acid sequence (Trp(258)-Tyr(279)) following the crystallized LRRD comprises a pattern typical for leucine-rich repeats with conserved hydrophobic side chains stabilizing the repeat fold. Moreover, functional data for amino acids between the LRRD and the transmembrane domain were fragmentary. We therefore investigated systematically these TSHR regions by mutagenesis to reveal insights into their functional contribution and potential structural features. We found that mutations of conserved hydrophobic residues between Thr(257) and Tyr(279) cause TSHR misfold, which supports a structural fold of this peptide, probably as an additional leucine-rich repeat. Furthermore, we identified several new mutations of hydrophilic amino acids in the entire hinge region leading to partial TSHR inactivation, indicating that these positions are important for intramolecular signal transduction. In summary, we provide new information regarding the structural features and functionalities of extracellular TSHR regions. Based on these insights and in context with previous results, we suggest an extracellular activation mechanism that supports an intramolecular agonistic unit as a central switch for activating effects at the extracellular region toward the serpentine domain.


Asunto(s)
Espacio Extracelular/metabolismo , Receptores de Tirotropina/química , Receptores de Tirotropina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Células COS , Bovinos , Chlorocebus aethiops , Secuencia Conservada , AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Fosfatos de Inositol/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Tirotropina/genética , Tirotropina/metabolismo
4.
Thyroid ; 32(4): 459-471, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35044245

RESUMEN

Background: The human adrenal cortex undergoes several rapid remodeling steps during its lifetime. In rodents, similar remodeling occurs postnatally in the "X-zone" layer through unknown mechanisms. Furthermore, little is known regarding the impact of thyroid hormone (TH) on adrenal glands in humans. Methods: To investigate the impact of TH on adrenal pathophysiology, we created two genetic murine models mimicking human nonautoimmune hypothyroidism and hyperthyroidism. Moreover, we analyzed serum thyrotropin (TSH) and steroid hormone concentrations in patients diagnosed with congenital hypothyroidism and premature adrenarche (PA). Results: We found that TH receptor beta-mediated hypertrophy of the X-zone significantly elevated the adrenal weights of hyperthyroid women. In the hypothyroid model, the X-zone was poorly developed in both sexes. Moreover, large reciprocal changes in the expression levels of genes that regulate adrenal cortical function were observed with both models. Unexpectedly, up- and downregulation of several genes involved in catecholamine synthesis were detected in the adrenal glands of the hypothyroid and hyperthyroid models, respectively. Furthermore, TSH and adrenal steroid concentrations correlated positively in pediatric patients with congenital hypothyroidism and PA. Conclusions: Our results revealed that congenital hypothyroidism and hyperthyroidism functionally affect adrenal gland development and related steroidogenic activity, as well as the adrenal medulla.


Asunto(s)
Hipotiroidismo Congénito , Hipertiroidismo , Animales , Niño , Hipotiroidismo Congénito/genética , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Hormonas Tiroideas , Tirotropina
5.
Clin Endocrinol (Oxf) ; 73(6): 815-20, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20846293

RESUMEN

OBJECTIVE: Constitutively activating mutations (CAMs) of the TSHR are the major cause for nonautoimmune hyperthyroidism. Re-examination of constitutive activity previously determined in CHO cell lines recently demonstrated the caveats for the in vitro determination of constitutive TSHR activity, which leads to false positive conclusions regarding the molecular origin of hyperthyroidism or hot thyroid carcinomas. DESIGN: Mutations L677V and T620I identified in hot thyroid carcinomas were previously characterized in CHO and in 3T3-Vill cell lines, respectively, stably expressing the mutant without determination of TSHR expression. F666L identified in a patient with hot thyroid nodules, I691F in a family with nonautoimmune hyperthyroidism and F631I identified in a hot thyroid carcinoma were not characterized for their in vitro function. Therefore, we decided to (re)evaluate the in vitro function of these five TSHR variants by determination of cell surface expression, and intracellular cAMP and inositol phosphate levels and performed additionally linear regression analyses to determine basal activity independently from the mutant's cell surface expression in COS-7 and HEK(GT) cells. RESULTS AND CONCLUSIONS: Only one (F631I) of the five investigated TSHR variants displayed constitutive activity for G(α) s signalling and showed correlation with the clinical phenotype. The previous false classification of T620I and L677V as CAMs is most likely related to the fact that both mutations were characterized in cell lines stably expressing the mutated receptor construct without assessing the respective receptor number per cell. Other molecular aetiologies for the nonautoimmune hyperthyroidism and/or hot thyroid carcinomas in these three patients and one family should be elucidated.


Asunto(s)
Hipertiroidismo/genética , Receptores de Tirotropina/genética , Receptores de Tirotropina/metabolismo , Neoplasias de la Tiroides/genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , AMP Cíclico/metabolismo , Citometría de Flujo , Humanos , Mutagénesis Sitio-Dirigida , Mutación
6.
FASEB J ; 22(8): 2798-808, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18381815

RESUMEN

The mechanisms governing transition of the thyroid stimulating hormone (TSH) receptor (TSHR) from basal to active conformations are poorly understood. Considering that constitutively activating mutations (CAMs) and inactivating mutations in each of the extracellular loops (ECLs) trigger only partial TSHR activation or inactivation, respectively, we hypothesized that full signaling occurs via multiple extracellular signal propagation events. Therefore, individual CAMs in the extracellular region were combined to create double and triple mutants. In support of our hypothesis, combinations of mutants in the ECLs are in some cases additive, while in others they are even synergistic, with triple mutant I486A/I568V/V656F exhibiting a 70-fold increase in TSH-independent signaling. The proximity but likely different spatial orientation of the residues of activating and inactivating mutations in each ECL supports a dual functionality to facilitate signal induction and conduction, respectively. This is the first report for G-protein coupled receptors, suggesting that multiple and cooperative signal propagating events at all three ECLs are required for full receptor activation. Our findings provide new insights concerning molecular signal transmission from extracellular domains toward the transmembrane helix bundle of the glycoprotein hormone receptors.


Asunto(s)
Receptores de Tirotropina/química , Receptores de Tirotropina/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Células COS , Chlorocebus aethiops , AMP Cíclico/biosíntesis , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Estructura Terciaria de Proteína , Receptores de Tirotropina/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
7.
Thyroid ; 28(10): 1372-1386, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30132406

RESUMEN

BACKGROUND: Constitutively active thyrotropin receptor (TSHR) mutations are the most common etiology of non-autoimmune hyperthyroidism (NAH). Thus far, the functionality of these mutations has been tested in vitro, but the in vivo models are lacking. METHODS: To understand the pathophysiology of NAH, the patient-derived constitutively active TSHR D633H mutation was introduced into the murine Tshr by homologous recombination. RESULTS: In this model, both subclinical and overt hyperthyroidism was observed, depending on the age, sex, and genotype. Homozygous mice presented hyperthyroidism at two months of age, while heterozygous animals showed only suppressed thyrotropin. Interestingly, at six months of age, thyroid hormone concentrations in all mutant mice were analogous to wild-type mice, and they showed colloid goiter with flattened thyrocytes. Strikingly, at one year of age, nearly all homozygous mice presented large papillary thyroid carcinomas. Mechanistically, this papillary thyroid carcinoma phenotype was associated with an overactive thyroid and strongly increased stainings of proliferation-, pERK-, and NKX2-1 markers, but no mutations in the "hot-spot" areas of common oncogenes (Braf, Nras, and Kras) were found. CONCLUSIONS: This is the first study to reveal the dynamic age-, sex-, and genotype-dependent development of NAH. Furthermore, the study shows that a constitutively active TSHR can trigger a malignant transformation of thyrocytes.


Asunto(s)
Bocio/genética , Hipertiroidismo/genética , Receptores de Tirotropina/genética , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética , Animales , Bocio/patología , Hipertiroidismo/patología , Ratones , Mutación , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología
8.
J Med Chem ; 49(13): 3888-96, 2006 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-16789744

RESUMEN

The substituted thieno[2,3-d]pyrimidine 3 (Org 41841), a partial agonist for the luteinizing hormone/choriogonadotropin receptor (LHCGR) and the closely related thyroid-stimulating hormone receptor (TSHR), was fundamentally altered, and the resulting analogues were analyzed for their potencies, efficacies, and specificities at LHCGR and TSHR. Chemical modification of the parent compound combined with prior mutagenesis of TSHR provided compelling experimental evidence in support of computational models of 3 binding to TSHR and LHCGR within their transmembrane cores. Biochemical analysis of a specific modification to the chemical structure of 3 provides additional evidence of a H-bond between the ligand and a glutamate residue in transmembrane helix 3, which is conserved in both receptors. Several key interactions were surveyed to determine their respective biochemical roles in terms of both van der Waals dimensions and hydrogen bond capacity and the respective relationship to biological activity.


Asunto(s)
Gonadotropina Coriónica/química , Hormona Luteinizante/química , Modelos Moleculares , Pirimidinas/síntesis química , Receptores de Tirotropina/química , Tiofenos/síntesis química , Sitios de Unión , Gonadotropina Coriónica/agonistas , Enlace de Hidrógeno , Ligandos , Hormona Luteinizante/agonistas , Pirimidinas/química , Receptores de Tirotropina/agonistas , Relación Estructura-Actividad , Tiofenos/química
9.
Thyroid ; 26(9): 1215-24, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27373559

RESUMEN

BACKGROUND: Congenital hypothyroidism (CH) is defined as the lack of thyroid hormones at birth. Mutations in at least 15 different genes have been associated with this disease. While up to 20% of CH cases are hereditary, the majority of cases are sporadic with unknown etiology. Apart from a monogenic pattern of inheritance, multigenic mechanisms have been suggested to play a role in CH. The genetics of CH has not been studied in Finland so far. Therefore, multigenic sequencing of CH candidate genes was performed in a Finnish patient cohort with both familial and sporadic CH. METHODS: A targeted next-generation sequencing (NGS) panel, covering all exons of the major CH genes, was applied for 15 patients with sporadic and 11 index cases with familial CH. RESULTS: Among the familial cases, six pathogenic mutations were found in the TPO, PAX8, and TSHR genes. Furthermore, pathogenic NKX2.1 and TG mutations were identified from sporadic cases, together with likely pathogenic variants in the TG, NKX2.5, SLC26A4, and DUOX2 genes. All identified novel pathogenic mutations were confirmed by Sanger-sequencing and characterized in silico and/or in vitro. CONCLUSION: In summary, the CH panel provides an efficient, cost-effective, and multigenic screening tool for both known and novel CH gene mutations. Hence, it may be a useful method to identify accurately the genetic etiology for dyshormogenic, familial, or syndromic forms of CH.


Asunto(s)
Autoantígenos/genética , Hipotiroidismo Congénito/genética , Yoduro Peroxidasa/genética , Proteínas de Unión a Hierro/genética , Mutación , Factor de Transcripción PAX8/genética , Receptores de Tirotropina/genética , Estudios de Cohortes , Femenino , Finlandia , Pruebas Genéticas , Humanos , Recién Nacido , Masculino , Linaje
10.
J Clin Invest ; 126(9): 3383-8, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27500488

RESUMEN

Autonomous thyroid adenomas (ATAs) are a frequent cause of hyperthyroidism. Mutations in the genes encoding the TSH receptor (TSHR) or the Gs protein α subunit (GNAS) are found in approximately 70% of ATAs. The involvement of other genes and the pathogenesis of the remaining cases are presently unknown. Here, we performed whole-exome sequencing in 19 ATAs that were paired with normal DNA samples and identified a recurrent hot-spot mutation (c.1712A>G; p.Gln571Arg) in the enhancer of zeste homolog 1 (EZH1) gene, which codes for a catalytic subunit of the polycomb complex. Targeted screening in an independent cohort confirmed that this mutation occurs with high frequency (27%) in ATAs. EZH1 mutations were strongly associated with known (TSHR, GNAS) or presumed (adenylate cyclase 9 [ADCY9]) alterations in cAMP pathway genes. Furthermore, functional studies revealed that the p.Gln571Arg EZH1 mutation caused increased histone H3 trimethylation and increased proliferation of thyroid cells. In summary, this study revealed that a hot-spot mutation in EZH1 is the second most frequent genetic alteration in ATAs. The association between EZH1 and TSHR mutations suggests a 2-hit model for the pathogenesis of these tumors, whereby constitutive activation of the cAMP pathway and EZH1 mutations cooperate to induce the hyperproliferation of thyroid cells.


Asunto(s)
Mutación , Complejo Represivo Polycomb 2/genética , Neoplasias de la Tiroides/genética , Adulto , Anciano , Dominio Catalítico , Diferenciación Celular , Proliferación Celular , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Receptores de Tirotropina/genética , Programas Informáticos , Glándula Tiroides/patología
11.
Endocrinology ; 146(12): 5197-203, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16150909

RESUMEN

Previous reports on the follicle-stimulating hormone receptor and choriogonadotropic/LH receptor, which belong to the glycoprotein hormone receptor family, suggest that the extracellular loop (ECL) 3 could be a key domain for ligand binding and intramolecular receptor signaling. In contrast to ECLs 1 and 2 of glycoprotein hormone receptors, the ECL3 displays high sequence homology, particularly in the central portion of the loop. Therefore, we opted to identify amino acids with functional importance within ECL3 of the TSH receptor (TSHR). Single alanine substitutions of all residues in ECL3 were generated. Functional characterization revealed the importance of five amino acids in the central portion of ECL3 and K660 at the ECL3/transmembrane helix (TMH) 7 junction for TSHR signaling. Decrease of G(s) activation and loss of G(q) activation by substitutions of K660 demonstrates a role for this position for TSHR conformation and signal transduction. By molecular modeling, we predicted potential interaction partners of K660:E409 and D410 in the N terminus of TMH1 and D573 in the ECL2. Complementary double mutants did not reconstitute G(s)/G(q)-mediated signaling, suggesting that K660 is not directly involved in a structural unit between ECL3 and the N terminus of TMH1. These results support a TSHR model in which the side chain of K660 is orientated toward the backbone of ECL2. Moreover, our findings provide evidence that a hydrophobic cluster, comprising residues 652-656 of ECL3, strongly influences intramolecular signal transduction and G protein activation of the TSHR.


Asunto(s)
Interacciones Hidrofóbicas e Hidrofílicas , Receptores de Tirotropina/química , Receptores de Tirotropina/genética , Transducción de Señal/genética , Alanina , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Humanos , Lisina , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Conformación Proteica , Estructura Terciaria de Proteína
12.
PLoS One ; 9(10): e111570, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25340405

RESUMEN

Glycoprotein hormone receptors (GPHR) have a large extracellular domain (ECD) divided into the leucine rich repeat (LRR) domain for binding of the glycoprotein hormones and the hinge region (HinR), which connects the LRR domain with the transmembrane domain (TMD). Understanding of the activation mechanism of GPHRs is hindered by the unknown interaction of the ECD with the TMD and the structural changes upon ligand binding responsible for receptor activation. Recently, our group showed that the HinR of the thyrotropin receptor (TSHR) can be replaced by those of the follitropin (FSHR) and lutropin receptor (LHCGR) without effects on surface expression and hTSH signaling. However, differences in binding characteristics for bovine TSH at the various HinRs were obvious. To gain further insights into the interplay between LRR domain, HinR and TMD we generated chimeras between the TSHR and FSHR. Our results obtained by the determination of cell surface expression, ligand binding and G protein activation confirm the similar characteristics of GPHR HinRs but they also demonstrate an involvement of the HinR in ligand selectivity indicated by the observed promiscuity of some chimeras. While the TSHR HinR contributes to specific binding of TSH and its variants, no such contribution is observed for FSH and its analog TR4401 at the HinR of the FSHR. Furthermore, the charge distribution at the poorly characterized LRR domain/HinR transition affected ligand binding and signaling even though this area is not in direct contact with the ligand. In addition our results also demonstrate the importance of the TMD/HinR interface. Especially the combination of the TSHR HinR with the FSHR-TMD resulted in a loss of cell surface expression of the respective chimeras. In conclusion, the HinRs of GPHRs do not only share similar characteristics but also behave as ligand specific structural and functional entities.


Asunto(s)
Receptores de HFE/química , Receptores de Tirotropina/química , Animales , Unión Competitiva , Células COS , Bovinos , Membrana Celular/metabolismo , Separación Celular , Chlorocebus aethiops , Cristalografía por Rayos X , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Hormona Folículo Estimulante/química , Humanos , Ligandos , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Transducción de Señal , Tirotropina/química
13.
J Clin Endocrinol Metab ; 99(10): E2051-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24947036

RESUMEN

OBJECTIVE: New in vivo mutations in G protein-coupled receptors open opportunities for insights into the mechanism of receptor activation. Here we describe the molecular mechanism of constitutive TSH receptor (TSHR) activation in an Austrian family with three generations of familial nonautoimmune hyperthyroidism. PATIENTS: The index patient was diagnosed with hyperthyroidism during her first pregnancy. Her first two children were diagnosed with hyperthyroidism at the age of 11 and 10 years, respectively. TSH suppression was also observed in the third child at the age of 8 years, who has normal free T4 levels until now. TSH suppression in infancy was observed in the fourth child. The mother of the index patient was diagnosed with toxic multinodular goiter at the age of 36 years. METHODS: DNA was extracted from blood samples from the index patient, her mother, and her four children. Screening for TSHR mutations was performed by high-resolution melting assays and subsequent sequencing. Elucidation of the underlying mechanism of TSHR activation was carried out by generation and structural analysis of TSHR transmembrane homology models and verification of model predictions by functional characterization of receptor mutations. RESULTS AND CONCLUSIONS: A newly discovered TSHR mutation L665F in transmembrane helix 7 of the receptor was detected in six members of this family. Functional characterization of L665F revealed constitutive activation for the Gs pathway and thus represents the molecular cause for hyperthyroidism in this family. The constitutive activation is possibly linked to a steric clash introduced by the L665F mutation between transmembrane helices 1 and 7.


Asunto(s)
Bocio Nodular/genética , Hipertiroidismo/congénito , Complicaciones del Embarazo/genética , Receptores de Tirotropina/genética , Adulto , Austria , Secuencia de Bases , Salud de la Familia , Femenino , Humanos , Hipertiroidismo/genética , Linaje , Mutación Puntual , Embarazo , Receptores de Tirotropina/química , Estereoisomerismo
14.
Mol Cell Endocrinol ; 393(1-2): 39-45, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-24915144

RESUMEN

Hot thyroid nodules (HTNs) in children are rare. Their reported malignancy rate is higher than in adults. However molecular data are rare. We present clinical and molecular data for 33 consecutive (29 benign and 4 malignant) HTNs. 17/29 Benign HTNs (59%) harbored somatic TSHR mutations. The most commonly observed mutation was M453T (in 8/29 samples). T632I and D633Y mutations were each detected twice. All other TSHR mutations were each found in one sample, including the new A538T mutation. One NRAS mutation was detected in a benign HTN with a M453T mutation. A PAX8/PPARG rearrangement was found in one malignant HTN. A T632I mutation was detected in one hot papillary thyroid carcinoma. The percentage of TSHR mutation positive HTNs in children and adolescents is within the range observed in adults. Contrary to adults, the M453T mutation is the predominant TSHR mutation in HTNs of children and adolescents. The increased malignancy rate of HTNs of children does not appear to be associated with RAS, BRAF, PAX8/PPARG and RET/PTC mutations.


Asunto(s)
Mutación/genética , Nódulo Tiroideo/genética , Adolescente , Animales , Células COS , Niño , Preescolar , Chlorocebus aethiops , Femenino , Humanos , Masculino , Nódulo Tiroideo/clasificación
15.
Endocrinology ; 152(10): 3986-96, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21846801

RESUMEN

The hinge region (HinR) is a variable structure of glycoprotein hormone receptors. Its amino acid composition and length is different for glycoprotein hormone receptors and connects the ligand binding domain with the serpentine domain. The role of the HinR of the receptors for TSH, follitropin (FSH), and LH/choriogonadotropin (LHCG) in receptor and signaling specificity is unknown. To investigate the role of the HinR for ligand binding, signal generation, and for the transmission of the signal towards the serpentine domain, we replaced the HinR of the TSH receptor (TSHR) by those of LHCG receptor and FSH receptor and introduced constitutively activating mutations and one mutation deficient for bovine (b)TSH binding in these chimeras. Functional characterization of the TSHR variants was carried out after transient transfection of COS-7 cells by determination of the cell surface expression, ligand binding, and recombinant human (rh)TSH or bTSH activation of second messengers. We show that the HinR of the TSHR stabilizes hormone binding regarding ligand affinity and retention time of the bound ligand as determined by dissociation experiments. Introduction of a constitutively activating extracellular loop mutation in these constructs lead to partially restored binding patterns. These findings indicate that the HinR-extracellular loop interface is besides signaling also important for bTSH binding. Furthermore, data for G protein signaling reveal that the activity of bTSH, but not of rhTSH, depends on the TSHR HinR, which was indicated by a significant right shift in the dose-response curves for G(s) and G(q) activation for TSHR chimeras harboring the LHCG receptor and FSH receptor HinR, respectively. Moreover, we identified different G protein signaling profiles for bTSH and rhTSH, which cannot be explained by the characterized HinR. For future studies regarding structure and function of the TSHR, it will be necessary to characterize TSHR variants with both or more ligands.


Asunto(s)
Receptores de Tirotropina/química , Receptores de Tirotropina/fisiología , Tirotropina/metabolismo , Animales , Unión Competitiva , Células COS , Bovinos , Chlorocebus aethiops , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/fisiología , Humanos , Ligandos , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología
16.
Endocrinology ; 152(8): 3268-78, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21628383

RESUMEN

In which ways the binding of the thyroid stimulating hormone to the extracellular domain of its receptor leads to activation of the thyroid-stimulating hormone receptor (TSHR) is currently only incompletely understood. It is known that TSH binding to the TSHR depends on the interaction with the leucine-rich repeat and sulfation at Y385 of the hinge region. Recently it was also shown that electrostatic interactions between positive charges of bovine (b) TSH and the residues E297, E303, and D382 of the hinge region contribute to hormone-TSHR binding. After the identification of these first TSH binding sites in the hinge region, it was apparent that multiple positions in this region remained to be characterized for their roles in hormone binding. The goal of this study was therefore to clarify whether further contact points of TSH exist in the structurally undefined hinge region. Therefore, we systematically analyzed 41 uncharacterized residues of the TSHR hinge region as single mutants regarding differences between cell surface expression and bTSH binding. Indeed, we identified further amino acids of the hinge region with influence on bTSH binding. Some of these contribute to a new binding domain from human TSHR position F381 to D386. These hinge mutants with influence on bTSH binding were also analyzed for binding of the superagonistic human TSH analog TR1401 demonstrating that these positions also have an impact on TR1401 binding. Moreover, side chain variations revealed that different amino acid properties like the negative charge, aromatic as well as hydrophilic characteristics, contribute to maintain the hormone-TSHR hinge interaction.


Asunto(s)
Receptores de Tirotropina/química , Tirotropina/metabolismo , Animales , Sitios de Unión , Bovinos , Humanos , Receptores de Tirotropina/metabolismo , Tirotropina/química
17.
Thyroid ; 21(3): 221-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21190443

RESUMEN

BACKGROUND: Germline thyrotropin receptor (TSHR) mutations are associated with sporadic congenital nonautoimmune hyperthyroidism and familial nonautoimmune hyperthyroidism. Somatic TSHR mutations are associated with toxic thyroid nodules (TTNs). The objective of the study was to define a relation of the clinical appearance and the in vitro activity (IVA) of the TSHR mutations described by several authors for these thyroid disorders. METHODS: We analyzed the IVAs published as linear regression analysis (LRA) of the constitutive activity as a function of the TSHR expression and the basal cyclic adenosine monophosphate (cAMP) values to determine differences between exclusively somatic, exclusively familial, and shared sporadic and somatic TSHR-mutations. Further, we investigated correlations of the LRAs/basal cAMP values with clinical activity characteristics (CACs) of TTNs, such as largest diameter of the TTN and the age of the patient at thyroid surgery. RESULTS: Shared sporadic and somatic mutations showed higher median LRA (14.5) and higher median basal cAMP values (fivefold) than exclusively familial mutations (6.1, p = 0.0002; 2.9-fold, p < 0.0001, respectively). Moreover, mutations shared between sporadic congenital nonautoimmune hyperthyroidism and toxic thyroid nodules (TTNs) showed higher median LRA/basal cAMP values (p < 0.0001) than exclusively somatic mutations in TTNs (5.1; 3.89-fold, respectively). Exclusively somatic mutations and exclusively familial mutations showed no significant difference in their median LRA values (p = 0.786) but a significant difference for basal cAMP values (p = 0.0006). The two examined CACs showed no correlation with the IVA characterized by LRA/basal cAMP values or with the presence or absence of a TSHR-mutation. CONCLUSIONS: This systematic analysis of published constitutively activating TSHR-mutations, their CACs, and their IVA provides evidence for higher IVA of shared sporadic and somatic TSHR mutations as compared with familial TSHR mutations. CACs of somatic TSHR mutations in TTNs did not have a clear association with the IVA as characterized by LRA or basal cAMP values.


Asunto(s)
Receptores de Tirotropina/genética , AMP Cíclico/metabolismo , Mutación de Línea Germinal , Humanos , Hipertiroidismo/congénito , Hipertiroidismo/genética , Mutación , Receptores de Tirotropina/metabolismo , Análisis de Regresión , Nódulo Tiroideo/genética , Nódulo Tiroideo/patología
18.
Methods Enzymol ; 485: 421-36, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21050930

RESUMEN

Constitutively activating mutations of the TSHR are the major cause for nonautoimmune hyperthyroidism, which is based on ligand independent, permanent receptor activation. Several reports have highlighted the difficulties to determine whether a TSHR mutation is constitutively active or not especially for borderline cases with only a slight increase of the basal cAMP activity. Current methods to precisely classify such mutants as constitutively active or not, are limited. In some cases, in vitro characterization of TSHR mutants has led to false positive conclusions regarding constitutive TSHR activity and subsequently the molecular origin of hyperthyroidism. For characterization of constitutive TSHR activity, a particular point to consider is that basal receptor activity tightly correlates with the receptor number expressed on the cell surface. Therefore, a comparison of the receptors basal activity in relation to the wild type is only possible with determination of the receptor cell surface expression. Thus, the experimental approaches to determine constitutive TSHR activity should consider the receptor's cell surface expression. We here provide a description of three methods for the determination of constitutive TSHR activity: (A) the evaluation of constitutive TSHR activity under conditions of equal receptor expression; (B) computation of the specific constitutive activity; and (C) the linear regression analysis (LRA). To date, LRA is the best experimental approach to characterize the mutant's basal activity as a function of TSHR cell surface expression. This approach utilizes a parallel measurement of basal cAMP values and receptor cell surface expression and therefore provides a more reliable decision with respect to the presence or absence of constitutive activity.


Asunto(s)
Receptores de Tirotropina/genética , Receptores de Tirotropina/metabolismo , Animales , Células COS , Chlorocebus aethiops , AMP Cíclico/metabolismo , Citometría de Flujo/métodos , Expresión Génica , Humanos , Modelos Lineales , Mutación , Ensayo de Unión Radioligante/métodos
19.
Trends Endocrinol Metab ; 21(2): 111-22, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19819720

RESUMEN

Glycoprotein hormone receptors (GPHRs) are members of the seven-transmembrane-spanning receptor family characterized by a large ectodomain. The hinge region belongs to a part of the GPHR ectodomain for which the three-dimensional structure has not yet been deciphered, leaving important questions unanswered concerning ligand binding and GPHR activation. Recent publications indicate that specific residues of the hinge region mediate hormone binding, receptor activation and/or intramolecular signaling for the three GPHRs, emphasizing the importance of this region. Based on these findings, the hinge region is involved at least in part in hormone binding and receptor activation. This review summarizes functional data regarding the hinge region, demonstrating that this receptor portion represents a link between ligand binding and subsequent GPHR activation.


Asunto(s)
Receptores de la Hormona Hipofisaria/química , Receptores de la Hormona Hipofisaria/fisiología , Secuencia de Aminoácidos , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Pliegue de Proteína , Estructura Terciaria de Proteína/fisiología , Receptores de HFE/química , Receptores de HFE/fisiología , Receptores de HL/química , Receptores de HL/fisiología , Receptores de la Hormona Hipofisaria/inmunología , Receptores de Tirotropina/química , Receptores de Tirotropina/inmunología , Receptores de Tirotropina/fisiología , Homología de Secuencia de Aminoácido
20.
PLoS One ; 5(3): e9745, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20305779

RESUMEN

In this study we wanted to gain insights into selectivity mechanisms between G-protein-coupled receptors (GPCR) and different subtypes of G-proteins. The thyrotropin receptor (TSHR) binds G-proteins promiscuously and activates both Gs (cAMP) and Gq (IP). Our goal was to dissect selectivity patterns for both pathways in the intracellular region of this receptor. We were particularly interested in the participation of poorly investigated receptor parts.We systematically investigated the amino acids of intracellular loop (ICL) 1 and helix 8 using site-directed mutagenesis alongside characterization of cAMP and IP accumulation. This approach was guided by a homology model of activated TSHR in complex with heterotrimeric Gq, using the X-ray structure of opsin with a bound G-protein peptide as a structural template.We provide evidence that ICL1 is significantly involved in G-protein activation and our model suggests potential interactions with subunits G alpha as well as G betagamma. Several amino acid substitutions impaired both IP and cAMP accumulation. Moreover, we found a few residues in ICL1 (L440, T441, H443) and helix 8 (R687) that are sensitive for Gq but not for Gs activation. Conversely, not even one residue was found that selectively affects cAMP accumulation only. Together with our previous mutagenesis data on ICL2 and ICL3 we provide here the first systematically completed map of potential interfaces between TSHR and heterotrimeric G-protein. The TSHR/Gq-heterotrimer complex is characterized by more selective interactions than the TSHR/Gs complex. In fact the receptor interface for binding Gs is a subset of that for Gq and we postulate that this may be true for other GPCRs coupling these G-proteins. Our findings support that G-protein coupling and preference is dominated by specific structural features at the intracellular region of the activated GPCR but is completed by additional complementary recognition patterns between receptor and G-protein subtypes.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Rodopsina/química , Tirotropina/química , Alanina/química , Animales , Células COS , Dominio Catalítico , Separación Celular , Chlorocebus aethiops , Biología Computacional/métodos , AMP Cíclico/metabolismo , Humanos , Fosfatos de Inositol/química , Modelos Moleculares , Mutación , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA