Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Anticancer Res ; 38(5): 2793-2802, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29715101

RESUMEN

BACKGROUND/AIM: Extra-thyroid expression of thyroid stimulating hormone (TSH) receptor (TSHR) has been reported in normal liver tissues, but never assessed in hepatocellular carcinoma (HCC). PATIENTS AND METHODS: Paired cancerous and non-cancerous HCC tissues were analyzed with TSHR expression assays. TSHR functional assessments and sequence analysis for the TSHR exon-10 were performed. RESULTS: TSHR overexpression was found in 150/197 (76.1%) HCCs. Higher TSHR expression was associated with unfavorable postoperative outcomes. Immunohistochemical analysis revealed predominantly nuclei/peri-nuclei localization of TSHR in cancerous tissues but cell membrane localization in non-cancerous parts. TSH stimulation on hepatoma cells resulted in increased cyclic adenosine monophosphate levels with altered cell sensitivity to cisplatin. Gene mutations leading to TSHR truncation were detected in 8/81 (9.9%) HCC tissues. CONCLUSION: Overexpression of TSHR was found in a great majority of HCC tissues and associated with unfavorable prognosis. Cell-based experiments and gene mutation analysis suggested that TSHR in HCCs was functional.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Receptores de Tirotropina/genética , Adulto , Anciano , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/metabolismo , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Modelos de Riesgos Proporcionales , Receptores de Tirotropina/biosíntesis
2.
Thyroid ; 24(6): 1051-61, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24635198

RESUMEN

Using gene expression profiling, we detected differential thyrotropin receptor (TSH-R) expression during human T-cell development in the thymus. This expression pattern indicated a potential role for the TSH-R within the thymus, independent of its function in the thyroid gland. Here, we demonstrate that TSH-R expression is thymus-specific within the immune system. TSH was able to bind and activate the TSH-R present on thymocytes, thereby activating calcium signaling and cyclic adenosine monophosphate signaling pathways. Mice lacking functional TSH-R expression (hyt/hyt mice) were shown to have lower frequencies of DP and SP thymocytes compared to their heterozygous littermates. Moreover, addition of TSH to co-cultures of human thymocytes enhanced T-cell development. Thus, TSH acts as a previously unrecognized growth factor for developing T cells, with potential clinical use to enhance thymic output and thereby the functional T-cell repertoire in the periphery. The direct effects of TSH on thymocytes may also explain the thus far enigmatic thymic hyperplasia in Graves' disease.


Asunto(s)
Linfocitos T/fisiología , Timo/metabolismo , Tirotropina/fisiología , Animales , Línea Celular , Niño , Preescolar , Técnicas de Cocultivo , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones Endogámicos BALB C , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/metabolismo , Transducción de Señal , Tirotropina/farmacología
3.
Res Vet Sci ; 93(1): 108-13, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21737109

RESUMEN

Thyrotropin receptor (TSH-R), thyroglobulin (Tg), thyroperoxidase (TPO), thyroid specific transcription factor-1 (TTF-1), paired box 8 transcription factor (PAX-8), insulin like growth factor-1 (IGF-1) and estrogen receptor alpha (ERα) transcripts were determined by real-time PCR in follicular carcinoma and contralateral (CL) lobes, and healthy thyroid canine glands. Concentrations of TSH-R, PAX-8, and ERα mRNA were not different among groups; the carcinoma group had lower Tg and TPO mRNA than healthy and CL groups, while no differences were found between the two latter groups, suggesting that the carcinoma tissue presents an altered capacity to synthesize thyroid hormones. The transcription factor that promotes thyrocytes proliferation, TTF-1 as well as IGF-1, presented a greater mRNA expression in the CL group, suggesting that the CL lobe may function in a compensatory state.


Asunto(s)
Enfermedades de los Perros/metabolismo , Receptor alfa de Estrógeno/biosíntesis , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Yoduro Peroxidasa/biosíntesis , Proteínas Nucleares/biosíntesis , Factores de Transcripción Paired Box/biosíntesis , Receptores de Tirotropina/biosíntesis , Tiroglobulina/biosíntesis , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo , Factores de Transcripción/metabolismo , Animales , Perros , Femenino , Masculino , Factor de Transcripción PAX8 , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Factor Nuclear Tiroideo 1 , Factores de Transcripción/biosíntesis
4.
Endocrinology ; 147(6): 2789-800, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16543368

RESUMEN

Immunization with adenovirus encoding the TSH receptor (TSHR) or its A-subunit induces Graves' hyperthyroidism in BALB/c and BALB/c x C57BL/6 offspring but not C57BL/6 mice. High-resolution genetic maps are available for 13 recombinant inbred CXB strains generated from BALB/c x C57BL/6 progeny by repeated brother x sister matings to establish fully inbred lines. CXB strains were studied before and after A-subunit adenovirus immunization for TSHR antibodies (TBI, inhibition of TSH binding), serum T4, and thyroid histology. All strains developed TBI activity (at variable levels), six strains became hyperthyroid, and one was overtly thyrotoxic. No low TBI responders became hyperthyroid, but high TBI did not predict hyperthyroidism. Preimmunization T4 levels varied in different CXB strains and was unrelated to subsequent T4 elevation. Linkage analysis indicated that different chromosomes were involved in generating TSHR antibodies and serum T4 before and after immunization. TBI activity was linked in part with major histocompatibility (MHC) genes on chromosome 17 (Chr 17) but induced Graves' disease involved non-MHC genes (Chr 19 and 10). The Chr 10 locus is close to the Trhde gene that encodes TSH-releasing hormone degrading enzyme. Expression of Trhde is controlled by thyroid hormones and linkage with a thyroid function-related gene is intriguing. Our data, the first genome scan in murine Graves' disease, provides insight into the role of MHC and non-MHC genes in human and murine Graves' disease. Finally, our study demonstrates the potential of recombinant inbred mice for discriminating between immune-response genes and thyroid function susceptibility genes in Graves' disease.


Asunto(s)
Autoanticuerpos/biosíntesis , Hipertiroidismo/genética , Receptores de Tirotropina/inmunología , Adenoviridae/genética , Animales , Formación de Anticuerpos/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Ligamiento Genético , Hipertiroidismo/etiología , Inmunoglobulinas Estimulantes de la Tiroides , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Subunidades de Proteína , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/genética , Recombinación Genética , Tiroxina/sangre
5.
J Immunol ; 176(1): 668-76, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16365463

RESUMEN

The thyrotropin receptor (TSHR), the major autoantigen in Graves' disease, is posttranslationally modified by intramolecular cleavage to form disulfide-linked A- and B-subunits. Because Graves' hyperthyroidism is preferentially induced in BALB/c mice using adenovirus encoding the free A-subunit rather than full-length human TSHR, the shed A-subunit appears to drive the disease-associated autoimmune response. To further investigate this phenomenon, we generated transgenic mice with the human A-subunit targeted to the thyroid. Founder transgenic mice had normal thyroid function and were backcrossed to BALB/c. The A-subunit mRNA expression was confirmed in thyroid tissue. Unlike wild-type littermates, transgenic mice immunized with low-dose A-subunit adenovirus failed to develop TSHR Abs, hyperthyroidism, or splenocyte responses to TSHR Ag. Conventional immunization with A-subunit protein and adjuvants induced TSHR Abs lacking the characteristics of human autoantibodies. Unresponsiveness was partially overcome using high-dose, full-length human TSHR adenovirus. Although of low titer, these induced Abs recognized the N terminus of the A-subunit, and splenocytes responded to A-subunit peptides. Therefore, "non-self" regions in the B-subunit did not contribute to inducing responses. Indeed, transgenic mice immunized with high-dose A-subunit adenovirus developed TSHR Abs with thyrotropin-binding inhibitory activity, although at lower titers than wild-type littermates, suggesting down-regulation in the transgenic mice. In conclusion, in mice expressing a human A-subunit transgene in the thyroid, non-self human B-subunit epitopes are not necessary to induce responses to the A-subunit. Our findings raise the possibility that autoimmunity to the TSHR in humans may not involve epitopes on a cross-reacting protein, but rather, strong adjuvant signals provided in bystander immune responses.


Asunto(s)
Autoinmunidad , Subunidades de Proteína/inmunología , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/inmunología , Glándula Tiroides/inmunología , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Enfermedad de Graves/inmunología , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores de Tirotropina/genética , Homología de Secuencia de Aminoácido , Glándula Tiroides/patología
6.
J Clin Endocrinol Metab ; 90(10): 5663-71, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16030158

RESUMEN

CONTEXT: Two classes of repeated genomic elements, retrotransposons and endogenous retroviruses, encode for endogenous nontelomeric reverse transcriptase (RT), a gene that is down-regulated in differentiated cells but is highly expressed in embryonic and transformed tissues. Two nonnucleosidic RT inhibitors, efavirenz and nevirapine, currently used in HIV treatment, reversibly down-regulate tumor growth and induce differentiation in several human tumor cell models. OBJECTIVES: Aggressive biological behavior and loss of specific thyroid cell functions, such as thyroglobulin, thyroid peroxidase, TSH receptor, Na/I symporter expression, and iodine uptake are features of anaplastic thyroid cancer. Thus, we evaluated the use of RT inhibitors as a potentially differentiating and molecular-targeted treatment of this neoplasm. RESULTS: Our findings showed that nevirapine and efavirenz reversibly inhibit cell proliferation without triggering cell death in the undifferentiated thyroid carcinoma ARO and FRO cells, which exhibited high levels of endogenous RT activity. Inhibition of cell growth was correlated with accumulation of cells in the G0/G1 phase of the cell cycle, with a concomitant decrease in the S phase. Moreover, treated cells demonstrated a differentiated phenotype and a significant reprogramming of gene expression characterized by up-regulation of the TSH receptor, thyroglobulin, thyroid peroxidase, and Na/I symporter genes. Interestingly, RT inhibition reestablished the ability to uptake iodine in response to TSH either in vitro or in vivo and reversibly down-regulated tumor growth in mice xenografts of ARO cells. CONCLUSIONS: These findings support the need for clinical trials to clarify whether RT inhibitors may restore the sensitivity to radiometabolic therapy in anaplastic thyroid tumors.


Asunto(s)
Carcinoma/metabolismo , Carcinoma/fisiopatología , Proliferación Celular/efectos de los fármacos , Yodo/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/fisiopatología , Tirotropina/fisiología , Animales , Apoptosis/efectos de los fármacos , Carcinoma/enzimología , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Immunoblotting , Ratones , Ratones Desnudos , Necrosis , Trasplante de Neoplasias , ADN Polimerasa Dirigida por ARN/metabolismo , Receptores de Tirotropina/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de la Tiroides/enzimología , Trasplante Heterólogo
7.
Thyroid ; 15(5): 422-6, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15929662

RESUMEN

Thyroid transcription factor-1 (TTF-1) is required for maximal expression of thyrotropin receptor (TSHR) in the thyroid. Extrathyroidal TSHR expression is detectable in normal orbital adipose tissues, with increased levels found in orbital tissues from patients with Graves' ophthalmopathy (GO), and in orbital preadipocyte cultures following differentiation. In order to determine whether TTF-1 might be involved in orbital TSHR expression, we used quantitative real-time polymerase chain reaction (PCR) to assess relative expression of this and other thyroid-associated transcription factors (TTF-2 and Pax-8) in GO orbital tissue specimens (n = 28) and cultures (n = 3), and in normal orbital tissues (n = 19) and cultures (n = 3). We detected TTF-1 and TTF-2 mRNA in GO and normal orbital tissue samples, with no difference in levels noted between the tissues. In the GO orbital cultures, TTF-1 mRNA was higher in differentiated than in control (undifferentiated) cultures (p < 0.05), while TTF-2 was unchanged. In the normal cultures, neither TTF-1 nor TTF-2 mRNA levels increased in differentiated cultures. Pax8 was undetectable in all orbital tissues and cell cultures. The presence of mRNA encoding TTF-1 in orbital tissues and cultures suggest that this transcription factor may play an important role in extrathyroidal, as it does in thyroidal, TSHR expression.


Asunto(s)
Tejido Adiposo/metabolismo , Proteínas Nucleares/metabolismo , Órbita/metabolismo , Receptores de Tirotropina/biosíntesis , Factores de Transcripción/metabolismo , Adenosina Trifosfatasas , Adipocitos/metabolismo , Diferenciación Celular , Células Cultivadas , Cartilla de ADN/farmacología , ADN Complementario/biosíntesis , ADN Complementario/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor Nuclear Tiroideo 1 , Transactivadores/metabolismo
8.
J Endocrinol Invest ; 27(3): 216-20, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15164996

RESUMEN

The TSH receptor (TSHr) is the autoantigen responsible for the hyperthyroidism of Graves' disease. Recent studies suggest that this receptor may also be an autoimmune target in Graves' ophthalmopathy (GO) and pretibial dermopathy (PTD). Its involvement in the pathogenesis of these conditions would help to explain the close clinical associations between hyperthyroidism, GO and PTD. TSHr has been shown to be present in normal orbital and dermal tissues and evidence supports the conviction that expression may be increased in tissues involved in GO and PTD. In the setting of Graves' disease, the expression of this antigen in connective tissues throughout the body may lead to systemic, subclinical connective tissue inflammation. Given this background, local or environmental factors such as circulating or local cytokines, gravitational dependency, anatomic constraint of the bony orbit, or trauma, may augment clinical disease involvement within the orbit and pretibial skin. Alternately, locally enhanced expression of this protein at the sites of clinical disease may not be directly involved in pathogenesis, but could be secondary to the ongoing process, and nonetheless important in disease progression.


Asunto(s)
Enfermedad de Graves/metabolismo , Órbita/metabolismo , Receptores de Tirotropina/biosíntesis , Adipocitos/fisiología , Animales , AMP Cíclico/biosíntesis , Enfermedad de Graves/etiología , Enfermedad de Graves/genética , Humanos , Ratones , Ratones Endogámicos BALB C
9.
Eur J Endocrinol ; 150(3): 269-76, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15012610

RESUMEN

OBJECTIVE: The malignancy of thyroid oncocytic tumours, or oncocytomas, is higher than that of follicular tumours. The aim of this study was to investigate the role of thyroid-specific genes in oncocytic tumours and papillary carcinomas. DESIGN AND METHODS: We compared 29 oncocytic tumours with 12 papillary carcinomas. Real-time quantitative PCR was used to measure the expression of thyroid-specific differentiation markers (thyrotrophin-stimulation hormone receptor (TSHR), thyroglobulin (TG) and Na(+)/I(-) symporter (NIS)), transcription factors (thyroid transcription factor-1 (TTF-1) and paired box gene-8 (PAX8)) and nuclear receptors (peroxisome proliferator-activated receptor (PPARgamma1) and thyroid hormone receptor (TRbeta1)) involved in thyroid carcinogenesis. RESULTS: TSHR, TTF-1 and TRbeta1 levels were significantly lower in oncocytic tumours than in papillary carcinomas, as a result of specific biological changes in oncocytic tumours. However, PAX8 and PPARgamma1 did not seem to be involved in the process. Applying the criterion of the underexpression of the thyroid-specific differentiation markers, TSHR, TG and NIS, we classified the oncocytic tumours and papillary carcinomas into three groups. In the first, all three markers were underexpressed; in the second, TSHR was normal while TG and NIS were underexpressed; and in the third, only NIS was underexpressed. The expression patterns revealed that 13 of the 24 oncocytic adenomas underexpressing TSHR in our study, as did four of the five oncocytic carcinomas. CONCLUSION: Cases of oncocytic adenoma associated with low levels of TSHR could be putative oncocytic carcinomas and should therefore receive adequate follow-up [corrected].


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/genética , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Adenocarcinoma Folicular/genética , Adenocarcinoma Folicular/metabolismo , Adenocarcinoma Folicular/patología , Adenocarcinoma Papilar/genética , Adenocarcinoma Papilar/metabolismo , Adenocarcinoma Papilar/patología , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box , Reacción en Cadena de la Polimerasa , ARN Neoplásico/química , ARN Neoplásico/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Simportadores/genética , Simportadores/metabolismo , Tiroglobulina/metabolismo , Neoplasias de la Tiroides/metabolismo , Factor Nuclear Tiroideo 1 , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Autoimmunity ; 36(6-7): 381-7, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14669945

RESUMEN

An animal model of Graves' disease (GD) will help us to clearly understand the role of thyroid-stimulating hormone receptor (TSHR)-specific T cells and TSHR-Abs during the development of GD and to develop TSHR-specific immunotherapy. This review focuses on four different recent approaches towards the development of an animal model of GD. These approaches are: (1) Immunization of AKR/N mice with fibroblasts coexpressing syngeneic major histocompatibility complex (MHC) class II and TSHR. (2) Immunization of selected strains of mice with an expression vector containing TSHR cDNA. (3) Immunization of BALB/c mice with syngeneic M12 cells or xenogenic HEK-293 cells expressing full-length or extracellular domain of TSHR (ETSHR). (4) Injection of adenovirus-expressing TSHR into BALB/c mice.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Graves , Adenoviridae/genética , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Epítopos de Linfocito T/inmunología , Fibroblastos/inmunología , Vectores Genéticos , Enfermedad de Graves/inmunología , Enfermedad de Graves/patología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Inmunización , Ratones , Ratones Endogámicos , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/genética , Receptores de Tirotropina/inmunología , Células TH1/inmunología , Células Th2/inmunología
11.
J Biol Chem ; 278(42): 40621-30, 2003 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-12902333

RESUMEN

TSH activates its specific receptor in thyroid cells and induces cAMP, a robust stimulator of thyroid cell proliferation. Conversely, cAMP is a potent inhibitor of growth in mouse fibroblasts. To dissect the signals mediating cAMP-dependent growth, we have expressed in mouse fibroblasts the human thyrotropin receptor (TSHR) or a constitutively active mutant, under the control of the tetracyclin promoter. Both TSHR and cAMP levels were modulated by tetracyclin. In the presence of serum, activation of TSHR by TSH induced growth arrest. In the absence of serum, cells expressing TSHR stimulated with TSH, replicated their DNA, but underwent apoptosis. Co-expression of cAMP-dependent protein kinase (PKA) regulatory subunit type II (RIIbeta) inhibited apoptosis and stimulated the growth of cells only in the presence of TSH. Expression of RIIbeta-PKA, in the absence of TSHR, induced apoptosis, which was reversed by cAMP. Growth, stimulated by TSHR-RIIbeta-PKA in mouse fibroblasts, was also dependent on Rap1 activity, indicating cAMP-dependent growth in thyroid cells. As for the molecular mechanism underlying these effects, we found that in normal fibroblasts, TSH induced AKT and ERK1/2 only in cells expressing TSHR and RII. Similarly, activation of TSHR increased cAMP levels greatly, but was unable to stimulate CREB phosphorylation and transcription of cAMP-induced genes in the absence of RII. These data provide a simple explanation for the anti-proliferative and proliferative effects of cAMP in different cell types and indicate that RII-PKAII complements TSHR action by stably propagating robust cAMP signals in cell compartments.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/química , Receptores de Tirotropina/biosíntesis , Adenoviridae/genética , Animales , Apoptosis , Northern Blotting , Western Blotting , División Celular , Núcleo Celular/metabolismo , Separación Celular , AMP Cíclico/metabolismo , Proteína Quinasa Tipo II Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Citometría de Flujo , Genes Dominantes , Etiquetado Corte-Fin in Situ , Ratones , Células 3T3 NIH , Fosforilación , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Tetraciclina/farmacología , Factores de Tiempo , Transcripción Genética , Transfección
12.
Cancer Res ; 63(9): 2316-21, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12727856

RESUMEN

Thyroid-stimulating hormone receptor (TSHR) expression is frequently silenced in epithelial thyroid cancers associated with decreased or absent TSH-promoted iodine uptake. To study the underlying molecular mechanism of decreased TSHR expression, we examined the methylation status of the TSHR gene promoter by sequencing bisulfite-treated DNA from thyroid tumors. After identification of methylated sites by sequencing bisulfite-treated DNA, we used methylation-specific polymerase chain reaction and found frequent CpG methylation in papillary thyroid cancer (23 of 39 patients; 59%) and follicular thyroid cancers (7 of 15 patients; 47%). In contrast, we saw no methylation in normal thyroid tissues and benign adenomas (0 of 8 patients; 0%). In human thyroid tumor cell lines, we observed that TSHR was normally expressed at the protein and mRNA level in cells where the TSHR gene was unmethylated, whereas it was silenced in cell lines where the TSHR promoter was hypermethylated. Treatment of the latter cells with a demethylating agent partially restored TSHR expression. We thus demonstrate aberrant methylation of human TSHR as a likely molecular pathway responsible for the silencing of this gene in thyroid cancers. We propose that methylation of TSHR may provide a novel diagnostic marker of malignancy and a basis for potential use of demethylating agents in conjunction with TSH-promoted radioiodine therapy for epithelial thyroid cancers.


Asunto(s)
Metilación de ADN , Silenciador del Gen/fisiología , Receptores de Tirotropina/genética , Neoplasias de la Tiroides/genética , Adenoma/genética , Adenoma/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Marcadores Genéticos/genética , Regiones Promotoras Genéticas , Receptores de Tirotropina/biosíntesis , Neoplasias de la Tiroides/metabolismo , Células Tumorales Cultivadas
13.
Thyroid ; 13(3): 233-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12729471

RESUMEN

The etiology of Graves' disease is multifactorial. We investigated the role of genetic and environmental factors on the susceptibility to Graves' hyperthyroidism using a new murine model. Intramuscular injection of recombinant adenovirus expressing the thyrotropin receptor (AdCMVTSHR) induces Graves'-like hyperthyroidism (thyrotropin receptor [TSHR] antibodies, elevated thyroxine, and diffuse goiter) in more than 50% of female BALB/c mice. The relative contributions of major histocompatibility complex (MHC) and non-MHC genes on the susceptibility to hyperthyroidism were studied by immunizing BALB/c (H-2d), BALB.K (H-2k), and DBA/2J (H-2d) mice with AdCMVTSHR. Hyperthyroidism developed in approximately 50% of BALB/c and BALB.K mice but only 5% of DBA/2J mice, indicating a major role for non-MHC genes in disease development. The effect of environmental microorganisms was evaluated by comparing disease incidence in BALB/c mice maintained in pathogen-free conditions versus those in nonsterile, conventional housing, as well as by coadministering microorganism components (Escherichia coli lipopolysaccharide or yeast zymosan A) as adjuvants with AdCMVTSHR. Neither type of exposure to environmental pathogens influenced disease induction. In conclusion, non-MHC genes, but not infectious organisms, play a major role in the etiology of this novel murine model of Graves' disease.


Asunto(s)
Enfermedad de Graves/genética , Enfermedad de Graves/inmunología , Receptores de Tirotropina/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Anticuerpos/análisis , Anticuerpos/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/química , Femenino , Enfermedad de Graves/microbiología , Enfermedad de Graves/patología , Inmunoglobulinas/inmunología , Inyecciones Intramusculares , Lipopolisacáridos/inmunología , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos DBA , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/genética , Tiroxina/análisis , Tiroxina/inmunología , Levaduras/química , Zimosan/inmunología
14.
J Immunol ; 170(7): 3522-7, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12646613

RESUMEN

Graves' hyperthyroidism has long been considered to be a Th2-type autoimmune disease because it is directly mediated by autoantibodies against the thyrotropin receptor (TSHR). However, several lines of evidence have recently challenged this concept. The present study evaluated the Th1/Th2 paradigm in Graves' disease using a recently established murine model involving injection of adenovirus expressing the TSHR (AdCMVTSHR). Coinjection with adenovirus expressing IL-4 (AdRGDCMVIL-4) decreased the ratio of Th1/Th2-type anti-TSHR Ab subclasses (IgG2a/IgG1) and suppressed the production of IFN-gamma by splenocytes in response to TSHR Ag. Importantly, immune deviation toward Th2 was accompanied by significant inhibition of thyroid-stimulating Ab production and reduction in hyperthyroidism. However, in a therapeutic setting, injection of AdRGDCMVIL-4 alone or in combination with AdCMVTSHR into hyperthyroid mice had no beneficial effect. In contrast, coinjection of adenoviruses expressing IL-12 and the TSHR promoted the differentiation of Th1-type anti-TSHR immune responses as demonstrated by augmented Ag-specific IFN-gamma secretion from splenocytes without changing disease incidence. Coinjection of adenoviral vectors expressing IL-4 or IL-12 had no effect on the titers of anti-TSHR Abs determined by ELISA or thyroid-stimulating hormone-binding inhibiting Ig assays, suggesting that Ab quality, not quantity, is responsible for disease induction. Our observations demonstrate the critical role of Th1 immune responses in a murine model of Graves' hyperthyroidism. These data may raise a cautionary note for therapeutic strategies aimed at reversing Th2-mediated autoimmune responses in Graves' disease in humans.


Asunto(s)
Autoanticuerpos/fisiología , Modelos Animales de Enfermedad , Enfermedad de Graves/inmunología , Enfermedad de Graves/prevención & control , Interleucina-4/uso terapéutico , Células Th2/inmunología , Células Th2/metabolismo , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Células COS , Epítopos de Linfocito T/inmunología , Femenino , Vectores Genéticos , Humanos , Inmunoglobulinas Estimulantes de la Tiroides/administración & dosificación , Inmunoglobulinas Estimulantes de la Tiroides/biosíntesis , Inmunoglobulinas Estimulantes de la Tiroides/genética , Inyecciones Intramusculares , Interleucina-12/biosíntesis , Interleucina-12/genética , Interleucina-4/administración & dosificación , Interleucina-4/biosíntesis , Interleucina-4/genética , Ratones , Ratones Endogámicos BALB C , Receptores de Tirotropina/administración & dosificación , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/genética , Receptores de Tirotropina/inmunología , Células TH1/inmunología , Células TH1/metabolismo
15.
J Immunol ; 170(4): 2195-204, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12574393

RESUMEN

In autoimmune Graves' disease (GD), autoantibodies bind to the thyrotropin receptor (TSHR) and cause hyperthyroidism. We studied the effects of fms-like tyrosine kinase receptor 3 ligand (Flt3-L) or GM-CSF treatment on the development of experimental autoimmune GD (EAGD) in mice, a slowly progressing Ab-mediated organ-specific autoimmune disease of the thyroid induced by immunization with syngeneic cells expressing TSHR. Flt3-L and GM-CSF treatment resulted in up-regulation of CD8a(+) and CD8a(-) dendritic cells, and skewing of cytokine and immune responses to TSHR in favor of Th1 and Th2, respectively. However, this skewing did not persist until the later stages, and thus failed to affect the course or severity of the disease. To determine whether the total absence of either IL-4 or IFN-gamma could affect the development of EAGD, we immunized wild-type, IFN-gamma(-/-) and IL-4(-/-) BALB/c mice with TSHR. Nearly 100% of the wild-type and IFN-gamma(-/-) mice developed EAGD with optimal TSHR-specific immune responses, while IL-4(-/-) mice completely resisted disease and showed delayed and suboptimal pathogenic Ab response. These data demonstrated that skewing immune responses to TSHR, using either Flt3-L or GM-CSF, in favor of Th1 or Th2, respectively, may not be sufficient to alter the course of the disease, while the complete absence of IL-4, but not IFN-gamma, can prevent the development of EAGD.


Asunto(s)
Enfermedad de Graves/inmunología , Enfermedad de Graves/prevención & control , Interleucina-4/deficiencia , Interleucina-4/genética , Autotolerancia , Células Th2/inmunología , Animales , Células CHO , Línea Celular , Cricetinae , Citocinas/biosíntesis , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Progresión de la Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Enfermedad de Graves/genética , Humanos , Inmunofenotipificación , Inyecciones Intraperitoneales , Interferón gamma/deficiencia , Interferón gamma/genética , Interleucina-4/fisiología , Ligandos , Proteínas de la Membrana/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Estructura Terciaria de Proteína/fisiología , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/fisiología , Autotolerancia/genética , Células TH1/inmunología , Células Th2/metabolismo
16.
J Immunol ; 168(6): 2789-94, 2002 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11884447

RESUMEN

In this work we report a novel method to efficiently induce a murine model of Graves' hyperthyroidism. Inbred mice of different strains were immunized by i.m. injection with adenovirus expressing thyrotropin receptor (TSHR) or beta-galactosidase (1 x 10(11) particles/mouse, three times at 3-wk intervals) and followed up to 8 wk after the third immunization. Fifty-five percent of female and 33% of male BALB/c (H-2(d)) and 25% of female C57BL/6 (H-2(b)) mice developed Graves'-like hyperthyroidism with elevated serum thyroxine (T(4)) levels and positive anti-TSHR autoantibodies with thyroid-stimulating Ig (TSI) and TSH-binding inhibiting Ig (TBII) activities. In contrast, none of female CBA/J (H-2(k)), DBA/1J (H-2(q)), or SJL/J (H-2(s)) mice developed Graves' hyperthyroidism or anti-TSHR autoantibodies except SJL/J, which showed strong TBII activities. There was a significant positive correlation between TSI values and T(4) levels, but the correlations between T(4) and TBII and between TSI and TBII were very weak. TSI activities in sera from hyperthyroid mice measured with some chimeric TSH/lutropin receptors suggested that their epitope(s) on TSHR appeared similar to those in patients with Graves' disease. The thyroid glands from hyperthyroid mice displayed diffuse enlargement with hypertrophy and hypercellularity of follicular epithelia with occasional protrusion into the follicular lumen, characteristics of Graves' hyperthyroidism. Decreased amounts of colloid were also observed. However, there was no inflammatory cell infiltration. Furthermore, extraocular muscles from hyperthyroid mice were normal. Thus, the highly efficient means that we now report to induce Graves' hyperthyroidism in mice will be very useful for studying the pathogenesis of autoimmunity in Graves' disease.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Enfermedad de Graves/genética , Enfermedad de Graves/inmunología , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/genética , Animales , Modelos Animales de Enfermedad , Femenino , Enfermedad de Graves/patología , Enfermedad de Graves/virología , Humanos , Inyecciones Intramusculares , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Plásmidos/administración & dosificación , Plásmidos/biosíntesis , Plásmidos/inmunología , Receptores de Tirotropina/administración & dosificación , Glándula Tiroides/patología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología
17.
Life Sci ; 68(25): 2781-7, 2001 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-11432444

RESUMEN

Thymic hyperplasia is associated with Graves' disease. It has been demonstrated that thyrotropin receptors are expressed in human thymus, and thymic thyrotropin receptors are suggested to be involved in the pathophysiology of Graves' disease. We have studied whether thyrotropin receptors are expressed in rat thymic tissue. Thyrotropin receptor mRNA was demonstrated in 5-day-old, 10-day-old, 20-day-old and adult rat thymus by reverse transcription polymerase chain reaction. Thyrotropin receptor mRNA was also demonstrated in cultured rat thymic epithelial cells. Thyrotropin stimulated cyclic AMP production in cultured rat thymic epithelial cells, suggesting the expression of functional thyrotropin receptors. The present results indicate that thyrotropin receptors are expressed in rat thymus.


Asunto(s)
Receptores de Tirotropina/genética , Timo/metabolismo , Animales , Animales Recién Nacidos , Southern Blotting , Células Cultivadas , AMP Cíclico/metabolismo , Cartilla de ADN/química , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Masculino , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Receptores de Tirotropina/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timo/crecimiento & desarrollo , Glándula Tiroides/citología
18.
Clin Chem Lab Med ; 39(1): 25-8, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11256796

RESUMEN

Detection of autoantibodies to the thyrotropin receptor by radioreceptor assays is largely requested in clinical practice for the diagnosis of Graves' disease and its differentiation from diffuse thyroid autonomy. Additionally, thyrotropin receptor antibodies (TRAb) measurement during antithyroid drug treatment can be useful to evaluate the risk of relapse after discontinuation of the therapy. Nevertheless, some patients affected by Graves' disease are TRAb-negative when a 1st generation assay is used. In this study we evaluated the diagnostic performance of a newly developed 2nd generation TRAb assay in 46 patients with Graves' disease with negative 1st generation TRAb assay results. A control group of 50 Graves' disease patients with positive 1st generation TRAb assay results, 50 patients with Hashimoto's thyroiditis and 50 patients with nodular goiter were also examined. Forty one of 46 patients with Graves' disease with negative 1st generation TRAb assay results showed a positive 2nd generation test. No differences were seen in control groups. In conclusion, the 2nd generation TRAb assay is more sensitive than the 1st generation test and should be used in clinical practice. Long-term prospective studies are needed to evaluate the prognostic role of the 2nd generation TRAb assay in Graves' disease.


Asunto(s)
Enfermedad de Graves/sangre , Receptores de Tirotropina/biosíntesis , Receptores de Tirotropina/inmunología , Anticuerpos/metabolismo , Antitiroideos/farmacología , Estudios de Casos y Controles , Química Clínica/métodos , Bocio/sangre , Humanos , Recurrencia , Factores de Riesgo , Sensibilidad y Especificidad , Tiroiditis Autoinmune/sangre
19.
Biotechnol Prog ; 16(5): 703-9, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11027160

RESUMEN

For the detection of autoantibodies to thyroid stimulating hormone receptors (TSH-R) in Graves' disease based on a novel coated tube assay system, human TSH-R is needed in large amounts. Whereas expression of TSH-R in bacteria, yeast, or insect cells results in nonfunctional, denaturated receptor, mammalian cells such as COS, CHO, and HeLa are able to express functional TSH-R, but only in very low amounts. Furthermore, for all of these cultivations expensive standard media containing 10% fetal calf serum are needed to obtain functional receptor. Here we report on the development of a serum-free production-scale process based on a stable transformed and highly productive human leukemia cell line K562 (1). Starting with K562-TSH-R cells growing in medium containing 10% fetal calf serum the cell line was adapted to serum-free medium. The adaptation medium was optimized in regards to amino acid and protein concentrations, since the use of unadjusted medium caused cell death after 2 days. The adapted cells were stable and could be cultivated without antibiotics for more than 50 cell doublings without losing their productivity. The obtained receptor showed improved TSH binding. The process development was based on cultivations in a 2-L bench-scale bioreactor. Cultivations in batch mode and chemostat mode and perfusion cultivation with the usage of an internal microfiltration device and a spin-filter device were compared. After process optimization a continuous process using spin-filter was set up and run in a 20 L-pilot-scale bioreactor. The presented results were the prerequisite for the production of the novel assay for the diagnosis of autoantibodies to TSH-R in Graves' disease.


Asunto(s)
Reactores Biológicos , Receptores de Tirotropina/biosíntesis , Medio de Cultivo Libre de Suero , Humanos , Células K562
20.
Invest Ophthalmol Vis Sci ; 41(11): 3249-55, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11006210

RESUMEN

PURPOSE: Adipogenesis contributes to the pathogenesis of thyroid eye disease (TED). Thyrotropin receptor (TSHR) transcripts are present in orbital fat. This study was conducted to determine whether they are expressed as functional protein, and if so, whether this is restricted to TED orbits or to a particular stage in adipocyte differentiation. METHODS: Samples of fat were obtained from 18 TED-affected orbits and 4 normal orbits, and 9 were obtained from nonorbital locations. Frozen sections were examined by immunocytochemistry using monoclonal antibodies specific for the human TSHR. Samples were disaggregated and the preadipocytes separated from the mature by differential centrifugation and cultured in serum-free or DM and examined for morphologic changes, oil red O and TSHR staining, and TSH-induced cyclic adenosine monophosphate (cAMP) production. RESULTS: Marked immunoreactivity was observed in frozen sections from all three TED samples and faint staining in both normal orbital fat samples. In vitro, 1% to 5% of preadipocytes displayed TSHR immunoreactivity in five of six TED and two of three normal orbital samples and in three of five nonorbital samples. Differentiation, was induced in all 14 orbital samples. Three of four nonorbital samples contained occasional differentiated cells. Fifty percent to 70% of differentiating cells demonstrated receptor immunoreactivity. Two of three TED and four of four nonorbital preadipocytes in DM and/or mature adipocytes displayed a TSH-mediated increase in cAMP. CONCLUSIONS: The results indicate that orbital fat TSHR transcripts are expressed as protein, which can be functional. This is not aberrant in TED orbits, although expression may be upregulated. The majority of preadipocytes undergoing differentiation express the receptor, indicating a key role for this population in one mechanism for increasing orbital volume.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Enfermedad de Graves/metabolismo , Receptores de Tirotropina/biosíntesis , Adipocitos/patología , Tejido Adiposo/patología , Anticuerpos Monoclonales , Diferenciación Celular , Separación Celular , Células Cultivadas , AMP Cíclico/biosíntesis , Femenino , Enfermedad de Graves/patología , Enfermedad de Graves/cirugía , Humanos , Técnicas para Inmunoenzimas , Masculino , Enfermedades Orbitales/metabolismo , Enfermedades Orbitales/patología , Enfermedades Orbitales/cirugía , Tirotropina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA