Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 287
Filter
1.
Clin Exp Immunol ; 162(3): 438-46, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21070207

ABSTRACT

Thyroid-stimulating immunoglobulins (TSI) are a functional biomarker of Graves' disease (GD). To develop a novel TSI bioassay, a cell line (MC4-CHO-Luc) was bio-engineered to constitutively express a chimeric TSH receptor (TSHR) and constructed with a cyclic adenosine monophosphate (cAMP)-dependent luciferase reporter gene that enables TSI quantification. Data presented as percentage of specimen-to-reference ratio (SRR%) were obtained from 271 patients with various autoimmune and thyroid diseases and 180 controls. Sensitivity of 96% and specificity of 99% for untreated GD were attained by receiver operating characteristic analysis, area under the curve 0·989, 95% confidence interval 0·969-0·999, P = 0·0001. Precision testing of manufactured reagents of high, medium, low and negative SRR% gave a percentage of coefficient-of-variation of 11·5%, 12·8%, 14·5% and 15·7%, respectively. There was no observed interference by haemoglobin, lipids and bilirubin and no non-specific stimulation by various hormones at and above physiological concentrations. TSI levels from GD patients without (SRR% 406 ± 134, mean ± standard deviation) or under anti-thyroid treatment (173 ± 147) were higher (P < 0·0001) compared with TSI levels of patients with Hashimoto's thyroiditis (51 ± 37), autoimmune diseases without GD (24 ± 10), thyroid nodules (30 ± 26) and controls (35 ± 18). The bioassay showed greater sensitivity when compared with anti-TSHR binding assays. In conclusion, the TSI-Mc4 bioassay measures the functional biomarker accurately in GD with a standardized protocol and could improve substantially the diagnosis of autoimmune diseases involving TSHR autoantibodies.


Subject(s)
Graves Disease/diagnosis , Receptors, Thyrotropin/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Biological Assay , CHO Cells , Cricetinae , Cricetulus , Graves Disease/blood , Graves Disease/immunology , Graves Disease/physiopathology , Humans , Immunoglobulins, Thyroid-Stimulating/blood , Protein Binding/genetics , Protein Engineering , Receptors, Thyrotropin/genetics , Recombinant Fusion Proteins/genetics , Reproducibility of Results , Sensitivity and Specificity , Transgenes/genetics
2.
J Clin Endocrinol Metab ; 95(5): 2123-31, 2010 May.
Article in English | MEDLINE | ID: mdl-20237164

ABSTRACT

CONTEXT: Immunoglobulins stimulating the TSH receptor (TSI) influence thyroid function and likely mediate extrathyroidal manifestations of Graves' disease (GD). OBJECTIVES: The aim of this study was to assess the clinical relevance of TSI in GD patients with or without Graves' orbitopathy (GO), to correlate the TSI levels with activity/severity of GO, and to compare the sensitivity/specificity of a novel TSI bioassay with TSH receptor (TSH-R) binding methods (TRAb). DESIGN: TSI were tested in two reporter cell lines designed to measure Igs binding the TSH-R and transmitting signals for cAMP/CREB/cAMP regulatory element complex-dependent activation of luciferase gene expression. Responsiveness to TSI of the novel chimeric (Mc4) TSH-R (amino acid residues 262-335 of human TSH-R replaced by rat LH-R) was compared with the wild-type (wt) TSH-R. RESULTS: All hyperthyroid GD/GO patients were TSI-positive. TSI were detected in 150 of 155 (97%, Mc4) and 148 of 155 (95%, wt) GO patients, in six of 45 (13%, Mc4) and 20 of 45 (44%, wt) mostly treated GD subjects, and in 0 of 40 (Mc4) and one of 40 (wt) controls. Serum TSI titers were 3- and 8-fold higher in GO vs. GD and control, respectively. All patients with diplopia and optic neuropathy and smokers were TSI-positive. TSI strongly correlated with GO activity (r = 0.87 and r = 0.7; both P < 0.001) and severity (r = 0.87 and r = 0.72; both P < 0.001) in the Mc4 and wt bioassays, respectively. Clinical sensitivity (97 vs. 77%; P < 0.001) and specificity (89 vs. 43%; P < 0.001) of the Mc4/TSI were greater than TRAb in GO. All 11 of 200 (5.5%) TSI-positive/TRAb-negative patients had GO, whereas all seven of 200 (3.5%) TSI-negative/TRAb-positive subjects had GD only. CONCLUSION: The novel Mc4/TSI is a functional indicator of GO activity and severity.


Subject(s)
Graves Ophthalmopathy/blood , Immunoglobulins, Thyroid-Stimulating/blood , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antithyroid Agents/therapeutic use , Female , Graves Disease/blood , Graves Disease/drug therapy , Graves Disease/radiotherapy , Graves Disease/surgery , Graves Ophthalmopathy/drug therapy , Graves Ophthalmopathy/genetics , Graves Ophthalmopathy/radiotherapy , Graves Ophthalmopathy/surgery , Humans , Hyperthyroidism/blood , Hyperthyroidism/drug therapy , Iodine Radioisotopes/therapeutic use , Male , Methimazole/therapeutic use , Middle Aged , Rats , Receptors, Thyrotropin/metabolism , Reference Values , Severity of Illness Index , Thyroidectomy , Young Adult
3.
J Endocrinol ; 189(3): 605-15, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16731791

ABSTRACT

Increased major histocompatibility complex (MHC) class I gene expression in nonimmune cell 'target tissues' involved in organ-specific diseases may be important in the pathogenesis of autoimmune diseases. This possibility in part evolves from studies of cultured thyrocytes where properties appear relevant to the development of thyroid autoimmune disease. In FRTL-5 rat thyroid cells in continuous culture, hormones and growth factors that regulate cell growth and function specifically decrease MHC class I gene expression. We hypothesized that this could reflect a mechanism to preserve self-tolerance and prevent autoimmune disease. The mechanisms of action of some of these hormones, namely TSH and hydrocortisone, have been already characterized. In this report, we show that IGF-I transcriptionally downregulates MHC class I gene expression and that its action is similar to that of insulin. The two hormones have a complex effect on the promoter of the MHC class I gene, PD1. In fact, they decrease the full promoter activity, but upregulate the activity of deleted mutants that have lost an upstream, tissue-specific regulatory region but still retain the enhancer A region. We show that insulin/IGF-I promotes the interactions of the p50/p65 subunits of NF-kappaB and AP-1 family members with these two regions, and that the tissue-specific region acts as a dominant silencer element on insulin/IGF-I regulation of promoter activity. These observations may be important to understand how MHC class I gene transcription is regulated in the cells.


Subject(s)
Gene Expression Regulation/drug effects , Genes, MHC Class I , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Thyroid Gland/metabolism , Transcription, Genetic , Animals , Cell Line , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , Flow Cytometry , Fluorescent Antibody Technique , NF-kappa B/genetics , Promoter Regions, Genetic , Rats , Thyroid Gland/drug effects , Transcription Factor AP-1/genetics
4.
Clin Exp Immunol ; 127(2): 234-42, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11876745

ABSTRACT

Although infection of single-stranded RNA viruses can enhance expression of major histocompatibility complex (MHC) class I genes, the mechanism underlying this process remains unclear. Recent studies have indicated that exposure of non-immune cells to double-stranded deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) of viral origin can directly increase the expression of MHC class I and related molecules without immune cell interaction. In this report, we show that transfection of single-stranded hepatitis A virus RNA into cultured hepatocytes results in the induction of genes for MHC class I, LMP2 and transporter for antigen processing (TAP1), in addition to the generation of viral proteins. We suggest that this stimulatory effect is due to the double-stranded RNA formed during replication of single-stranded viral RNA, and involves both double-stranded, RNA-dependent protein kinase PKR and the secretion of IFNbeta.


Subject(s)
Gene Expression Regulation, Viral , Genes, MHC Class I , Hepatitis A virus/genetics , Hepatocytes/immunology , Histocompatibility Antigens Class I/biosynthesis , I-kappa B Proteins , RNA, Viral/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Cells, Cultured/immunology , DNA-Binding Proteins/metabolism , Hepatitis A virus/physiology , Hepatoblastoma/pathology , Humans , Interferon-beta/metabolism , Liver Neoplasms/pathology , NF-kappa B/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational , RNA, Double-Stranded/genetics , RNA, Double-Stranded/physiology , RNA, Messenger/biosynthesis , RNA, Viral/genetics , Transfection , Tumor Cells, Cultured/immunology , Viral Matrix Proteins/biosynthesis , Viral Matrix Proteins/genetics , Viral Proteins/biosynthesis , Viral Proteins/genetics , Virus Replication , eIF-2 Kinase/physiology
5.
J Clin Endocrinol Metab ; 86(8): 3647-53, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11502791

ABSTRACT

Spontaneous remission of Graves' disease during pregnancy is thought to be due to a reduction of thyroid-stimulating antibody activity. We suspected, however, that a broader change in TSH receptor antibody characteristics might play an important role in modulating disease activity during pregnancy. We measured TSH binding inhibitory Ig, thyroid-stimulating antibody, and thyroid stimulating-blocking antibody activities in 13 pregnant Graves' disease patients at first, second, and third trimesters and 4 months postpartum. To measure and epitope-map thyroid-stimulating antibody and thyroid stimulating-blocking antibody activities, we used CHO cells transfected with wild-type human TSH receptor or with several TSH receptor-LH/hCG receptor chimeras: Mc1+2, Mc2, and Mc4. These chimeric cells have their respective TSH receptor residues 9-165, 90-165, and 261-370 substituted with equivalent residues of the LH/hCG receptor. Overall thyroid-stimulating antibody decreased, whereas thyroid stimulating-blocking antibody increased progressively during pregnancy. TSH binding inhibitory Ig fluctuated in individual patients, but overall the activities remained statistically unchanged. Thyroid stimulating-blocking antibody appeared in subjects who were either negative for thyroid-stimulating antibody or whose thyroid-stimulating antibody activity increased or decreased during pregnancy. Epitope mapping showed that the thyroid-stimulating antibodies were mainly directed against residues 9-165 of the N-terminus of the TSH receptor extracellular domain. All thyroid stimulating-blocking antibodies had blocking activities against residues 261-370 of the C-terminus of the ectodomain. However, the majority of the thyroid stimulating-blocking antibodies had a hybrid conformational epitope directed against N-terminal residues 9-89 or 90-165 as well. Despite a change in the activity level, we did not observe any change in the epitope of either the stimulatory or blocking Abs as pregnancy advanced. In conclusion, a change in the specificity of TSH receptor antibody from stimulatory to blocking activity was observed during pregnancy, and the appearance of thyroid stimulating-blocking antibody may contribute to the remission of Graves' disease during pregnancy.


Subject(s)
Graves Disease/immunology , Immunoglobulins, Thyroid-Stimulating/blood , Pregnancy Complications/immunology , Receptors, Thyrotropin/immunology , Adult , Analysis of Variance , Antithyroid Agents/therapeutic use , Epitopes/analysis , Female , Graves Disease/blood , Graves Disease/drug therapy , Humans , Immunoglobulin G/blood , Postpartum Period/blood , Postpartum Period/immunology , Pregnancy , Pregnancy Complications/blood , Thyroid Function Tests , Thyrotropin/blood , Thyrotropin/immunology , Thyroxine/blood , Time Factors , Triiodothyronine/blood
6.
Cancer Res ; 61(16): 6105-11, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507060

ABSTRACT

Enhanced activation of Akt occurs in Cowden's disease, an inherited syndrome of follicular thyroid, breast, colon, and skin tumors, via inactivation of its regulatory protein, PTEN. Whereas PTEN inactivation is uncommon in sporadic thyroid cancer, activation of growth factor pathways that signal through Akt is frequently identified. We hypothesized that Akt overactivation could be a common finding in sporadic thyroid cancer and might be important in thyroid cancer biology. We examined thyroid cancer cells lines and benign and malignant thyroid tissue for total Akt activation and isoform-specific Akt expression. In thyroid cancer cells, Akt 1, 2, and 3 proteins were expressed, total Akt was activated by insulin phosphatidylinositol 3'-kinase, and inhibition of phosphatidylinositol 3'-kinase reduced cell viability. In human thyroid tissue, increased levels of phosphorylated total Akt were identified in follicular but not papillary cancers compared with normal tissue. Levels of Akt 1 and 2 proteins and Akt 2 RNA were elevated only in the follicular cancers. In paired samples, Akt 1, 2, 3, and phospho-Akt levels were higher in five of six cancers, including three of three follicular cancers. These data suggest that Akt activation may play a role in the pathogenesis or progression of sporadic thyroid cancer.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Thyroid Neoplasms/enzymology , Adenocarcinoma, Follicular/enzymology , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Carcinoma, Papillary/enzymology , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Cell Survival/physiology , Enzyme Activation , Gene Expression , Humans , Insulin/pharmacology , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/physiology , Thyroid Gland/enzymology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyrotropin/pharmacology , Tumor Cells, Cultured
7.
J Immunol ; 167(5): 2602-7, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11509601

ABSTRACT

Mature APCs play a key role in the induction of Ag-specific immunity. This work examines whether genomic DNA released by dying cells provides a stimulus for APC maturation. Double-stranded but not single-stranded genomic DNA triggered APC to up-regulate expression of MHC class I/II and various costimulatory molecules. Functionally, dsDNA enhanced APC function in vitro and improved primary cellular and humoral immune responses in vivo. These effects were dependent on the length and concentration of the dsDNA but were independent of nucleotide sequence. The maturation of APC induced by dsDNA may promote host survival by improving immune surveillance at sites of tissue injury/infection.


Subject(s)
Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Cell Death/immunology , DNA/immunology , DNA/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Base Sequence , Cell Death/drug effects , Cell Differentiation , Cell Line , DNA/genetics , DNA/metabolism , DNA Fragmentation , DNA, Bacterial/genetics , DNA, Bacterial/immunology , DNA, Bacterial/pharmacology , Female , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
8.
Biochem Biophys Res Commun ; 285(3): 795-9, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11453662

ABSTRACT

Thyroglobulin (Tg), has recently been identified as a transcriptional regulator of thyroid-restricted genes. The extrathyroidal expression of several of these genes (including the transcription factor Pax-8) together with the occurrence of specific Tg binding sites suggests a secondary role for Tg as a circulating hormone. In this study, we demonstrate using Northern analysis that Pax-8 is expressed in the mouse mesangial cell, and that its transcript levels are suppressed by Tg. These cells also express an asialoglycoprotein receptor, a receptor involved in Tg endocytosis in the thyroid, and a Tg transcript smaller than the 8.3-kb thyroidal form. Reverse transcriptase PCR showed that suppression of Pax-8 by Tg is correlated with reduced expression of bcl-2 apoptosis suppressor. Tg, but not triiodothyronine (T(3)) significantly increased MC proliferation above control as determined by DNA content of MC cultures. The effect of Tg on proliferation was not duplicated by either bovine serum albumin, gamma-globulins, lactoferrin, or the ASGPR-specific ligand,orosomucoid. These results suggest a possible endocrine role for Tg in regulating both Pax-8 related gene transcription and cell division in the mesangial cell.


Subject(s)
DNA-Binding Proteins/metabolism , Glomerular Mesangium/drug effects , Glomerular Mesangium/metabolism , Nuclear Proteins , Thyroglobulin/pharmacology , Trans-Activators/metabolism , Animals , Asialoglycoprotein Receptor , Blotting, Northern , Cell Division/drug effects , Cells, Cultured , DNA/metabolism , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Glomerular Mesangium/cytology , Mice , PAX8 Transcription Factor , Paired Box Transcription Factors , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Triiodothyronine/pharmacology
9.
Thyroid ; 11(4): 339-51, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11349832

ABSTRACT

Thyrotropin (TSH)-initiated cell cycle progression from G1 to S phase in FRTL-5 thyroid cells requires serum, insulin, or insulin-like growth factor 1 (IGF-1) and involves activation of 3-hydroxy-3-methylglutaryl-CoA reductase, geranylgeranylation of RhoA, p27Kip1 degradation, and activation of cyclin-dependent kinase (cdk) 2. In the present report, we show that the serine-threonine kinase Akt is an important mediator of insulin/IGF-1/serum effects on cell cycle progression in FRTL-5 thyroid cells. The phosphoinositol (OH) 3 kinase inhibitors, Wortmannin (WM) and Ly294002 (LY), block the ability of insulin/IGF-1 to reduce p27 expression, to induce expression of cyclins E, D1, and A as well as cdk 2 and 4, and to phosphorylate retinoblastoma protein. They also inhibit insulin/IGF-1-increased DNA synthesis and cell cycle entrance (S+G2/M). Insulin/IGF-1 rapidly induced activation of Aktl in a PI3 kinase-dependent manner, and increased Aktl RNA levels. Most importantly, FRTL-5 cells transfected with a constitutively active form of Aktl have higher basal rates of DNA synthesis and no longer require exogenous insulin/IGF-1 or serum for TSH-induced growth. In sum, Aktl appears to have an important role in insulin/IGF-1 regulation of FRTL-5 thyroid cell growth and cell cycle progression.


Subject(s)
Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Thyroid Gland/cytology , Animals , Cell Cycle , Cell Division , Cell Line , DNA/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats , Thyrotropin/pharmacology
10.
Trends Endocrinol Metab ; 12(1): 10-6, 2001.
Article in English | MEDLINE | ID: mdl-11137035

ABSTRACT

Iodide transport by thyrocytes involves porters on the apical and basal surfaces of the cell facing the follicular lumen and bloodstream, respectively. Recent work identifies pendrin as an apical porter and shows that follicular thyroglobulin is a transcriptional regulator of the gene encoding pendrin and other thyroid-restricted genes. For example, whereas follicular thyroglobulin suppresses the gene expression and activity of the sodium iodide symporter (NIS), it increases pendrin gene expression. A potential new dynamic for iodide flux and thyroid hormone formation in thyrocytes has thus emerged and is supported by in vivo data.


Subject(s)
Carrier Proteins/physiology , Iodides/metabolism , Membrane Transport Proteins , T-Lymphocytes/metabolism , Thyroglobulin/physiology , Animals , Carrier Proteins/biosynthesis , Homeostasis/physiology , Humans , Sulfate Transporters , Thyroglobulin/biosynthesis
11.
Int Rev Immunol ; 19(6): 619-31, 2000.
Article in English | MEDLINE | ID: mdl-11129118

ABSTRACT

Mice immunized with fibroblasts expressing an MHC class II molecule and human thyrotropin receptor (TSHR), but not either alone, develop major features characteristic of Graves' disease (GD), such as thyroid-stimulating autoantibodies directed against TSHR, increased serum thyroid hormone levels, and enlarged thyroid glands. The results indicate the need for the simultaneous expression of a class II molecule and the TSHR on the surface of the fibroblasts to develop stimulating anti-TSHR antibodies and full-blown GD in our model. A T cell line established from a mouse with hyperthyroidism proliferates in response to fibroblasts expressing a class II molecule and TSHR, but not to the fibroblasts expressing only TSHR, indicating that the class II molecules on the fibroblasts present TSHR-derived peptide(s) to T cells. These results strongly suggest that the acquisition of antigen-presenting ability by thyrocytes can lead to the induction or progression of GD. We identified a T cell epitope of TSHR by the proliferative response of spleen cells from mice immunized with fibroblasts expressing a class II molecule and TSHR to 80 overlapping peptides spanning the extracellular domain of human TSHR. The identification of a major T cell epitope provides an important clue to a novel therapy of GD.


Subject(s)
Graves Disease/immunology , Graves Disease/pathology , Histocompatibility Antigens Class II/immunology , Receptors, Thyrotropin/immunology , Animals , Antibody Formation , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Fibroblasts/immunology , Graves Disease/therapy , Histocompatibility Antigens Class II/genetics , Humans , Immunization , Immunotherapy , Mice , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Thyrotropin/genetics , T-Lymphocytes/immunology , Transfection
12.
Int Rev Immunol ; 19(6): 633-64, 2000.
Article in English | MEDLINE | ID: mdl-11129119

ABSTRACT

In this report we summarize evidence to support a model for the development of Graves' disease. The model suggests that Graves' disease is initiated by an insult to the thyrocyte in an individual with a normal immune system. The insult, infectious or otherwise, causes double strand DNA or RNA to enter the cytoplasm of the cell. This causes abnormal expression of major histocompatibility (MHC) class I as a dominant feature, but also aberrant expression of MHC class II, as well as changes in genes or gene products needed for the thyrocyte to become an antigen presenting cell (APC). These include increased expression of proteasome processing proteins (LMP2), transporters of antigen peptides (TAP), invariant chain (Ii), HLA-DM, and the co-stimulatory molecule, B7, as well as STAT and NF-kappaB activation. A critical factor in these changes is the loss of normal negative regulation of MHC class I, class II, and thyrotropin receptor (TSHR) gene expression, which is necessary to maintain self-tolerance during the normal changes in gene expression involved in hormonally-increased growth and function of the cell. Self-tolerance to the TSHR is maintained in normals because there is a population of CD8- cells which normally suppresses a population of CD4+ cells that can interact with the TSHR if thyrocytes become APCs. This is a host self-defense mechanism that we hypothesize leads to autoimmune disease in persons, for example, with a specific viral infection, a genetic predisposition, or even, possibly, a TSHR polymorphism. The model is suggested to be important to explain the development of other autoimmune diseases including systemic lupus or diabetes.


Subject(s)
Graves Disease/immunology , Animals , Antigen-Presenting Cells/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Gene Expression Regulation , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immune System/immunology , Self Tolerance/immunology , Thymus Gland/cytology , Thyroiditis, Autoimmune/immunology , Thyrotropin/immunology
13.
Biochem Biophys Res Commun ; 278(1): 58-62, 2000 Nov 11.
Article in English | MEDLINE | ID: mdl-11071855

ABSTRACT

Class II transactivator (CIITA) is the master regulator of MHC class II genes, and mediates their induction by interferon gamma (IFN gamma). To study the role of CIITA in modulating the expression of thyroid-specific genes, we cloned the full-length rat CIITA and use it to transfect a rat thyroid cell line. We found that only one type of CIITA, type IV, is induced in thyroid cells upon IFN gamma stimulation, and that CIITA is capable not only of inducing the expression of MHC genes in the thyroid, but also of differentially suppressing the expression of thyroid-specific genes. These findings suggest new avenues for the development of thyroid autoimmune diseases.


Subject(s)
Genes, MHC Class II/genetics , Nuclear Proteins , Thyroid Gland/metabolism , Trans-Activators/physiology , Transcription, Genetic , Transcriptional Activation , Animals , Antigen Presentation , Blotting, Northern , Cell Line , Cells, Cultured , Cloning, Molecular , DNA, Complementary/metabolism , Exons , Gene Library , Genes, MHC Class I/genetics , Interferon-gamma/metabolism , Models, Genetic , Promoter Regions, Genetic , Protein Isoforms , Rats , Spleen/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , Transfection
14.
Thyroid ; 10(10): 909-17, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11081257

ABSTRACT

Hypothyroidism after radioactive iodine (RAI) therapy for Graves' disease can be transient or permanent. The cause for early transient hypothyroidism is unknown. We evaluated 11 patients who developed transient hypothyroidism within 6 months of RAI and 12 who remained euthyroid after RAI. Approximately equal numbers of patients in each group had thyroid-stimulating antibody (TSAb) that increased cyclic adenosine monophosphate (cAMP) levels in Chinese hamster ovary (CHO) cells transfected with the recombinant human thyrotropin receptor (TSHR) (WT cells). Approximately equal numbers of patients from both groups had an increase in TSAb activity post-RAI. All TSAbs had their dominant functional epitope on the N-terminus of the TSHR extracellular domain, requiring residues 90-165 for activity because they, but not TSH, completely lost stimulating activity in a receptor chimera, wherein TSHR residues 90-165 were substituted by equivalent residues of the lutropin/choriogonadotropin receptor (LH/CGR). Although equal numbers of patients in both groups had thyrotropin-binding inhibiting immunoglobulin activity (TBII), as measured by radioreceptor assay before RAI, patients with transient hypothyroidism had a surge in TBII activity and all except one became positive for thyroid-stimulating blocking antibodies (TSBAb), as measured by inhibition of TSH-stimulated cAMP from WT cells. When immunoglobulin G (IgGs) were epitope-mapped using TSHR/LH-CGR chimeras with different substitutions, 8 hypothyroid subjects had TSBAbs directed against residues 90-165 of the TSHR, as well as TSHR residues 261-370. Two had functional epitopes directed at residues 9-89 as well as TSHR residues 261-370. None of the euthyroid control patients developed TSBAbs and their TBII activity decreased post-RAI. When patients with transient hypothyroidism reverted to a euthyroid state, TSAb was still detectable in 5; however, TBII was present in all and TSBAb, although decreased, was still positive in 9. In summary, RAI therapy was associated with a change in thyroid antibody characteristics in most patients. Additionally, patients with a surge in TBII and the appearance of TSBAb developed transient hypothyroidism after RAI.


Subject(s)
Antibodies, Blocking/immunology , Hypothyroidism/immunology , Immunoglobulins, Thyroid-Stimulating/immunology , Iodine Radioisotopes/therapeutic use , Thyrotoxicosis/radiotherapy , Adult , Aged , Animals , Antibodies, Blocking/metabolism , Autoantibodies/immunology , Autoantibodies/metabolism , CHO Cells , Cricetinae , Epitopes/immunology , Female , Humans , Immunoglobulin G/immunology , Immunoglobulins, Thyroid-Stimulating/metabolism , Male , Middle Aged , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/metabolism , Thyroid Gland/immunology , Thyrotoxicosis/immunology , Transfection
15.
Mol Cell Biol ; 20(22): 8499-512, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11046146

ABSTRACT

Follicular thyroglobulin (TG) selectively suppresses the expression of thyroid-restricted transcription factors, thereby altering the expression of thyroid-specific proteins. In this study, we investigated the molecular mechanism by which TG suppresses the prototypic thyroid-restricted transcription factor, thyroid transcription factor 1 (TTF-1), in rat FRTL-5 thyrocytes. We show that the region between bp -264 and -153 on the TTF-1 promoter contains two nuclear factor I (NFI) elements whose function is involved in TG-mediated suppression. Thus, NFI binding to these elements is critical for constitutive expression of TTF-1; TG decreases NFI binding to the NFI elements in association with TG repression. NFI is a family of transcription factors that is ubiquitously expressed and contributes to constitutive and cell-specific gene expression. In contrast to the contribution of NFI proteins to constitutive gene expression in other systems, we demonstrate that follicular TG transcriptionally represses all NFI RNAs (NFI-A, -B, -C, and -X) in association with decreased NFI binding and that the RNA levels decrease as early as 4 h after TG treatment. Although TG treatment for 48 h results in a decrease in NFI protein-DNA complexes measured in DNA mobility shift assays, NFI proteins are still detectable by Western analysis. We show, however, that the binding of all NFI proteins is redox regulated. Thus, diamide treatment of nuclear extracts strongly reduces the binding of NFI proteins, and the addition of higher concentrations of dithiothreitol to nuclear extracts from TG-treated cells restores NFI-DNA binding to levels in extracts from untreated cells. We conclude that NFI binding to two NFI elements, at bp -264 to -153, positively regulates TTF-1 expression and controls constitutive TTF-1 levels. TG mediates the repression of TTF-1 gene expression by decreasing NFI RNA and protein levels, as well as by altering the binding activity of NFI, which is redox controlled.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins , Nuclear Proteins/genetics , Thyroglobulin/metabolism , Transcription Factors/genetics , Amino Acid Sequence , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins/genetics , Cattle , Cells, Cultured , DNA/metabolism , Diamide/chemistry , Diamide/pharmacology , Gene Expression Regulation , Molecular Sequence Data , NFI Transcription Factors , Nuclear Proteins/metabolism , Oligonucleotides/metabolism , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Regulatory Sequences, Nucleic Acid , Thyroglobulin/genetics , Thyroglobulin/pharmacology , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/physiology , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism , Y-Box-Binding Protein 1
16.
Thyroid ; 10(7): 579-86, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10958310

ABSTRACT

The goal of this study was to evaluate the clinical significance of the blocking thyrotropin receptor antibodies (TSHRAb) in Graves' disease. The amount of blocking and stimulating TSHRAb were measured in 200 patients with untreated hyperthyroid Graves' disease using several cell lines carrying different TSHR chimera. Stimulating TSHRAb were measured in Chinese hamster ovary (CHO) cells with wild-type human TSHR (CHO-hTSHR) or a TSHR chimera with residues 90-165 (Mc2) or 8-165 (Mc1+2) substituted by equivalent residues of rat luteinizing hormone/chorionic gonadotrophin (LH/CG) receptor or in FRTL-5 cells. Blocking TSHRAb were measured in Mc2 cells. The activities of different TSHRAb were assessed and clinical features were compared to patients who were positive or negative for blocking TSHRAb antibodies. Blocking TSHRAbs were detected in 18.5% of patients (37/200) with hyperthyroid Graves' disease. Patients with blocking antibodies had significantly lower mean stimulating TSHRAb activities than those without blocking antibodies in wild-type CHO-hTSHR cells (301 +/- 179 vs. 446% +/- 537%, p = 0.005). Mean stimulating TSHRAb activities measured by FRTL-5, Mc1+2, or Mc2 cells and mean thyrotropin receptor inhibitor immunoglobulin (TBII) activities were not different between the two groups. The patients with blocking antibodies were not different from those without blocking antibodies in age, gender ratio, initial serum free thyroxine (T4) levels, or goiter size. However, the prevalence of exophthalmos was higher (35.1% vs. 17.5%, p = 0.024) in the patients with blocking antibodies than those without. In summary, the presence of blocking TSHRAb is not rare in patients with hyperthyroid Graves' disease when measured with chimeric receptor expressing cells. Blocking TSHRAb in Graves' sera do not strongly antagonize the action of stimulating TSHRAb in vivo, but could be a major factor responsible for underestimation of stimulating TSHRAb activities measured by CHO-hTSHR. The association of blocking TSHRAb with ophthalmopathy suggests that the TSHRAb repertoire of Graves' patients is different in those who do and who do not have ophthalmopathy.


Subject(s)
Autoantibodies/blood , Graves Disease/immunology , Receptors, Thyrotropin/blood , Adult , Animals , CHO Cells , Cell Line , Cricetinae , Female , Humans , Immunoglobulins, Thyroid-Stimulating , Male , Middle Aged , Rats , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Recombinant Fusion Proteins/immunology , Thyroid Gland/metabolism
17.
J Clin Endocrinol Metab ; 85(6): 2116-21, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10852437

ABSTRACT

Blocking-type TSH-binding inhibitor Igs (TBIIs) are known to cause hypothyroidism and an atrophic thyroid gland in patients with primary myxedema. They can block the activity of thyroid-stimulating antibodies (TSAbs) in Graves' patients as well as the activity of TSH. The majority of the epitopes for these blocking-type TBIIs have been, and are shown herein, to be present on the C-terminal region of the extracellular domain of the human TSH receptor (TSHR), whereas those for Graves' TSAbs are on the N-terminus. We report on a patient with Hashimoto's thyroiditis who suffered from mild hypothyroidism and a moderately sized goiter. Her serum had a potent blocking-type TBII and a weak TSAb in human and porcine TSHR systems. Using human TSHR/lutropin-CG receptor chimeras, we determined that the functional epitope of her blocking-type TBII was uniquely present on the N-terminal, rather than the C-terminal, region of the extracellular domain of the TSHR, unlike the case for blocking-type TBIIs in primary myxedema patients. The epitope of her TSAb was also unusual. Although the functional epitopes of most TSAbs are known to involve the N-terminal region of the receptor, her TSAb epitope did not seem to be present solely on the N- or C-terminus of the extracellular domain of the receptor. Blocking-type TBIIs from patients with primary myxedema blocked her TSAb activity as well as stimulation by TSH; her blocking-type TBII was able to only partially block her TSAb. In contrast, her blocking-type TBII almost completely blocked TSAbs from Graves' patients. Thus, we suggest that the unique epitopes of this patient's heterogeneous population of TSH receptor antibodies, at least in part, contribute to regulation of her thyroid function.


Subject(s)
Autoantibodies/blood , Immunoglobulins, Thyroid-Stimulating/blood , Receptors, Thyrotropin/blood , Receptors, Thyrotropin/physiology , Thyroid Gland/physiopathology , Thyroiditis, Autoimmune/immunology , Thyroiditis, Autoimmune/physiopathology , Animals , CHO Cells , Cricetinae , Epitopes/immunology , Female , Goiter/etiology , Goiter/immunology , Humans , Hypothyroidism/etiology , Hypothyroidism/immunology , Middle Aged , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Swine , Thyroid Gland/immunology , Thyroiditis, Autoimmune/blood , Transfection
18.
Thyroid ; 10(4): 295-303, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10807057

ABSTRACT

Thyrotropin (TSH), via its cyclic adenosine monophosphate (cAMP) signal, decreases thyrotropin receptor (TSHR) gene expression in FRTL-5 thyroid cells, whereas it increases expression of the thyroglobulin (Tg) gene. Despite the opposite effects of TSH on TSHR and Tg expression, both genes are positively controlled by thyroid transcription factor-1 (TTF-1) and evidence has accumulated that TSH can decrease TTF-1 mRNA levels. In this report, we further characterize the action of TSH on TTF-1 in order to understand its different activities on the TSHR and Tg genes better. The effect of TSH on the TSHR requires the presence of insulin and serum and we show here that also both factors are necessary for the TSH effect to decrease TTF-1 mRNA levels. The decrease is paralleled by a downregulation of TTF-1 protein levels as well as by a decrease in TTF-1/DNA complex when the TTF-1 site of the TSHR promoter was used as probe. Again, the decrease requires insulin and serum. The TSH downregulation of TTF-1 mRNA levels is due to a decrease in its transcription rate. Using a luciferase-linked chimera construct spanning 5.18 kb of the TTF-1 5'-flanking region, we show that TSH decreases TTF-1 promoter activity and that this effect depends on insulin and serum. These data contrast with the action of TSH on Tg and Pax-8 gene expression. TSH increases Pax-8 mRNA levels and the increase is evident whether insulin and serum are present or not. Moreover, this increase is paralleled by an increase in Pax-8 protein binding to an oligonucleotide derived from the C site of the Tg promoter, which can bind both TTF-1 and Pax-8. The present data thus show that TTF-1 gene expression is interdependently regulated by TSH and serum growth factors including insulin. They also show this interdependent-regulation is not duplicated in the case of Pax-8. We suggest that these differences may contribute to the distinct ability of TSH to regulate TSHR versus Tg gene expression in FRLT-5 thyroid cells.


Subject(s)
DNA-Binding Proteins/genetics , Insulin/metabolism , Nuclear Proteins/genetics , Thyroid Gland/metabolism , Thyrotropin/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Base Sequence , Cell Line , Culture Media , Cyclic AMP/pharmacology , DNA/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Nuclear Proteins/metabolism , PAX8 Transcription Factor , Paired Box Transcription Factors , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Thyroid Gland/drug effects , Thyroid Nuclear Factor 1 , Thyrotropin/pharmacology , Trans-Activators/metabolism , Transcription Factors/metabolism
19.
Thyroid ; 10(3): 219-25, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10779136

ABSTRACT

Thyroid disease has been associated with the occurrence of pathophysiologic changes in the vasculature that may result in part from altered serum thyroid hormone and serum lipid levels. Thyrotropin (TSH) levels are also altered in thyroid disease, but a direct effect of TSH on vascular smooth muscle has not previously been considered. In the present study, human coronary artery smooth muscle cells (CASMC) were induced into two morphologically distinct forms by culturing in either (1) growth factor supplemented, 0.5% serum medium (SmGM-3) or (2) basal medium (SmBM) plus 10% fetal bovine serum (FBS). Intracellular cyclic adenosine monophosphate (cAMP) accumulation was determined by radioimmunoassay after exposure to increasing doses of bovine TSH. Cells grown in SmBM/10% FBS for 3 days exhibited a dose-dependent increase in intracellular cAMP that reached a level 10 times higher than baseline at the highest dose examined (100 mIU/mL). In contrast, cells grown in SmGM-3 medium exhibited no change in intracellular cAMP on exposure to increasing TSII. Low serum (0.5% FBS) reduced the ability of TSH to stimulate cAMP above the control value in CASMC. Pretreatment of CASMC with either transforming growth factor-beta1 (TGF-beta1) or tumor necrosis factor-alpha (TNF-alpha) lowered basal levels of cAMP production, but did not inhibit the ability of TSH to stimulate cAMP production. Human, but not rat aortic smooth muscle cells in culture also responded to TSH with a significant increase in cAMP. The results of this study suggest that TSH may exert direct effects on vascular smooth muscle mediated by adenylate cyclase activation that could conceivably affect the progression of vascular disease associated with thyroid dysfunction.


Subject(s)
Coronary Vessels/metabolism , Cyclic AMP/metabolism , Growth Substances/pharmacology , Muscle, Smooth, Vascular/metabolism , Thyrotropin/pharmacology , Actins/genetics , Animals , Cattle , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Culture Media, Serum-Free , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Rats , Receptors, Thyrotropin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
20.
Mol Endocrinol ; 14(4): 486-505, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10770487

ABSTRACT

Transforming growth factor (TGF)-beta1-decreased major histocompatibility complex (MHC) class I gene expression in thyrocytes is transcriptional; it involves trans factors and cis elements important for hormone- as well as iodide-regulated thyroid growth and function. Thus, in rat FRTL-5 thyrocytes, TGF-beta1 regulates two elements within -203 bp of the transcription start site of the MHC class I 5'-flanking region: Enhancer A, -180 to -170 bp, and a downstream regulatory element (DRE), -127 to -90 bp, that contains a cAMP response element (CRE)-like sequence. TGF-beta1 reduces the interaction of a NF-kappaB p50/fra-2 heterodimer (MOD-1) with Enhancer A while increasing its interaction with a NF-kappaB p50/p65 heterodimer. Both reduced MOD-1 and increased p50/p65 suppresses class I expression. Decreased MOD-1 and increased p50/p65 have been separately associated with the ability of autoregulatory (high) concentrations of iodide to suppress thyrocyte growth and function, as well as MHC class I expression. TGF-beta1 has two effects on the downstream regulatory element (DRE). It increases DRE binding of a ubiquitously expressed Y-box protein, termed TSEP-1 (TSHR suppressor element binding protein-1) in rat thyroid cells; TSEP-1 has been shown separately to be an important suppressor of the TSH receptor (TSHR) in addition to MHC class I and class II expression. It also decreases the binding of a thyroid-specific trans factor, thyroid transcription factor-1 (TTF-1), to the DRE, reflecting the ability of TGF-beta1 to decrease TTF-1 RNA levels. TGF-beta1-decreased TTF-1 expression accounts in part for TGF-beta1-decreased thyroid growth and function, since decreased TTF-1 has been shown to decrease thyroglobulin, thyroperoxidase, sodium iodide symporter, and TSHR gene expression, coincident with decreased MHC class I. Finally, we show that TGF-beta1 increases c-jun RNA levels and induces the formation of new complexes involving c-jun, fra-2, ATF-1, and c-fos, which react with Enhancer A and the DRE. TGF-beta1 effects on c-jun may be a pivotal fulcrum in the hitherto unrecognized coordinate regulation of Enhancer A and the DRE.


Subject(s)
Antimicrobial Cationic Peptides , CCAAT-Enhancer-Binding Proteins , Gene Expression Regulation/drug effects , Genes, MHC Class I , Thyroid Gland/immunology , Transcription Factors/pharmacology , Transforming Growth Factor beta/pharmacology , Animals , Cell Line , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Enhancer Elements, Genetic , Humans , NF-kappa B/metabolism , NF-kappa B/pharmacology , NFI Transcription Factors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Regulatory Sequences, Nucleic Acid , Response Elements , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolism , Y-Box-Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...