Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
J Endocrinol Invest ; 44(5): 969-977, 2021 May.
Article in English | MEDLINE | ID: mdl-32808162

ABSTRACT

BACKGROUND: The homeostatic euthyroid set point of the hypothalamus-pituitary-thyroid axis of any given individual is unique and oscillates narrowly within substantially broader normal population ranges of circulating free thyroxine (FT4) and thyroid-stimulating hormone (TSH), otherwise termed 'thyroid function test (TFT)'. We developed a mathematical algorithm codenamed Thyroid-SPOT that effectively reconstructs the personalized set point in open-loop situations and evaluated its performance in a retrospective patient sample. METHODS: We computed the set points of 101 patients who underwent total thyroidectomy for non-functioning thyroid disease using Thyroid-SPOT on each patient's own serial post-thyroidectomy TFT. Every predicted set point was compared against its respective healthy pre-operative euthyroid TFT per individual and their separation (i.e. predicted-observed TFT) quantified. RESULTS: Bland-Altman analysis to measure the agreement between each pair of an individual's predicted and actual set points revealed a mean difference in FT4 and TSH of + 3.03 pmol/L (95% CI 2.64, 3.43) and - 0.03 mIU/L (95% CI - 0.25, 0.19), respectively. These differences are small compared to the width of the reference intervals. Thyroid-SPOT can predict the euthyroid set point remarkably well, especially for TSH with a 10-16-fold spread in magnitude between population normal limits. CONCLUSION: Every individual's equilibrium euthyroid set point is unique. Thyroid-SPOT serves as an accurate, precise and reliable targeting system for optimal personalized restoration of euthyroidism. This algorithm can guide clinicians in L-thyroxine dose titrations to resolve persistent dysthyroid symptoms among challenging cases harbouring "normal TFT" within the laboratory ranges but differing significantly from their actual euthyroid set points.


Subject(s)
Thyroid Diseases/surgery , Thyroid Gland , Thyroidectomy , Thyrotropin-Releasing Hormone/blood , Thyrotropin/blood , Thyroxine , Algorithms , Drug Dosage Calculations , Female , Hormone Replacement Therapy/methods , Humans , Hypothalamo-Hypophyseal System/metabolism , Male , Middle Aged , Postoperative Period , Reference Values , Thyroid Diseases/diagnosis , Thyroid Diseases/physiopathology , Thyroid Function Tests/methods , Thyroid Gland/metabolism , Thyroid Gland/surgery , Thyroidectomy/adverse effects , Thyroidectomy/methods , Thyroxine/blood , Thyroxine/pharmacology
2.
J Endocrinol Invest ; 26(8): 780-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14669837

ABSTRACT

Thyroid hormone plays important roles in metabolism, growth, and differentiation. Germline mutations in thyroid hormone receptor beta (TRbeta) have been identified in many individuals with resistance to thyroid hormone (RTH), a syndrome of hyposensitivity to T3. However, it has become increasingly apparent that somatic mutations can also occur in individual tissues, and are associated with tumors and malignancies in man. Herein we review the occurrence and identification of germline and somatic TR mutations and characterization of their pathological effects on hormone resistance and tumorigenesis.


Subject(s)
Germ-Line Mutation/genetics , Mutation/physiology , Receptors, Thyroid Hormone/genetics , Animals , Humans , Pituitary Neoplasms/genetics , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Neoplasms/genetics , Transcription, Genetic
3.
Thyroid ; 12(1): 81-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11838736

ABSTRACT

Resistance to thyroid hormone (RTH) is a clinical syndrome characterized by elevated serum thyroid hormone (TH) levels, unsuppressed thyrotropin (TSH) levels, and tissue hyposensitivity to TH. In almost all cases, the genetic basis of RTH lies in mutation of one of the two TH receptor beta (TRbeta) alleles. Recently, patients from several families with phenotypic manifestations of RTH in the absence of TR mutations have been described. We report a case of a 31-year-old woman who presented with goiter, tachycardia, elevated TH levels, unsuppressed TSH, and "inappropriately normal" levels of peripheral TH action markers. In two separate clinical evaluations, the patient exhibited typical clinical and biochemical evidence for peripheral and pituitary RTH. Surprisingly, reverse transcriptase-polymerase chain reaction (RT-PCR) of full-length TRalpha and TRbeta mRNAs, and genomic PCR using primers flanking exons encoding the carboxy-terminal region of TRbeta failed to demonstrate mutations in the TRalpha or TRbeta genes. It is likely that defects in the regulation of TR genes or mutations in transcriptional cofactors involved in TR signaling account for this patient's phenotype.


Subject(s)
Mutation , Receptors, Thyroid Hormone/genetics , Thyroid Hormone Receptors beta , Thyroid Hormone Resistance Syndrome/genetics , Adult , DNA-Binding Proteins/genetics , Fatigue , Female , Goiter , Humans , Intelligence Tests , National Institutes of Health (U.S.) , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tachycardia , Thyroid Hormone Resistance Syndrome/diagnosis , Thyroid Hormones/blood , Thyrotropin/blood , Thyrotropin-Releasing Hormone , Triiodothyronine , United States , Weight Loss
4.
J Clin Endocrinol Metab ; 86(11): 5142-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11701667

ABSTRACT

Resistance to thyroid hormone (RTH) is a syndrome in which patients have elevated thyroid hormone (TH) levels and decreased sensitivity to its action. We describe a child with extreme RTH and a severe phenotype. A 22-month-old female presented to the NIH with goiter, growth retardation, short stature, and deafness. Additionally, the patient had hypotonia, mental retardation, visual impairment, and a history of seizures. Brain magnetic resonance imaging showed evidence of demyelination and bilateral ventricular enlargement. The patient had markedly elevated free T3 and free T4 levels of more than 2000 pg/dl (normal, 230-420 pg/dl) and more than 64 pmol/liter (normal, 10.3-20.6 pmol/liter), respectively, and TSH of 6.88 mU/liter (normal, 0.6-6.3 mU/liter). These are the highest TH levels reported for a heterozygous RTH patient. A T3 stimulation test confirmed the diagnosis of RTH in the pituitary and peripheral tissues. Molecular analyses of the patient's genomic DNA by PCR identified a single base deletion in exon 10 of her TRbeta gene that resulted in a frameshift and early stop codon. This, in turn, encoded a truncated receptor that lacked the last 20 amino acids. Cotransfection studies showed that the mutant TR was transcriptionally inactive even in the presence of 10(-6) M T3 and had strong dominant negative activity over the wild-type receptor. It is likely that the severely defective TRbeta mutant contributed to the extreme RTH phenotype and resistance in our patient.


Subject(s)
Thyroid Hormone Resistance Syndrome/genetics , Bone Development/physiology , Brain/pathology , Cells, Cultured , Female , Frameshift Mutation , Gene Deletion , Humans , Infant , Magnetic Resonance Imaging , Phenotype , Thyroxine/blood , Transcription, Genetic/genetics , Transfection , Triiodothyronine/blood
5.
J Clin Endocrinol Metab ; 86(11): 5572-6, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11701737

ABSTRACT

In patients with TSH-secreting tumors (TSHomas), serum TSH is poorly suppressed by thyroid hormone. The mechanism for this defect in negative regulation of TSH secretion is not known. To investigate the possibility of a somatic mutation of TR causing this defect, we performed mutational analysis of TRbeta by RT-PCR using RNA obtained from five surgically resected TSHomas. In one TSHoma, we identified a somatic mutation in the ligand-binding domain of TRbeta that caused a His to Tyr substitution at codon 435 of TRbeta1 corresponding to codon 450 of TRbeta2. Interestingly, this mutation occurred in the same codon as two mutations (TRbetaH435L and H435Q) previously identified in patients with the syndrome of resistance to thyroid hormone. This mutant TRbeta had impaired T3 binding and T3-mediated negative regulation. It also blocked the negative regulation by wild-type TRbeta2 on glycoprotein hormone alpha-subunit and TSHbeta reporter genes in cotransfection studies. Our results demonstrate that somatic mutation of TRbeta occurred in a TSHoma and was probably responsible for the defect in negative regulation of TSH by thyroid hormone in the tumor.


Subject(s)
Mutation/genetics , Mutation/physiology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Receptors, Thyroid Hormone/genetics , Thyrotropin/metabolism , Adult , Aged , Cells, Cultured , DNA/genetics , DNA Mutational Analysis , Female , Genetic Vectors , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Hormone Resistance Syndrome/genetics , Transfection , Triiodothyronine/metabolism
6.
Mol Endocrinol ; 15(9): 1529-38, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11518802

ABSTRACT

Patients with TSH-secreting pituitary tumors (TSHomas) have high serum TSH levels despite elevated thyroid hormone levels. The mechanism for this defect in the negative regulation of TSH secretion is not known. We performed RT-PCR to detect mutations in TRbeta from a surgically resected TSHoma. Analyses of the RT-PCR products revealed a 135-bp deletion within the sixth exon that encodes the ligand-binding domain of TRbeta2. This deletion was caused by alternative splicing of TRbeta2 mRNA, as near-consensus splice sequences were found at the junction site and no deletion or mutations were detected in the tumoral genomic DNA. This TRbeta variant (TRbeta2spl) lacked thyroid hormone binding and had impaired T3-dependent negative regulation of both TSHbeta and glycoprotein hormone alpha-subunit genes in cotransfection studies. Furthermore, TRbeta2spl showed dominant negative activity against the wild-type TRbeta2. These findings strongly suggest that aberrant alternative splicing of TRbeta2 mRNA generated an abnormal TR protein that accounted for the defective negative regulation of TSH in the TSHoma. This is the first example of aberrant alternative splicing of a nuclear hormone receptor causing hormonal dysregulation. This novel posttranscriptional mechanism for generating abnormal receptors may occur in other hormone-resistant states or tumors in which no receptor mutation is detected in genomic DNA.


Subject(s)
Alternative Splicing/genetics , Gene Expression Regulation, Neoplastic , Pituitary Neoplasms/genetics , Receptors, Thyroid Hormone/genetics , Thyrotropin/metabolism , Adenoma , Aged , Animals , Female , Genes, Reporter , Humans , Pituitary Neoplasms/metabolism , Protein Isoforms , Receptors, Thyroid Hormone/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thyrotropin/blood , Triiodothyronine/administration & dosage , Two-Hybrid System Techniques
7.
Physiol Rev ; 81(3): 1097-142, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11427693

ABSTRACT

Thyroid hormones (THs) play critical roles in the differentiation, growth, metabolism, and physiological function of virtually all tissues. TH binds to receptors that are ligand-regulatable transcription factors belonging to the nuclear hormone receptor superfamily. Tremendous progress has been made recently in our understanding of the molecular mechanisms that underlie TH action. In this review, we present the major advances in our knowledge of the molecular mechanisms of TH action and their implications for TH action in specific tissues, resistance to thyroid hormone syndrome, and genetically engineered mouse models.


Subject(s)
Thyroid Hormones/physiology , Amino Acid Sequence , Animals , DNA/metabolism , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Receptors, Thyroid Hormone/physiology , Structure-Activity Relationship
8.
J Endocrinol Invest ; 24(5): 361-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11407657

ABSTRACT

The measurement of plasma CT has an important role as a screening test for medullary thyroid carcinoma (MTC) in patients with thyroid nodules. However, elevated plasma CT levels should be interpreted within the context of the overall clinical picture in each individual case and carefully validated before therapeutic decisions are made. We present the case of a 17-yr-old girl who was referred to us with a thyroid nodule and elevated plasma CT levels, as measured by a one-site RIA not involving prior plasma extraction. Plasma CT was re-measured using two different methods, a RIA with prior plasma extraction and a two-site immunochemiluminometric assay (ICMA), and was either very low or undetectable. Subsequently, samples were re-assayed using the initially applied CT RIA; plasma CT levels were again found to be elevated. These elevations were of a spurious nature, probably caused by the presence of an unidentified substance in the patient's plasma interfering with the measurement of CT in the initially used RIA. Our patient was eventually diagnosed with Hashimoto's thyroiditis, and had no evidence of MTC. As several conditions can cause either true or spurious hypercalcitoninemia, we suggest that elevated plasma CT levels should be confirmed at least once before other extensive diagnostic investigations are initiated or thyroidectomy is recommended. Finally, the assay selected should detect only the mature CT molecule.


Subject(s)
Calcitonin/blood , Carcinoma, Medullary/diagnosis , Thyroid Neoplasms/diagnosis , Thyroid Nodule/blood , Adolescent , Antibodies, Anti-Idiotypic/blood , Biopsy, Needle , False Positive Reactions , Female , Humans , Immunoassay , Luminescent Measurements , Magnetic Resonance Imaging , Multiple Endocrine Neoplasia Type 2a/genetics , Radioimmunoassay , Thyroiditis, Autoimmune/diagnosis , Tomography, X-Ray Computed , Ultrasonography
9.
J Biol Chem ; 276(18): 15066-72, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11328825

ABSTRACT

Unliganded thyroid hormone receptors (TRs) interact with corepressors and repress basal transcription of target genes in cotransfection and in vitro studies. Currently, little is known about the function of corepressors in vivo. We thus used a mouse albumin promoter to generate several transgenic mouse lines that overexpressed a dominant negative mutant corepressor, NCoRi, in liver. The transgenic mice had normal liver weight, appearance, and minimal changes in enzyme activity. To study the effects of NCoRi on transcription of hepatic target genes, we examined T3-regulated gene expression of hypo- and hyperthyroid transgenic mice. In hypothyroid mice, hepatic expression of Spot 14, Bcl-3, glucose 6-phosphatase, and 5'-deiodinase mRNA was higher in transgenic mice than littermate controls whereas these genes were induced to similar levels in T3-treated mice. Derepression was not observed for malic enzyme mRNA expression in hypothyroid mice. Thus, NCoRi selectively blocked basal transcription of several thyroid hormone-responsive genes but had no effect on ligand-mediated transcription. Additionally, compensatory increases in endogenous SMRT and NCoR mRNA were observed in hypothyroid transgenic mice. Interestingly, hepatocyte proliferation as detected by BrdUrd incorporation was increased in transgenic mice. The gene profile in transgenic mouse livers was studied by cDNA microarray, and several genes related to cell proliferation were induced. In summary, our studies show that NCoR plays important roles in mediating basal repression by TRs and may prevent cellular proliferation in vivo.


Subject(s)
Cell Division/physiology , Liver/metabolism , Receptors, Thyroid Hormone/physiology , Repressor Proteins/metabolism , Animals , Gene Expression , Mice , Mice, Transgenic , Repressor Proteins/genetics
10.
Mol Endocrinol ; 15(4): 485-500, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11266502

ABSTRACT

The glucocorticoid receptor interacting protein-1 (GRIP1) is a member of the steroid receptor coactivator (SRC) family of transcriptional regulators. Green fluorescent protein (GFP) fusions were made to full-length GRIP1, and a series of GRIP1 mutants lacking the defined regulatory regions and the intracellular distribution of these proteins was studied in HeLa cells. The distribution of GRIP1 was complex, ranging from diffuse nucleoplasmic to discrete intranuclear foci. Formation of these foci was dependent on the C-terminal region of GRIP1, which contains the two characterized transcriptional activation domains, AD1 and AD2. A subpopulation of GRIP1 foci associate with ND10s, small nuclear bodies that contain several proteins including PML, SP100, DAXX, and CREB-binding protein (CBP). Association with the ND10s is dependent on the AD1 of GRIP1, a region of the protein previously described as a CBP-interacting domain. The GRIP1 foci are enriched in components of the 26S proteasome, including the core 20S proteasome, PA28alpha, and ubiquitin. In addition, the irreversible proteasome inhibitor lactacystin induced an increase in the total fluorescence intensity of the GFP-GRIP1 expressing cells, demonstrating that GRIP1 is degraded by the proteasome. These findings suggest the intriguing possibility that degradation of GRIP1 by the 26S proteasome may be a key component of its regulation.


Subject(s)
Acetylcysteine/analogs & derivatives , Antigens, Nuclear , Cell Nucleus Structures/metabolism , Intracellular Signaling Peptides and Proteins , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex , Transcription Factors/metabolism , Acetylcysteine/pharmacology , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Autoantigens/metabolism , Base Sequence , Binding Sites , CREB-Binding Protein , Carrier Proteins/metabolism , Co-Repressor Proteins , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Chaperones , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 2 , Nuclear Receptor Coactivator 3 , Peptide Hydrolases/drug effects , Promyelocytic Leukemia Protein , Protease Inhibitors/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Trans-Activators/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins , Ubiquitins/metabolism
11.
J Biol Chem ; 276(14): 11237-45, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11152480

ABSTRACT

In this report, we have studied the intracellular dynamics and distribution of the thyroid hormone receptor-beta (TRbeta) in living cells, utilizing fusions to the green fluorescent protein. Wild-type TRbeta was mostly nuclear in both the absence and presence of triiodothyronine; however, triiodothyronine induced a nuclear reorganization of TRbeta. By mutating defined regions of TRbeta, we found that both nuclear corepressor and retinoid X receptor are involved in maintaining the unliganded receptor within the nucleus. A TRbeta mutant defective in DNA binding had only a slightly altered nuclear/cytoplasmic distribution compared with wild-type TRbeta; thus, site-specific DNA binding is not essential for maintaining TRbeta within the nucleus. Both ATP depletion studies and heterokaryon analysis demonstrated that TRbeta rapidly shuttles between the nuclear and the cytoplasmic compartments. Cotransfection of nuclear corepressor and retinoid X receptor markedly decreased the shuttling by maintaining unliganded TRbeta within the nucleus. In summary, our findings demonstrate that TRbeta rapidly shuttles between the nucleus and the cytoplasm and that protein-protein interactions of TRbeta with various cofactors, rather than specific DNA interactions, play the predominant role in determining the intracellular distribution of the receptor.


Subject(s)
Receptors, Thyroid Hormone/metabolism , Biological Transport , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA/metabolism , HeLa Cells , Humans , Mutation , Protein Binding , Receptors, Thyroid Hormone/genetics , Signal Transduction , Transfection
12.
Mol Endocrinol ; 14(7): 947-55, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10894146

ABSTRACT

The liver is an important target organ of thyroid hormone. However, only a limited number of hepatic target genes have been identified, and little is known about the pattern of their regulation by thyroid hormone. We used a quantitative fluorescent cDNA microarray to identify novel hepatic genes regulated by thyroid hormone. Fluorescent-labeled cDNA prepared from hepatic RNA of T3-treated and hypothyroid mice was hybridized to a cDNA microarray, representing 2225 different mouse genes, followed by computer analysis to compare relative changes in gene expression. Fifty five genes, 45 not previously known to be thyroid hormone-responsive genes, were found to be regulated by thyroid hormone. Among them, 14 were positively regulated by thyroid hormone, and unexpectedly, 41 were negatively regulated. The expression of 8 of these genes was confirmed by Northern blot analyses. Thyroid hormone affected gene expression for a diverse range of cellular pathways and functions, including gluconeogenesis, lipogenesis, insulin signaling, adenylate cyclase signaling, cell proliferation, and apoptosis. This is the first application of the microarray technique to study hormonal regulation of gene expression in vivo and should prove to be a powerful tool for future studies of hormone and drug action.


Subject(s)
Gene Expression Regulation , Liver/physiology , Oligonucleotide Array Sequence Analysis/methods , Thyroid Hormones/metabolism , Adenylyl Cyclases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , DNA, Complementary , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Glucose-6-Phosphatase/drug effects , Glucose-6-Phosphatase/genetics , Insulin/metabolism , Lipid Metabolism , Liver/drug effects , Mice , Thyroid Hormones/pharmacology , Triiodothyronine/metabolism , Triiodothyronine/pharmacology
13.
J Clin Endocrinol Metab ; 85(1): 237-44, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634393

ABSTRACT

The optimal treatment of metastatic thyroid cancer that produces high amounts of thyroid hormone has not been well defined. A 46-yr-old woman presented with a follicular thyroid carcinoma arising from a struma ovarii with hepatic metastases. After the removal of both the struma and the thyroid gland, the liver metastases showed evidence of a high degree of hormonogenesis. Brain, chest, abdomen, and bone imaging was negative for additional metastases. Because iodine uptake by most thyroid carcinomas is quite low in the absence of high levels of ambient TSH, we used recombinant human TSH (rhTSH) (Thyrogen) to achieve a concentration of 131I activity in the tumor high enough for a significant cytotoxic effect. After rhTSH administration (0.9 mg im daily for 2 consecutive days), a 131I diagnostic whole body scan confirmed the existence of 17 discrete hepatic foci of 131I uptake. To calculate the amount of 131I that would deliver an absorbed radiation dose that would be optimally cytotoxic to the metastases (>8000 rad/lesion) and not to the normal liver, we performed lesion dosimetry. Analysis of dosimetric data showed that 15 of 17 lesions would receive an adequate radiation dose following the administration of 65 mCi of 131I. Additionally, we performed whole body dosimetry to assure that this dose would not cause bone marrow toxicity. The patient was reevaluated 6 months after therapy; the liver metastases showed significant, but partial, response. In conclusion, we used the combination of rhTSH with lesional and whole body dosimetry for the treatment of highly functional metastases from follicular thyroid carcinoma arising within a struma ovarii. This strategy can be applied to determine a safe and effective dose of 131I for the treatment of any thyroid cancer metastases that produce enough TH to preclude stimulation of endogenous pituitary TSH secretion.


Subject(s)
Ovarian Neoplasms/secondary , Struma Ovarii/secondary , Teratoma/pathology , Thyroid Neoplasms/pathology , Thyrotropin/therapeutic use , Female , Humans , Iodine Radioisotopes/therapeutic use , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Magnetic Resonance Imaging , Middle Aged , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/radiotherapy , Recombinant Proteins/therapeutic use , Struma Ovarii/diagnosis , Struma Ovarii/radiotherapy
15.
EMBO J ; 18(19): 5389-98, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10508171

ABSTRACT

We have investigated ligand-dependent negative regulation of the thyroid-stimulating hormone beta (TSHbeta) gene. Thyroid hormone (T3) markedly repressed activity of the TSHbeta promoter that had been stably integrated into GH(3 )pituitary cells, through the conserved negative regulatory element (NRE) in the promoter. By DNA affinity binding assay, we show that the NRE constitutively binds to the histone deacetylase 1 (HDAC1) present in GH(3 )cells. Significantly, upon addition of T3, the NRE further recruited the thyroid hormone receptor (TRbeta) and another deacetylase, HDAC2. This recruitment coincided with an alteration of in vivo chromatin structure, as revealed by changes in restriction site accessibility. Supporting the direct interaction between TR and HDAC, in vitro assays showed that TR, through its DNA binding domain, strongly bound to HDAC2. Consistent with the role for HDACs in negative regulation, an inhibitor of the enzymes, trichostatin A, attenuated T3-dependent promoter repression. We suggest that ligand-dependent histone deacetylase recruitment is a mechanism of the negative-feedback regulation, a critical function of the pituitary-thyroid axis.


Subject(s)
Feedback , Histone Deacetylases/metabolism , Thyrotropin/genetics , Base Sequence , Chromatin/chemistry , Cyclic AMP/pharmacology , DNA , Gene Expression Regulation/drug effects , Genes, Reporter , Ligands , Protein Binding , Regulatory Sequences, Nucleic Acid , Sequence Deletion , Sequence Homology, Nucleic Acid , Thyrotropin/metabolism , Triiodothyronine/pharmacology
16.
Graefes Arch Clin Exp Ophthalmol ; 237(3): 231-40, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10090587

ABSTRACT

PURPOSE: To determine whether human trabecular meshwork cells (HTM) are a potential target tissue for thyroid hormone (3,3',5-triiodothyronine, T3). METHODS: Cultured HTM were assayed for the presence of thyroid hormone receptors (TRs) and retinoid X receptors (RXRs) by reverse-transcriptase polymerase chain reaction (RT-PCR) to detected TR and RXR mRNA, and by immunohistochemistry to detect nuclear TR and RXR proteins. Functionality of the TR was determined by analysis of 125I-T3 binding affinity and capacity in HTM nuclear extracts. Effects of T3 on modulation of hyaluronic acid (HA) levels in HTM were measured as a function of dose and duration of T3 administration. RESULTS: Analysis of RT-PCR and immunohistochemistry demonstrated that cultured HTM expressed TRalpha1, TRalpha2, and TRbeta1 but not TRbeta2; and RXRalpha but not RXRbeta and RXRgamma isoforms. Saturation binding and analysis of 125I-T3 to HTM nuclear extracts revealed Kd of 57 pM. The number of T3 binding sites extrapolated from a Scatchard plot was 7.3 x 10(10)/microg of HTM nuclear protein extract. T3 supplementation reduced the concentration of HA in the cell medium by 32-43% compared to cells grown in the absence of T3. CONCLUSIONS: Cultured HTM express three TR isoforms and one RXR isoform, bind T3 with an affinity similar to that of TR in responsive cells, and modulate their HA production in response to T3. These findings indicate that the human trabecular meshwork tissue has the capacity to respond to thyroid hormones.


Subject(s)
Receptors, Thyroid Hormone/metabolism , Trabecular Meshwork/metabolism , Adult , Cell Count/drug effects , Cells, Cultured/drug effects , DNA Primers/chemistry , Enzyme-Linked Immunosorbent Assay , Humans , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/analysis , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/drug effects , Receptors, Thyroid Hormone/genetics , Retinoid X Receptors , Reverse Transcriptase Polymerase Chain Reaction , Trabecular Meshwork/cytology , Trabecular Meshwork/drug effects , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Triiodothyronine/pharmacology
17.
J Biomed Sci ; 6(2): 71-8, 1999.
Article in English | MEDLINE | ID: mdl-10087437

ABSTRACT

Thyroid hormones (L-triiodothyronine, T3; L-tetraiodothyronine, T4) regulate normal cellular growth and development, and general metabolism as well. Their various actions are mediated by the thyroid hormone receptor, a ligand-dependent transcriptional factor belonging to the nuclear hormone receptor superfamily. The recent discovery of coregulators (coactivators, corepressors, and cointegrators) has greatly enhanced our understanding of thyroid hormone receptor functions. Hence we review and discuss, in brief, the potential role of thyroid hormone receptor coregulators involved in diverse cellular activities.


Subject(s)
Receptors, Thyroid Hormone/physiology , Transcription Factors/physiology , Animals , Humans
18.
Endocrinology ; 140(3): 1356-64, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10067863

ABSTRACT

This study is designed to clarify the role of an orphan nuclear hormone receptor, ROR alpha, on thyroid hormone (TH) receptor (TR)-mediated transcription on a TH-response element (TRE). A transient transfection study using various TREs [i.e., F2 (chick lysozyme TRE), DR4 (direct repeat), and palindrome TRE] and TR and ROR alpha1 was performed. When ROR alpha1 and TR were cotransfected into CV1 cells, ROR alpha1 enhanced the transactivation by liganded-TR on all TREs tested without an effect on basal repression by unliganded TR. By electrophoretic mobility shift assay, on the other hand, although ROR alpha bound to all TREs tested as a monomer, no (or weak) TR and ROR alpha1 heterodimer formation was observed on various TREs except when a putative ROR-response element was present. The transactivation by ROR alpha1 on a ROR-response element, which does not contain a TRE, was not enhanced by TR. The effect of ROR alpha1 on the TREs is unique, because, whereas other nuclear hormone receptors (such as vitamin D receptor) may competitively bind to TRE to exert dominant negative function, ROR alpha1 augmented TR action. These results indicate that ROR alpha1 may modify the effect of liganded TR on TH-responsive genes. Because TR and ROR alpha are coexpressed in cerebellar Purkinje cells, and perinatal hypothyroid animals and ROR alpha-disrupted animals show similar abnormalities of this cell type, cross-talk between these two receptors may play a critical role in Purkinje cell differentiation.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Response Elements/physiology , Thyrotropin-Releasing Hormone/physiology , Trans-Activators/physiology , Transcriptional Activation , Animals , Humans , Mice , Nuclear Receptor Subfamily 1, Group F, Member 1 , Rats , Sequence Analysis, DNA
19.
J Biol Chem ; 273(50): 33166-73, 1998 Dec 11.
Article in English | MEDLINE | ID: mdl-9837884

ABSTRACT

We previously reported that the responsiveness of hepatocytes to thyroid hormone is markedly attenuated when they were cultured as monolayers rather than spheroids. To elucidate the mechanisms underlying the altered responsiveness, thyroid hormone receptor auxiliary proteins in the hepatocytes were analyzed by electrophoretic mobility shift assay. The major thyroid hormone receptor auxiliary protein was identified as 9-cis-retinoic acid receptor alpha (RXRalpha) in the hepatocytes regardless of the culture conditions. The cytoplasmic fraction was shown to contain a protease(s) that cleaves RXRalpha at its amino terminus. The presence of the protease in the cytosol, but not in the nucleus, was ascertained by incubating full-length 35S-labeled RXRalpha with each fraction. Using various protease inhibitors, it was shown that cathepsin L-type protease could participate in the cleavage of the RXRalpha. The enzyme activity was much higher in the monolayers than the spheroids. Inhibition of this enzyme activity in the monolayer hepatocyte resulted in the increase of nuclear RXRalpha protein and the augmentation of T3-dependent induction of spot 14 mRNA. These results suggest that the changes in cathepsin L-type protease activity in the cytosol may alter the turnover of RXRalpha in the nucleus and modify the function of steroid receptor superfamilies that heterodimerize with RXRalpha.


Subject(s)
Cathepsins/metabolism , Endopeptidases , Receptors, Retinoic Acid/metabolism , Thyroid Hormones/metabolism , Transcription Factors/metabolism , Animals , Cathepsin L , Cell Nucleus/metabolism , Cells, Cultured , Cysteine Endopeptidases , Hydrolysis , Liver/cytology , Liver/metabolism , Male , Rats , Rats, Wistar , Retinoid X Receptors
20.
Endocrinology ; 139(10): 4197-204, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9751500

ABSTRACT

We studied the interactions of two natural thyroid hormone receptor (TR) mutants from patients with resistance to thyroid hormone (RTH) and an artificial TR mutant with a nuclear receptor corepressor, N-CoR, and a steroid receptor coactivator, SRC-1. In electrophoretic mobility shift assays, wild-type TRbeta-1 interacted with N-CoR in the absence of ligand, whereas T3 caused dissociation of the TRbeta-1/N-CoR complex and formation of TRbeta-1/SRC-1 complex. In contrast, a natural mutant (G345R) with poor T3-binding affinity formed TRbeta-1/N-CoR complex, both in the absence and presence of T3, but could not form TRbeta-1/SRC-1 complex. Another TR mutant, which bound T3 with normal affinity and containing a mutation in the AF-2 region (E457D), had normal interactions with N-CoR but could not bind SRC-1. Both these mutants had strong dominant negative activity on wild-type TR transactivation. Studies with a TR mutant that had slightly decreased T3-binding affinity (R320H) showed a T3-dependent decrease in binding to N-CoR and increase in binding to SRC-1 that reflected its decreased ligand binding affinity. Additionally, when N-CoR and SRC-1 were added to these receptors at various T3 concentrations in electrophoretic mobility shift assays, TR/N-CoR and TR/SRC-1 complexes, but not intermediate complexes were observed, suggesting that N-CoR release is necessary before SRC-1 binding to TR. Our data provide new insight on the molecular mechanisms of dominant negative activity in RTH and suggest that the inability of mutant TRs to interact with coactivators such as SRC-1, which results from reduced T3-binding affinity, is a determinant of dominant negative activity.


Subject(s)
Nuclear Proteins/physiology , Receptors, Thyroid Hormone/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , DNA/metabolism , Histone Acetyltransferases , Humans , Mutation , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Coactivator 1 , Receptors, Thyroid Hormone/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...