Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 280
Filter
1.
J Endocrinol Invest ; 43(1): 31-41, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31352644

ABSTRACT

There are three important thyroid hormone-binding proteins in human serum, thyroxine-binding globulin, transthyretin, and albumin. Genetic variation in these proteins can lead to altered thyroid hormone binding and abnormalities in serum tests of thyroid hormone. Importantly, patients harboring these mutations are euthyroid; thus, the recognition of these conditions is crucial to prevent unnecessary repeated testing and treatment. This article provides an updated overview of serum thyroid hormone transport biology and reviews the underlying genetic alterations, clinical presentation, and appropriate evaluation of patients with suspected mutations in serum thyroid hormone-binding proteins.


Subject(s)
Carrier Proteins/genetics , Membrane Proteins/genetics , Mutation , Thyroid Diseases/diagnosis , Thyroid Diseases/genetics , Thyroid Hormones/genetics , Evaluation Studies as Topic , Humans , Thyroid Hormone-Binding Proteins
2.
BMC Med Genet ; 19(1): 69, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720101

ABSTRACT

BACKGROUND: Congenital hypothyroidism (CH) has an incidence of approximately 1:3000, but only 15% have mutations in the thyroid hormone synthesis pathways. Genetic analysis allows for the precise diagnosis. CASE PRESENTATION: A 3-week old girl presented with a large goiter, serum TSH > 100 mIU/L (reference range: 0.7-5.9 mIU/L); free T4 < 3.2 pmol/L (reference range: 8.7-16 pmol/L); thyroglobulin (TG) 101 µg/L. Thyroid Tc-99 m scan showed increased radiotracer uptake. One brother had CH and both affected siblings have been clinically and biochemically euthyroid on levothyroxine replacement. Another sibling had normal thyroid function. Both Sudanese parents reported non-consanguinity. Peripheral blood DNA from the proposita was subjected to whole exome sequencing (WES). WES identified a novel homozygous missense mutation of the TG gene: c.7021G > A, p.Gly2322Ser, which was subsequently confirmed by Sanger sequencing and present in one allele of both parents. DNA samples from 354 alleles in four Sudanese ethnic groups (Nilotes, Darfurians, Nuba, and Halfawien) failed to demonstrate the presence of the mutant allele. Haplotyping showed a 1.71 centiMorgans stretch of homozygosity in the TG locus suggesting that this mutation occurred identical by descent and the possibility of common ancestry of the parents. The mutation is located in the cholinesterase-like (ChEL) domain of TG. CONCLUSIONS: A novel rare missense mutation in the TG gene was identified. The ChEL domain is critical for protein folding and patients with CH due to misfolded TG may present without low serum TG despite the TG gene mutations.


Subject(s)
Congenital Hypothyroidism/genetics , Exome Sequencing/methods , Mutation, Missense , Thyroglobulin/genetics , Australia/ethnology , Congenital Hypothyroidism/blood , Female , Genetic Predisposition to Disease , Humans , Infant, Newborn , Pedigree , Protein Folding , Sudan , Thyroglobulin/blood , Thyroglobulin/chemistry
3.
Acta Endocrinol (Buchar) ; 13(4): 515-518, 2017.
Article in English | MEDLINE | ID: mdl-31149227

ABSTRACT

OBJECTIVE: The non-effectiveness of levothyroxine administration in hypothyroidism depends on many factors and mechanisms influencing its absorption in small intestins or bounding of circulating hormone with different active molecules. METHODS: Thyroid hormones, TSH, rT3, TGl, TPO-Ab, TG-Ab, were measured using commercially available assays. For anti-T4ab, radioiodine-labeled T4 was added to the patient's serum and the IgG fraction subsequently precipitated by addition of 15% polyethylene glycol. Background was determined by testing 100 control sera from individuals without autoimmune thyroid disease. RESULTS: A 42-year old woman (71.5 kg) with Hashimoto thyroiditis receiving levothyroxine (L-T4) 150 µg and liothyronine (L-T3) 37.5 µg was admitted to the hospital with clinical data of hypothyroidism, TSH-23.8 mU/L, FT4-6.18 pmol/L (n.range 9-19 pmol/L), TPO-Ab 696 IU/mL, TG-Ab 818 IU/mL, circulating T4- antibodies positive. She has a good adherence to medication, malabsorption or administration of other drugs were excluded. L-T4 absorption test revealed 44% increase of serum FT4 at 120 min after ingestion of 150 mcg L-T4 (2.1 mcg/kg). Methylprednisolone pulses of 500 mg i.v. administered in three consecutive days at equal doses of L-T4/L-T3 resulted in a rapid increase of FT4 to 14.5 pmol/L, fall of TSH to 0.18 mU/L and decrease of anti-T4 antibodies to referent range; TPO-Ab and TG-Ab also decreased significantly. Monotherapy by 150 mcg L-T4 was continued in the next three months. A recurrence of hypothyroidism with increase of circulating T4-Ab was observed 100 days later. New administrations of methylprednisolone two pulses of 500 mg revealed a similar normalization of thyroid hormones and anti-T4 antibodies. CONCLUSION: The data showed that T4-antibodies might be a cause of insufficient effects of levothyroxine therapy in autoimmune hypothyroidism. This could be overcome by glucocorticoid administration probably resulting in FT4 release from circulating immune complexes.

4.
Endocrinology ; 157(4): 1363-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26812160

ABSTRACT

Hypothyroidism is the most frequent and earliest endocrine complication in cystinosis, a multisystemic lysosomal storage disease caused by defective transmembrane cystine transporter, cystinosin (CTNS gene). We recently demonstrated in Ctns(-/-) mice that altered thyroglobulin biosynthesis associated with endoplasmic reticulum stress, combined with defective lysosomal processing, caused hypothyroidism. In Ctns(-/-) kidney, hematopoietic stem cell (HSC) transplantation provides long-term functional and structural protection. Tissue repair involves transfer of cystinosin-bearing lysosomes from HSCs differentiated as F4/80 macrophages into deficient kidney tubular cells, via tunneling nanotubes that cross basement laminae. Here we evaluated the benefit of HSC transplantation for cystinotic thyroid and investigated the underlying mechanisms. HSC engraftment in Ctns(-/-) thyroid drastically decreased cystine accumulation, normalized the TSH level, and corrected the structure of a large fraction of thyrocytes. In the thyroid microenvironment, HSCs differentiated into a distinct, mixed macrophage/dendritic cell lineage expressing CD45 and major histocompatibility complex II but low CD11b and F4/80. Grafted HSCs closely apposed to follicles and produced tunneling nanotube-like extensions that crossed follicular basement laminae. HSCs themselves further squeezed into follicles, allowing extensive contact with thyrocytes, but did not transdifferentiate into Nkx2.1-expressing cells. Our observations revealed significant differences of basement lamina porosity between the thyroid and kidney and/or intrinsic macrophage invasive properties once in the thyroid microenvironment. The contrast between extensive thyrocyte protection and low HSC abundance at steady state suggests multiple sequential encounters and/or remanent impact. This is the first report demonstrating the potential of HSC transplantation to correct thyroid disease and supports a major multisystemic benefit of stem cell therapy for cystinosis.


Subject(s)
Cystinosis/therapy , Disease Models, Animal , Hematopoietic Stem Cell Transplantation/methods , Thyroid Gland/physiopathology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Cell Differentiation , Cystine/metabolism , Cystinosis/genetics , Cystinosis/physiopathology , Female , Hematopoietic Stem Cells/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Thyroid Gland/metabolism , Thyrotropin/metabolism , Transplantation, Homologous
5.
Endocrinology ; 156(6): 2349-64, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25811319

ABSTRACT

Thyroid hormones are released from thyroglobulin (Tg) in lysosomes, which are impaired in infantile/nephropathic cystinosis. Cystinosis is a lysosomal cystine storage disease due to defective cystine exporter, cystinosin. Cystinotic children develop subclinical and then overt hypothyroidism. Why hypothyroidism is the most frequent and earliest endocrine complication of cystinosis is unknown. We here defined early alterations in Ctns(-/-) mice thyroid and identified subcellular and molecular mechanisms. At 9 months, T4 and T3 plasma levels were normal and TSH was moderately increased (∼4-fold). By histology, hyperplasia and hypertrophy of most follicles preceded colloid exhaustion. Increased immunolabeling for thyrocyte proliferation and apoptotic shedding indicated accelerated cell turnover. Electron microscopy revealed endoplasmic reticulum (ER) dilation, apical lamellipodia indicating macropinocytic colloid uptake, and lysosomal cystine crystals. Tg accumulation in dilated ER contrasted with mRNA down-regulation. Increased expression of ER chaperones, glucose-regulated protein of 78 kDa and protein disulfide isomerase, associated with alternative X-box binding protein-1 splicing, revealed unfolded protein response (UPR) activation by ER stress. Decreased Tg mRNA and ER stress suggested reduced Tg synthesis. Coordinated increase of UPR markers, activating transcription factor-4 and C/EBP homologous protein, linked ER stress to apoptosis. Hormonogenic cathepsins were not altered, but lysosome-associated membrane protein-1 immunolabeling disclosed enlarged vesicles containing iodo-Tg and impaired lysosomal fusion. Isopycnic fractionation showed iodo-Tg accumulation in denser lysosomes, suggesting defective lysosomal processing and hormone release. In conclusion, Ctns(-/-) mice showed the following alterations: 1) compensated primary hypothyroidism and accelerated thyrocyte turnover; 2) impaired Tg production linked to ER stress/UPR response; and 3) altered endolysosomal trafficking and iodo-Tg processing. The Ctns(-/-) thyroid is useful to study disease progression and evaluate novel therapies.


Subject(s)
Cystinosis/metabolism , Cystinosis/pathology , Endoplasmic Reticulum Stress/physiology , Lysosomes/metabolism , Thyroglobulin/biosynthesis , Unfolded Protein Response/physiology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Female , Male , Mice
6.
Ann Endocrinol (Paris) ; 72(2): 95-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21511232

ABSTRACT

Intracellular metabolism of thyroid hormone and availability of the active hormone, triiodothyronine is regulated by three selenoprotein iodothyronine deiodinases (Ds). While acquired changes in D activities are common, inherited defects in humans have not been identified. Selenium (Se) is an essential trace element required for the biosynthesis of selenoproteins, and selenocysteine insertion sequence (SECIS) binding protein 2 (SBP2) represents a key trans-acting factor for the cotranslational insertion of selenocysteine into selenoproteins. In 2005 we reported the first mutations in the SBP2 gene in two families in which the probands presented with transient growth retardation associated with abnormal thyroid function tests, low triiodothyronine (T3), high thyroxine (T4) and reverse T3, and slightly elevated thyrotropin. Affected children were either homozygous or compound heterozygous for SBP2 gene mutations and the relatively mild phenotype was due to partial SBP2 deficiency, affecting the expression of a subset of selenoproteins. In vivo studies of these subjects have explored the effects of Se and thyroid hormone supplementation. In vitro experiments have provided new insights into the effect of SBP2 mutations. A broader and more complex phenotype was brought to light by the subsequent identification of three new cases from different families with SBP2 gene mutations. These mutations caused a severe SBP2 deficiency resulting in reduced synthesis of most of the 25 known human selenoproteins. Here we summarize the clinical presentation of SBP2 mutations, their effect on SBP2 function and downstream consequences for selenoprotein synthesis and function.


Subject(s)
RNA-Binding Proteins/genetics , Selenoproteins/biosynthesis , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Dietary Supplements , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Male , Mutation , RNA-Binding Proteins/metabolism , Selenium/metabolism , Selenoproteins/genetics , Selenoproteins/metabolism , Thyroid Gland/metabolism , Thyroid Hormones/blood
7.
Exp Clin Endocrinol Diabetes ; 119(2): 81-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20827662

ABSTRACT

CONTEXT: Thyroid hormone (TH) mediated changes in gene expression were thought to be primarily initiated by the nuclear TH receptor (TR) binding to a thyroid hormone response element in the promoter of target genes. A recently described extranuclear mechanism of TH action consists of the association of TH-liganded TRß with phosphatidylinositol 3-kinase (PI3K) in the cytosol and subsequent activation of the PI3K pathway. OBJECTIVE: The aim of this study was to examine the effect of TH, TRß and PI3K on stanniocalcin 1 (STC1) expression in human cells. DESIGN: We treated human skin fibroblasts with triiodothyronine (T3) in the absence or presence of the PI3K inhibitor LY294002, a dominant negative PI3K subunit, Δp85α, and the protein synthesis inhibitor cycloheximide (CHX). The role of the TRß was studied in cells from patients with resistance to thyroid hormone (RTH). STC-1 mRNA expression was measured by real-time PCR. RESULTS: We found an induction of STC1 by T3 in normal cells, but less in cells from subjects with RTH (2.7 ± 0.2 vs. 1.6 ± 0.04, P < 0.01). The effect of T3 was completely abrogated by blocking PI3K with LY294002 (3.9 ± 0.5 vs. 0.85 ± 0.5; P < 0.05) and greatly reduced after transfection of a dominant negative PI3K subunit, demonstrating dependency on the PI3K pathway. CONCLUSION: These results establish STC1 as a TH target gene in humans. Furthermore, we show that STC1 induction by TH depends on both TRß and PI3K activation.


Subject(s)
Glycoproteins/genetics , Phosphatidylinositol 3-Kinase/metabolism , Thyroid Hormone Receptors beta/agonists , Triiodothyronine/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/physiology , Gene Expression Regulation/drug effects , Glycoproteins/metabolism , Humans , Mutant Proteins/metabolism , Mutant Proteins/physiology , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormone Receptors beta/physiology , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormone Resistance Syndrome/metabolism , Thyroid Hormone Resistance Syndrome/pathology , Transcriptional Activation/drug effects
8.
Oncogene ; 29(42): 5678-86, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20676139

ABSTRACT

It is well known that thyroid disease is more frequent in women than in men; however, the molecular basis for this gender-based difference is still poorly understood. The activation of phosphoinositide 3-kinase (PI3K), through different mechanisms including loss of the PTEN tumor suppressor, is being increasingly recognized as a major player in the development of thyroid neoplastic lesions. Loss of Pten in the mouse thyroid results in a significant increase in the thyrocyte proliferative index, which is more prominent in the female mice. In this study, we show that 52% of the Pten(-/-) female mice, but only 12% of the males, develop follicular adenomas by 1 year of age. In addition, 50% of female mutants, but only 35% of males older than 1 year of age develop invasive, and often metastatic, follicular carcinomas. Mutant females have a significantly shorter overall survival compared with male mutants. Hormonal manipulation experiments established a direct role of estrogens in controlling the increased thyrocyte proliferation index in mutant females. Furthermore, while genetic ablation of one Cdkn1b allele accelerated the development of neoplastic lesions, it also abolished the gender differences in survival and reduced the difference in neoplastic lesion development rate, underlining a key role of p27 in mediating estrogen action in the thyroid follicular cells. These data, based on a clinically relevant model of thyroid follicular carcinoma, provide, to the best of our knowledge, for the first time in vivo evidence that circulating estrogens are directly responsible for the increased female susceptibility to thyroid disease, at least on activation of the PI3K pathway, and provide new insights into the gender-based differences characterizing thyroid neoplastic disorders.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Estrogens/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptor Cross-Talk/physiology , Sex Characteristics , Thyroid Neoplasms/metabolism , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Animals , Blotting, Western , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Female , Gene Expression , Gene Expression Profiling , Incidence , Male , Mice , Mice, Mutant Strains , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
9.
Exp Clin Endocrinol Diabetes ; 117(1): 34-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18561095

ABSTRACT

BACKGROUND: Resistance to thyroid hormone (RTH) is a dominantly inherited syndrome characterized by decreased responsiveness of target tissues to thyroid hormone. Two members of a Turkish family, a mother and son, had thyroid function tests suggestive of resistance to thyroid hormone (RTH). METHODS: The clinical presentation was, however, different. The mother (proposita) had palpitation, weakness, tiredness, nervousness, dry mouth and was misdiagnosed as having multinodular toxic goiter which was treated with antithyroid drugs and partial thyroidectomy. Her younger son had attention deficit hyperactivity disorder and primary encopresis, but normal intellectual quotient. Both had elevated serum iodothyronine levels with nonsuppressed thyrotropin. RESULTS: A mutation in one allele of the thyroid hormone receptor beta gene (P453A) was identified, providing a genetic confirmation for the diagnosis of RTH. CONCLUSION: Mutational analysis of the TRss gene allows definitive diagnosis of RTH, potentially avoiding the need for protracted and expensive pituitary function testing.


Subject(s)
Mutation , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Resistance Syndrome/genetics , Adolescent , Adult , Aged , Amino Acid Substitution , Child , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Nuclear Family , Pedigree , Propylthiouracil/therapeutic use , Thyroid Hormone Resistance Syndrome/surgery , Thyroidectomy , Thyroxine/therapeutic use
10.
J Endocrinol Invest ; 28(4): 379-83, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15966514

ABSTRACT

The syndrome of resistance to thyroid hormone (RTH) is characterized by impaired tissue responses to thyroid hormone. Hashimoto's thyroiditis is the most common thyroid autoimmune disease. We present a Turkish family with both RTH and Hashimoto's thyroiditis. RTH was detected through the presence of point mutation in thyroid hormone receptor (TR), and Hashimoto's thyroiditis was diagnosed due to the presence of thyroid autoantibodies. The proposita, her affected mother as well as her unaffected sister have thyroid autoantibodies consistent with Hashimoto's thyroiditis, and a heterozygous point mutation in exon 10 encoding the ligand (3,3',5-L-T3)-binding domain of the TRbeta gene was detected in both the proposita and the mother. The mutation is a replacement of cytosine for guanine in codon 453 (CCT->GCT) producing a missense mutation substituting a normal proline with an alanine (P453A), which reduces the affinity for T3 to 17% of that of the normal TRbeta. Both also have modest elevation of serum TSH levels. In severe RTH, marked elevation of thyroid hormone concentrations in the absence of suppressed TSH supports the laboratory diagnosis of RTH. However, when RTH is mild and associated with thyroiditis, even a modest thyroid gland insufficiency can obliterate the serum T4 and T3 elevations, typical of RTH. This will manifest as elevated serum TSH. Demonstration of TRbeta gene mutation is then necessary to establish the diagnosis. In addition, under these circumstances, treatment with thyroid hormone should be considered.


Subject(s)
Thyroid Hormone Receptors alpha/genetics , Thyroid Hormones/pharmacology , Thyroiditis, Autoimmune/drug therapy , Adolescent , Autoantibodies , Drug Resistance , Female , Humans , Male , Middle Aged , Mutation, Missense , Pedigree , Thyroiditis, Autoimmune/ethnology , Thyroiditis, Autoimmune/genetics , Turkey
11.
Ir J Med Sci ; 174(4): 60-4, 2005.
Article in English | MEDLINE | ID: mdl-16445164

ABSTRACT

BACKGROUND: Resistance to Thyroid Hormone (RTH) is a condition caused by tissue hyposensitivity to the effects of circulating thyroid hormone, and may be misdiagnosed as hyperthyroidism. AIMS: We report the first case of RTH in an Irish patient highlighting the clinical features and the pathophysiological mechanism underlying the characteristic laboratory abnormalities found in the condition. METHODS: We describe an isolated case of RTH initially misdiagnosed as hyperthyroidism, and detail the investigations which ultimately led to the correct diagnosis. Genetic screening of the thyroid hormone receptor beta gene was performed. RESULTS: Thyroid function tests including T3 suppression test and TRH-stimulation test suggested a diagnosis of RTH. Genetic testing failed to demonstrate a mutation in the thyroid hormone receptor. CONCLUSION: RTH is a rare inherited condition that may be misdiagnosed as hyperthyroidism. The case we describe most likely results from a de novo mutation in an as yet undiscovered gene. RTH should be considered in patients with elevated thyroid hormone levels and normal TSH so that unnecessary and potentially harmful treatment can be avoided.


Subject(s)
Hyperthyroidism/diagnosis , Pituitary Neoplasms/diagnosis , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Resistance Syndrome/diagnosis , Adult , DNA Mutational Analysis , Diagnosis, Differential , Female , Humans , Magnetic Resonance Imaging , Mutation , Risk Assessment , Severity of Illness Index , Thyroid Function Tests , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormones/metabolism
12.
J Endocrinol Invest ; 27(4): 318-22, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15233549

ABSTRACT

The thyroid hormone receptor beta (TRbeta) gene generates two different proteins by use of a different promoter (beta1 and beta2). We now report a novel short TRbeta1 RNA splice variant in humans lacking 35 nucleotides at the 3' end of the non-coding exon 1 due to an alternative 5' splice donor site. This short variant was first identified in sequences of cDNA obtained from cultured human fibroblasts. Both variants were found in human fibroblasts, brain, pituitary, adrenal gland, placenta, muscle, thyroid and lymphocytes. These TRbeta1 variants possess splice donor sites with a sequence score slightly favoring the TRbeta1 long variant. Variant-specific real-time polymerase chain reaction (PCR) showed that their relative proportions were equal except in pituitary and muscle, in which the long form was 3- and 5-fold in excess. T3 treatment of fibroblasts grown in thyroid hormone depleted medium did not affect the absolute or relative expression of the two variants. Furthermore, the expression level in fibroblasts from patients with resistance to thyroid hormone with or without TRbeta gene mutations was not different to that in fibroblasts from normal controls.


Subject(s)
5' Untranslated Regions/genetics , Alternative Splicing/genetics , Thyroid Hormone Receptors beta/genetics , Base Sequence , Cells, Cultured , Fibroblasts/chemistry , Fibroblasts/drug effects , Genetic Variation , Humans , Molecular Sequence Data , Mutation , Polymerase Chain Reaction , RNA, Messenger/analysis , Thyroid Hormone Resistance Syndrome/genetics , Triiodothyronine/pharmacology
13.
J Endocrinol Invest ; 26(8): 770-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14669836

ABSTRACT

Resistance to TSH is a syndrome of reduced sensitivity to a biologically active TSH molecule. Subjects have elevated TSH levels but no goiter. However, thyroid hormone concentration may vary from normal to very high, depending on the severity of the resistance. Individuals with very high TSH, low T4 and hypoplastic thyroid glands can be mistakenly diagnosed as having primary hypothyroidism due to a defective development of the thyroid gland. Those with normal or slightly decreased T4 can be misdiagnosed as having central hypothyroidism especially if their serum TSH concentration is only slightly elevated. Mutations in the TSH receptor (TSHr) gene have been reported in 16 families with homozygous or compound heterozygous inheritance. The mutant TSHrs show reduced or no function due to either altered ligand binding or defect in membrane targeting. Some individuals, heretozygous for a TSHr gene mutation can present mild resistance to TSH manifesting as euthyroidism with slight hyperthyrotropinemia. A larger proportion of families express the phenotype of resistance to TSH in the absence of a TSHr defect. In many the inheritance is dominant and the genetic cause has not been yet determined.


Subject(s)
Thyroid Hormone Resistance Syndrome , Thyrotropin/physiology , Amino Acid Sequence , Animals , Genotype , Humans , Molecular Sequence Data , Mutation/physiology , Phenotype , Receptors, Thyrotropin/physiology , Thyroid Hormones/physiology , Thyrotropin/genetics
15.
J Endocrinol ; 172(1): 177-85, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11786385

ABSTRACT

The maintenance of thyroid hormone (TH) homeostasis is dependent on the synthesis and secretion of TH regulated by TSH. This is achieved, in turn, by the negative feedback of TH on TSH secretion and synthesis, which requires the interaction with TH receptors (TRs). Derived by alternative splicing of two gene transcription products, three TRs (TRbeta1, TRbeta2 and TRalpha1) interact with TH while another, TRalpha2, binds to DNA but not to TH. In this study we compare the results of thyroid function tests in mice with deletions of the TRalpha and TRbeta genes alone and present novel data on mice that are double homozygous and combined heterozygous. Homozygous deletions of both the TRalpha and TRbeta in the same mouse (TRalphao/o; TRbeta-/-) resulted in serum TSH values only slightly lower than those in athyreotic, Pax8 knockout mice. Whereas the absence of TRalpha alone does not cause resistance to TH, the absence of TRbeta in the presence of TRalpha results in a 205, 169, 544% increase in serum thyroxine (T(4)), triiodothyronine (T(3)) and TSH concentrations respectively. However, in the absence of TRbeta, loss of one TRalpha allele can worsen the resistance to TH with a 243 and 307% increase in T(4) and T(3) respectively. Similarly, while the heterozygous mouse with a single TRbeta allele shows no alteration in thyroid function, the concomitant deletion of TRalpha brings about mild but significant resistance to TH. Furthermore, the severity of the resistance to TH was noted to decrease with age in parallel with the decrease in serum free T(4) values also seen in wild-type mice. These results demonstrate that (1) unliganded TRalpha or TRbeta are not absolutely necessary for the upregulation of TSH; (2) TRbeta but not TRalpha is sufficient for TH-mediated downregulation of TSH; and (3) TRalpha may partially substitute for TRbeta in mediating a partial TH-dependent TSH suppression.


Subject(s)
Aging/physiology , Receptors, Thyroid Hormone/genetics , Thyroid Gland/physiology , Alternative Splicing , Animals , Heterozygote , Homozygote , Male , Mice , Mice, Transgenic , Thyroid Function Tests , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood
16.
J Pediatr ; 139(6): 887-91, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11743520

ABSTRACT

We found familial dysalbuminemic hyperthyroxinemia (FDH) in a 5-month-old boy with congenital hypothyroidism (CH) who had a blood thyrotropin (TSH) level of 479 mU/L but normal total serum thyroxine (T4) and higher than normal total triiodothyronine (T3) levels. Thyroid hormone substitution began at 5 weeks of age when T4 and T3 concentrations were below normal. Until the age of 5 months, treatment with levothyroxine was suboptimal on the basis of high serum TSH levels despite above-normal T4 levels. FDH was confirmed by isoelectric focusing and testing of other family members. DNA analysis of the patient revealed R218H, a mutation in the serum albumin gene associated with FDH, which was also present in the patient's euthyroid father and brother. Thyroid scans, serum thyroglobulin measurements, and free T4 measurements using equilibrium dialysis or 2-step immunoassay methods can identify thyroid hormone-binding protein defects and simplify the diagnosis and treatment of infants with CH.


Subject(s)
Albumins/genetics , Albuminuria/genetics , Congenital Hypothyroidism , Hyperthyroxinemia/genetics , Hypothyroidism/genetics , Mutation/genetics , Albuminuria/blood , Humans , Hyperthyroxinemia/blood , Hypothyroidism/blood , Infant , Male , Thyrotropin/blood , Thyrotropin/genetics , Thyroxine/blood , Thyroxine/genetics
17.
Endocrinology ; 142(12): 5131-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713206

ABSTRACT

Through the cAMP signaling pathway, TSH stimulates thyroid follicular cell proliferation, differentiation, and function. Although the autocrine production of IGF-I in the thyroid gland suggests an important physiological function for this factor in these processes, the exact role of the IGF-I/IGF-I receptor system in vivo remains unclear. Although the mitogenic action of TSH requires the presence of IGF-I or insulin in primary culture of dog and human thyroid cells, IGF-I has an effect equal to and independent of the effect of TSH on cell proliferation in rat thyroid cell lines and may even be the main growth regulator in this case. To investigate the in vivo function of the IGF-I/IGF-I receptor system, transgenic mice overexpressing human IGF-I, IGF-I receptor, or both in the thyroid were generated. Adult transgenic mice did not present external signs of thyroid dysfunction, but mice overexpressing both transgenes had significantly increased gland weight and follicular lumen area. A decreased TSH level together with a slightly increased serum T(4) concentration and increased thyroidal iodine uptake were also observed, suggesting that IGF-I and IGF-I receptor stimulate thyroid function to some extent in vivo.


Subject(s)
Goiter/etiology , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Thyroid Gland/physiopathology , Thyrotropin/physiology , Animals , Goiter/genetics , Goiter/pathology , Humans , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/genetics , Iodine/metabolism , Mice , Mice, Transgenic/genetics , Organ Size , Receptor, IGF Type 1/genetics , Reference Values , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyrotropin/blood , Thyroxine/blood
18.
J Clin Endocrinol Metab ; 86(10): 5039-44, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11600582

ABSTRACT

Inherited T(4)-binding globulin deficiency is caused by mutations in the T(4)-binding globulin gene located on the X chromosome. We describe herein three novel mutations in three different families producing complete T(4)-binding globulin deficiency. The proposita of a family from Harwichport is a female with XO Turner's syndrome who expressed only the mutant T(4)-binding globulin allele. Her T(4)-binding globulin sequence has a 19-nucleotide deletion in the distal portion of exon 4. This causes a frameshift and a premature stop at codon 384 of the mature protein. Structure analysis with the Swiss PDB-Viewer revealed that this mutation removes beta-strand s5B from the core of the T(4)-binding globulin molecule, leading to a severe folding defect that is likely to prevent synthesis and secretion. The propositi of complete T(4)-binding globulin deficiency 7 and 8 were 7-month-old and 3-wk-old male infants who were identified because of low serum T(4) levels detected during neonatal screening. Sequencing of complete T(4)-binding globulin deficiency 7 revealed a single nucleotide deletion, a G at position 2690 in exon 3. This leads to an alteration of the amino acid sequence starting at codon 283 and a premature stop at codon 301. Complete T(4)-binding globulin deficiency 8 also has a deletion of the first nucleotide of exon 4, a G at position 3358. This leads to a frameshift and a premature stop at codon 374. As in the case of complete T(4)-binding globulin deficiency J, which has also a nucleotide deletion but downstream (position 3421) and a stop at codon 374, these two T(4)-binding globulin mutants undoubtedly have a defect in intracellular transport and therefore fail to be secreted. This explains the lack of T(4)-binding globulin in the hemizygous affected subjects.


Subject(s)
Mutation , Thyroxine-Binding Proteins/genetics , Adolescent , Amino Acid Sequence , Female , Humans , Infant , Infant, Newborn , Male , Thyroxine/blood , Thyroxine-Binding Proteins/chemistry , Thyroxine-Binding Proteins/deficiency , Triiodothyronine/blood
19.
J Clin Endocrinol Metab ; 86(9): 4273-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549661

ABSTRACT

We report the occurrence of transient thyrotoxicosis during pregnancy in a subject with resistance to thyroid hormone. Before pregnancy, the subject was euthyroid, with normal serum TSH and elevated levels of free T(3) and free T(4) caused by a mutation in the TRbeta gene (R243Q). Beginning at the fourth week of gestation serum levels of free T(3) and T(4) increased in parallel with an increase in hCG. At 6-7 wk gestation she manifested hypermetabolic features, with mild nausea and vomiting. Peak levels of serum hCG and thyroid hormone concentrations were attained at 12 wk gestation, when serum TSH was fully suppressed. In the following weeks of gestation, thyroid hormone levels declined, with amelioration of the symptoms. A baby boy also affected with resistance to thyroid hormone harboring the same TRbeta gene mutation was born by normal vaginal delivery.


Subject(s)
Pregnancy Complications/physiopathology , Thyroid Hormone Resistance Syndrome/physiopathology , Thyrotoxicosis/physiopathology , Adult , Chorionic Gonadotropin/blood , Female , Humans , Pedigree , Pregnancy , Pregnancy Complications/diagnostic imaging , Thyroid Function Tests , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormones/blood , Thyrotoxicosis/etiology , Thyrotoxicosis/genetics , Ultrasonography
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...