Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Eur J Endocrinol ; 186(1): 73-82, 2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34727089

ABSTRACT

OBJECTIVE: Resistance to thyroid hormone beta (RTHß) is an inherited syndrome caused by mutations in the thyroid hormone receptor ß (THRB) gene. Patients with RTHß typically have elevated thyroid hormone levels with non-suppressed serum thyroid-stimulating hormone (TSH). We aimed to elucidate the clinical, laboratory, and imaging findings of RTHß patients and further to explore their association with THRB gene mutations. DESIGN AND METHODS: We retrospectively reviewed the clinical charts and compared the clinical findings of 68 RTHß patients (45 probands and 23 relatives) and 30 unaffected relatives in Kuma Hospital. RESULTS: Genetic testing revealed 35 heterozygous THRB gene mutations. Among all RTHß patients, autoimmune thyroid disease (AITD) was detected in 42.1% of men and 40.9% of women, showing that the prevalence of AITD in affected males was significantly higher than in unaffected relatives (P = 0.019). During the follow-up of 44 patients, 13 patients (29.5%; 8 (42.1%) with AITD and 5 (20%) without AITD) temporarily showed thyroid function test results inconsistent with RTHß. Two patients with the R383H mutation, which has little dominant-negative effect, temporarily showed normal thyroid hormone and TSH levels without AITD. CONCLUSIONS: The frequency of AITD in male RTHß patients was significantly higher compared to unaffected relatives. More than 20% of RTHß patients temporarily showed laboratory findings atypical of RTHß during their follow-up, and patients with AITD and specific THRB mutations were prone to display such findings. Therefore, genetic testing should be performed even for patients with fluctuations in thyroid function test results to avoid misdiagnosis and inappropriate treatment.


Subject(s)
Thyroid Gland/physiopathology , Thyroid Hormone Resistance Syndrome , Thyroiditis, Autoimmune , Adult , Case-Control Studies , DNA Mutational Analysis , Female , Humans , Japan , Male , Middle Aged , Mutation , Retrospective Studies , Thyroid Function Tests , Thyroid Gland/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Resistance Syndrome/blood , Thyroid Hormone Resistance Syndrome/complications , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormone Resistance Syndrome/physiopathology , Thyroid Hormones/blood , Thyroiditis, Autoimmune/blood , Thyroiditis, Autoimmune/complications , Thyroiditis, Autoimmune/genetics , Thyroiditis, Autoimmune/physiopathology , Thyrotropin/blood
2.
Endocr J ; 68(5): 509-517, 2021 May 28.
Article in English | MEDLINE | ID: mdl-33827995

ABSTRACT

Confirmation of sustained syndrome of inappropriate secretion of thyrotropin (SITSH) is a milestone in diagnosis of ß type of resistance to thyroid hormone (RTHß). The differential diagnoses of RTHß include TSH-producing pituitary adenoma (TSHoma) and familial dysalbuminemic hyperthyroxinemia (FDH), which also present SITSH. Recently, patients with RTHα caused by a mutation in thyroid hormone receptor α were reported and they did not present SITSH but a decline in the serum T4/T3 ratio. This review was aimed to overview thyroid function tests in RTH and related disorders. First, the characteristics of the thyroid function in RTHß, TSHoma, and FDH obtained from a Japanese database are summarized. Second, the degrees of SITSH in patients with truncations and frameshifts were compared with those in patients with single amino acid deletions and single amino acid substitutions obtained from the literature. Third, the degrees of SITSH in homozygous patients were compared with those in heterozygous patients with cognate mutations. Finally, the FT3/FT4 ratios in RTHα are summarized. In principle, the TSH values in FDH were within the normal range and apparent FT4 values in FDH were much higher than in RTHß and TSHoma. The FT3/FT4 values in RTHß were significantly lower than in TSHoma. The degrees of SITSH in patients with truncations and frameshifts were more severe than those in patients with single amino acid deletions and single amino acid substitutions, and those in homozygous patients were more severe than those in heterozygous patients with cognate mutations. The FT3/FT4 ratios in RTHα were higher than 1.0.


Subject(s)
Adenoma/diagnosis , Hyperpituitarism/diagnosis , Pituitary Neoplasms/diagnosis , Thyroid Gland/physiopathology , Thyroid Hormone Resistance Syndrome/diagnosis , Thyroid Hormones/blood , Adenoma/blood , Adenoma/physiopathology , Diagnosis, Differential , Humans , Hyperpituitarism/blood , Hyperpituitarism/physiopathology , Pituitary Neoplasms/blood , Pituitary Neoplasms/physiopathology , Thyroid Function Tests , Thyroid Hormone Resistance Syndrome/blood , Thyroid Hormone Resistance Syndrome/physiopathology
3.
Thyroid ; 31(4): 678-691, 2021 04.
Article in English | MEDLINE | ID: mdl-32924834

ABSTRACT

Background: In humans, resistance to thyroid hormone (RTH) caused by mutations in the thyroid hormone receptor alpha (THRA) gene, RTHα, manifests as tissue-specific hypothyroidism and circulating thyroid hormone levels exhibit hypothyroid-like clinical features. Before the identification of patients with RTHα, several Thrα1 knock-in mouse models were generated to clarify the function of TRα1. However, the phenotypes of these mice were not consistent with the clinical presentation of RTHα in humans. For the present study, we generated an RTHα mouse model that carries the Thra1E403X mutation found in human RTHα patients. Here, we report the gross phenotypes of this mouse RTHα model. Methods: Traditional homologous recombination gene targeting techniques were used to introduce a mutation (Thra1E403X) in the mouse Thra gene. The phenotypes of the resulting mice were studied and compared with clinical features observed for RTHα with THRAE403X. Results: Thrα1E403X/E403X homozygous mice exhibited severe neurological phenotypes, such as spasticity and motor ataxia, which were similar to those observed in endemic cretinism. Thrα1E403X/+ heterozygous mice reproduced most clinical manifestations of patient with RTHα, such as a normal survival rate and male fertility, as well as delayed postnatal growth and development, neurological and motor coordination deficits, and anemia. The mice had typical thyroid function with a modest increase in serum triiodothyronine (T3) levels, a low thyroxine (T4)/T3 ratio, and low reverse T3 (rT3) levels. Conclusions: The Thrα1E403X/+ mice faithfully recapitulate the clinical features of human RTHα and thus can provide a useful tool to dissect the role of TRα1 in development and to determine the pathological mechanisms of RTHα.


Subject(s)
Mutation , Thyroid Gland/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormones/blood , Animals , Ataxia/genetics , Ataxia/metabolism , Ataxia/physiopathology , Bone Development , Brain/growth & development , Disease Models, Animal , Fertility , Genetic Predisposition to Disease , Heterozygote , Homozygote , Mice, Inbred C57BL , Mice, Mutant Strains , Motor Activity , Muscle Spasticity/genetics , Muscle Spasticity/metabolism , Muscle Spasticity/physiopathology , Muscle Strength , Phenotype , Thyroid Gland/physiopathology , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Resistance Syndrome/blood , Thyroid Hormone Resistance Syndrome/physiopathology
4.
Thyroid ; 31(6): 1003-1005, 2021 06.
Article in English | MEDLINE | ID: mdl-33198587

ABSTRACT

Resistance to thyroid hormone alpha (RTHα) is caused by mutations in thyroid hormone receptor α (THRA). Little is known about the natural history and treatment of RTHα, and diagnosis before the age of 1 year has not been previously reported. A de novo heterozygous THRA mutation (pC380SfsX9) was identified in a 10-month-old female investigated for developmental delay, hypotonia, macrocephaly, and severe constipation. Treatment with levothyroxine was accompanied by an appropriate rise in thyroxine (T4), triiodothyronine (T3), as well as decrease in thyrotropin levels and in the T3/T4 ratio with a trend toward normalization of peripheral markers of thyroid hormone action. THRA pC380SfsX9 results in extreme RTHα.


Subject(s)
Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Resistance Syndrome/drug therapy , Thyroxine/therapeutic use , Constipation/physiopathology , Developmental Disabilities/physiopathology , Early Diagnosis , Early Medical Intervention , Female , Humans , Infant , Megalencephaly/physiopathology , Muscle Hypotonia/physiopathology , Mutation , Thyroid Hormone Resistance Syndrome/diagnosis , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormone Resistance Syndrome/physiopathology
6.
Best Pract Res Clin Endocrinol Metab ; 31(5): 487-503, 2017 10.
Article in English | MEDLINE | ID: mdl-29223283

ABSTRACT

Thyroid hormones are essential for growth, differentiation and metabolism during prenatal and postnatal life. The hypothalamus-pituitary-thyroid (HPT)-axis is optimized for these actions. Knowledge of this hormonal axis is derived from decades of experiments in animals and man, and more recently from spontaneous mutations in man and constructed mutations in mice. This review examines the HPT-axis in relation to 24 h TSH profiles in men in various physiological and pathophysiological conditions, including obesity, age, longevity, and primary as well as central hypothyroidism. Hormone rhythms can be analyzed by quantitative methods, e.g. operator-independent deconvolution, approximate entropy and fitting the 24-h component by Cosinor analysis or related procedures. These approaches have identified some of the regulatory components in (patho)physiological conditions.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Thyroid Gland/physiology , Female , Humans , Hypothyroidism/physiopathology , Male , Sleep/physiology , Thyroid Hormone Resistance Syndrome/physiopathology , Thyrotropin/blood
7.
J Clin Endocrinol Metab ; 102(9): 3517-3525, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28911146

ABSTRACT

Context: Patients with resistance to thyroid hormone (TH) α (RTHα) are characterized by growth retardation, macrocephaly, constipation, and abnormal thyroid function tests. In addition, almost all RTHα patients have mild anemia, the pathogenesis of which is unknown. Animal studies suggest an important role for TH and TH receptor (TR)α in erythropoiesis. Objective: To investigate whether a defect in TRα affects the maturation of red blood cells in RTHα patients. Design, Setting, and Patients: Cultures of primary human erythroid progenitor cells (HEPs), from peripheral blood of RTHα patients (n = 11) harboring different inactivating mutations in TRα (P398R, F397fs406X, C392X, R384H, A382fs388X, A263V, A263S), were compared with healthy controls (n = 11). During differentiation, erythroid cells become smaller, accumulate hemoglobin, and express different cell surface markers. We assessed cell number and cell size, and used cell staining and fluorescence-activated cell sorter analysis to monitor maturation at different time points. Results: After ∼14 days of ex vivo expansion, both control and patient-derived progenitors differentiated spontaneously. However, RTHα-derived cells differentiated more slowly. During spontaneous differentiation, RTHα-derived HEPs were larger, more positive for c-Kit (a proliferation marker), and less positive for glycophorin A (a differentiation marker). The degree of abnormal spontaneous maturation of RTHα-derived progenitors did not correlate with severity of underlying TRα defect. Both control and RTHα-derived progenitors responded similarly when differentiation was induced. T3 exposure accelerated differentiation of both control- and RTHα patient-derived HEPs. Conclusions: Inactivating mutations in human TRα affect the balance between proliferation and differentiation of progenitor cells during erythropoiesis, which may contribute to the mild anemia seen in most RTHα patients.


Subject(s)
Anemia/genetics , Erythropoiesis/genetics , Gene Expression Regulation , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Resistance Syndrome/genetics , Adolescent , Adult , Anemia/epidemiology , Anemia/physiopathology , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Erythrocytes/metabolism , Female , Humans , Incidence , Male , Middle Aged , Mutation , Prognosis , Reference Values , Role , Stem Cells/cytology , Stem Cells/physiology , Thyroid Hormone Resistance Syndrome/epidemiology , Thyroid Hormone Resistance Syndrome/physiopathology , Young Adult
8.
J Clin Endocrinol Metab ; 102(9): 3234-3240, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28586435

ABSTRACT

Context: Fetuses exposed to the high thyroid hormone (TH) levels of mothers with resistance to thyroid hormone beta (RTH-ß), due to mutations in the THRB gene, have low birth weight and suppressed TSH. Objective: Determine if such exposure to high TH levels in embryonic life has a long-term effect into adulthood. Design: Observations in humans with a parallel design on animals to obtain a preliminary information regarding mechanism. Setting: University research centers. Patients or other participants: Humans and mice with no RTH-ß exposed during intrauterine life to high TH levels from mothers who were euthyroid due to RTH-ß. Controls were humans and mice of the same genotype but born to fathers with RTH-ß and mothers without RTH-ß and thus, with normal serum TH levels. Interventions: TSH responses to stimulation with thyrotropin-releasing hormone (TRH) during adult life in humans and male mice before and after treatment with triiodothyronine (T3). We also measured gene expression in anterior pituitaries, hypothalami, and cerebral cortices of mice. Results: Adult humans and mice without RTH-ß, exposed to high maternal TH in utero, showed persistent central resistance to TH, as evidenced by reduced responses of serum TSH to TRH when treated with T3. In mice, anterior pituitary TSH-ß and deiodinase 3 (D3) mRNAs, but not hypothalamic and cerebral cortex D3, were increased. Conclusions: Adult humans and mice without RTH-ß exposed in utero to high maternal TH levels have persistent central resistance to TH. This is likely mediated by the increased expression of D3 in the anterior pituitary, enhancing local T3 degradation.


Subject(s)
Fetal Diseases/blood , Hyperthyroidism/blood , Maternal-Fetal Exchange/physiology , Thyroid Hormone Resistance Syndrome/etiology , Thyroid Hormones/blood , Adult , Age Factors , Analysis of Variance , Animals , Disease Models, Animal , Female , Fetal Diseases/etiology , Follow-Up Studies , Genes, erbA , Humans , Hyperthyroidism/complications , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Placental Circulation/physiology , Pregnancy , Prenatal Exposure Delayed Effects , Random Allocation , Risk Assessment , Sampling Studies , Thyroid Hormone Resistance Syndrome/physiopathology
9.
J Clin Endocrinol Metab ; 102(7): 2620-2627, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28379567

ABSTRACT

Context: In animal models, disruption of thyroid hormone (TH) receptor-ß (TRß) reduces the long/medium wavelength (L/M) and increases the short-wavelength (S) cones. Retinal photoreceptor (RP) functions are unknown in patients with resistance to TH syndrome (RTHß) with dominant-negative TRß mutations. Objective: To investigate RP functions in RTHß. Design, Setting, and Participants: Case-control study involving 27 RTHß patients and 31 age/sex-matched controls, conducted in two tertiary referral centers in Italy. Main Outcome Measures: Color vision sensitivity assessed by Farnsworth; central macular thickness (CMT) of the outer retinal layer measured by spectral-domain optical coherence tomography; and retinal function tested by full-field electroretinogram (ERG) and S-cone ERG. Results: Color sensitivity was worse in RTHß patients than controls (P = 0.002). CMT was overlapping between the study groups but directly correlated with sex hormone-binding globuline levels in RTHß. We found a significant reduction in amplitude of the cone (P = 0.024) and of the rod response (P = 0.006) in the ERG of RTHß patients compared with controls. The response of the L/M cones measured by a specialized ERG test was lower in RTHß than controls (P = 0.027), whereas no differences were found in the S-cone response. No correlations were found between TH levels, total error score, or electrophysiological results. Furthermore, no differences were found between patients with maternal or de novo/paternal inheritance. Conclusions: We report, to our knowledge, the first in vivo evidence of functional defects of RP in RTHß. These changes occur independently of endogenous TH levels or the prenatal exposure to high or normal maternal TH.


Subject(s)
Color Vision Defects/diagnosis , Photoreceptor Cells, Vertebrate/pathology , Thyroid Hormone Resistance Syndrome/physiopathology , Adult , Case-Control Studies , Color Vision/physiology , Electrophysiology , Electroretinography/methods , Female , Humans , Italy , Male , Middle Aged , Reference Values , Statistics, Nonparametric , Tertiary Care Centers , Thyroid Function Tests , Tomography, Optical Coherence/methods , Young Adult
10.
Pediatr Endocrinol Diabetes Metab ; 23(4): 209-214, 2017.
Article in English | MEDLINE | ID: mdl-29574474

ABSTRACT

Thyroid hormone resistance syndrome, also known as Refetoff syndrome, is a rare disease associated with decreased reaction of body tissues to thyroid hormones (TH). Patients with Refetoff syndrome tend to have elevated free TH concentrations and normal or inadequately elevated TSH (caused by thyrotropic cells in the pituitary gland insensitivity to TH). The cause of the disease is the mutation in TR-beta receptor gene. Depending on the clinical presentation generalised and pituitary resistance to TH are described. The syndrome is often misdiagnosed as hyperthyroidism and unnecessarily treated with anti-thyroid drugs. Some patients receive l-thyroxine treatment for apparent hypothyroidism. In this report, three patients are presented with a long history in our Clinic and Outpatient Clinic.


Subject(s)
Hyperthyroidism/drug therapy , Hypothyroidism/drug therapy , Pituitary Gland/metabolism , Thyroid Hormone Resistance Syndrome/drug therapy , Thyroid Hormone Resistance Syndrome/physiopathology , Thyroid Hormones/blood , Thyroxine/therapeutic use , Adolescent , Child , Female , Humans , Male , Treatment Outcome , Young Adult
12.
J Clin Endocrinol Metab ; 101(2): 349-52, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26652765

ABSTRACT

CONTEXT: A majority of patients with resistance to thyroid hormone (RTH) are asymptomatic, whereas some patients show signs of hyperthyroidism, or hypothyroidism, or both. Thyrotoxic periodic paralysis is the most common form of acquired periodic paralysis. However, it has not been reported in a patient with RTH up to now. OBJECTIVE: We evaluated a 36-year-old male patient from China with elevated serum free T4 and free T3 and inappropriately high TSH who presented with periodic paralysis. STUDY DESIGN: Clinical, biochemical, and radiological assessments, as well as DNA sequencing, were performed. RESULTS: The patient's laboratory tests revealed the following: TSH, 6.14 mIU/L (0.27-4.2 mIU/L); free T3, 12.85 pmol/L (2.8-7.1 pmol/L); free T4, 33.62 pmol/L (9.05-25.5 pmol/L); and serum SHBG, 19.4 nmol/L (18.3-54.1 nmol/L). No significant suppression of TSH was observed in the rapid TSH suppression test with somatostatin analogs. Compound muscle action potential after exercise of the patient was reduced by 58%. Sequencing of thyroid hormone receptor genes confirmed a C446S mutation in the THRß gene. CONCLUSIONS: This is the first report of periodic paralysis as a new phenotype of RTH syndrome.


Subject(s)
Paralyses, Familial Periodic/genetics , Thyroid Hormone Resistance Syndrome/genetics , Adult , DNA/genetics , Hormone Antagonists/pharmacology , Humans , Male , Mutation/genetics , Paralyses, Familial Periodic/physiopathology , Sex Hormone-Binding Globulin/analysis , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Resistance Syndrome/physiopathology , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood
13.
Gac Med Mex ; 150(5): 465-9, 2014.
Article in Spanish | MEDLINE | ID: mdl-25275849

ABSTRACT

Thyroid hormone resistance is a syndrome characterized by a reduced response to thyroid hormone with different degrees of resistance at target tissues. We present the clinical features, physical findings, and study protocol in a woman with thyroid hormone resistance. An arginine to tryptophan mutation on the ß isoform of the thyroid hormone receptor gene was demonstrated. Thyroid hormone resistance is an uncommon cause of thyroid dysfunction. It is necessary to perform an adequate study and confirmation to avoid an inadequate and ineffective treatment of this condition.


Subject(s)
Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Resistance Syndrome/physiopathology , Adult , Female , Humans , Mutation , Thyroid Hormone Resistance Syndrome/genetics
14.
Lancet Diabetes Endocrinol ; 2(8): 619-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24969835

ABSTRACT

BACKGROUND: The thyroid hormone receptor α gene (THRA) transcript is alternatively spliced to generate either thyroid hormone receptor (TR)α1 or a non-hormone-binding variant protein, TRα2, the function of which is unknown. Here, we describe the first patients identified with a mutation in THRA that affects both TRα1 and TRα2, and compare them with patients who have resistance to thyroid hormone owing to a mutation affecting only TRα1, to delineate the relative roles of TRα1 and TRα2. METHODS: We did clinical, biochemical, and genetic analyses of an index case and her two sons. We assessed physical and radiological features, thyroid function, physiological and biochemical markers of thyroid hormone action, and THRA sequence. FINDINGS: The patients presented in childhood with growth failure, developmental delay, and constipation, which improved after treatment with thyroxine, despite normal concentrations of circulating thyroid hormones. They had similar clinical (macrocephaly, broad faces, skin tags, motor dyspraxia, slow speech), biochemical (subnormal ratio of free thyroxine:free tri-iodothyronine [T3], low concentration of total reverse T3, high concentration of creatine kinase, mild anaemia), and radiological (thickened calvarium) features to patients with TRα1-mediated resistance to thyroid hormone, although our patients had a heterozygous mis-sense mutation (Ala263Val) in both TRα1 and TRα2 proteins. The Ala263Val mutant TRα1 inhibited the transcriptional function of normal receptor in a dominant-negative fashion. By contrast, function of Ala263Val mutant TRα2 matched its normal counterpart. In vitro, high concentrations of T3 restored transcriptional activity of Ala263Val mutant TRα1, and reversed the dominant-negative inhibition of its normal counterpart. High concentrations of T3 restored expression of thyroid hormone-responsive target genes in patient-derived blood cells. INTERPRETATION: TRα1 seems to be the principal functional product of the THRA gene. Thyroxine treatment alleviates hormone resistance in patients with mutations affecting this gene, possibly ameliorating the phenotype. These findings will help the diagnosis and treatment of other patients with resistance to thyroid hormone resulting from mutations in THRA. FUNDING: Wellcome Trust, NIHR Cambridge Biomedical Research Centre, Marie Curie Actions, Foundation for Development of Internal Medicine in Europe.


Subject(s)
Alternative Splicing , Mutation, Missense , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Resistance Syndrome/genetics , Adult , Amino Acid Substitution , Family Health , Female , Gait Apraxia/etiology , Heterozygote , Humans , Male , Megalencephaly/etiology , Middle Aged , Polyps/etiology , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Skin Neoplasms/etiology , Speech Disorders/etiology , Thyroid Hormone Receptors alpha/agonists , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Resistance Syndrome/drug therapy , Thyroid Hormone Resistance Syndrome/physiopathology , Thyroxine/therapeutic use , Treatment Outcome
15.
Eur J Clin Nutr ; 68(2): 234-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24281313

ABSTRACT

BACKGROUND/OBJECTIVES: The objective of this study was to develop approaches to expressing resting energy expenditure (REE) and lean body mass (LM) phenotypes of metabolic disorders in terms of Z-scores relative to their predicted healthy values. SUBJECTS/METHODS: Body composition and REE were measured in 135 healthy participants. Prediction equations for LM and REE were obtained from linear regression and the range of normality by the standard deviation of residuals. Application is demonstrated in patients from three metabolic disorder groups (lipodystrophy, n=7; thyrotoxicosis, n=16; and resistance to thyroid hormone (RTH), n=46) in which altered REE and/or LM were characterised by departure from the predicted healthy values, expressed as a Z-score. RESULTS: REE (kJ/min) = -0.010 × age (years)+0.016 × FM (kg)+0.054 × fat-free mass (kg)+1.736 (R2 = 0.732, RSD = 0.36 kJ/min). LM (kg)=5.30 × bone mineral content (kg)+10.66 × height2 (m)+6.40 (male). LM (kg)=0.20 × fat (kg)+14.08 × height2 (m)-2.93 (female).(male R2=0.55, RSD = 3.90 kg; female R2 = 0.59, RSD=3.85 kg).We found average Z-scores for REE and LM of 1.77 kJ/min and -0.17 kg in the RTH group, 5.82 kJ/min and -1.23 kg in the thyrotoxic group and 2.97 kJ/min and 4.20 kg in the LD group. CONCLUSION: This approach enables comparison of data from individuals with metabolic disorders with those of healthy individuals, describing their departure from the healthy mean by a Z-score.


Subject(s)
Body Composition , Energy Metabolism , Metabolic Diseases/physiopathology , Adolescent , Adult , Female , Humans , Linear Models , Lipodystrophy/physiopathology , Male , Middle Aged , Sex Factors , Thyroid Hormone Resistance Syndrome/physiopathology , Thyrotoxicosis/physiopathology
16.
J Clin Endocrinol Metab ; 98(7): 3029-38, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23633213

ABSTRACT

CONTEXT: Recently the first patients with inactivating mutations in T3 receptor (TR)-α1 have been identified. These patients have low free T4, low T4, high T3, low rT3, and normal TSH serum levels, in combination with growth retardation, delayed bone development, and constipation. OBJECTIVE: The aim of the current study was to report the effects of levothyroxine (LT4) treatment on the clinical phenotype of 2 patients (father and daughter) with a heterozygous inactivating mutation in TRα1. SETTING AND PARTICIPANTS: Both patients were treated with LT4 for the last 5 years. To evaluate the effect of LT4 treatment, LT4 was withdrawn for 35 days and subsequently reinitiated. Data were collected from medical records, by reanalysis of serum collected over the last 6 years, and by a detailed clinical evaluation. RESULTS: Treatment with LT4 resulted in a suppression of serum TSH and normalization of serum free T4 and rT3, whereas T3 levels remained elevated in both patients. In addition, there was a normalization of the dyslipidemia as well as a response in serum IGF-I, SHBG, and creatine kinase in the index patient. All these parameters returned to pretreatment values when LT4 was briefly stopped. LT4 also resulted in an improvement of certain clinical features, such as constipation and nerve conductance. However, cognitive and fine motor skill defects remained. CONCLUSION: This study reports the consequences of LT4 treatment over a prolonged period of time in 2 of the first patients with a heterozygous mutation in TRα1. LT4 therapy leads to an improvement of certain but not all features of the clinical phenotype.


Subject(s)
Frameshift Mutation , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Resistance Syndrome/drug therapy , Thyroid Hormone Resistance Syndrome/genetics , Thyroxine/therapeutic use , Child , Constipation/etiology , Constipation/prevention & control , Drug Resistance , Dyslipidemias/etiology , Dyslipidemias/prevention & control , Female , Heterozygote , Humans , Male , Middle Aged , Neural Conduction/drug effects , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Resistance Syndrome/metabolism , Thyroid Hormone Resistance Syndrome/physiopathology , Thyroid Hormones/blood , Treatment Outcome
17.
Endocr Rev ; 34(5): 619-57, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23575764

ABSTRACT

Thyroid hormones are extremely important for metabolism, development, and growth during the lifetime. The hypothalamo-pituitary-thyroid axis is precisely regulated for these purposes. Much of our knowledge of this hormonal axis is derived from experiments in animals and mutations in man. This review examines the hypothalamo-pituitary-thyroid axis particularly in relation to the regulated 24-hour serum TSH concentration profiles in physiological and pathophysiological conditions, including obesity, primary hypothyroidism, pituitary diseases, psychiatric disorders, and selected neurological diseases. Diurnal TSH rhythms can be analyzed with novel and precise techniques, eg, operator-independent deconvolution and approximate entropy. These approaches provide indirect insight in the regulatory components in pathophysiological conditions.


Subject(s)
Health Status , Thyrotropin/metabolism , Adult , Animals , Circadian Rhythm , Female , Humans , Hypothalamo-Hypophyseal System/physiopathology , Hypothyroidism/physiopathology , Infant, Newborn , Male , Mental Disorders/physiopathology , Middle Aged , Nervous System Diseases/physiopathology , Obesity/physiopathology , Pituitary Diseases/physiopathology , Pregnancy , Sex Factors , Thyroid Diseases/physiopathology , Thyroid Gland/physiopathology , Thyroid Hormone Resistance Syndrome/physiopathology , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood
18.
Biochim Biophys Acta ; 1830(7): 4004-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23528896

ABSTRACT

BACKGROUND: Thyroid hormone acts via receptor subtypes (TRα1, TRß1, TRß2) with differing tissue distributions, encoded by distinct genes (THRA, THRB). THRB mutations cause a disorder with central (hypothalamic-pituitary) resistance to thyroid hormone action with markedly elevated thyroid hormone and normal TSH levels. SCOPE OF REVIEW: This review describes the clinical features, genetic and molecular pathogenesis of a homologous human disorder mediated by defective THRA. Clinical features include growth retardation, skeletal dysplasia and constipation associated with low-normal T4 and high-normal T3 levels and a low T4/T3 ratio, together with subnormal reverse T3 levels. Heterozygous TRa1 mutations in affected individuals generate defective mutant receptors which inhibit wild-type receptor action in a dominant negative manner. MAJOR CONCLUSIONS: Mutations in human TRα1 mediate RTH with features of hypothyroidism in particular tissues (e.g. skeleton, gastrointestinal tract), but are not associated with a markedly dysregulated pituitary-thyroid axis. GENERAL SIGNIFICANCE: Human THRA mutations could be more common but may have eluded discovery due to the absence of overt thyroid dysfunction. Nevertheless, in the appropriate clinical context, a thyroid biochemical signature (low T4/T3 ratio, subnormal reverse T3 levels), may enable future identification of cases. This article is part of a Special Issue entitled Thyroid hormone signalling.


Subject(s)
Thyroid Hormone Receptors alpha/physiology , Thyroid Hormone Resistance Syndrome/physiopathology , Thyroid Hormones/physiology , Humans , Mutation , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormone Resistance Syndrome/metabolism , Thyroid Hormones/genetics , Thyroid Hormones/metabolism
19.
Biochim Biophys Acta ; 1830(7): 3987-4003, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22986150

ABSTRACT

BACKGROUND: Six known steps are required for the circulating thyroid hormone (TH) to exert its action on target tissues. For three of these steps, human mutations and distinct phenotypes have been identified. SCOPE OF REVIEW: The clinical, laboratory, genetic and molecular characteristics of these three defects of TH action are the subject of this review. The first defect, recognized 45years ago, produces resistance to TH and carries the acronym, RTH. In the majority of cases it is caused by TH receptor ß gene mutations. It has been found in over 3000 individuals belonging to approximately 1000 families. Two relatively novel syndromes presenting reduced sensitivity to TH involve membrane transport and metabolism of TH. One of them, caused by mutations in the TH cell-membrane transporter MCT8, produces severe psychomotor defects. It has been identified in more than 170 males from 90 families. A defect of the intracellular metabolism of TH in 10 individuals from 8 families is caused by mutations in the SECISBP2 gene required for the synthesis of selenoproteins, including TH deiodinases. MAJOR CONCLUSIONS: Defects at different steps along the pathway leading to TH action at cellular level can manifest as reduced sensitivity to TH. GENERAL SIGNIFICANCE: Knowledge of the molecular mechanisms involved in TH action allows the recognition of the phenotypes caused by defects of TH action. Once previously known defects have been ruled out, new molecular defects could be sought, thus opening the avenue for novel insights in thyroid physiology. This article is part of a Special Issue entitled Thyroid hormone signaling.


Subject(s)
Thyroid Hormone Resistance Syndrome/physiopathology , Thyroid Hormones/physiology , Biological Transport/genetics , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation , Phenotype , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Receptors, Thyroid Hormone/physiology , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormone Resistance Syndrome/metabolism , Thyroid Hormones/genetics , Thyroid Hormones/metabolism
20.
Nihon Rinsho ; 70(11): 1951-7, 2012 Nov.
Article in Japanese | MEDLINE | ID: mdl-23214067

ABSTRACT

Resistance to thyroid hormone (RTH) is a syndrome in which the responsiveness of end organs to thyroid hormone (TH) is reduced. Given that the TH-responsive end-organs include pituitary thyrotrophs, almost all patients with RTH manifest unsuppressed thyrotropin (TSH) despite elevated free-T4 and free-T3 levels. This abnormal finding in the thyroid function test is termed "syndrome of inappropriate secretion of TSH" (SITSH) or "central hyperthyroidism". Patients with TSH-secreting pituitary tumors(TSHoma) also manifest SITSH. Thus, the differential diagnosis of RTH vs. TSHoma is sometimes difficult and challenging. In this review article, the etiology of RTH and diagnostic approach for SITSH are explained and an algorithm for differential diagnosis of RTH vs. TSHoma is proposed.


Subject(s)
Hyperpituitarism/diagnosis , Thyroid Hormone Resistance Syndrome/diagnosis , Thyroid Hormones/metabolism , Humans , Hyperpituitarism/etiology , Hyperpituitarism/physiopathology , Hyperthyroidism/etiology , Hyperthyroidism/physiopathology , Pituitary Neoplasms/diagnosis , Thyroid Gland/metabolism , Thyroid Hormone Resistance Syndrome/etiology , Thyroid Hormone Resistance Syndrome/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL