Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 273
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pituitary ; 21(1): 76-83, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29255988

RESUMEN

PURPOSE: Mutation frequencies of PROP1, POU1F1 and HESX1 in patients with combined pituitary hormone deficiencies (CPHD) vary substantially between populations. They are low in sporadic CPHD patients in Western Europe. However, most clinicians still routinely send DNA of their CPHD patients for genetic screening of these pituitary transcription factors. Before we can recommend against screening of PROP1, POU1F1 and HESX1 as part of routine work-up for Western-European sporadic CPHD patients, it is crucial to rule out possible defects in regulatory regions of these genes, which could also disturb the complex process of pituitary organogenesis. METHODS: The regulatory regions of PROP1, POU1F1 and HESX1 are not covered by Whole Exome Sequencing as they are largely located outside the coding regions. Therefore, we manually sequenced the regulatory regions, previously defined in the literature, of PROP1, POU1F1 and HESX1 among 88 Dutch patients with CPHD. We studied promoter SNPs in relation to phenotypic data. RESULTS: We found six known SNPs in the PROP1 promoter. In the POU1F1 promoter, we found one new variant and two known SNPs. We did not find any variant in the HESX1 promoter. CONCLUSION: Although the new POU1F1 variant might explain the phenotype of one patient, the general conclusion of this study is that variants in regulatory regions of PROP1, POU1F1 and HESX1 are rare in patients with sporadic CPHD in the Netherlands. We recommend that genetic screening of these pituitary transcription factors should no longer be part of routine work-up for Western-European, and especially Dutch, sporadic CPHD patients.


Asunto(s)
Secuenciación del Exoma , Pruebas Genéticas/métodos , Proteínas de Homeodominio/genética , Hipopituitarismo/genética , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Factor de Transcripción Pit-1/genética , Femenino , Estudios de Asociación Genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Hipopituitarismo/diagnóstico , Masculino , Países Bajos , Fenotipo , Valor Predictivo de las Pruebas , Procedimientos Innecesarios
2.
Hum Mutat ; 38(3): 260-264, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27805744

RESUMEN

Mutations in the thyroid hormone transporter SLC16A2 (MCT8) cause the Allan-Herndon-Dudley Syndrome (AHDS), characterized by severe psychomotor retardation and peripheral thyrotoxicosis. Here, we report three newly identified AHDS patients. Previously documented mutations were identified in probands 1 (p.R271H) and 2 (p.G564R), resulting in a severe clinical phenotype. A novel mutation (p.G564E) was identified in proband 3, affecting the same Gly564 residue, but resulting in a relatively mild clinical phenotype. Functional analysis in transiently transfected COS-1 and JEG-3 cells showed a near-complete inactivation of TH transport for p.G564R, whereas considerable cell-type-dependent residual transport activity was observed for p.G564E. Both mutants showed a strong decrease in protein expression levels, but differentially affected Vmax and Km values of T3 transport. Our findings illustrate that different mutations affecting the same residue may have a differential impact on SLC16A2 transporter function, which translates into differences in severity of the clinical phenotype.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/diagnóstico , Discapacidad Intelectual Ligada al Cromosoma X/genética , Transportadores de Ácidos Monocarboxílicos/genética , Hipotonía Muscular/diagnóstico , Hipotonía Muscular/genética , Atrofia Muscular/diagnóstico , Atrofia Muscular/genética , Mutación , Fenotipo , Biomarcadores , Niño , Preescolar , Humanos , Imagen por Resonancia Magnética , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/terapia , Hipotonía Muscular/terapia , Atrofia Muscular/terapia , Linaje , Simportadores
3.
Eur J Epidemiol ; 32(2): 135-144, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27709449

RESUMEN

Human chorionic gonadotropin (hCG) is a pregnancy-specific hormone that regulates placental development. hCG concentrations vary widely throughout gestation and differ based on fetal sex. Abnormal hCG concentrations are associated with adverse pregnancy outcomes including fetal growth restriction. We studied the association of hCG concentrations with fetal growth and birth weight. In addition, we investigated effect modification by gestational age of hCG measurement and fetal sex. Total serum hCG (median 14.4 weeks, 95 % range 10.1-26.2), estimated fetal weight (measured by ultrasound during 18-25th weeks and >25th weeks) and birth weight were measured in 7987 mother-child pairs from the Generation R cohort and used to establish fetal growth. Small for gestational age (SGA) was defined as a standardized birth weight lower than the 10th percentile of the study population. There was a non-linear association of hCG with birth weight (P = 0.009). However, only low hCG concentrations measured during the late first trimester (11th and 12th week) were associated with birth weight and SGA. Low hCG concentrations measured in the late first trimester were also associated with decreased fetal growth (P = 0.0002). This was the case for both male and female fetuses. In contrast, high hCG concentrations during the late first trimester were associated with increased fetal growth amongst female, but not male fetuses. Low hCG in the late first trimester is associated with lower birth weight due to a decrease in fetal growth. Fetal sex differences exist in the association of hCG concentrations with fetal growth.


Asunto(s)
Peso al Nacer/fisiología , Gonadotropina Coriónica/sangre , Desarrollo Fetal/fisiología , Primer Trimestre del Embarazo , Adulto , Femenino , Humanos , Recién Nacido , Masculino , Embarazo , Factores Sexuales
4.
Pituitary ; 20(5): 499-508, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28593555

RESUMEN

INTRODUCTION: Growth hormone is secreted by the pituitary gland, which forms part of the craniofacial midline. IRF6 encodes a transcription factor involved in the development of the craniofacial midline and mutations in IRF6 are known to disturb craniofacial development. Craniofacial and pituitary development are closely related. After whole exome sequencing revealed a new mutation in IRF6 in a family with Idiopathic Growth Hormone Deficiency (IGHD), we screened the remainder of our IGHD cohort for mutations in this gene and related their genotypes to pituitary and craniofacial morphology. MATERIALS AND METHODS: We sequenced all coding exons and exon-intron boundaries of IRF6 in 81 patients with IGHD. We performed a multiplex ligation-dependent probe amplification (MLPA) in order to exclude copy number variations in IRF6. We analyzed facial measurements taken from standardized digital pictures of 48 patients. RESULTS: We found two new variants and eleven polymorphisms. Apart from the new mutation found in the index family (p.Arg233Cys), we found one other new heterozygous missense mutation in IRF6 (Pro456Ser). p.Arg233Cys was reported as extremely rare in exome databases (1 allele out of 120.852 alleles sequenced), strictly conserved among species and was predicted deleterious by all variant predictor programs. Pro456Ser was predicted to be benign. MLPA did not reveal any exon deletions or duplications in any of the patients. CONCLUSION: This is the first report of IRF6 analysis in an IGHD cohort. We found one new mutation which, based on in silico analysis, could be of functional relevance. However, we did not find any mutations in the other patients. Therefore, we conclude that IRF6 defects are rare in IGHD patients and further research should focus on new candidate genes.


Asunto(s)
Enanismo Hipofisario/genética , Factores Reguladores del Interferón/genética , Enfermedades de la Hipófisis/genética , Hipófisis/metabolismo , Variaciones en el Número de Copia de ADN/genética , Exones/genética , Femenino , Pruebas Genéticas , Hormona de Crecimiento Humana/genética , Humanos , Masculino , Mutación , Linaje , Hipófisis/patología
5.
Clin Endocrinol (Oxf) ; 85(6): 902-909, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27384268

RESUMEN

BACKGROUND: Low maternal thyroid function during early pregnancy is associated with various adverse outcomes including impaired neurocognitive development of the offspring, premature delivery and abnormal birthweight. AIM: To aid doctors in the risk assessment of thyroid dysfunction during pregnancy, we set out to investigate clinical risk factors and derive a prediction model based on easily obtainable clinical variables. METHODS: In total, 9767 women during early pregnancy (≤18 week) were selected from two population-based prospective cohorts: the Generation R Study (N = 5985) and the ABCD study (N = 3782). We aimed to investigate the association of easily obtainable clinical subject characteristics such as maternal age, BMI, smoking status, ethnicity, parity and gestational age at blood sampling with the risk of low free thyroxine (FT4) and elevated thyroid stimulating hormone (TSH), determined according to the 2·5th-97·5th reference range in TPOAb negative women. RESULTS: BMI, nonsmoking and ethnicity were risk factors for elevated TSH levels; however, the discriminative ability was poor (range c-statistic of 0·57-0·60). Sensitivity analysis showed that addition of TPOAbs to the model yielded a c-statistic of 0·73-0·75. Maternal age, BMI, smoking, parity and gestational age at blood sampling were risk factors for low FT4, which taken together provided adequate discrimination (range c-statistic of 0·72-0·76). CONCLUSIONS: Elevated TSH levels depend predominantly on TPOAb levels, and prediction of elevated TSH levels is not possible with clinical characteristics only. In contrast, the validated clinical prediction model for FT4 had high discriminative value to assess the likelihood of low FT4 levels.


Asunto(s)
Hipotiroidismo/diagnóstico , Modelos Biológicos , Valor Predictivo de las Pruebas , Complicaciones del Embarazo , Adulto , Autoanticuerpos/sangre , Autoantígenos/inmunología , Estudios de Cohortes , Femenino , Edad Gestacional , Humanos , Yoduro Peroxidasa/inmunología , Proteínas de Unión a Hierro/inmunología , Persona de Mediana Edad , Embarazo , Pronóstico , Estudios Prospectivos , Factores de Riesgo , Tirotropina/sangre , Tiroxina/sangre , Adulto Joven
6.
Clin Endocrinol (Oxf) ; 85(5): 757-763, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27187054

RESUMEN

AIM: We aimed to investigate TT4 physiological aspects and associations with clinical end-points. BACKGROUND: Total T4 (TT4) has been suggested as a marker for maternal thyroid function during pregnancy because as compared to FT4 (i) TT4 measurement is not affected by binding protein interference, (ii) TT4 is considered to be more stable from the second trimester onwards, and (iii) TT4 better reflects changes in the hypothalamic-pituitary-thyroid axis. However, this is based on data from small studies, and, more importantly, it is unknown whether TT4 is associated with adverse pregnancy or child outcomes. METHODS: We selected 5647 mother-child pairs from a large population-based prospective cohort with data on maternal TSH, FT4 and TT4 during early pregnancy (median 13·2 weeks, 95% range 9·8-17·6). We used multivariable (non)linear and logistic regression models to study the association of maternal TT4 with pre-eclampsia, premature delivery, birthweight and offspring IQ and compare the results with previously obtained results for FT4. RESULTS: The change of mean TT4 levels was 27·5% compared to 20·2% for FT4. There was a log-linear association of TT4 and FT4 with TSH, but the explained variability of TSH was much lower for TT4 than for FT4 (R-squared TT4: 2·5% vs 8·0% for FT4). In contrast to FT4, there was no independent association of maternal TT4 with pre-eclampsia, premature delivery, birthweight or offspring IQ. CONCLUSION: Maternal TT4 levels are highly variable in the first half of pregnancy and are poorly related to maternal TSH. This study shows that maternal TT4 levels are either not associated, or not better associated as compared to FT4, with adverse pregnancy or child outcomes. This suggests that the maternal TT4 is inferior to FT4 in the assessment of maternal thyroid function during the first half of pregnancy.


Asunto(s)
Complicaciones del Embarazo/inducido químicamente , Segundo Trimestre del Embarazo/sangre , Tiroxina/efectos adversos , Tiroxina/sangre , Adulto , Niño , Estudios de Cohortes , Femenino , Humanos , Sistema Hipotálamo-Hipofisario , Recién Nacido de Bajo Peso , Inteligencia , Pruebas de Inteligencia , Madres , Sistema Hipófiso-Suprarrenal , Embarazo , Complicaciones del Embarazo/sangre , Estudios Prospectivos , Pruebas de Función de la Tiroides , Adulto Joven
7.
Clin Chem ; 61(5): 704-13, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25829408

RESUMEN

BACKGROUND: Gestational thyroid dysfunction is common and associated with maternal and child morbidity and mortality. During pregnancy, profound changes in thyroid physiology occur, resulting in different thyroid-stimulating hormone (TSH) and free thyroxine (FT4) reference intervals compared to the nonpregnant state. Therefore, international guidelines recommend calculating trimester- and assay-specific reference intervals per center. If these reference intervals are unavailable, TSH reference intervals of 0.1-2.5 mU/L for the first trimester and 0.2-3.0 mU/L for the second trimester are recommended. In daily practice, most institutions do not calculate institution-specific reference intervals but rely on these fixed reference intervals for the diagnosis and treatment of thyroid disorders during pregnancy. However, the calculated reference intervals for several additional pregnancy cohorts have been published in the last few years and show substantial variation. CONTENT: We provide a detailed overview of the available studies on thyroid function reference intervals during pregnancy, different factors that contribute to these reference intervals, and the maternal and child complications associated with only minor variations in thyroid function. SUMMARY: There are large differences in thyroid function reference intervals between different populations of pregnant women. These differences can be explained by variations in assays as well as population-specific factors, such as ethnicity and body mass index. The importance of using correct reference intervals is underlined by the fact that even small subclinical variations in thyroid function have been associated with detrimental pregnancy outcomes, including low birth weight and pregnancy loss. It is therefore crucial that institutions do not rely on fixed universal cutoff concentrations, but calculate their own pregnancy-specific reference intervals.


Asunto(s)
Complicaciones del Embarazo/fisiopatología , Pruebas de Función de la Tiroides , Femenino , Humanos , Embarazo , Estándares de Referencia
8.
Eur J Epidemiol ; 30(9): 1057-66, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25963653

RESUMEN

Human chorionic gonadotropin (hCG) is a pregnancy hormone secreted by the placental synctiotrophoblast cell layer that has been linked to fetal growth and various placental, uterine and fetal functions. In order to investigate the effects of hCG on clinical endpoints, knowledge on reference range (RR) methodology and determinants of gestational hCG levels is crucial. Moreover, a better understanding of gestational hCG physiology can improve current screening programs and future clinical management. Serum total hCG levels were determined in 8195 women participating in the Generation R Study. Gestational age specific RRs using 'ultrasound derived gestational age' (US RRs) were calculated and compared with 'last menstrual period derived gestational age' (LMP RRs) and a model-based RR. We also investigated which pregnancy characteristics were associated with hCG levels. Compared to the US RRs, the LMP RRs were lower, most notably for the median and lower limit levels. No considerable differences were found between RRs calculated in the general population or in uncomplicated pregnancies only. Maternal smoking, BMI, parity, ethnicity, fetal gender, placental weight and hyperemesis gravidarum symptoms were associated with total hCG. We provide gestational RRs for total hCG and show that total hCG values and RR cut-offs during pregnancy vary depending on pregnancy dating methodology. This is likely due to the influence of hCG on embryonic growth, suggesting that ultrasound based pregnancy dating might be less reliable in women with high/low hCG levels. Furthermore, we identify different pregnancy characteristics that influence total hCG levels considerably and should therefore be accounted for in clinical studies.


Asunto(s)
Gonadotropina Coriónica/sangre , Proteína Plasmática A Asociada al Embarazo/análisis , Embarazo/sangre , Adulto , Biomarcadores/sangre , Índice de Masa Corporal , Femenino , Edad Gestacional , Humanos , Países Bajos , Placenta , Diagnóstico Prenatal , Estudios Prospectivos , Valores de Referencia , Factores Socioeconómicos
9.
Biochim Biophys Acta ; 1830(7): 3974-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22543196

RESUMEN

BACKGROUND: As a prerequisite for thyroid hormone (TH) metabolism and action TH has to be transported into cells where TH deiodinases and receptors are located. The trans-membrane passage of TH is facilitated by TH transporters of which the monocarboxylate transporter MCT8 has been most intensively studied. Inactivating mutations in the gene encoding MCT8 are associated with a severe form of psychomotor retardation and abnormal serum TH levels (Allan-Herndon-Dudley syndrome). In order to define the underlying pathogenic mechanisms, Mct8 knockout mice have been generated and intensively studied. Most surprisingly, Mct8 ko mice do not show any neurological symptoms but fully replicate the abnormal serum thyroid state. SCOPE OF REVIEW: We will summarize the findings of these mouse studies that shed light on various aspects of Mct8 deficiency and unambiguously demonstrated the pivotal role of Mct8 in mediating TH transport in various tissues. These studies have also revealed the presence of the complex interplay between different pathogenic mechanisms that contribute to the generation of the abnormal TH serum profile. MAJOR CONCLUSIONS: Most importantly, studies of Mct8 ko mice indicated the presence of additional TH transporters that act in concert with Mct8. Interesting candidates for such a function are the L-type amino acid transporters Lat1 and Lat2 as well as the organic anion transporting polypeptide Oatp1c1. GENERAL SIGNIFICANCE: Overall, the analysis of Mct8 deficient mice has greatly expanded our knowledge about the (patho-) physiological function of this transporter and established a sound basis for the characterization of additional TH transporter candidates. This article is part of a Special Issue entitled Thyroid hormone signalling.


Asunto(s)
Proteínas de Transporte de Membrana/deficiencia , Hormonas Tiroideas/metabolismo , Animales , Transporte Biológico , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Transportadores de Ácidos Monocarboxílicos/deficiencia , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores , Hormonas Tiroideas/genética
10.
Clin Endocrinol (Oxf) ; 80(4): 598-606, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23992400

RESUMEN

OBJECTIVE: Iodine deficiency during pregnancy results in thyroid dysfunction and has been associated with adverse obstetric and foetal effects, leading to worldwide salt iodization programmes. As nowadays 69% of the world's population lives in iodine-sufficient regions, we investigated the effects of variation in iodine status on maternal and foetal thyroid (dys)function in an iodine-sufficient population. DESIGN, PARTICIPANTS AND MEASUREMENTS: Urinary iodine, serum TSH, free T4 (FT4) and TPO-antibody levels were determined in early pregnancy (13·3 (1·9) week; mean (SD)) in 1098 women from the population-based Generation R Study. Newborn cord serum TSH and FT4 levels were determined at birth. RESULTS: The median urinary iodine level was 222·5 µg/l, indicating an iodine-sufficient population. 30·8% and 11·5% had urinary iodine levels <150 and >500 µg/l, respectively. When comparing mothers with urinary iodine levels <150 vs ≥150 µg/l, and >500 vs ≤500 µg/l, there were no differences in the risk of maternal increased or decreased TSH, hypothyroxinaemia or hyperthyroidism. Mothers with urinary iodine levels >500 µg/l had a higher risk of a newborn with decreased cord TSH levels (5·6 ± 1·4 (mean ± SE) vs 2·1 ± 0·5%, P = 0·04), as well as a higher risk of a hyperthyroid newborn (3·1 ± 0·9 vs 0·6 ± 0·3%, P = 0·02). These mothers had newborns with higher cord FT4 levels (21·7 ± 0·3 vs 21·0 ± 0·1 pm, P = 0·04). Maternal urinary iodine levels <150 µg/l were not associated with newborn thyroid dysfunction. CONCLUSIONS: In an iodine-sufficient population, higher maternal urinary iodine levels are associated with an increased risk of a hyperthyroid newborn.


Asunto(s)
Hipertiroidismo/sangre , Enfermedades del Recién Nacido/sangre , Yodo/orina , Femenino , Sangre Fetal , Humanos , Recién Nacido , Yoduros , Madres , Embarazo , Tirotropina/sangre , Tiroxina/sangre
11.
Hum Mutat ; 34(7): 1018-25, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23568789

RESUMEN

SLC 16A2, the gene for the second member of the solute carrier family 16 (monocarboxylic acid transporter), located on chromosome Xq13.2, encodes a very efficient thyroid hormone transporter: monocarboxylate transporter 8, MCT8. Its loss of function is responsible in males for a continuum of psychomotor retardation ranging from severe (no motor acquisition, no speech) to mild (ability to walk with help and a few words of speech). Triiodothyronine uptake measurement in transfected cells and, more recently, patient fibroblasts, has been described to study the functional consequences of MCT8 mutations. Here, we describe three novel MCT8 mutations, including one missense variation not clearly predicted to be damaging but found in a severely affected patient. Functional studies in fibroblasts and JEG3 cells demonstrate the usefulness of both cellular models in validating the deleterious effects of a new MCT8 mutation if there is still a doubt as to its pathogenicity. Moreover, the screening of fibroblasts from a large number of patient fibroblasts and of transfected mutations has allowed us to demonstrate that JEG3 transfected cells are more relevant than fibroblasts in revealing a genotype-phenotype correlation.


Asunto(s)
Estudios de Asociación Genética , Transportadores de Ácidos Monocarboxílicos/genética , Mutación , Trastornos Psicomotores/genética , Trastornos Psicomotores/fisiopatología , Adolescente , Línea Celular Tumoral , Células Cultivadas , Niño , Preescolar , Fibroblastos/metabolismo , Humanos , Masculino , Transportadores de Ácidos Monocarboxílicos/metabolismo , Índice de Severidad de la Enfermedad , Simportadores , Hormonas Tiroideas/metabolismo
12.
Clin Endocrinol (Oxf) ; 79(5): 595-605, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23834164

RESUMEN

Normal thyroid hormone (TH) metabolism and action require adequate cellular TH signalling. This entails proper function of TH transporters in the plasma membrane, intracellular deiodination of TH and action of the bioactive hormone T3 at its nuclear receptors (TRs). The present review summarizes the discoveries of different syndromes with reduced sensitivity at the cellular level. Mutations in the TH transporter MCT8 cause psychomotor retardation and abnormal thyroid parameters. Mutations in the SBP2 protein, which is required for normal deiodination, give rise to a multisystem disorder including abnormal thyroid function tests. Mutations in TRß1 are a well-known cause of resistance to TH with mostly a mild phenotype, while only recently, patients with mutations in TRα1 were identified. The latter patients have slightly abnormal TH levels, growth retardation and cognitive defects. This review will describe the mechanisms of disease, clinical phenotype, diagnostic testing and suggestions for treatment strategies for each of these syndromes.


Asunto(s)
Hormonas Tiroideas/metabolismo , Membrana Celular/metabolismo , Humanos , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Simportadores , Glándula Tiroides/metabolismo
13.
Clin Endocrinol (Oxf) ; 78(3): 415-23, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22897141

RESUMEN

OBJECTIVE: Combined pituitary hormone deficiency (CPHD) is characterized by deficiencies of two or more anterior pituitary hormones. Its genetic cause is unknown in the majority of cases. The Hedgehog (Hh) signalling pathway has been implicated in disorders associated with pituitary development. Mutations in Sonic Hedgehog (SHH) have been described in patients with holoprosencephaly (with or without pituitary involvement). Hedgehog interacting protein (HHIP) has been associated with variations in adult height in genome wide association studies. We investigated whether mutations in these two genes of the Hh pathway, SHH and HHIP, could result in 'idiopathic' CPHD. DESIGN/PATIENTS: We directly sequenced the coding regions and exon - intron boundaries of SHH and HHIP in 93 CPHD patients of the Dutch HYPOPIT study in whom mutations in the classical CPHD genes PROP1, POU1F1, HESX1, LHX3 and LHX4 had been ruled out. We compared the expression of Hh genes in Hep3B transfected cells between wild-type proteins and mutants. RESULTS: We identified three single-nucleotide variants (p.Ala226Thr, c.1078C>T and c.*8G>T) in SHH. The function of the latter was severely affected in our in vitro assay. In HHIP, we detected a new activating variant c.-1G>C, which increases HHIP's inhibiting function on the Hh pathway. CONCLUSIONS: Our results suggest involvement of the Hedgehog pathway in CPHD. We suggest that both SHH and HHIP are investigated as a second screening in CPHD, after mutations in the classical CPHD genes have been ruled out.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Hedgehog/genética , Hipopituitarismo/genética , Glicoproteínas de Membrana/genética , Adolescente , Adulto , Femenino , Humanos , Hipopituitarismo/etiología , Masculino , Mutación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
14.
Clin Endocrinol (Oxf) ; 78(2): 310-5, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22924588

RESUMEN

OBJECTIVE: Monocarboxylate transporter 8 (MCT8) is an essential thyroid hormone (TH) transporter as humans with MCT8 mutations have severe neurological and endocrine abnormalities. The objectives are (i) to identify novel MCT8 mutations and (ii) to assess their functional relevance; (iii) to describe the effects of block-and-replace treatment in an MCT8 patient. DESIGN: The TOP-R study is a cross-sectional nation-wide multicentre study. PATIENTS: Subjects with unexplained mental retardation (MR) were screened for MCT8 mutations. RESULTS: We identified three mutations: p.F501del (previously described), p.L492P and p.T162T. The F501del and L492P mutants, but not the T162T mutant, showed diminished T3, T4 and rT3 transport in transfected cells. TH transport in T162T fibroblasts was also not affected. One patient was treated with block-and-replace therapy to normalize serum TH levels. The results indicated a slow onset of the decrease in serum T4 and T3 by successive treatment with methimazole and PTU, and eventually their complete normalization by administration of LT4 with PTU but not with methimazole. The frequency of MCT8 mutations in males with X-linked MR approximately 3·9%. CONCLUSIONS: We identified several MCT8 mutations in a cohort of subjects with unexplained MR. We demonstrated the pathogenicity of two missense mutations. The synonymous variant did not affect TH transport. Block-and-replace therapy of one patient reversed the TH abnormalities. Our data suggest a decreased TH secretion rate and an increased T4 to T3 conversion by the type I deiodinase in patients with MCT8 mutations. Our study indicates that MCT8 mutations are a relatively frequent cause of X-linked MR.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Adulto , Transporte Biológico , Estudios de Cohortes , Estudios Transversales , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Transportadores de Ácidos Monocarboxílicos/genética , Mutación , Simportadores , Tiroxina/metabolismo
15.
Hum Mol Genet ; 19(21): 4189-200, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20705735

RESUMEN

Thyroid hormone (TH) is crucial for normal brain development. TH transporters control TH homeostasis in brain as evidenced by the complex endocrine and neurological phenotype of patients with mutations in monocarboxylate transporter 8 (MCT8). We investigated the mechanisms of disease by analyzing gene expression profiles in fibroblasts from patients with MCT8 mutations. Studying MCT8 and its transcriptional context in different comprehensive spatial and temporal human brain transcriptome data sets revealed distinct region-specific MCT8 expression. Furthermore, MCT8 demonstrated a clear age-dependent decrease, suggesting its importance in early brain development. Performing comparative transcriptome analysis, we linked the genes differentially expressed (DE) in patient fibroblasts to the human brain transcriptome. DE genes in patient fibroblasts were strongly over-represented among genes highly correlated with MCT8 expression in brain. Furthermore, using the same approach we identified which genes in the classical TH signaling pathway are affected in patients. Finally, we provide evidence that the TRα2 receptor variant is closely connected to MCT8. The present study provides a molecular basis for understanding which pathways are likely affected in the brains of patients with mutations in MCT8. Our data regarding a functional relationship between MCT8 and TRα2 suggest an unanticipated role for TRα2 in the (patho)physiology of TH signaling in the brain. This study demonstrates how genome-wide expression data from patient-derived non-neuronal tissue related to the human brain transcriptome may be successfully employed to improve our understanding of neurological disease.


Asunto(s)
Encéfalo/metabolismo , Perfilación de la Expresión Génica , Transportadores de Ácidos Monocarboxílicos/genética , Mutación , Transcripción Genética , Humanos , Simportadores
16.
Clin Endocrinol (Oxf) ; 76(5): 706-12, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22117696

RESUMEN

OBJECTIVES: Growth hormone insensitivity syndrome (GHIS) is characterized by extreme short stature and resistance to the actions of growth hormone (GH). The heterogeneity ranges from the most severe form, known as Laron syndrome, to less severe phenotypes like idiopathic short stature and partial GH insensitivity. Here, we aimed to identify and characterize the molecular cause of severe short stature in a patient with resistance to GH treatment. PATIENT: We describe a male patient born small for gestational age [38 weeks gestation, length 38·5 cm; -7·8 standard deviation score (SDS), weight 1350 g; -4·84 SDS]. At the age of 7 years (109·7 cm; -2·89 SD), he received GH treatment (1 mg/m(2)/day) for 1 year without any increase in height SDS, IGF-I or IGFBP-3 levels. Double-GH-dose treatment for another year did not result in any improvement in growth factor level either. The patient does not have the typical Laron craniofacial and somatic features. RESULTS: Analysis of GHR showed a heterozygous nonsense mutation (c.703C>T; p.Arg217X). Extensive mutation screening as well as copy number variation analysis of other candidate genes in the GH-IGF-I axis excluded any additional genetic defects. Analysis of the patient's fibroblasts showed that growth hormone receptor (GHR) messenger ribonucleic acid (mRNA) expressed from the mutant allele was degraded by a mechanism called nonsense-mediated mRNA decay (NMD). CONCLUSIONS: GHIS in this patient is because of a heterozygous nonsense mutation in GHR. Our study is the first to demonstrate that NMD is involved in the phenotypic variability of GHIS caused by GHR mutations.


Asunto(s)
Codón sin Sentido , Predisposición Genética a la Enfermedad/genética , Síndrome de Laron/genética , Receptores de Somatotropina/genética , Adolescente , Secuencia de Bases , Estatura/efectos de los fármacos , Estatura/genética , Niño , Análisis Mutacional de ADN , Fibroblastos/metabolismo , Estudios de Seguimiento , Heterocigoto , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Síndrome de Laron/tratamiento farmacológico , Síndrome de Laron/metabolismo , Masculino , Degradación de ARNm Mediada por Codón sin Sentido , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
Clin Endocrinol (Oxf) ; 76(4): 577-81, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22017394

RESUMEN

OBJECTIVE: The effects of long-term TSH-suppressive levothyroxine (LT4) therapy on thyroid hormone metabolism in patients with differentiated thyroid carcinoma (DTC) are unknown. The aim of the study was to investigate the changes in thyroid hormone metabolism after long-term TSH-suppressive LT4 therapy in patients with DTC. PATIENTS AND METHODS: Sixty one patients with DTC were followed. For each patient, frozen remnant sera from two time points were selected: time1 (drawn within 1 year of I-131 ablation; TSH on file <0·3 mIU/l; recruitment period 1999-2002) and time2 (last available sample with TSH on file <0·3 mIU/l; minimum of 3 years of continuous TSH-suppressive LT4-therapy on record). TSH, reverse triiodothyronine (rT3), total triiodothyronine (TT3) and total thyroxine (TT4) levels were measured at both time1 and time2, and relationships between these parameters were analysed. RESULTS: Total triiodothyronine, TT4 and TSH levels were significantly reduced at time2 (P < 0·001), whereas LT4 dose, bodyweight and rT3 levels remained constant between time1 and time2. There were no significant changes in the relationship between the dose of LT4/kg bodyweight and TT4 levels (P = 0·14). TT4/TT3 was increased at time2 (P < 0·001), whereas TT4/rT3 and TT3/rT3 were significantly decreased at time2. There appeared to be no relationship of the effects found and advancing age. CONCLUSION: After long-term TSH-suppressive LT4 therapy for DTC, there are significant changes in thyroid hormone metabolism, which are best explained by a combined downregulation of deiodinases subtypes 1 and 2 and an upregulation of deiodinase subtype 3.


Asunto(s)
Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Tiroxina/uso terapéutico , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Tiroides/tratamiento farmacológico , Tirotropina/sangre , Tiroxina/sangre , Triyodotironina/sangre , Adulto Joven
18.
J Nutr ; 142(12): 2167-74, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23077186

RESUMEN

The rare but deleterious effects of severe iodine deficiency during pregnancy on cognitive functioning of children are well known. Reports on possible associations between mild-to-moderate maternal iodine deficiency and child development, however, are scarce. In a population-based cohort we examined the association between maternal urinary iodine during early pregnancy and executive functioning in children at 4 y of age. In addition, we investigated the modification of this association by maternal diet and thyroid function. During pregnancy, we measured urinary iodine and thyroid hormone concentrations in 1156 women. In 692 of their children impairment of executive functioning was assessed by the Behavior Rating Inventory of Executive Function. Five hundred mothers of Dutch national origin completed an FFQ. Analyses were performed by using regression models. The children of mothers with low urinary iodine showed higher scores on the problem scales of inhibition [ß = 0.05 (95% CI: 0.01, 0.10), P = 0.03] and working memory [ß = 0.07 (95% CI: 0.02, 0.12), P = 0.003]. Although maternal dietary intake and thyroid hormone concentration did not significantly modify these associations, the associations between urinary iodine and problems of inhibition were attenuated after adjustment for maternal psychological symptoms. In addition, the consumption of bread [ß = 0.61 (95% CI: 0.27, 0.95), P < 0.001] and eggs (ß = 1.87 (95% CI: 0.13, 3.62), P = 0.04] was associated with higher urinary iodine. Thus, low maternal urinary iodine during pregnancy is associated with impaired executive functioning in children. Because these symptoms were subclinical and occurred at an early age, future studies are needed to show whether these children are more vulnerable to develop later clinical disorders.


Asunto(s)
Función Ejecutiva , Yodo/orina , Embarazo/orina , Adulto , Preescolar , Femenino , Humanos , Recién Nacido , Yodo/administración & dosificación , Yodo/deficiencia , Masculino
19.
J Clin Invest ; 118(3): 975-83, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18259611

RESUMEN

Thyroid hormone is a critical determinant of cellular metabolism and differentiation. Precise tissue-specific regulation of the active ligand 3,5,3'-triiodothyronine (T3) is achieved by the sequential removal of iodine groups from the thyroid hormone molecule, with type 3 deiodinase (D3) comprising the major inactivating pathway that terminates the action of T3 and prevents activation of the prohormone thyroxine. Using cells endogenously expressing D3, we found that hypoxia induced expression of the D3 gene DIO3 by a hypoxia-inducible factor-dependent (HIF-dependent) pathway. D3 activity and mRNA were increased both by hypoxia and by hypoxia mimetics that increase HIF-1. Using ChIP, we found that HIF-1alpha interacted specifically with the DIO3 promoter, indicating that DIO3 may be a direct transcriptional target of HIF-1. Endogenous D3 activity decreased T3-dependent oxygen consumption in both neuronal and hepatocyte cell lines, suggesting that hypoxia-induced D3 may reduce metabolic rate in hypoxic tissues. Using a rat model of cardiac failure due to RV hypertrophy, we found that HIF-1alpha and D3 proteins were induced specifically in the hypertrophic myocardium of the RV, creating an anatomically specific reduction in local T3 content and action. These results suggest a mechanism of metabolic regulation during hypoxic-ischemic injury in which HIF-1 reduces local thyroid hormone signaling through induction of D3.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Hipoxia/metabolismo , Yoduro Peroxidasa/fisiología , Isquemia/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Inducción Enzimática , Hipertrofia Ventricular Derecha/metabolismo , Masculino , Ratas , Ratas Wistar , Transducción de Señal , Triyodotironina/fisiología
20.
N Engl J Med ; 358(17): 1811-8, 2008 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-18434651

RESUMEN

DEHAL1 has been identified as the gene encoding iodotyrosine deiodinase in the thyroid, where it controls the reuse of iodide for thyroid hormone synthesis. We screened patients with hypothyroidism who had features suggestive of an iodotyrosine deiodinase defect for mutations in DEHAL1. Two missense mutations and a deletion of three base pairs were identified in four patients from three unrelated families; all the patients had a dramatic reduction of in vitro activity of iodotyrosine deiodinase. Patients had severe goitrous hypothyroidism, which was evident in infancy and childhood. Two patients had cognitive deficits due to late diagnosis and treatment. Thus, mutations in DEHAL1 led to a deficiency in iodotyrosine deiodinase in these patients. Because infants with DEHAL1 defects may have normal thyroid function at birth, they may be missed by neonatal screening programs for congenital hypothyroidism.


Asunto(s)
Bocio/genética , Hipotiroidismo/genética , Yoduro Peroxidasa/genética , Mutación Missense , Eliminación de Secuencia , Adulto , Secuencia de Aminoácidos , Niño , Análisis Mutacional de ADN , Femenino , Mutación del Sistema de Lectura , Bocio/enzimología , Homocigoto , Humanos , Hipotiroidismo/tratamiento farmacológico , Hipotiroidismo/enzimología , Yoduro Peroxidasa/deficiencia , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Monoyodotirosina/metabolismo , Sistemas de Lectura Abierta , Fenotipo , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA