Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Hum Mol Genet ; 26(3): 599-610, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28025328

RESUMO

Congenital hypothyroidism is the most common neonatal endocrine disorder and is primarily caused by developmental abnormalities otherwise known as thyroid dysgenesis (TD). We performed whole exome sequencing (WES) in a consanguineous family with TD and subsequently sequenced a cohort of 134 probands with TD to identify genetic factors predisposing to the disease. We identified the novel missense mutations p.S148F, p.R114Q and p.L177W in the BOREALIN gene in TD-affected families. Borealin is a major component of the Chromosomal Passenger Complex (CPC) with well-known functions in mitosis. Further analysis of the missense mutations showed no apparent effects on mitosis. In contrast, expression of the mutants in human thyrocytes resulted in defects in adhesion and migration with corresponding changes in gene expression suggesting others functions for this mitotic protein. These results were well correlated with the same gene expression pattern analysed in the thyroid tissue of the patient with BOREALIN-p.R114W. These studies open new avenues in the genetics of TD in humans.


Assuntos
Proteínas de Ciclo Celular/genética , Predisposição Genética para Doença , Mutação de Sentido Incorreto/genética , Disgenesia da Tireoide/genética , Proteínas de Ciclo Celular/biossíntese , Movimento Celular/genética , Exoma/genética , Feminino , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mitose/genética , Linhagem , Disgenesia da Tireoide/patologia
2.
J Med Genet ; 49(6): 366-72, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22636604

RESUMO

BACKGROUND: Deletions including chromosome 14 band q13 have been linked to variable phenotypes. With current molecular methods the authors aim to elucidate a genotype-phenotype correlation by accurately determining the size and location of the deletions and the associated phenotype. METHODS: Here the authors report the molecular karyotyping and phenotypic description of seven patients with overlapping deletions including chromosome 14q13. RESULTS: The authors show that deletions including 14q13 result in a recognisable phenotype mainly due to haploinsufficiency of two genes (NKX2-1, PAX9). FOXG1 (on chromosome band 14q12) involvement seems to be the main determinant of phenotype severity. The patients in this study without FOXG1 involvement and deletions of up to 10 Mb have a relatively mild phenotype. The authors cannot explain why some patients in literature with overlapping but smaller deletions appear to have a more severe phenotype. A previously presumed association with holoprosencephaly could not be confirmed as none of the patients in this series had holoprosencephaly. CONCLUSIONS: FOXG1 appears the main determinant of the severity of phenotypes resulting from deletions including 14q13. The collected data show no evidence for a locus for holoprosencephaly in the 14q13 region, but a locus for agenesis of the corpus callosum cannot be excluded.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 14 , Fatores de Transcrição Forkhead/genética , Holoprosencefalia/genética , Proteínas do Tecido Nervoso/genética , Adulto , Criança , Mapeamento Cromossômico , Feminino , Haploinsuficiência , Humanos , Recém-Nascido , Cariotipagem , Masculino , Proteínas Nucleares/genética , Fator de Transcrição PAX9/genética , Fenótipo , Análise de Sequência de DNA , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética
3.
Horm Res Paediatr ; 96(4): 426-431, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36513041

RESUMO

INTRODUCTION: Sensenbrenner syndrome, or cranioectodermal dysplasia (OMIM #218330), is a rare genetic condition inherited as an autosomal recessive with less than 70 reported cases worldwide. It results in multiorgan abnormalities along with ectodermal structural defects. No previous reported cases demonstrated primary hypothyroidism in a matter of Sensenbrenner syndrome. CASE PRESENTATION: Herein, we report a 6-year-old girl who suffered from progressive liver failure and end-stage renal disease secondary to Sensenbrenner syndrome, which was associated with severe primary hypothyroidism that completely recovered after a combined renal and liver transplant. CONCLUSION: For the first time in the literature, we report an association of Sensenbrenner syndrome with hypothyroidism that resolved after a combined renal and liver transplant. Such findings expand the clinical spectrum of this syndrome. However, a larger cohort is needed to confirm or exclude such an association. Our case highlights the importance of thyroid function monitoring in any patient with renal and liver failure prior to and after a hepatorenal transplant.


Assuntos
Craniossinostoses , Displasia Ectodérmica , Falência Hepática , Feminino , Humanos , Criança , Displasia Ectodérmica/complicações , Displasia Ectodérmica/genética , Craniossinostoses/complicações , Craniossinostoses/genética , Osso e Ossos , Falência Hepática/complicações
4.
Front Endocrinol (Lausanne) ; 14: 1286747, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37964961

RESUMO

Background: BOREALIN/CDCA8 mutations are associated with congenital hypothyroidism and thyroid dysgenesis. Borealin is involved in mitosis as part of the Chromosomal Passenger Complex. Although BOREALIN mutations decrease thyrocyte adhesion and migration, little is known about the specific role of Borealin in the thyroid. Methods: We characterized thyroid development and function in Borealin-deficient (Borealin +/-) mice using histology, transcriptomic analysis, and quantitative PCR. Results: Thyroid development was impaired with a hyperplastic anlage on embryonic day E9.5 followed by thyroid hypoplasia from E11.5 onward. Adult Borealin +/- mice exhibited euthyroid goiter and defect in thyroid hormone synthesis. Borealin +/- aged mice had disorganized follicles and papillary-like structures in thyroids due to ERK pathway activation and a strong increase of Braf-like genes described by The Cancer Genome Atlas (TCGA) network of papillary thyroid carcinoma. Moreover, Borealin +/- thyroids exhibited structural and transcriptomic similarities with papillary thyroid carcinoma tissue from a human patient harboring a BOREALIN mutation, suggesting a role in thyroid tumor susceptibility. Conclusion: These findings demonstrate Borealin involvement in critical steps of thyroid structural development and function throughout life. They support a role for Borealin in thyroid dysgenesis with congenital hypothyroidism. Close monitoring for thyroid cancer seems warranted in patients carrying BOREALIN mutations.


Assuntos
Hipotireoidismo Congênito , Disgenesia da Tireoide , Neoplasias da Glândula Tireoide , Animais , Camundongos , Proteínas de Ciclo Celular/genética , Hipotireoidismo Congênito/genética , Câncer Papilífero da Tireoide/genética , Disgenesia da Tireoide/genética , Neoplasias da Glândula Tireoide/genética
5.
Pediatr Investig ; 6(2): 123-134, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35774517

RESUMO

Congenital hypothyroidism (CH) is the most common neonatal endocrine disorder and one of the most common preventable causes of intellectual disability in the world. CH may be due to developmental or functional thyroid defects (primary or peripheral CH) or be hypothalamic-pituitary in origin (central CH). In most cases, primary CH is caused by a developmental malformation of the gland (thyroid dysgenesis, TD) or by a defect in thyroid hormones synthesis (dyshormonogenesis, DH). TD represents about 65% of CH and a genetic cause is currently identified in fewer than 5% of patients. The remaining 35% are cases of DH and are explained with certainty at the molecular level in more than 50% of cases. The etiology of CH is mostly unknown and may include contributions from individual and environmental factors. In recent years, the detailed phenotypic description of patients, high-throughput sequencing technologies, and the use of animal models have made it possible to discover new genes involved in the development or function of the thyroid gland. This paper reviews all the genetic causes of CH. The modes by which CH is transmitted will also be discussed, including a new oligogenic model. CH is no longer simply a dominant disease for cases of CH due to TD and recessive for cases of CH due to DH, but a far more complex disorder.

6.
Med Sci (Paris) ; 38(3): 263-273, 2022 Mar.
Artigo em Francês | MEDLINE | ID: mdl-35333163

RESUMO

Congenital hypothyroidism (CH) is the most frequent neonatal endocrine disorder. CH is due to thyroid development or thyroid function defects (primary) or may be of hypothalamic-pituitary origin (central). Primary CH is caused essentially by abnormal thyroid gland morphogenesis (thyroid dysgenesis, TD) or defective thyroid hormone synthesis (dyshormonogenesis, DH). DH accounts for about 35% of CH and a genetic cause is identified in 50% of patients. However, TD accounts for about 65% of CH, and a genetic cause is identified in less than 5% of patients. The pathogenesis of CH is largely unknown and may include the contribution of individual and environmental factors. During the last years, detailed phenotypic description of patients, next-generation sequence technologies and use of animal models allowed the discovery of novel candidate genes in thyroid development and function. We provide an overview of recent genetic causes of primary and central CH. In addition, mode of inheritance and the oligogenic model of CH are discussed.


Title: Génétique de l'hypothyroïdie congénitale. Abstract: L'hypothyroïdie congénitale (HC) est la maladie endocrinienne néonatale la plus fréquente. Elle peut être due à des défauts de développement ou de la fonction de la thyroïde (HC primaire ou périphérique) ou d'origine hypothalamo-hypophysaire (HC centrale). L'HC primaire est causée dans la majorité des cas par une anomalie du développement de la glande (dysgénésie thyroïdienne, DT) ou par un défaut de synthèse des hormones thyroïdiennes (dyshormonogenèse, DH). Une origine génétique est identifiée chez 50 % des patients présentant une HCDH mais dans moins de 5 % des patients présentant une HCDT. Cette revue fait le point sur l'ensemble des causes génétiques des HC et sur les différents modes de transmission. L'HC n'est plus simplement une maladie dominante pour les dysgénésies thyroïdiennes et récessive pour les dyshormonogenèses, mais est devenue une maladie plus complexe.


Assuntos
Hipotireoidismo Congênito , Disgenesia da Tireoide , Hipotireoidismo Congênito/genética , Bases de Dados Genéticas , Humanos , Mutação , Disgenesia da Tireoide/genética , Hormônios Tireóideos
7.
J Clin Endocrinol Metab ; 107(12): 3418-3427, 2022 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-36107810

RESUMO

Congenital hypothyroidism (CH) is the most frequent neonatal endocrine disorder and the most common preventable cause of development delay and growth failure if diagnosed and treated early. The thyroid is the first endocrine gland to develop during embryonic life and to be recognizable in humans. Thyroid development and maturation can be divided into 2 phases: a first phase of embryogenesis and a second phase of folliculogenesis and differentiation with thyroid hormone production at the final steps. Regulation of the thyroid function requires normal development of the hypothalamic-pituitary-thyroid axis, which occurs during the embryonic and neonatal period. Defects in any of steps of thyroid development, differentiation, and regulation lead to permanent CH. Newborn screening programs, established in only one-third of countries worldwide, detect CH and are cost-effective and highly sensitive and specific. During the last decade, epidemiology of CH has changed with increased frequency of thyroid in situ in primary CH. Advances in molecular testing have expanded knowledge and understanding of thyroid development and function. However, a molecular cause is identified in only 5% of CH due to thyroid dysgenesis. The purpose of this article is to describe the clinical approach to the child with CH, focusing on diagnostic work-up and future challenges on optimizing thyroid replacement therapy and regenerative medicine. The review is written from the perspective of the case of 2 girls referred for CH after newborn screening and diagnosed with thyroid ectopy. The genetic work-up revealed novel mutations in TUBB1 gene, associated with large platelets and abnormal platelet physiology.


Assuntos
Hipotireoidismo Congênito , Disgenesia da Tireoide , Criança , Feminino , Humanos , Recém-Nascido , Hipotireoidismo Congênito/diagnóstico , Hipotireoidismo Congênito/genética , Hipotireoidismo Congênito/terapia , Triagem Neonatal , Disgenesia da Tireoide/complicações , Hormônios Tireóideos
8.
Hum Mol Genet ; 18(12): 2266-76, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19336474

RESUMO

Thyroid transcription factor 1 (NKX2-1/TITF1) mutations cause brain-lung-thyroid syndrome, characterized by congenital hypothyroidism (CH), infant respiratory distress syndrome (IRDS) and benign hereditary chorea (BHC). The objectives of the present study were (i) detection of NKX2-1 mutations in patients with CH associated with pneumopathy and/or BHC, (ii) functional analysis of new mutations in vitro and (iii) description of the phenotypic spectrum of brain-lung-thyroid syndrome. We identified three new heterozygous missense mutations (L176V, P202L, Q210P), a splice site mutation (376-2A-->G), and one deletion of NKX2-1 at 14q13. Functional analysis of the three missense mutations revealed loss of transactivation capacity on the human thyroglobulin enhancer/promoter. Interestingly, we showed that deficient transcriptional activity of NKX2-1-P202L was completely rescued by cotransfected PAX8-WT, whereas the synergistic effect was abolished by L176V and Q210P. The clinical spectrum of 6 own and 40 published patients with NKX2-1 mutations ranged from the complete triad of brain-lung-thyroid syndrome (50%), brain and thyroid disease (30%), to isolated BHC (13%). Thyroid morphology was normal (55%) and compensated hypothyroidism occurred in 61%. Lung disease occurred in 54% of patients (IRDS at term 76%; recurrent pulmonary infections 24%). On follow-up, 20% developed severe chronic interstitial lung disease, and 16% died. In conclusion, we describe five new NKX2.1 mutations with, for the first time, complete rescue by PAX8 of the deficient transactivating capacity in one case. Additionally, our review shows that the majority of affected patients display neurological and/or thyroidal problems and that, although less frequent, lung disease is responsible for a considerable mortality.


Assuntos
Proteínas Nucleares/genética , Fatores de Transcrição Box Pareados/metabolismo , Fatores de Transcrição/genética , Sequência de Bases , Encéfalo/metabolismo , Linhagem Celular , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Lactente , Pulmão/metabolismo , Masculino , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/metabolismo , Fator de Transcrição PAX8 , Fatores de Transcrição Box Pareados/genética , Glândula Tireoide/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/metabolismo , Ativação Transcricional
9.
Endocrine ; 71(3): 696-705, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33650047

RESUMO

INTRODUCTION: Congenital hypothyroidism (CH) is the most frequent neonatal endocrine disorder and one of the most common preventable forms of mental retardation worldwide. CH is due to thyroid development or thyroid function defects (primary) or may be of hypothalamic-pituitary origin (central). Primary CH is caused essentially by abnormal thyroid gland morphogenesis (thyroid dysgenesis, TD) or defective thyroid hormone synthesis (dyshormonogenesis, DH). TD accounts for about 65% of CH, however a genetic cause is identified in less than 5% of patients. PURPOSE: The pathogenesis of CH is largely unknown and may include the contribution of individual and environmental factors. During the last years, detailed phenotypic description of patients, next-generation sequence technologies and use of animal models allowed the discovery of novel candidate genes in thyroid development, function and pathways. RESULTS AND CONCLUSION: We provide an overview of recent genetic causes of primary and central CH. In addition, mode of inheritance and the oligogenic model of CH are discussed.


Assuntos
Hipotireoidismo Congênito , Disgenesia da Tireoide , Hipotireoidismo Congênito/genética , Bases de Dados Genéticas , Humanos , Recém-Nascido , Mutação , Disgenesia da Tireoide/genética , Hormônios Tireóideos
10.
Hum Mutat ; 31(2): E1146-62, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20020530

RESUMO

NKX2-1 (NK2 homeobox 1) is a critical regulator of transcription for the surfactant protein (SP)-B and -C genes (SFTPB and SFTPC, respectively). We identified and functionally characterized two new de novo NKX2-1 mutations c.493C>T (p.R165W) and c.786_787del2 (p.L263fs) in infants with closely similar severe interstitial lung disease (ILD), hypotonia, and congenital hypothyroidism. Functional analyses using A549 and HeLa cells revealed that NKX2-1-p.L263fs induced neither SFTPB nor SFTPC promoter activation and had a dominant negative effect on wild-type (WT) NKX2-1. In contrast,NKX2-1-p.R165W activated SFTPC, to a significantly greater extent than did WTNKX2-1, while SFTPB activation was only significantly reduced in HeLa cells. In accordance with our in vitro data, we found decreased amounts of SP-B and SP-C by western blot in bronchoalveolar lavage fluid (patient with p.L263fs) and features of altered surfactant protein metabolism on lung histology (patient with NKX2-1-p.R165W). In conclusion, ILD in patients with NKX2-1 mutations was associated with altered surfactant protein metabolism, and both gain and loss of function of the mutated NKX2-1 genes on surfactant protein promoters were associated with ILD in "Brain-Lung-Thyroid syndrome".


Assuntos
Anormalidades Múltiplas/genética , Regulação da Expressão Gênica , Doenças Pulmonares Intersticiais/genética , Mutação/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Proteínas Associadas a Surfactantes Pulmonares/genética , Fatores de Transcrição/genética , Anormalidades Múltiplas/diagnóstico por imagem , Anormalidades Múltiplas/patologia , Sequência de Aminoácidos , Sequência de Bases , Líquido da Lavagem Broncoalveolar , Linhagem Celular Tumoral , Criança , Pré-Escolar , DNA , Evolução Fatal , Feminino , Humanos , Lactente , Recém-Nascido , Doenças Pulmonares Intersticiais/complicações , Doenças Pulmonares Intersticiais/diagnóstico por imagem , Doenças Pulmonares Intersticiais/patologia , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Especificidade de Órgãos/genética , Gravidez , Ligação Proteica , Radiografia , Síndrome , Glândula Tireoide/patologia , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
11.
Mol Cell Endocrinol ; 510: 110834, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32360566

RESUMO

Terminal thyroid gland differentiation - the last developmental step needed to enable thyroid hormone (T4) synthesis - involves profound structural and biochemical changes in the thyroid follicular cells (TFCs). We aimed to develop an ex vivo thyroid model of embryonic mouse thyroid that would replicate the in vivo TFC differentiation program. E13.5 thyroid explants were cultured ex vivo in chemically defined medium for 7 days. Immunostaining and qPCR of thyroid explants showed thyroglobulin production onset, follicle formation, and T4 synthesis onset in 1-, 3-, and 5-day-old cultures, respectively. Differentiation was maintained and follicular growth continued throughout the 7-day culture period. Pharmacological approaches to culture inhibition were performed successfully in the ex vivo thyroids. Our robust and well described ex vivo thyroid culture model replicates the sequence of thyroid differentiation to T4 synthesis seen in vivo. This model can be used to test the effects of pharmacological inhibitors on thyroid hormone production.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/anatomia & histologia , Modelos Biológicos , Glândula Tireoide/anatomia & histologia , Glândula Tireoide/fisiologia , Animais , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Microdissecção , Receptores Notch/metabolismo , Tireoglobulina/metabolismo , Glândula Tireoide/embriologia , Sobrevivência de Tecidos/fisiologia
12.
Eur J Endocrinol ; 183(5): K1-K5, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32805706

RESUMO

BACKGROUND: Among patients with congenital hypothyroidism, 35% have dyshormonogenesis (DH) with thyroid gland in situ with or without goiter. The majority of DH cases are due to mutations in genes involved in thyroid hormone production as TG, TPO, SLC5A5/NIS, SLC26A4/PDS, IYD/DEHAL1, DUOX2, and DUOXA2, and are usually inherited on an autosomal recessive basis. Most previously reported cases of fetal hypothyroidism and goiter were related to TG or TPO mutations and recently DUOXA2. PATIENT: In a male patient with antenatal goiter treated with intraamniotic levothyroxine injections, whose long-term follow-up is described in detail, two novel NIS mutations were detected. Mutations of NIS were located in exon 1 (c.52G>A, p.G18R) and exon 13 (c.1546C>T, p.R516X), each mutation was inherited from parents, who are healthy carriers. The p.G18R mutation affecting the first transmembrane domain of the protein can be responsible for deficient iodide uptake. However, the second is a nonsense mutation leading probably to mRNA degradation. In addition, the patient has undergone a thyroidectomy and we have studied the thyroid tissue. The thyroid histology showed heterogeneity with large follicles, epithelial hyperplasia and many areas of fibrosis. Immunohistochemistry with NIS specific antibody showed NIS staining at the basolateral plasma membrane of the thyrocytes. CONCLUSIONS: We report the first case of fetal goitrous hypothyroidism due to two novel NIS mutations with access to thyroid tissue of the patient, specific histology studies and long-term follow-up. This case expands our knowledge and provides further insights on molecular causes of fetal goiter in humans.


Assuntos
Hipotireoidismo Congênito/genética , Bócio/genética , Mutação , Simportadores/genética , Adolescente , Criança , Pré-Escolar , Hipotireoidismo Congênito/tratamento farmacológico , Bócio/tratamento farmacológico , Humanos , Lactente , Recém-Nascido , Masculino , Diagnóstico Pré-Natal , Tiroxina/uso terapêutico , Resultado do Tratamento , Adulto Jovem
13.
Front Endocrinol (Lausanne) ; 11: 545339, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33692749

RESUMO

Objective: To elucidate the molecular cause in a well-characterized cohort of patients with Congenital Hypothyroidism (CH) and Dyshormonogenesis (DH) by using targeted next-generation sequencing (TNGS). Study design: We studied 19 well-characterized patients diagnosed with CH and DH by targeted NGS including genes involved in thyroid hormone production. The pathogenicity of novel mutations was assessed based on in silico prediction tool results, functional studies when possible, variant location in important protein domains, and a review of the recent literature. Results: TNGS with variant prioritization and detailed assessment identified likely disease-causing mutations in 10 patients (53%). Monogenic defects most often involved TG, followed by DUOXA2, DUOX2, and NIS and were usually homozygous or compound heterozygous. Our review shows the importance of the detailed phenotypic description of patients and accurate analysis of variants to provide a molecular diagnosis. Conclusions: In a clinically well-characterized cohort, TNGS had a diagnostic yield of 53%, in accordance with previous studies using a similar strategy. TG mutations were the most common genetic defect. TNGS identified gene mutations causing DH, thereby providing a rapid and cost-effective genetic diagnosis in patients with CH due to DH.


Assuntos
Hipotireoidismo Congênito/diagnóstico , Hipotireoidismo Congênito/genética , Adolescente , Adulto , Criança , Pré-Escolar , Hipotireoidismo Congênito/fisiopatologia , Oxidases Duais/genética , Feminino , Predisposição Genética para Doença , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Mutação , Linhagem , Simportadores/genética , Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo , Adulto Jovem
14.
Thyroid ; 28(7): 941-944, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29790453

RESUMO

BACKGROUND: Primary congenital hypothyroidism (CH) affects about 1:3000 newborns worldwide and is mainly caused by defects in thyroid gland development (thyroid dysgenesis [TD]) or hormone synthesis. A genetic cause is identified in <10% of TD patients. The aim was to identify novel candidate genes in patients with TD using next-generation sequencing tools. PATIENT FINDINGS: Whole exome sequencing was used to study two families: a consanguineous Tunisian family (one child with severe thyroid hypoplasia) and a French family (two newborn siblings, with a thyroid in situ that was not enlarged on ultrasound at diagnosis). Variants in candidate genes were filtered according to type of variation, frequency in public and in-house databases, in silico prediction tools, and inheritance mode. Unexpectedly, three different variants of the thyroid peroxidase (TPO) gene were identified. A homozygous missense mutation (c.875C>T, p.S292F) was found in the Tunisian patient with severe thyroid hypoplasia. The two French siblings were compound heterozygotes (c.387delC/c.2578G>A, p.N129Kfs*80/p.G860R) for TPO mutations. All three mutations have been previously described in patients with goitrous CH. In these patients, treatment was initiated immediately after diagnosis, and the effect, if any, of thyrotropin stimulation of these thyroids remains unclear. CONCLUSIONS: The first cases are reported of thyroid hypoplasia at diagnosis during the neonatal period in patients with CH and TPO mutations. These cases highlight the importance of screening for TPO mutations not only in goitrous CH, but also in normal or small-size thyroids, and they broaden the clinical spectrum of described phenotypes.


Assuntos
Hipotireoidismo Congênito/genética , Iodeto Peroxidase/genética , Disgenesia da Tireoide/genética , Adolescente , Adulto , Alelos , Criança , Feminino , Frequência do Gene , Humanos , Masculino , Mutação , Testes de Função Tireóidea , Sequenciamento do Exoma
15.
EMBO Mol Med ; 10(12)2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30446499

RESUMO

The genetic causes of congenital hypothyroidism due to thyroid dysgenesis (TD) remain largely unknown. We identified three novel TUBB1 gene mutations that co-segregated with TD in three distinct families leading to 1.1% of TUBB1 mutations in TD study cohort. TUBB1 (Tubulin, Beta 1 Class VI) encodes for a member of the ß-tubulin protein family. TUBB1 gene is expressed in the developing and adult thyroid in humans and mice. All three TUBB1 mutations lead to non-functional α/ß-tubulin dimers that cannot be incorporated into microtubules. In mice, Tubb1 knock-out disrupted microtubule integrity by preventing ß1-tubulin incorporation and impaired thyroid migration and thyroid hormone secretion. In addition, TUBB1 mutations caused the formation of macroplatelets and hyperaggregation of human platelets after stimulation by low doses of agonists. Our data highlight unexpected roles for ß1-tubulin in thyroid development and in platelet physiology. Finally, these findings expand the spectrum of the rare paediatric diseases related to mutations in tubulin-coding genes and provide new insights into the genetic background and mechanisms involved in congenital hypothyroidism and thyroid dysgenesis.


Assuntos
Plaquetas/citologia , Plaquetas/patologia , Mutação , Agregação Plaquetária , Disgenesia da Tireoide/genética , Tubulina (Proteína)/genética , Animais , Humanos , Camundongos , Camundongos Knockout , Disgenesia da Tireoide/patologia
16.
J Clin Endocrinol Metab ; 92(1): 70-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17077129

RESUMO

CONTEXT: Terminal differentiation of the human thyroid is characterized by the onset of follicle formation and thyroid hormone synthesis at 11 gestational weeks (GW). OBJECTIVE: This study aimed to investigate the ontogeny of thyroglobulin (Tg), thyroid peroxidase (TPO), sodium/iodide symporter (NIS), pendrin (PDS), dual oxidase 2 (DUOX2), thyroid-stimulating hormone receptor (TSHR), and thyroid transcription factor 1 (TITF1), forkhead box E1 (FOXE1), and paired box gene 8 (PAX8) in the developing human thyroid. DESIGN: Thyroid tissues from human embryos and fetuses (7-33 GW; n = 45) were analyzed by quantitative PCR to monitor mRNA expression for each gene and by immunohistochemistry to determine the cellular distribution of TITF1, TSHR, Tg, TPO, NIS, and the onset of T4 production. A broken line regression model was fitted for each gene to compare the loglinear increase in expression before and after the onset of T4 synthesis. RESULTS: TITF1, FOXE1, PAX8, TSHR, and DUOX2 were stably expressed from 7 to 33 GW. Tg, TPO, and PDS expression was detectable as early as 7 GW and was correlated with gestational age (all, P < 0.01), and the slope of the regression line was significantly different before and after the onset of T4 synthesis at 11 GW (all, P < 0.01). NIS expression appeared last and showed the highest fit by the broken line regression model of all genes (correlation age P < 0.0001, broken line regression P < 0.0001). Immunohistochemical studies detected TITF1, TSHR, and Tg in unpolarized thyrocytes before follicle formation. T(4) and NIS labeling were only found in developing follicles from 11 GW on. CONCLUSION: These results imply a key role of NIS for the onset of human thyroid function.


Assuntos
Feto/metabolismo , Simportadores/genética , Glândula Tireoide/embriologia , Feminino , Idade Gestacional , Humanos , Imuno-Histoquímica , Iodeto Peroxidase/análise , Iodeto Peroxidase/genética , Gravidez , RNA Mensageiro/análise , Receptores da Tireotropina/análise , Simportadores/análise , Tireoglobulina/análise , Tireoglobulina/genética
17.
Hum Genet ; 122(5): 467-76, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17717707

RESUMO

Familial cases of congenital hypothyroidism from thyroid dysgenesis (TD) (OMIM 218700) occur with a frequency 15-fold higher than by chance, FOXE1 is one of the candidate genes for this genetic predisposition and contains an alanine tract. Our purpose is to assess the influence of length of the alanine tract of FOXE1 on genetic susceptibility to TD. A case-control association study (based on 115 patients affected by TD and 129 controls genotyped by direct sequencing) and transmission disequilibrium testing (TDT) analyses were performed. The transcriptional activities of FOXE1 constructs containing 14 or 16 alanines were also studied. In the case-control association study, the 16/16 and 16/14 genotypes were inversely associated with TD (OR = 0.39, 95%CI = 0.22-0.68, P = 0.0005), strongly suggesting that the presence of 16 alanines in the tract protect against the occurrence of TD. This association was stronger in the subgroup of patients with ectopic thyroid (OR = 0.28, 95%CI = 0.13-0.58, P = 0.00015). The protection was confirmed by the TDT analysis performed in 39 trios (chi(2) = 4.3, P = 0.0374). Alternatively, the presence of the 14/14 genotype is associated with an increase risk of TD (OR = 2.59, 95%CI = 1.56-4.62, P = 0.0005). The expression studies showed that the transcriptional activities of FOXE1 with 16 alanines were significantly higher (1.55-fold) than FOXE1 containing 14 alanines (P < 0.003), while the nuclear localisation of the proteins was not affected. We conclude that FOXE1 through its alanine containing stretch modulates significantly the risk of TD occurrence, enhancing a mechanism linking an alanine containing transcription factor to disease.


Assuntos
Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/genética , Disgenesia da Tireoide/genética , Sequência de Bases , Estudos de Casos e Controles , Linhagem Celular , Hipotireoidismo Congênito/genética , Primers do DNA/genética , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Razão de Chances , Peptídeos/química , Peptídeos/genética , Polimorfismo Genético , Transfecção
18.
Antioxid Redox Signal ; 26(15): 864-877, 2017 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-27401113

RESUMO

AIMS: The BRAFV600E oncogene, reported in 40%-60% of papillary thyroid cancer (PTC), has an important role in the pathogenesis of PTC. It is associated with the loss of thyroid iodide-metabolizing genes, such as sodium/iodide symporter (NIS), and therefore with radioiodine refractoriness. Inhibition of mitogen-activated protein kinase (MAPK) pathway, constitutively activated by BRAFV600E, is not always efficient in resistant tumors suggesting that other compensatory mechanisms contribute to a BRAFV600E adaptive resistance. Recent studies pointed to a key role of transforming growth factor ß (TGF-ß) in BRAFV600E-induced effects. The reactive oxygen species (ROS)-generating NADPH oxidase NOX4, which is increased in PTC, has been identified as a new key effector of TGF-ß in cancer, suggestive of a potential role in BRAFV600E-induced thyroid tumors. RESULTS: Here, using two human BRAFV600E-mutated thyroid cell lines and a rat thyroid cell line expressing BRAFV600E in a conditional manner, we show that NOX4 upregulation is controlled at the transcriptional level by the oncogene via the TGF-ß/Smad3 signaling pathway. Importantly, treatment of cells with NOX4-targeted siRNA downregulates BRAFV600E-induced NIS repression. Innovation and Conclusion: Our results establish a link between BRAFV600E and NOX4, which is confirmed by a comparative analysis of NOX4 expression in human (TCGA) and mouse thyroid cancers. Remarkably, analysis of human and murine BRAFV600E-mutated thyroid tumors highlights that the level of NOX4 expression is inversely correlated to thyroid differentiation suggesting that other genes involved in thyroid differentiation in addition to NIS might be silenced by a mechanism controlled by NOX4-derived ROS. This study opens a new opportunity to optimize thyroid cancer therapy. Antioxid. Redox Signal. 26, 864-877.


Assuntos
Carcinoma Papilar/metabolismo , Regulação para Baixo , NADPH Oxidase 4/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Simportadores/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Animais , Feminino , Humanos , Masculino , Camundongos , NADPH Oxidase 4/genética , Proteínas Proto-Oncogênicas B-raf/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas
19.
J Clin Endocrinol Metab ; 91(3): 933-40, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16403819

RESUMO

CONTEXT: Congenital hyperinsulinism (HI) is characterized by hypoglycemia related to inappropriate insulin secretion. Focal and diffuse forms of hyperinsulinism share a similar clinical presentation, but their treatment is dramatically different. Preoperative differential diagnosis was based on pancreatic venous sampling, a technically demanding technique. OBJECTIVE: Positron emission tomography (PET) after injection of [18F]fluoro-L-DOPA (L-dihydroxyphenylalanine) has been evaluated for the preoperative differentiation between focal and diffuse HI, by imaging uptake of radiotracer and the conversion of [18F]fluoro-L-dopa into dopamine by DOPA decarboxylase. We propose to validate this test by immunohistochemical approach. PATIENTS AND METHODS: Pancreatic surgical specimens of four focal and three diffuse HI were studied, using anti-DOPA decarboxylase and proinsulin antibodies. The effect of an inhibitor of DOPA decarboxylase (carbidopa) on insulin secretion was evaluated in vivo and in cultured INS-1 cells. RESULTS: Immunohistochemical detection of DOPA decarboxylase showed diffuse staining of Langerhans islets in the whole pancreas in all diffuse cases, in contrast with dense focal staining in all focal cases. Staining of Langerhans islets outside the focal lesion was diffusely but weakly positive. We correlated the localization of DOPA decarboxylase and proinsulin in normal pancreas and in both diffuse and focal HI tissues. The diffuse PET uptake found before treatment in one child with diffuse HI disappeared completely after carbidopa administration, suggesting in vivo that pancreatic cells can take up amine precursors and contain DOPA decarboxylase. The insulin secretion measured in the supernatant was the same whether INS-1 cells were treated by dopamine or Lodosyn or untreated. CONCLUSION: We validate PET with as a consistent test to differentiate diffuse and focal HI.


Assuntos
Di-Hidroxifenilalanina , Dopa Descarboxilase/metabolismo , Hiperinsulinismo/patologia , Insulina/metabolismo , Pâncreas/diagnóstico por imagem , Adolescente , Animais , Linhagem Celular , Criança , Pré-Escolar , Radioisótopos de Flúor , Humanos , Hiperinsulinismo/diagnóstico por imagem , Hiperinsulinismo/enzimologia , Hiperinsulinismo/cirurgia , Imuno-Histoquímica , Lactente , Secreção de Insulina , Ilhotas Pancreáticas/diagnóstico por imagem , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Pâncreas/enzimologia , Pâncreas/patologia , Tomografia por Emissão de Pósitrons , Ratos
20.
Endocrinol Metab Clin North Am ; 45(2): 243-54, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27241962

RESUMO

Thyroid dysgenesis (TD) is the most common cause of congenital hypothyroidism in iodine-sufficient regions and includes a spectrum of developmental anomalies. The genetic components of TD are complex. Although a sporadic disease, advances in developmental biology have revealed monogenetic forms of TD. Inheritance is not based on a simple Mendelian pattern and additional genetic elements might contribute to the phenotypic spectrum. This article summarizes the key steps of normal thyroid development and provides an update on responsible genes and underlying mechanisms of TD. Up-to-date technologies in genetics and biology will allow us to advance in our knowledge of TD.


Assuntos
Hipotireoidismo Congênito/genética , Genes Controladores do Desenvolvimento , Disgenesia da Tireoide/genética , Humanos , Iodo/deficiência , Glândula Tireoide/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA