Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
País/Região como assunto
Intervalo de ano de publicação
1.
Mol Biol Rep ; 50(3): 2351-2356, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36577834

RESUMO

BACKGROUND: Thyroid hormones play a vital function in the maturation in the course of mind development. Regarding the well-known effects of choline on brain-derived neurotrophic factor (BDNF), the study examined the effects of choline on hippocampal BDNF gene expression in maternal hypothyroidism rats. METHODS AND RESULTS: To induce the hypothyroidism, 6-propyl-2-thiouracil was introduced to the ingesting water from the sixth day of gestation to twenty-first postnatal day (PND). Choline-treatment started twice a day on the first day of gestation until PND 21. On PND28, pups were sacrificed. The expression of BDNF gene was evaluated after the hippocampus was harvested. Our results demonstrated that both male and female pre-pubertal offspring rats' BDNF gene expression was decreased by developmental hypothyroidism. Choline increases the ratio of relative gene expression of BDNF in the hippocampus of males and females in the control/hypothyroidism group, especially in males. CONCLUSIONS: It can be concluded that maternal choline supplementation on the first day of gestation until PND 21 improves brain development and cognitive function in pre-pubertal offspring rats regarding control/hypothyroidism groups.


Assuntos
Hipotireoidismo , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Masculino , Ratos , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Colina/farmacologia , Colina/metabolismo , Suplementos Nutricionais , Expressão Gênica , Hipocampo/metabolismo , Hipotireoidismo/tratamento farmacológico , Hipotireoidismo/genética , Hipotireoidismo/metabolismo , Mães , Efeitos Tardios da Exposição Pré-Natal/metabolismo
2.
Int J Mol Sci ; 22(18)2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34576178

RESUMO

The transcription factor GATA2 regulates gene expression in several cells and tissues, including hematopoietic tissues and the central nervous system. Recent studies revealed that loss-of-function mutations in GATA2 are associated with hematological disorders. Our earlier in vitro studies showed that GATA2 plays an essential role in the hypothalamus-pituitary-thyroid axis (HPT axis) by regulating the genes encoding prepro-thyrotropin-releasing hormone (preproTRH) and thyroid-stimulating hormone ß (TSHß). However, the effect of GATA2 mutants on the transcriptional activity of their promoters remains unelucidated. In this study, we created five human GATA2 mutations (R308P, T354M, R396Q, R398W, and S447R) that were reported to be associated with hematological disorders and analyzed their functional properties, including transactivation potential and DNA-binding capacity toward the preproTRH and the TSHß promoters. Three mutations (T354M, R396Q, and R398W) within the C-terminal zinc-finger domain reduced the basal GATA2 transcriptional activity on both the preproTRH and the TSHß promoters with a significant loss of DNA binding affinity. Interestingly, only the R398W mutation reduced the GATA2 protein expression. Subsequent analysis demonstrated that the R398W mutation possibly facilitated the GATA2 degradation process. R308P and S447R mutants exhibited decreased transcriptional activity under protein kinase C compared to the wild-type protein. In conclusion, we demonstrated that naturally occurring GATA2 mutations impair the HPT axis through differential functional mechanisms in vitro.


Assuntos
Fator de Transcrição GATA2/genética , Hipotálamo/metabolismo , Mutação/genética , Hipófise/metabolismo , Glândula Tireoide/metabolismo , Western Blotting , Haploinsuficiência/genética , Haploinsuficiência/fisiologia , Humanos , Hipotireoidismo/genética , Regiões Promotoras Genéticas/genética , Tireotropina Subunidade beta/genética , Tireotropina Subunidade beta/metabolismo , Ativação Transcricional/genética , Ativação Transcricional/fisiologia
3.
Invest New Drugs ; 38(6): 1717-1729, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32500465

RESUMO

Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), the first immune checkpoint to be targeted clinically, has provided an effective treatment option for various malignancies. However, the clinical advantages associated with CTLA-4 inhibitors can be offset by the potentially severe immune-related adverse events (IRAEs), including autoimmune thyroid dysfunction. To investigate the candidate genes and signaling pathways involving in autoimmune thyroid dysfunction related to anti-CTLA-4 therapy, integrated differentially expressed genes (DEGs) were extracted from the intersection of genes from Gene Expression Omnibus (GEO) datasets and text mining. The functional enrichment was performed by gene ontology (GO) annotation and Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis. Protein-protein interaction (PPI) network, module enrichment, and hub gene identification were constructed and visualized by the online Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape software. A total of 22 and 17 integrated human DEGs in hypothyroidism and hyperthyroidism group related to anti-CTLA-4 therapy were identified, respectively. Functional enrichment analysis revealed 24 GO terms and 1 KEGG pathways in the hypothyroid group and 21 GO terms and 2 KEGG pathways in the hyperthyroid group. After PPI network construction, the top five hub genes associated with hypothyroidism were extracted, including ALB, MAPK1, SPP1, PPARG, and MIF, whereas those associated with hyperthyroidism were ALB, FCGR2B, CD44, LCN2, and CD74. The identification of the candidate key genes and enriched signaling pathways provides potential biomarkers for autoimmune thyroid dysfunction related to anti-CTLA-4 therapy and might contribute to the future diagnosis and management of IRAEs for cancer patients.


Assuntos
Doenças Autoimunes/genética , Antígeno CTLA-4/antagonistas & inibidores , Hipertireoidismo/genética , Hipotireoidismo/genética , Inibidores de Checkpoint Imunológico/efeitos adversos , Animais , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/metabolismo , Biomarcadores , Biologia Computacional , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Humanos , Hipertireoidismo/induzido quimicamente , Hipotireoidismo/induzido quimicamente , Camundongos , Mapas de Interação de Proteínas , Transdução de Sinais
4.
J Med Genet ; 55(10): 693-700, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30061370

RESUMO

BACKGROUND: Four genetic causes of isolated congenital central hypothyroidism (CeH) have been identified, but many cases remain unexplained. We hypothesised the existence of other genetic causes of CeH with a Mendelian inheritance pattern. METHODS: We performed exome sequencing in two families with unexplained isolated CeH and subsequently Sanger sequenced unrelated idiopathic CeH cases. We performed clinical and biochemical characterisation of the probands and carriers identified by family screening. We investigated IRS4 mRNA expression in human hypothalamus and pituitary tissue, and measured serum thyroid hormones and Trh and Tshb mRNA expression in hypothalamus and pituitary tissue of Irs4 knockout mice. RESULTS: We found mutations in the insulin receptor substrate 4 (IRS4) gene in two pairs of brothers with CeH (one nonsense, one frameshift). Sequencing of IRS4 in 12 unrelated CeH cases negative for variants in known genes yielded three frameshift mutations (two novel) in three patients and one male sibling. All male carriers (n=8) had CeH with plasma free thyroxine concentrations below the reference interval. MRI of the hypothalamus and pituitary showed no structural abnormalities (n=12). 24-hour thyroid-stimulating hormone (TSH) secretion profiles in two adult male patients showed decreased basal, pulsatile and total TSH secretion. IRS4 mRNA was expressed in human hypothalamic nuclei, including the paraventricular nucleus, and in the pituitary gland. Female knockout mice showed decreased pituitary Tshb mRNA levels but had unchanged serum thyroid hormone concentrations. CONCLUSIONS: Mutations in IRS4 are associated with isolated CeH in male carriers. As IRS4 is involved in leptin signalling, the phenotype may be related to disrupted leptin signalling.


Assuntos
Hipotireoidismo/genética , Proteínas Substratos do Receptor de Insulina/genética , Leptina/metabolismo , Transdução de Sinais , Tiroxina/sangue , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Feminino , Heterozigoto , Humanos , Hipotálamo/metabolismo , Lactente , Masculino , Camundongos , Pessoa de Meia-Idade , Mutação , Linhagem , Hipófise/metabolismo , Adulto Jovem
5.
Thyroid ; 26(12): 1701-1705, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27762734

RESUMO

BACKGROUND: Hemizygous mutations in the immunoglobulin superfamily member 1 (IGSF1) gene have been demonstrated to cause congenital central hypothyroidism in males. This study reports a family with a novel mutation in the IGSF1 gene located on the long arm of the X chromosome. PATIENT FINDINGS: A two-month-old boy was diagnosed with central hypothyroidism because of prolonged jaundice. A thyrotropin-releasing hormone (TRH) stimulation test indicated dysfunction in both the hypothalamus and the pituitary gland, and prompted the IGSF1 gene to be analyzed. The patient had a novel nonsense variant, c.2713C>T (p.Q905X), in exon 14 of the IGSF1 gene. Studies of the family revealed that the patient's sister and mother were heterozygous carriers of the IGSF1 mutation. The patient's maternal uncle carried the same mutation as the proband but had no overt symptoms. The mother and uncle started levothyroxine supplementation because of subclinical hypothyroidism. SUMMARY: A novel mutation (c.2713C>T, p.Q905X) of the IGSF1 gene was identified that causes congenital central hypothyroidism in a Japanese family. The findings further expand the clinical heterogeneity of this entity.


Assuntos
Hipotireoidismo Congênito/genética , Hipotireoidismo/genética , Imunoglobulinas/genética , Proteínas de Membrana/genética , Mutação , Adulto , Hipotireoidismo Congênito/tratamento farmacológico , Análise Mutacional de DNA , Terapia de Reposição Hormonal , Humanos , Hipotireoidismo/tratamento farmacológico , Lactente , Masculino , Linhagem , Tiroxina/uso terapêutico
6.
J Clin Endocrinol Metab ; 101(12): 4564-4573, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27603907

RESUMO

CONTEXT: Isolated congenital central hypothyroidism (CeH) can result from mutations in TRHR, TSHB, and IGSF1, but its etiology often remains unexplained. We identified a missense mutation in the transducin ß-like protein 1, X-linked (TBL1X) gene in three relatives diagnosed with isolated CeH. TBL1X is part of the thyroid hormone receptor-corepressor complex. OBJECTIVE: The objectives of the study were the identification of TBL1X mutations in patients with unexplained isolated CeH, Sanger sequencing of relatives of affected individuals, and clinical and biochemical characterization; in vitro investigation of functional consequences of mutations; and mRNA expression in, and immunostaining of, human hypothalami and pituitary glands. DESIGN: This was an observational study. SETTING: The study was conducted at university medical centers. PATIENTS: Nineteen individuals with and seven without a mutation participated in the study. MAIN OUTCOME MEASURES: Outcome measures included sequencing results, clinical and biochemical characteristics of mutation carriers, and results of in vitro functional and expression studies. RESULTS: Sanger sequencing yielded five additional mutations. All patients (n = 8; six males) were previously diagnosed with CeH (free T4 [FT4] concentration below the reference interval, normal thyrotropin). Eleven relatives (two males) also carried mutations. One female had CeH, whereas 10 others had low-normal FT4 concentrations. As a group, adult mutation carriers had 20%-25% lower FT4 concentrations than controls. Twelve of 19 evaluated carriers had hearing loss. Mutations are located in the highly conserved WD40-repeat domain of the protein, influencing its expression and thermal stability. TBL1X mRNA and protein are expressed in the human hypothalamus and pituitary. CONCLUSIONS: TBL1X mutations are associated with CeH and hearing loss. FT4 concentrations in mutation carriers vary from low-normal to values compatible with CeH.


Assuntos
Perda Auditiva/genética , Hipotireoidismo/genética , Hipófise/metabolismo , Tiroxina/sangue , Transducina/genética , Adolescente , Adulto , Criança , Feminino , Perda Auditiva/etiologia , Heterozigoto , Humanos , Hipotálamo/metabolismo , Hipotireoidismo/sangue , Hipotireoidismo/complicações , Lactente , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , RNA Mensageiro/metabolismo , Adulto Jovem
7.
Endocrinology ; 157(8): 3266-77, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27254003

RESUMO

Mice deficient in the type 3 deiodinase (D3KO mice) manifest impaired clearance of thyroid hormone (TH), leading to elevated levels of TH action during development. This alteration causes reduced neonatal viability, growth retardation, and central hypothyroidism. Here we examined how these phenotypes are affected by a deficiency in the monocarboxylate transporter 8 (MCT8), which is a major contributor to the transport of the active thyroid hormone, T3, into the cell. MCT8 deficiency eliminated the neonatal lethality of type 3 deiodinase (D3)-deficient mice and significantly ameliorated their growth retardation. Double-mutant newborn mice exhibited similar peripheral thyrotoxicosis and increased brain expression of T3-dependent genes as mice with D3 deficiency only. Later in neonatal life and adulthood, double-mutant mice manifested central and peripheral TH status similar to mice with single MCT8 deficiency, with low serum T4, elevated serum TSH and T3, and decreased T3-dependent gene expression in the hypothalamus. In double-mutant adult mice, both thyroid gland size and the hypothyroidism-induced rise in TSH were greater than those in mice with single D3 deficiency but less than those in mice with MCT8 deficiency alone. Our results demonstrate that the marked phenotypic abnormalities observed in the D3-deficient mouse, including perinatal mortality, growth retardation, and central hypothyroidism in adult animals, require expression of MCT8, confirming the interdependent relationship between the TH transport into cells and the deiodination processes.


Assuntos
Viabilidade Fetal , Crescimento e Desenvolvimento , Iodeto Peroxidase/genética , Proteínas de Membrana Transportadoras/genética , Animais , Animais Recém-Nascidos , Retardo do Crescimento Fetal/genética , Viabilidade Fetal/genética , Crescimento e Desenvolvimento/genética , Hipotálamo/fisiologia , Hipotireoidismo/genética , Masculino , Camundongos , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos , Fenótipo , Simportadores , Glândula Tireoide/fisiologia
8.
PLoS Biol ; 12(9): e1001952, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25248098

RESUMO

Characterization of the genetic defects causing gonadotropic deficiency has made a major contribution to elucidation of the fundamental role of Kisspeptins and Neurokinin B in puberty onset and reproduction. The absence of puberty may also reveal neurodevelopmental disorders caused by molecular defects in various cellular pathways. Investigations of these neurodevelopmental disorders may provide information about the neuronal processes controlling puberty onset and reproductive capacity. We describe here a new syndrome observed in three brothers, which involves gonadotropic axis deficiency, central hypothyroidism, peripheral demyelinating sensorimotor polyneuropathy, mental retardation, and profound hypoglycemia, progressing to nonautoimmune insulin-dependent diabetes mellitus. High-throughput sequencing revealed a homozygous in-frame deletion of 15 nucleotides in DMXL2 in all three affected patients. This homozygous deletion was associated with lower DMXL2 mRNA levels in the blood lymphocytes of the patients. DMXL2 encodes the synaptic protein rabconnectin-3α, which has been identified as a putative scaffold protein for Rab3-GAP and Rab3-GEP, two regulators of the GTPase Rab3a. We found that rabconnectin-3α was expressed in exocytosis vesicles in gonadotropin-releasing hormone (GnRH) axonal extremities in the median eminence of the hypothalamus. It was also specifically expressed in cells expressing luteinizing hormone (LH) and follicle-stimulating hormone (FSH) within the pituitary. The conditional heterozygous deletion of Dmxl2 from mouse neurons delayed puberty and resulted in very low fertility. This reproductive phenotype was associated with a lower number of GnRH neurons in the hypothalamus of adult mice. Finally, Dmxl2 knockdown in an insulin-secreting cell line showed that rabconnectin-3α controlled the constitutive and glucose-induced secretion of insulin. In conclusion, this study shows that low levels of DMXL2 expression cause a complex neurological phenotype, with abnormal glucose metabolism and gonadotropic axis deficiency due to a loss of GnRH neurons. Our findings identify rabconectin-3α as a key controller of neuronal and endocrine homeostatic processes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Diabetes Mellitus Tipo 1/genética , Hipoglicemia/genética , Hipotireoidismo/genética , Infertilidade Masculina/genética , Deficiência Intelectual/genética , Proteínas do Tecido Nervoso/genética , Polineuropatias/genética , Deleção de Sequência , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Adolescente , Animais , Sequência de Bases , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Hormônio Foliculoestimulante/genética , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Haploinsuficiência , Homozigoto , Humanos , Hipoglicemia/metabolismo , Hipoglicemia/patologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotireoidismo/metabolismo , Hipotireoidismo/patologia , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/deficiência , Neurônios/metabolismo , Neurônios/patologia , Hipófise/crescimento & desenvolvimento , Hipófise/metabolismo , Hipófise/patologia , Polineuropatias/metabolismo , Polineuropatias/patologia , Maturidade Sexual , Síndrome , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Testículo/patologia , Adulto Jovem
9.
Thyroid ; 24(11): 1575-93, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25087834

RESUMO

BACKGROUND: The thyroid hormone triiodothyronine (T3) is known to affect energy balance. Recent evidence points to an action of T3 in the hypothalamus, a key area of the brain involved in energy homeostasis, but the components and mechanisms are far from understood. The aim of this study was to identify components in the hypothalamus that may be involved in the action of T3 on energy balance regulatory mechanisms. METHODS: Sprague Dawley rats were made hypothyroid by giving 0.025% methimazole (MMI) in their drinking water for 22 days. On day 21, half the MMI-treated rats received a saline injection, whereas the others were injected with T3. Food intake and body weight measurements were taken daily. Body composition was determined by magnetic resonance imaging, gene expression was analyzed by in situ hybridization, and T3-induced gene expression was determined by microarray analysis of MMI-treated compared to MMI-T3-injected hypothalamic RNA. RESULTS: Post mortem serum thyroid hormone levels showed that MMI treatment decreased circulating thyroid hormones and increased thyrotropin (TSH). MMI treatment decreased food intake and body weight. Body composition analysis revealed reduced lean and fat mass in thyroidectomized rats from day 14 of the experiment. MMI treatment caused a decrease in circulating triglyceride concentrations, an increase in nonesterified fatty acids, and decreased insulin levels. A glucose tolerance test showed impaired glucose clearance in the thyroidectomized animals. In the brain, in situ hybridization revealed marked changes in gene expression, including genes such as Mct8, a thyroid hormone transporter, and Agrp, a key component in energy balance regulation. Microarray analysis revealed 110 genes to be up- or downregulated with T3 treatment (± 1.3-fold change, p<0.05). Three genes chosen from the differentially expressed genes were verified by in situ hybridization to be activated by T3 in cells located at or close to the hypothalamic ventricular ependymal layer and differentially expressed in animal models of long- and short-term body weight regulation. CONCLUSION: This study identified genes regulated by T3 in the hypothalamus, a key area of the brain involved in homeostasis and neuroendocrine functions. These include genes hitherto not known to be regulated by thyroid status.


Assuntos
Glicemia/metabolismo , Metabolismo Energético/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hipotireoidismo/genética , Tri-Iodotironina/farmacologia , Animais , Composição Corporal/efeitos dos fármacos , Composição Corporal/fisiologia , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Hipotireoidismo/induzido quimicamente , Hipotireoidismo/metabolismo , Masculino , Metimazol , Ratos , Ratos Sprague-Dawley
10.
Hum Mol Genet ; 23(10): 2651-64, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24381310

RESUMO

Mutations of the thyroid hormone receptor α gene (THRA) cause hypothyroidism in patients with growth and developmental retardation, and skeletal dysplasia. Genetic evidence indicates that the dominant negative activity of TRα1 mutants underlies pathological manifestations. Using a mouse model of hypothyroidism caused by a dominant negative TRα1PV mutant and its derived mouse model harboring a mutated nuclear receptor corepressor (NCOR1ΔID) (Thra1(PV/+)Ncor1(ΔID/ΔID) mice), we recently showed that aberrant release of TRα1 mutants from the NCOR1 repressor complex mediates dominant negative actions of TRα1 mutants in vivo. We tested the hypothesis that deacetylation of nucleosomal histones associated with aberrant recruitment of corepressors by TRα1 mutants underlies pathological phenotypic expression. We treated Thra1(PV/+)and Thra1(PV/+)Ncor1(ΔID/ΔID) mice with a histone deacetylase (HDAC) inhibitor, suberoylanilide hydroxyamic acid (SAHA). SAHA significantly ameliorated the impaired growth, bone development and adipogenesis of Thra1(PV/+) mice. In Thra1(PV/+)Ncor1(ΔID/ΔID) mice, SAHA improved these abnormalities even further. We focused our molecular analyses on how SAHA improved the impaired adipogenesis leading to the lean phenotype. We found that SAHA reverted the impaired adipogenesis by de-repressing the expression of the two master regulators of adipogenesis, C/ebpα and Pparγ, as well as other adipogenic genes at both the mRNA and protein levels. Chromatin immunoprecipitation analyses indicated SAHA increased the extent of acetylation of nucleosomal H4K5 and H3 to re-activate adipogenic genes to reverting adipogenesis. Thus, HDAC confers in vivo aberrant actions of TRα1 mutants. Importantly, for the first time, the present studies show that HDAC inhibitors are clearly beneficial for hypothyroidism and could be therapeutics for treatment.


Assuntos
Inibidores de Histona Desacetilases/uso terapêutico , Ácidos Hidroxâmicos/uso terapêutico , Hipotireoidismo/tratamento farmacológico , Receptores alfa dos Hormônios Tireóideos/genética , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Avaliação Pré-Clínica de Medicamentos , Epigênese Genética/efeitos dos fármacos , Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/farmacologia , Hipotireoidismo/sangue , Hipotireoidismo/genética , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/patologia , Masculino , Camundongos Transgênicos , PPAR gama/genética , PPAR gama/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Hormônios Tireóideos/sangue , Vorinostat , Aumento de Peso/efeitos dos fármacos
11.
OMICS ; 17(11): 584-93, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24044365

RESUMO

The thyroid pathway represents a complex interaction of different glands for thyroid hormone synthesis. Thyrotropin releasing hormone is synthesized in the hypothalamus and regulates thyrotropin stimulating hormone gene expression in the pituitary gland. In order to understand the complexity of the thyroid pathways, and using experimental data retrieved from the biomedical literature (e.g., NCBI, HuGE Navigator, Protein Data Bank, and KEGG), we constructed a metabolic map of the thyroid hormone pathway at a molecular level and analyzed it topologically. A total of five hub nodes were predicted in regards to the transcription thyroid receptor (TR), cAMP response element-binding protein (CREB), signal transducer and activator of transcription 3 (STAT 3), nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB), and activator protein 1 (AP-1) as being potentially important in study of thyroid disorders and as novel putative therapeutic drug targets. Notably, the thyroid receptor is a highly connected hub node and currently used as a therapeutic target in hypothyroidism. Our analysis represents the first comprehensive description of the thyroid pathway, which pertains to understanding the function of the protein and gene interaction networks. The findings from this study are therefore informative for pathophysiology and rational therapeutics of thyroid disorders.


Assuntos
Hipotireoidismo/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Transdução de Sinais , Glândula Tireoide/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Bases de Dados de Proteínas , Regulação da Expressão Gênica , Humanos , Hipotálamo/metabolismo , Hipotireoidismo/genética , Hipotireoidismo/patologia , NF-kappa B/genética , NF-kappa B/metabolismo , Hipófise/metabolismo , Mapeamento de Interação de Proteínas , Receptores dos Hormônios Tireóideos/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Glândula Tireoide/patologia , Tireotropina/genética , Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/genética , Hormônio Liberador de Tireotropina/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo
12.
PLoS One ; 8(8): e71822, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951251

RESUMO

BACKGROUND: Protein-tyrosine sulfation is a post-translational modification of an unknown number of secreted and membrane proteins mediated by two known Golgi tyrosylprotein sulfotransferases (TPST-1 and TPST-2). We reported that Tpst2-/- mice have mild-moderate primary hypothyroidism, whereas Tpst1-/- mice are euthyroid. While using magnetic resonance imaging (MRI) to look at the thyroid gland we noticed that the salivary glands in Tpst2-/- mice appeared smaller than in wild type mice. This prompted a detailed analysis to compare salivary gland structure and function in wild type, Tpst1-/-, and Tpst2 -/- mice. METHODOLOGY/PRINCIPAL FINDINGS: Quantitative MRI imaging documented that salivary glands in Tpst2-/- females were (≈) 30% smaller than wild type or Tpst1-/- mice and that the granular convoluted tubules in Tpst2-/- submandibular glands were less prominent and were almost completely devoid of exocrine secretory granules compared to glands from wild type or Tpst1-/- mice. In addition, pilocarpine-induced salivary flow and salivary α-amylase activity in Tpst2-/- mice of both sexes was substantially lower than in wild type and Tpst1-/- mice. Anti-sulfotyrosine Western blots of salivary gland extracts and saliva showed no differences between wild type, Tpst1-/-, and Tpst2-/- mice, suggesting that the salivary gland hypofunction is due to factor(s) extrinsic to the salivary glands. Finally, we found that all indicators of hypothyroidism (serum T4, body weight) and salivary gland hypofunction (salivary flow, salivary α-amylase activity, histological changes) were restored to normal or near normal by thyroid hormone supplementation. CONCLUSIONS/SIGNIFICANCE: Our findings conclusively demonstrate that low body weight and salivary gland hypofunction in Tpst2-/- mice is due solely to primary hypothyroidism.


Assuntos
Hipotireoidismo/metabolismo , Glândulas Salivares/metabolismo , Sulfotransferases/metabolismo , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Peso Corporal/fisiologia , Suplementos Nutricionais , Feminino , Expressão Gênica , Hipotireoidismo/sangue , Hipotireoidismo/genética , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândulas Salivares/patologia , Glândulas Salivares/fisiopatologia , alfa-Amilases Salivares/metabolismo , Glândula Submandibular/metabolismo , Glândula Submandibular/patologia , Glândula Submandibular/fisiopatologia , Sulfotransferases/genética , Tireoide (USP)/administração & dosagem , Tireoide (USP)/farmacologia , Tiroxina/sangue
13.
Thyroid ; 23(9): 1057-65, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23517243

RESUMO

BACKGROUND: The atherogenic effects of hypothyroidism on lipid metabolism could result, in part, from the reduced clearance of remnant lipoproteins. In this study, we investigated the expression of hepatic low-density lipoprotein receptor-related protein 1 (LRP1), a receptor for remnant lipoproteins, in hypothyroidism and the effect of 3,3',5-triiodo-L-thyronine (T3) treatment on hepatic LRP1 expression. METHODS: C57BL/6 mice were fed a normal diet (control group) or a low-iodine diet supplemented with 0.15% propylthiouracil (PTU/LI group) for 4 weeks. Mice in the PTU/LI group were injected intraperitoneally with T3 (0, 30, and 150 µg/kg of body weight) for 7 days. HepG2 cells were incubated in fetal bovine serum or charcoal-stripped fetal bovine serum with various concentrations of T3. The expression and function of LRP1 in liver samples and cells were analyzed. RESULTS: Hypothyroidism was successfully induced in PTU/LI mice. Hepatic LRP1 protein expression was lower in the PTU/LI group than in the control group. T3 treatment upregulated hepatic LRP1 protein expression in PTU/LI mice. LRP1 expression in HepG2 cells was reduced after incubation in the medium containing charcoal-stripped fetal bovine serum, which mimics hypothyroidism in vitro, and was recovered by T3 treatment. The protein expression of LRP1 in HepG2 cells was increased by T3 treatment in a dose-dependent manner up to 2.0 nM T3. However, LRP1 mRNA transcription was not affected by hypothyroidism conditions or T3 treatment, either in liver samples or in HepG2 cells. T3 treatment on HepG2 cells increased cellular uptake of lipid-conjugated apolipoprotein E through LRP1. CONCLUSIONS: Our data demonstrate that hepatic LRP1 expression and function decrease in hypothyroidism and are regulated by the thyroid hormone. These results suggest that in hypothyroidism, decreased expression of hepatic LRP1 may be associated with reduced clearance of circulating remnant lipoproteins.


Assuntos
Aterosclerose/etiologia , Dislipidemias/etiologia , Hipotireoidismo/complicações , Fígado/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Dislipidemias/genética , Dislipidemias/metabolismo , Células Hep G2 , Humanos , Hipotireoidismo/induzido quimicamente , Hipotireoidismo/tratamento farmacológico , Hipotireoidismo/genética , Hipotireoidismo/metabolismo , Fígado/efeitos dos fármacos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Propiltiouracila , RNA Mensageiro/metabolismo , Receptores de LDL/genética , Tri-Iodotironina/farmacologia , Proteínas Supressoras de Tumor/genética
14.
Mol Biol Rep ; 39(11): 9849-61, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22733496

RESUMO

In the present study, regulatory role of vitamin E and curcumin on antioxidant gene (AOG) expression in hypothyroid rat liver is reported. Adult male rats were rendered hypothyroid by administration of 0.05 % 6-propyl-thiouracil in their drinking water, while vitamin E (200 mg/kg body weight) and curcumin (30 mg/kg body weight) were supplemented orally for 30 days. Expression of antioxidant genes (Cu/Zn-superoxide dismutase; SOD1, Mn superoxide dismutase; SOD2, catalase; CAT, glutathione peroxidase; GPx1 and glutathione reductase; GR) was evaluated using RT-PCR and Western blot analyses. The activities of antioxidant enzymes were measured in mitochondrial fraction (MF) and post-mitochondrial fraction (PMF) of rat liver. In addition measurement of glutathione redox status was also carried out in both the fractions. The enhanced transcripts of CAT, GPx1 and GR in hypothyroid rat liver were alleviated by administration of vitamin E and curcumin. Elevated levels of translated product of all AOGs in hypothyroid group were remained unchanged after antioxidant administration. However, enhanced SOD1, GPx1 and decreased GR activities in PMF were normalized by vitamin E and curcumin. Similarly the increased SOD2, GPx1 and decreased CAT activities in MF were also normalized by vitamin E and curcumin supplementation. Administration of vitamin E and curcumin enhanced mitochondrial GSH level; whereas the enhanced GSH level in PMF of hypothyroid rats was alleviated by vitamin E. Thus it can be concluded that besides the antioxidant role of vitamin E and curcumin, they also regulate hepatic antioxidant gene expression in hypothyroid rats.


Assuntos
Antioxidantes/farmacologia , Antitireóideos/efeitos adversos , Curcumina/farmacologia , Expressão Gênica , Fígado/metabolismo , Propiltiouracila/efeitos adversos , Vitamina E/farmacologia , Animais , Glutationa/metabolismo , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Hipotireoidismo/induzido quimicamente , Hipotireoidismo/genética , Hipotireoidismo/metabolismo , Fígado/enzimologia , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Estresse Oxidativo , Ratos , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Glutationa Peroxidase GPX1
15.
PLoS One ; 5(9): e12931, 2010 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-20877559

RESUMO

Impaired expression of selenium-containing proteins leads to perturbed thyroid hormone (TH) levels, indicating the central importance of selenium for TH homeostasis. Moreover, critically ill patients with declining serum selenium develop a syndrome of low circulating TH and a central downregulation of the hypothalamus-pituitary-thyroid axis. This prompted us to test the reciprocal effect, i.e., if TH status would also regulate selenoprotein expression and selenium levels. To investigate the TH dependency of selenium metabolism, we analyzed mice expressing a mutant TH receptor α1 (TRα1+m) that confers a receptor-mediated hypothyroidism. Serum selenium was reduced in these animals, which was a direct consequence of the mutant TRα1 and not related to their metabolic alterations. Accordingly, hyperthyroidism, genetically caused by the inactivation of TRß or by oral TH treatment of adult mice, increased serum selenium levels in TRα1+m and controls, thus demonstrating a novel and specific role for TRα1 in selenium metabolism. Furthermore, TH affected the mRNA levels for several enzymes involved in selenoprotein biosynthesis as well as serum selenoprotein P concentrations and the expression of other antioxidative selenoproteins. Taken together, our results show that TH positively affects the serum selenium status and regulates the expression of several selenoproteins. This demonstrates that selenium and TH metabolism are interconnected through a feed-forward regulation, which can in part explain the rapid parallel downregulation of both systems in critical illness.


Assuntos
Regulação da Expressão Gênica , Hipotireoidismo/metabolismo , Selênio/sangue , Selenoproteínas/genética , Tri-Iodotironina/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Hipotireoidismo/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Selenoproteínas/metabolismo , Receptores alfa dos Hormônios Tireóideos/genética , Receptores alfa dos Hormônios Tireóideos/metabolismo , Receptores beta dos Hormônios Tireóideos/genética , Receptores beta dos Hormônios Tireóideos/metabolismo
16.
Thyroid ; 20(8): 917-26, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20615127

RESUMO

BACKGROUND: We describe a rare case of congenital hypothyroidism and an extremely high serum thyrotropin (TSH) level caused by a combination of resistance to thyroid hormone (RTH) and a lingual thyroid. As the RTH mutant, R316C, was new, the optimum dose of levothyroxine was unclear. To aid in assessment of the therapy, we characterized the mutant R316C thyroid hormone receptor (TR) and compared it with a common mutant, R316H, using in vitro studies. SUMMARY: The patient was a newborn female having severe hypothyroidism with a free thyroxine level of 0.36 ng/dL and a serum TSH level of 177 microU/mL. A scintiscan showed ectopic lingual thyroid tissue without a normal thyroid gland. Supplementation with levothyroxine at a dose of >350 microg/day did not normalize the serum TSH level; however, the patient showed normal growth and intelligence at 14 years of age. Consistent with the results of a computer analysis, the binding of R316C to triiodothyronine (T3) was significantly decreased to 38% that of the wild type. Electrophoretic mobility shift assay demonstrated that like R316H, R316C did not form a homodimer, but formed a heterodimer with RXR. However, a glutathione-S-transferase pull-down assay showed reduced binding of R316C with NCoR in the absence of T3 and impaired release in the presence of T3. In addition, transient transfection experiments demonstrated that unlike R316H, R316C had severe impairment of transcriptional activity on genes both positively and negatively regulated by thyroid hormone. It also had a clear dominant negative effect on genes negatively, but not positively, regulated by thyroid hormone, including the TSH-releasing hormone and TSHbeta genes. CONCLUSION: This is the first reported case of a R316C TR mutation. The characteristics of the R316C mutant differed from those of the R316H mutant. Our findings suggest that R316C causes reduced association with and impaired release of NCoR, resulting in RTH predominantly at the pituitary level, and that slightly elevated serum TSH level with high dose of levothyroxine might be optimum for normal growth.


Assuntos
Hipotireoidismo/complicações , Hipotireoidismo/genética , Tireoide Lingual/complicações , Tireoide Lingual/genética , Mutação , Hormônios Tireóideos/metabolismo , Animais , Criança , Dimerização , Feminino , Humanos , Recém-Nascido , Receptores dos Hormônios Tireóideos/metabolismo , Receptores X de Retinoides/metabolismo , Tiroxina/metabolismo , Tiroxina/farmacologia , Tri-Iodotironina/metabolismo
17.
Nat Med ; 15(10): 1186-94, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19767733

RESUMO

Thyroid dysfunction is a global health concern, causing defects including neurodevelopmental disorders, dwarfism and cardiac arrhythmia. Here, we show that the potassium channel subunits KCNQ1 and KCNE2 form a thyroid-stimulating hormone-stimulated, constitutively active, thyrocyte K+ channel required for normal thyroid hormone biosynthesis. Targeted disruption of Kcne2 in mice impaired thyroid iodide accumulation up to eightfold, impaired maternal milk ejection, halved milk tetraiodothyronine (T4) content and halved litter size. Kcne2-deficient mice had hypothyroidism, dwarfism, alopecia, goiter and cardiac abnormalities including hypertrophy, fibrosis, and reduced fractional shortening. The alopecia, dwarfism and cardiac abnormalities were alleviated by triiodothyronine (T3) and T4 administration to pups, by supplementing dams with T(4) before and after they gave birth or by feeding the pups exclusively from Kcne2+/+ dams; conversely, these symptoms were elicited in Kcne2+/+ pups by feeding exclusively from Kcne2-/- dams. These data provide a new potential therapeutic target for thyroid disorders and raise the possibility of an endocrine component to previously identified KCNE2- and KCNQ1-linked human cardiac arrhythmias.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Deleção de Sequência , Glândula Tireoide/metabolismo , Hormônios Tireóideos/metabolismo , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cruzamentos Genéticos , Heterozigoto , Homozigoto , Hipotireoidismo/genética , Hipotireoidismo/metabolismo , Lactação/genética , Lactação/metabolismo , Camundongos , Camundongos Knockout , Leite/metabolismo , Miócitos Cardíacos/metabolismo , Glândula Tireoide/ultraestrutura , Hormônios Tireóideos/genética , Tri-Iodotironina/genética , Tri-Iodotironina/metabolismo
18.
Endocrinol. nutr. (Ed. impr.) ; 55(supl.1): 29-34, ene. 2008. tab
Artigo em Espanhol | IBECS | ID: ibc-61167

RESUMO

La deficiencia de yodo grave y moderada durante la gestación y la lactancia afecta a la función tiroidea de la madre y del neonato, así como al desarrollo neuropsicológico del niño. En la mayoría de los países de Europa el estado nutricional de yodo de gestantes y recién nacidos es deficiente, pues las necesidades de yodo en las mujeres embarazadas y en las que amamantan a sus hijos son al menos el doble de las recomendadas para la población adulta. Estudios realizados en España confirman que la mayoría de las mujeres se encuentran en yododeficiencia durante la gestación y la lactancia y muestran quela ingesta de sal yodada no es suficiente para conseguir un estado nutricional de yodo óptimo en esta población. Varios estudios demuestran el efecto beneficioso de la suplementación de yodo de las embarazadas sobre el estado nutricional de yodo y la función tiroidea de la madre y el recién nacido. Las mujeres embarazadas, las que amamantan a sus hijos y las que planifican su gestación deberían recibir suplementos de yodo en forma de tabletas de yoduro potásico (AU)


Severe and mild iodine deficiency during pregnancy and lactation affects thyroid function of the mother and neonate as well as the infant’s neuropsychological development. In most European countries, maternal and neonatal iodine status is deficient, since iodine requirements in pregnant and lactating women are almost double those recommended for non pregnantor non-lactating women. Studies performed in Spain confirm that most women are iodine deficient during pregnancy and lactation, and show that iodized salt intake is insufficient to achieve optimal iodine nutritional status in this population. Several studies have demonstrated the beneficial effect of iodine supplementation in pregnant women on the iodine and thyroid status of both mother and neonate. Pregnant and breastfeeding women and women planning to become pregnant should take iodine supplements in the form of potassium iodide tablets (AU)


Assuntos
Humanos , Masculino , Feminino , Gravidez , Criança , Adulto , Lactação , Lactação/fisiologia , Gravidez/metabolismo , Iodeto de Potássio/metabolismo , Iodeto de Potássio/uso terapêutico , Deficiência de Iodo/metabolismo , Deficiência de Iodo/terapia , Hipotireoidismo/dietoterapia , Estado Nutricional/genética , Estado Nutricional/fisiologia , Neuropsicologia/tendências , Lactação/metabolismo , Hipotireoidismo/metabolismo , Hipotireoidismo/genética , Gravidez/fisiologia , Receptores da Tireotropina/uso terapêutico , Estado Nutricional
19.
Endocrinology ; 148(12): 5680-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17823249

RESUMO

The type 3 deiodinase (D3) is a selenoenzyme that inactivates thyroid hormones and is highly expressed during development and in the adult central nervous system. We have recently observed that mice lacking D3 activity (D3KO mice) develop perinatal thyrotoxicosis followed in adulthood by a pattern of hormonal levels that is suggestive of central hypothyroidism. In this report we describe the results of additional studies designed to investigate the regulation of the thyroid axis in this unique animal model. Our results demonstrate that the thyroid and pituitary glands of D3KO mice do not respond appropriately to TSH and TRH stimulation, respectively. Furthermore, after induction of severe hypothyroidism by antithyroid treatment, the rise in serum TSH in D3KO mice is only 15% of that observed in wild-type mice. In addition, D3KO animals rendered severely hypothyroid fail to show the expected increase in prepro-TRH mRNA in the paraventricular nucleus of the hypothalamus. Finally, treatment with T(3) results in a serum T(3) level in D3KO mice that is much higher than that in wild-type mice. This is accompanied by significant weight loss and lethality in mutant animals. In conclusion, the absence of D3 activity results in impaired clearance of T(3) and significant defects in the mechanisms regulating the thyroid axis at all levels: hypothalamus, pituitary, and thyroid.


Assuntos
Hipotálamo/metabolismo , Iodeto Peroxidase/metabolismo , Hipófise/metabolismo , Glândula Tireoide/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotireoidismo/genética , Hipotireoidismo/metabolismo , Hibridização In Situ , Iodeto Peroxidase/genética , Masculino , Camundongos , Camundongos Knockout , Hipófise/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sobrevida , Glândula Tireoide/efeitos dos fármacos , Hormônios Tireóideos/administração & dosagem , Hormônios Tireóideos/farmacologia , Tireotropina/sangue , Tireotropina/farmacologia , Hormônio Liberador de Tireotropina/genética , Hormônio Liberador de Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/farmacologia
20.
Eur J Endocrinol ; 151 Suppl 3: U39-44, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15554885

RESUMO

Recently molecular genetic defects in some cases of congenital hypothyroidism (CH) as well as of rare cases of central hypothyroidism have been identified. These studies have led to the description of so far unexplained forms of these disorders. In some patients with CH early diagnosis by newborn screening and early treatment was not able to lead to a normal mental development. This could subsequently be explained by molecular defects of transcription factors (FOXE-1/FKHL15, NKX2.1) which are important not only for the embryonic development of the thyroid gland but also for other organs including the central nervous system (CNS). These findings will help in understanding the critical role of thyroid hormones in the pre-and postnatal CNS development. However, many questions regarding the molecular defects and their consequences in the majority of patients with CH still remain open and will be addressed in this article.


Assuntos
Hipotireoidismo Congênito , Hipotireoidismo/genética , Animais , Sistema Nervoso Central/embriologia , Feminino , Proteínas de Homeodomínio/genética , Humanos , Hipotálamo/anormalidades , Proteínas com Homeodomínio LIM , Camundongos , Hipófise/anormalidades , Glândula Tireoide/anormalidades , Glândula Tireoide/embriologia , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA