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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 117(36): 22544-22551, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32826330

RESUMO

Obesity is a major health problem worldwide, given its growing incidence and its association with a variety of comorbidities. Weight gain results from an increase in energy intake without a concomitant increase in energy expenditure. To combat the obesity epidemic, many studies have focused on the pathways underlying satiety and hunger signaling, while other studies have concentrated on the mechanisms involved in energy expenditure, most notably adaptive thermogenesis. Hypothyroidism in humans is typically associated with a decreased basal metabolic rate, lower energy expenditure, and weight gain. However, hypothyroid mouse models have been reported to have a leaner phenotype than euthyroid controls. To elucidate the mechanism underlying this phenomenon, we used a drug-free mouse model of hypothyroidism: mice lacking the sodium/iodide symporter (NIS), the plasma membrane protein that mediates active iodide uptake in the thyroid. In addition to being leaner than euthyroid mice, owing in part to reduced food intake, these hypothyroid mice show signs of compensatory up-regulation of the skeletal-muscle adaptive thermogenic marker sarcolipin, with an associated increase in fatty acid oxidation (FAO). Neither catecholamines nor thyroid-stimulating hormone (TSH) are responsible for sarcolipin expression or FAO stimulation; rather, thyroid hormones are likely to negatively regulate both processes in skeletal muscle. Our findings indicate that hypothyroidism in mice results in a variety of metabolic changes, which collectively lead to a leaner phenotype. A deeper understanding of these changes may make it possible to develop new strategies against obesity.


Assuntos
Hipotireoidismo/metabolismo , Músculo Esquelético/metabolismo , Termogênese/fisiologia , Animais , Modelos Animais de Doenças , Ingestão de Alimentos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Musculares/metabolismo , Fenótipo , Proteolipídeos/metabolismo , Simportadores/genética , Simportadores/metabolismo
2.
Annu Rev Physiol ; 79: 261-289, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28192058

RESUMO

Active iodide (I-) transport in both the thyroid and some extrathyroidal tissues is mediated by the Na+/I- symporter (NIS). In the thyroid, NIS-mediated I- uptake plays a pivotal role in thyroid hormone (TH) biosynthesis. THs are key during embryonic and postembryonic development and critical for cell metabolism at all stages of life. The molecular characterization of NIS in 1996 and the use of radioactive I- isotopes have led to significant advances in the diagnosis and treatment of thyroid cancer and provide the molecular basis for studies aimed at extending the use of radioiodide treatment in extrathyroidal malignancies. This review focuses on the most recent findings on I- homeostasis and I- transport deficiency-causing NIS mutations, as well as current knowledge of the structure/function properties of NIS and NIS regulatory mechanisms. We also discuss employing NIS as a reporter gene using viral vectors and stem cells in imaging, diagnostic, and therapeutic procedures.


Assuntos
Iodetos/metabolismo , Simportadores/metabolismo , Animais , Genes Reporter/genética , Humanos , Simportadores/genética , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia , Glândula Tireoide/fisiologia , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/fisiopatologia
3.
Proc Natl Acad Sci U S A ; 114(43): E9172-E9180, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-29073114

RESUMO

Hypothyroidism, a metabolic disease characterized by low thyroid hormone (TH) and high thyroid-stimulating hormone (TSH) levels in the serum, is strongly associated with nonalcoholic fatty liver disease (NAFLD). Hypothyroidism-induced NAFLD has generally been attributed to reduced TH signaling in the liver with a consequent decrease in lipid utilization. Here, we found that mildly hypothyroid mice develop NAFLD without down-regulation of hepatic TH signaling or decreased hepatic lipid utilization. NAFLD was induced by impaired suppression of adipose tissue lipolysis due to decreased insulin secretion and to a reduced response of adipose tissue itself to insulin. This condition leads to increased shuttling of fatty acids (FAs) to the liver, where they are esterified and accumulated as triglycerides. Lipid accumulation in the liver induces hepatic insulin resistance, which leads to impaired suppression of endogenous glucose production after feeding. Hepatic insulin resistance, synergistically with lowered insulin secretion, increases serum glucose levels, which stimulates de novo lipogenesis (DNL) in the liver. Up-regulation of DNL also contributes to NAFLD. In contrast, severely hypothyroid mice show down-regulation of TH signaling in their livers and profound suppression of adipose tissue lipolysis, which decreases delivery of FAs to the liver. The resulting lack of substrates for triglyceride esterification protects severely hypothyroid mice against NAFLD. Our findings demonstrate that NAFLD occurs when TH levels are mildly reduced, but, paradoxically, not when they are severely reduced. Our results show that the pathogenesis of hypothyroidism-induced NAFLD is both intra- and extrahepatic; they also reveal key metabolic differences between mild and severe hypothyroidism.


Assuntos
Hipotireoidismo/complicações , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Hepatopatia Gordurosa não Alcoólica/etiologia , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiopatologia , Animais , Modelos Animais de Doenças , Hipotireoidismo/etiologia , Insulina/metabolismo , Secreção de Insulina , Metabolismo dos Lipídeos , Lipólise/fisiologia , Fígado/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/complicações , Simportadores/genética
4.
Proc Natl Acad Sci U S A ; 108(44): 17933-8, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22011571

RESUMO

I(-) uptake in the thyroid, the first step in thyroid hormone biosynthesis, is mediated by the Na(+)/I(-) symporter (NIS) with an electrogenic 2Na(+):1I(-) stoichiometry. We have obtained mechanistic information on NIS by characterizing the congenital I(-) transport defect-causing NIS mutant G93R. This mutant is targeted to the plasma membrane but is inactive. Substitutions at position 93 show that the longer the side chain of the neutral residue at this position, the higher the K(m) for the anion substrates. Unlike WT NIS, which mediates symport of Na(+) and the environmental pollutant perchlorate electroneutrally, G93T/N/Q/E/D NIS, strikingly, do it electrogenically with a 21 stoichiometry. Furthermore, G93E/Q NIS discriminate between anion substrates, a discovery with potential clinical relevance. A 3D homology model of NIS based on the structure of the bacterial Na(+)/galactose transporter identifies G93 as a critical player in the mechanism of the transporter: the changes from an outwardly to an inwardly open conformation during the transport cycle use G93 as a pivot.


Assuntos
Iodetos/metabolismo , Sódio/metabolismo , Simportadores/metabolismo , Animais , Ânions , Células COS , Linhagem Celular , Chlorocebus aethiops , Humanos , Modelos Moleculares , Simportadores/química
5.
FASEB J ; 26(8): 3252-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22549510

RESUMO

The KCNQ1 α subunit and the KCNE2 ß subunit form a potassium channel in thyroid epithelial cells. Genetic disruption of KCNQ1-KCNE2 causes hypothyroidism in mice, resulting in cardiac hypertrophy, dwarfism, alopecia, and prenatal mortality. Here, we investigated the mechanistic requirement for KCNQ1-KCNE2 in thyroid hormone biosynthesis, utilizing whole-animal dynamic positron emission tomography. The KCNQ1-specific antagonist (-)-[3R,4S]-chromanol 293B (C293B) significantly impaired thyroid cell I(-) uptake, which is mediated by the Na(+)/I(-) symporter (NIS), in vivo (dSUV/dt: vehicle, 0.028 ± 0.004 min(-1); 10 mg/kg C293B, 0.009 ± 0.006 min(-1)) and in vitro (EC(50): 99 ± 10 µM C293B). Na(+)-dependent nicotinate uptake by SMCT, however, was unaffected. Kcne2 deletion did not alter the balance of free vs. thyroglobulin-bound I(-) in the thyroid (distinguished using ClO(4)(-), a competitive inhibitor of NIS), indicating that KCNQ1-KCNE2 is not required for Duox/TPO-mediated I(-) organification. However, Kcne2 deletion doubled the rate of free I(-) efflux from the thyroid following ClO(4)(-) injection, a NIS-independent process. Thus, KCNQ1-KCNE2 is necessary for adequate thyroid cell I(-) uptake, the most likely explanation being that it is prerequisite for adequate NIS activity.


Assuntos
Iodetos/metabolismo , Canal de Potássio KCNQ1/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Glândula Tireoide/metabolismo , Animais , Células COS , Chlorocebus aethiops , Humanos , Hipotireoidismo/genética , Canal de Potássio KCNQ1/antagonistas & inibidores , Canal de Potássio KCNQ1/genética , Camundongos , Tomografia por Emissão de Pósitrons , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Simportadores/antagonistas & inibidores , Simportadores/metabolismo , Glândula Tireoide/efeitos dos fármacos
6.
J Physiol ; 590(23): 6013-26, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23006481

RESUMO

Dietary I(-) absorption in the gastrointestinal tract is the first step in I(-) metabolism. Given that I(-) is an essential constituent of the thyroid hormones, its concentrating mechanism is of significant physiological importance. We recently described the expression of the Na(+)/I(-) symporter (NIS) on the apical surface of the intestinal epithelium as a central component of the I(-) absorption system and reported reduced intestinal NIS expression in response to an I(-)-rich diet in vivo. Here, we evaluated the mechanism involved in the regulation of NIS expression by I(-) itself in enterocytes. Excess I(-) reduced NIS-mediated I(-) uptake in IEC-6 cells in a dose- and time-dependent fashion, which was correlated with a reduction of NIS expression at the plasma membrane. Perchlorate, a competitive inhibitor of NIS, prevented these effects, indicating that an increase in intracellular I(-) regulates NIS. Iodide induced rapid intracellular recruitment of plasma membrane NIS molecules and NIS protein degradation. Lower NIS mRNA levels were detected in response to I(-) treatment, although no transcriptional effect was observed. Interestingly, I(-) decreased NIS mRNA stability, affecting NIS translation. Heterologous green fluorescent protein-based reporter constructs revealed a significant repressive effect of the I(-)-targeting NIS mRNA 3 untranslated region. In conclusion, excess I(-) downregulates NIS expression in enterocytes by virtue of a complex mechanism. Our data suggest that I(-) regulates intestinal NIS mRNA expression at the post-transcriptional level as part of an autoregulatory effect of I(-) on its own metabolism.


Assuntos
Iodeto de Potássio/farmacologia , Simportadores/fisiologia , Animais , Linhagem Celular , Dieta , Enterócitos/efeitos dos fármacos , Enterócitos/metabolismo , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Transcrição Gênica
7.
Thyroid ; 31(8): 1272-1281, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33779310

RESUMO

Background: The sodium/iodide (Na+/I-) symporter (NIS) mediates active transport of I- into the thyroid gland. Mutations in the SLC5A5 gene, which encodes NIS, cause I- transport defects (ITDs)-which, if left untreated, lead to congenital hypothyroidism and consequent cognitive and developmental deficiencies. The ITD-causing NIS mutation Y348D, located in transmembrane segment (TMS) 9, was reported in three Sudanese patients. Methods: We generated cDNAs coding for Y348D NIS and mutants with other hydrophilic and hydrophobic amino acid substitutions at position 348 and transfected them into cells. The activity of the resulting mutants was quantitated by radioiodide transport assays. NIS glycosylation was investigated by Western blotting after endoglycosidase H (Endo H) and PNGase-F glycosidase treatment. Subcellular localization of the mutant proteins was ascertained by flow cytometry analysis, cell surface biotinylation, and immunofluorescence. The intrinsic activity of Y348D was studied by measuring radioiodide transport in membrane vesicles prepared from Y348D-NIS-expressing cells. Our NIS homology models and molecular dynamics simulations were used to identify residues that interact with Y348 and investigate possible interactions between Y348 and the membrane. The sequences of several Slc5 family transporters were aligned, and a phylogenetic tree was generated in ClustalX. Results: Cells expressing Y348D NIS transport no I-. Furthermore, Y348D NIS is only partially glycosylated, is retained intracellularly, and is intrinsically inactive. Hydrophilic residues other than Asp at position 348 also yield NIS proteins that fail to be targeted to the plasma membrane (PM), whereas hydrophobic residues at this position, which we show do not interact with the membrane, rescue PM targeting and function. Conclusions: Y348D NIS does not reach the PM and is intrinsically inactive. Hydrophobic amino acid substitutions at position 348, however, preserve NIS activity. Our findings are consistent with our homology model's prediction that Y348 should face the side opposite the TMS9 residues that coordinate Na+ and participate in Na+ transport, and with the notion that Y348 interacts only with hydrophobic residues. Hydrophilic or charged residues at position 348 have deleterious effects on NIS PM targeting and activity, whereas a hydrophobic residue at this position rescues NIS activity.


Assuntos
Membrana Celular/metabolismo , Iodo/metabolismo , Mutação/genética , Simportadores/genética , Glândula Tireoide/metabolismo , Substituição de Aminoácidos , Transporte Biológico/genética , Biotinilação , Hipotireoidismo Congênito/genética , DNA/genética , Glicosilação , Humanos , Interações Hidrofóbicas e Hidrofílicas , Radioisótopos do Iodo , Mutagênese Sítio-Dirigida , Frações Subcelulares/metabolismo
8.
Proc Natl Acad Sci U S A ; 104(51): 20250-5, 2007 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-18077370

RESUMO

The Na(+)/I(-) symporter (NIS) is a key plasma membrane protein that mediates active I(-) uptake in the thyroid, lactating breast, and other tissues with an electrogenic stoichiometry of 2 Na(+) per I(-). In the thyroid, NIS-mediated I(-) uptake is the first step in the biosynthesis of the iodine-containing thyroid hormones, which are essential early in life for proper CNS development. In the lactating breast, NIS mediates the translocation of I(-) to the milk, thus supplying this essential anion to the nursing newborn. Perchlorate (ClO(4)(-)) is a well known competitive inhibitor of NIS. Exposure to food and water contaminated with ClO(4)(-) is common in the U.S. population, and the public health impact of such exposure is currently being debated. To date, it is still uncertain whether ClO(4)(-) is a NIS blocker or a transported substrate of NIS. Here we show in vitro and in vivo that NIS actively transports ClO(4)(-), including ClO(4)(-) translocation to the milk. A simple mathematical fluxes model accurately predicts the effect of ClO(4)(-) transport on the rate and extent of I(-) accumulation. Strikingly, the Na(+)/ ClO(4)(-) transport stoichiometry is electroneutral, uncovering that NIS translocates different substrates with different stoichiometries. That NIS actively concentrates ClO(4)(-) in maternal milk suggests that exposure of newborns to high levels of ClO(4)(-) may pose a greater health risk than previously acknowledged because ClO(4)(-) would thus directly inhibit the newborns' thyroidal I(-) uptake.


Assuntos
Poluentes Ambientais/metabolismo , Iodo/metabolismo , Percloratos/metabolismo , Simportadores/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Cães , Iodo/análise , Cinética , Leite/química , Modelos Teóricos , Percloratos/análise , Ratos , Transfecção
9.
Nat Struct Mol Biol ; 27(7): 683, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32472109

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Nat Struct Mol Biol ; 27(6): 533-539, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32451489

RESUMO

The Na+/I- symporter (NIS), the plasma membrane protein that actively transports I- (stoichiometry 2Na+:1I-) in thyroid physiology and radioiodide-based thyroid cancer treatment, also transports the environmental pollutant perchlorate (stoichiometry 1Na+:1ClO4-), which competes with I- for transport. Until now, the mechanism by which NIS transports different anion substrates with different stoichiometries has remained unelucidated. We carried out transport measurements and analyzed these using a statistical thermodynamics-based equation and electrophysiological experiments to show that the different stoichiometry of ClO4- transport is due to ClO4- binding to a high-affinity non-transport allosteric site that prevents Na+ from binding to one of its two sites. Furthermore, low concentrations of ClO4- inhibit I- transport not only by competition but also, critically, by changing the stoichiometry of I- transport to 1:1, which greatly reduces the driving force. The data reveal that ClO4- pollution in drinking water is more dangerous than previously thought.


Assuntos
Percloratos/metabolismo , Simportadores/química , Simportadores/metabolismo , Regulação Alostérica , Sítio Alostérico , Animais , Ânions/química , Ânions/metabolismo , Sítios de Ligação , Transporte Biológico , Cães , Eletrofisiologia/métodos , Feminino , Humanos , Iodo/metabolismo , Células Madin Darby de Rim Canino , Mutação , Oócitos/metabolismo , Oócitos/fisiologia , Percloratos/química , Ratos , Sódio/metabolismo , Simportadores/genética , Termodinâmica , Xenopus laevis
11.
Cell Rep ; 19(6): 1083-1090, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28494858

RESUMO

The mammalian target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth that is commonly deregulated in human diseases. Here we find that mTORC1 controls a transcriptional program encoding amino acid transporters and metabolic enzymes through a mechanism also used to regulate protein synthesis. Bioinformatic analysis of mTORC1-responsive mRNAs identified a promoter element recognized by activating transcription factor 4 (ATF4), a key effector of the integrated stress response. ATF4 translation is normally induced by the phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α) through a mechanism that requires upstream open reading frames (uORFs) in the ATF4 5' UTR. mTORC1 also controls ATF4 translation through uORFs, but independently of changes in eIF2α phosphorylation. mTORC1 instead employs the 4E-binding protein (4E-BP) family of translation repressors. These results link mTORC1-regulated demand for protein synthesis with an ATF4-regulated transcriptional program that controls the supply of amino acids to the translation machinery.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Processamento Pós-Transcricional do RNA , Regiões 5' não Traduzidas , Fator 4 Ativador da Transcrição/genética , Animais , Fator de Iniciação 2 em Eucariotos/metabolismo , Células HEK293 , Humanos , Camundongos , Fases de Leitura Aberta , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
12.
Sci Rep ; 7(1): 5329, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28706256

RESUMO

The sodium/iodide symporter (NIS) mediates active iodide (I-) accumulation in the thyroid, the first step in thyroid hormone (TH) biosynthesis. Mutations in the SLC5A5 gene encoding NIS that result in a non-functional protein lead to congenital hypothyroidism due to I- transport defect (ITD). ITD is a rare autosomal disorder that, if not treated promptly in infancy, can cause mental retardation, as the TH decrease results in improper development of the nervous system. However, in some patients, hypothyroidism has been ameliorated by unusually large amounts of dietary I-. Here we report the first NIS knockout (KO) mouse model, obtained by targeting exons 6 and 7 of the Slc5a5 gene. In NIS KO mice, in the thyroid, stomach, and salivary gland, NIS is absent, and hence there is no active accumulation of the NIS substrate pertechnetate (99mTcO4-). NIS KO mice showed undetectable serum T4 and very low serum T3 levels when fed a diet supplying the minimum I- requirement for rodents. These hypothyroid mice displayed oxidative stress in the thyroid, but not in the brown adipose tissue or liver. Feeding the mice a high-I- diet partially rescued TH biosynthesis, demonstrating that, at high I- concentrations, I- enters the thyroid through routes other than NIS.


Assuntos
Suplementos Nutricionais , Hipotireoidismo/prevenção & controle , Iodetos/administração & dosagem , Simportadores/deficiência , Animais , Transporte Biológico , Modelos Animais de Doenças , Iodetos/metabolismo , Camundongos Knockout , Hormônios Tireóideos/sangue , Resultado do Tratamento
13.
Oncotarget ; 7(34): 54811-54824, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27363025

RESUMO

Treating breast cancer brain metastases (BCBMs) is challenging. Na+/I- symporter (NIS) expression in BCBMs would permit their selective targeting with radioiodide (131I-). We show impressive enhancement of tumor response by combining131I- with gemcitabine (GEM), a cytotoxic radiosensitizer. Nude mice mammary fat-pad (MFP) tumors and BCBMs were generated with braintropic MDA-MB-231Br cells transduced with bicistronically-linked NIS and firefly luciferase cDNAs. Response was monitored in vivo via bioluminescent imaging and NIS tumor expression.131I-/GEM therapy inhibited MFP tumor growth more effectively than either agent alone. BCBMs were treated with: high or low-dose GEM (58 or 14.5 mg/Kg×4); 131I- (1mCi or 2×0.5 mCi 7 days apart); and 131I-/GEM therapy. By post-injection day (PID) 25, 82-86% of controls and 78-83% of 131I--treated BCBM grew, whereas 17% low-dose and 36% high-dose GEM regressed. The latter tumors were smaller than the controls with comparable NIS expression (~20% of cells). High and low-dose 131I-/ GEM combinations caused 89% and 57% tumor regression, respectively. High-dose GEM/131I- delayed tumor growth: tumors increased 5-fold in size by PID45 (controls by PID18). Although fewer than 25% of cells expressed NIS, GEM/131I- caused dramatic tumor regression in NIS-transduced BCBMs. This effect was synergistic, and supports the hypothesis that GEM radiosensitizes cells to 131I-.


Assuntos
Neoplasias Encefálicas/terapia , Neoplasias da Mama/terapia , Desoxicitidina/análogos & derivados , Radioisótopos do Iodo/uso terapêutico , Simportadores/metabolismo , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundário , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Terapia Combinada , Desoxicitidina/uso terapêutico , Feminino , Células HEK293 , Humanos , Radioisótopos do Iodo/farmacocinética , Camundongos Nus , Simportadores/genética , Transfecção , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Carga Tumoral/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
14.
J Clin Endocrinol Metab ; 100(10): E1353-61, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26204134

RESUMO

CONTEXT: Iodide (I(-)), an essential constituent of the thyroid hormones, is actively accumulated in the thyroid by the Na(+)/I(-) symporter (NIS), a key plasma membrane protein encoded by the slc5a5 gene. Mutations in slc5a5 cause I(-) transport defects (ITDs), autosomal-recessive disorders in which I(-) accumulation is totally or partially impaired, leading to congenital hypothyroidism. The characterization of NIS mutants has yielded significant insights into the molecular mechanism of NIS. OBJECTIVE: This study aimed to determine the basis of a patient's ITD clinical phenotype, by sequencing her slc5a5 gene. DESIGN: Genomic DNA was purified and the slc5a5 gene sequence determined. Functional in vitro studies were performed to characterize the V270E NIS mutant. PATIENT: The index patient was diagnosed with hypothyroidism with minimal radioiodide uptake in a normally located, although enlarged, thyroid gland. RESULTS: We identified a new NIS mutation: V270E. The patient had the compound heterozygous NIS mutation R124H/V270E. R124H NIS has been characterized previously. We show that V270E markedly reduces I(-) uptake via a pronounced (but not total) impairment of the protein's plasma membrane targeting. Remarkably, V270E is intrinsically active. Therefore, a negative charge at position 270 interferes with NIS cell surface trafficking. The patient's minimal I(-) uptake enabled sufficient thyroid hormone biosynthesis to prevent cognitive impairment. CONCLUSIONS: A nonpolar residue at position 270, which all members of the SLC5A family have, is required for NIS plasma membrane targeting.


Assuntos
Transtornos Cognitivos/genética , Transtornos do Crescimento/genética , Mutação , Simportadores/genética , Alelos , Criança , Hipotireoidismo Congênito/genética , Análise Mutacional de DNA , Feminino , Humanos , Fenótipo
15.
Nat Commun ; 6: 8070, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26437858

RESUMO

The aetiology of human fibrolamellar hepatocellular carcinomas (hFL-HCCs), cancers occurring increasingly in children to young adults, is poorly understood. We present a transplantable tumour line, maintained in immune-compromised mice, and validate it as a bona fide model of hFL-HCCs by multiple methods. RNA-seq analysis confirms the presence of a fusion transcript (DNAJB1-PRKACA) characteristic of hFL-HCC tumours. The hFL-HCC tumour line is highly enriched for cancer stem cells as indicated by limited dilution tumourigenicity assays, spheroid formation and flow cytometry. Immunohistochemistry on the hFL-HCC model, with parallel studies on 27 primary hFL-HCC tumours, provides robust evidence for expression of endodermal stem cell traits. Transcriptomic analyses of the tumour line and of multiple, normal hepatic lineage stages reveal a gene signature for hFL-HCCs closely resembling that of biliary tree stem cells--newly discovered precursors for liver and pancreas. This model offers unprecedented opportunities to investigate mechanisms underlying hFL-HCCs pathogenesis and potential therapies.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Proteínas Mutantes Quiméricas/genética , Células-Tronco Neoplásicas/metabolismo , RNA Mensageiro/metabolismo , Adolescente , Adulto , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Proteínas de Choque Térmico HSP40/genética , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Transplante de Neoplasias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de RNA , Esferoides Celulares , Células Tumorais Cultivadas , Adulto Jovem
16.
Brain Res Bull ; 58(6): 607-12, 2002 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-12372566

RESUMO

The molecular mechanisms involved in the regulation of synaptic plasticity and neuroprotection by estradiol (E(2)) and progesterone (P(4)) are unknown. Because these processes involve changes in cytoskeleton organization, we studied the effects of E(2) and P(4) in the expression of two cytoskeletal proteins: microtubule associated protein 2 (MAP2) and tau in the hippocampus and the frontal cortex of ovariectomized adult rats. While tau expression was unaffected by E(2) and P(4), an increase in MAP2 protein content in the hippocampus but not in the cortex was observed after E(2) and P(4) treatments. Interestingly, these steroids did not modify MAP2 mRNA content in the hippocampus. These data suggest that MAP2 is involved in the structural changes induced by E(2) and P(4) in the rat hippocampus, and that MAP2 expression is regulated by these steroid hormones at a postranscriptional level.


Assuntos
Estradiol/farmacologia , Hipocampo/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/biossíntese , Progesterona/farmacologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Estradiol/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipocampo/metabolismo , Ovariectomia , Progesterona/fisiologia , Ratos , Ratos Wistar , Proteínas tau/biossíntese
17.
Am J Physiol Cell Physiol ; 296(4): C654-62, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19052257

RESUMO

Absorption of dietary iodide, presumably in the small intestine, is the first step in iodide (I(-)) utilization. From the bloodstream, I(-) is actively taken up via the Na(+)/I(-) symporter (NIS) in the thyroid for thyroid hormone biosynthesis and in such other tissues as lactating breast, which supplies I(-) to the newborn in the milk. The molecular basis for intestinal I(-) absorption is unknown. We sought to determine whether I(-) is actively accumulated by enterocytes and, if so, whether this process is mediated by NIS and regulated by I(-) itself. NIS expression was localized exclusively at the apical surface of rat and mouse enterocytes. In vivo intestine-to-blood transport of pertechnetate, a NIS substrate, was sensitive to the NIS inhibitor perchlorate. Brush border membrane vesicles accumulated I(-) in a sodium-dependent, perchlorate-sensitive manner with kinetic parameters similar to those of thyroid cells. NIS was expressed in intestinal epithelial cell line 6, and I(-) uptake in these cells was also kinetically similar to that in thyrocytes. I(-) downregulated NIS protein expression and its own NIS-mediated transport both in vitro and in vivo. We conclude that NIS is functionally expressed on the apical surface of enterocytes, where it mediates active I(-) accumulation. Therefore, NIS is a significant and possibly central component of the I(-) absorption system in the small intestine, a system of key importance for thyroid hormone biosynthesis and thus systemic intermediary metabolism.


Assuntos
Enterócitos/metabolismo , Absorção Intestinal , Intestino Delgado/metabolismo , Iodetos/metabolismo , Simportadores/metabolismo , Animais , Linhagem Celular , Polaridade Celular , Enterócitos/efeitos dos fármacos , Absorção Intestinal/efeitos dos fármacos , Intestino Delgado/citologia , Intestino Delgado/efeitos dos fármacos , Radioisótopos do Iodo , Cinética , Masculino , Camundongos , Microvilosidades/metabolismo , Percloratos/farmacologia , Ratos , Ratos Sprague-Dawley , Pertecnetato Tc 99m de Sódio/metabolismo , Simportadores/antagonistas & inibidores , Glândula Tireoide/metabolismo
18.
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
19.
Comp Biochem Physiol A Mol Integr Physiol ; 147(3): 685-690, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16931081

RESUMO

We characterized the expression pattern of progesterone receptor (PR) in two regions of the oviduct (ampullae and isthmus), and the uterus (epithelium and stroma) of the rabbit (Oryctolagus cuniculus) during early pregnancy (1-4 days) by RT-PCR and immunohistochemistry. We observed a significant increase in the expression of PR at mRNA level in the uterus on days 1 and 2 of pregnancy, followed by a decrease on days 3 and 4. These changes were also observed at protein level in the uterine epithelium. Interestingly, PR immunoreactivity decreased in stromal cells in all days of pregnancy as compared with non-pregnant rabbits (NG). In the isthmus PR mRNA expression significantly increased on day 2 of pregnancy and diminished on days 3 and 4, whereas no significant changes were observed in the ampullae. In epithelial and stromal cells of the isthmus, PR immunostaining was reduced through pregnancy as compared with NG group. In contrast, a reduction in PR immunostaining was observed on days 1-3 with an increase on day 4 in epithelial and stromal cells of the ampullae. The overall results suggest that PR exhibit a differential expression pattern in the oviduct and the uterus during early pregnancy of the rabbit, and that these differences are related to different functions of PR in the reproductive tract during early pregnancy.


Assuntos
Tubas Uterinas/metabolismo , Regulação da Expressão Gênica , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Útero/metabolismo , Animais , Tubas Uterinas/citologia , Feminino , Imuno-Histoquímica , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coelhos , Útero/citologia
20.
J Neurobiol ; 60(4): 473-80, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15307151

RESUMO

The molecular mechanisms involved in the regulation of synaptic plasticity in the hippocampus during the estrous cycle of the rat are not completely understood. Because this process implicates changes in neuronal cytoskeleton organization, we analyzed the content of microtubule associated protein 2 (MAP2) and Tau in the hippocampus and the frontal cortex of the rat by Western blot, as well as the hippocampal distribution of MAP2 during the estrous cycle by immunohistochemistry. In the hippocampus the lowest content of MAP2 was found on diestrus day, and it significantly increased at proestrus. This increase was maintained on estrus and metestrus days. In the frontal cortex MAP2 content did not significantly change during the estrous cycle. In contrast, the content of Tau did not vary during the estrous cycle in either the hippocampus or the frontal cortex. The immunohistochemical analysis showed an increase in dendrite thickness and in dendritic branching in the CA1 region on proestrus day, as well as an aggregation of MAP2 in apical dendrites near to pyramidal somata on this day in comparison with diestrus. We suggest that changes in the content and neuronal distribution of MAP2 are involved in the structural changes that occur in the hippocampus of the rat during the estrous cycle, and that these variations are related to changes in estradiol and progesterone levels.


Assuntos
Ciclo Estral/fisiologia , Hipocampo/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Animais , Dendritos/metabolismo , Dendritos/ultraestrutura , Estradiol/metabolismo , Feminino , Hipocampo/citologia , Microtúbulos/ultraestrutura , Neurônios/ultraestrutura , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/metabolismo , Progesterona/metabolismo , Células Piramidais/metabolismo , Células Piramidais/ultraestrutura , Ratos , Ratos Wistar , Regulação para Cima/fisiologia , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA