Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Medicinas Complementares
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Metab ; 66: 101636, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36375792

RESUMO

OBJECTIVE: Thyroid transcription factor-1 (TTF-1), a homeodomain-containing transcription factor, is predominantly expressed in discrete areas of the hypothalamus, which acts as the central unit for the regulation of whole-body energy homeostasis. Current study designed to identify the roles of TTF-1 on the responsiveness of the hypothalamic circuit activity to circulating leptin and the development of obesity linked to the insensitivity of leptin. METHODS: We generated conditional knock-out mice by crossing TTF-1flox/flox mice with leptin receptor (ObRb)Cre or proopiomelanocortin (POMC)Cre transgenic mice to interrogate the contributions of TTF-1 in leptin signaling and activity. Changes of food intake, body weight and energy expenditure were evaluated in standard or high fat diet-treated transgenic mice by using an indirect calorimetry instrument. Molecular mechanism was elucidated with immunohistochemistry, immunoblotting, quantitative PCR, and promoter assays. RESULTS: The selective deletion of TTF-1 gene expression in cells expressing the ObRb or POMC enhanced the anorexigenic effects of leptin as well as the leptin-induced phosphorylation of STAT3. We further determined that TTF-1 inhibited the transcriptional activity of the ObRb gene. In line with these findings, the selective deletion of the TTF-1 gene in ObRb-positive cells led to protective effects against diet-induced obesity via the amelioration of leptin resistance. CONCLUSIONS: Collectively, these results suggest that hypothalamic TTF-1 participates in the development of obesity as a molecular component involved in the regulation of cellular leptin signaling and activity. Thus, TTF-1 may represent a therapeutic target for the treatment, prevention, and control of obesity.


Assuntos
Leptina , Pró-Opiomelanocortina , Fator Nuclear 1 de Tireoide , Animais , Camundongos , Hipotálamo/metabolismo , Leptina/genética , Leptina/metabolismo , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Fator Nuclear 1 de Tireoide/genética , Fator Nuclear 1 de Tireoide/metabolismo
2.
Reprod Biol Endocrinol ; 19(1): 30, 2021 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-33622350

RESUMO

BACKGROUND: TTF1 is a transcription factor that is expressed in the hypothalamus after birth and plays crucial roles in pubertal development. TTF1 may regulate the expression of the Kiss1 gene, which may drive puberty onset in the hypothalamic arcuate (ARC) and anterior ventral paraventricular (AVPV) nuclei. METHODS: A dual-luciferase reporter assay was used to detect binding between TTF1 and the Kiss1 gene promoter. To investigate the effects of TTF1, we modified TTF1 expression in cell lines and in the ARC or AVPV nucleus of 21-day-old female rats via lentivirus infection. TTF1 and other puberty onset-related genes were detected by qRT-PCR and western blot analyses. RESULTS: The in vitro data indicated that TTF1 knockdown (KD) significantly reduced Kiss1 and GnRH expression. Overexpression (OE) of TTF1 promoted Kiss1 expression. In vivo, the expression of Kiss1 and GnRH decreased significantly in the rats with hypothalamic ARC- or AVPV-specific TTF1 KD. The TTF1-KD rats showed vaginal opening delay. H&E staining revealed that the corpus luteum was obviously reduced at the early puberty and adult stages in the rats with ARC- or AVPV-specific TTF1 KD. CONCLUSION: TTF1 bound to the promoter of the Kiss1 gene and enhanced its expression. For 21-day-old female rats, decreased TTF1 in the hypothalamic ARC or AVPV nucleus resulted in delayed vaginal opening and ovarian abnormalities. These observations suggested that TTF1 regulates puberty onset by promoting the expression of Kiss1 and plays an important role in gonad development.


Assuntos
Hipotálamo/metabolismo , Maturidade Sexual/genética , Fator Nuclear 1 de Tireoide/genética , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Células Cultivadas , Regulação para Baixo , Feminino , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Células HEK293 , Humanos , Hipotálamo Anterior/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Ratos , Ratos Sprague-Dawley , Fator Nuclear 1 de Tireoide/metabolismo , Fatores de Tempo
3.
Sci China Life Sci ; 64(3): 419-433, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32803714

RESUMO

Cenpj is a centrosomal protein located at the centrosomes and the base of cilia, it plays essential roles in regulating neurogenesis and cerebral cortex development. Although centrosomal and cilium dysfunction are one of the causes of obesity, insulin resistance, and type 2 diabetes, the role that Cenpj plays in the regulation of body weight remains unclear. Here, we deleted Cenpj by crossing Cenpjflox/flox mice with Nkx2.1-Cre mice. Loss of the centrosomal protein Cenpj in Nkx2.1-expressing cells causes morbid obesity in mice at approximately 4 months of age with expended brain ventricles but no change of brain size. We found that hypothalamic cells exhibited reduced proliferation and increased apoptosis upon Cenpj depletion at the embryonic stages, resulting in a dramatic decrease in the number of Proopiomelanocortin (POMC) neurons and electrophysiological dysfunction of NPY neurons in the arcuate nucleus (ARC) in adults. Furthermore, depletion of Cenpj also reduced the neuronal projection from the ARC to the paraventricular nucleus (PVN), with decreased melanocortin-4 receptors (MC4R) expression in PVN neurons. The study defines the roles that Cenpj plays in regulating hypothalamus development and body weight, providing a foundation for further understanding of the pathological mechanisms of related diseases.


Assuntos
Técnicas de Silenciamento de Genes , Hipotálamo/fisiopatologia , Proteínas Associadas aos Microtúbulos/genética , Obesidade Mórbida/fisiopatologia , Animais , Apoptose , Linhagem Celular , Proliferação de Células , Hipotálamo/embriologia , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo , Obesidade Mórbida/genética , Pró-Opiomelanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Fator Nuclear 1 de Tireoide/genética
4.
BMJ Case Rep ; 13(12)2020 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-33370995

RESUMO

A 7-month-old-term male infant presented with cough, tachypnoea, hypoxaemia and post-tussive emesis. Clinical history was significant for respiratory failure and pulmonary hypertension in the neonatal period requiring assisted ventilation, congenital hypothyroidism, mild hypotonia, recurrent respiratory infections, hypoxaemia requiring supplemental oxygen and nasogastric tube feeds. Physical examination showed tachypnoea, coarse bilateral breath sounds and mild hypotonia. Chest radiograph revealed multifocal pulmonary opacities with coarse interstitial markings and right upper lobe atelectasis. Following antibiotic therapy for suspected aspiration pneumonia, chest CT scan was performed and showed multiple areas of pulmonary consolidation and scattered areas of bilateral ground-glass opacities. Genetic studies showed a large deletion of chromosome 14q13.1-14q21.1, encompassing the NK2 homeobox 1 (NKX2-1) gene consistent with a diagnosis of brain-thyroid-lung (BTL) syndrome. Our case highlights the importance of genetic studies to diagnose BTL syndrome in infants with hypothyroidism, hypotonia and lung disease.


Assuntos
Atetose/diagnóstico , Coreia/diagnóstico , Deleção Cromossômica , Cromossomos Humanos Par 14/genética , Hipotireoidismo Congênito/diagnóstico , Hipóxia/genética , Hipotonia Muscular/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/diagnóstico , Combinação Amoxicilina e Clavulanato de Potássio/administração & dosagem , Atetose/complicações , Atetose/genética , Atetose/terapia , Coreia/complicações , Coreia/genética , Coreia/terapia , Hipotireoidismo Congênito/complicações , Hipotireoidismo Congênito/genética , Hipotireoidismo Congênito/terapia , Nutrição Enteral , Hidratação , Testes Genéticos , Humanos , Hipóxia/diagnóstico , Hipóxia/terapia , Lactente , Intubação Gastrointestinal , Pulmão/diagnóstico por imagem , Masculino , Hipotonia Muscular/diagnóstico , Hipotonia Muscular/terapia , Oxigênio/administração & dosagem , Síndrome do Desconforto Respiratório do Recém-Nascido/complicações , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia , Fator Nuclear 1 de Tireoide/genética , Tomografia Computadorizada por Raios X
5.
Brain Struct Funct ; 225(9): 2857-2869, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33145610

RESUMO

During the development of the central nervous system, the immature neurons suffer different migration processes. It is well known that Nkx2.1-positive ventricular layer give rise to critical tangential migrations into different regions of the developing forebrain. Our aim was to study this phenomenon in the hypothalamic region. With this purpose, we used a transgenic mouse line that expresses the tdTomato reporter driven by the promotor of Nkx2.1. Analysing the Nkx2.1-positive derivatives at E18.5, we found neural contributions to the prethalamic region, mainly in the zona incerta and in the mes-diencephalic tegmental region. We studied the developing hypothalamus along the embryonic period. From E10.5 we detected that the Nkx2.1 expression domain was narrower than the reporter distribution. Therefore, the Nkx2.1 expression fades in a great number of the early-born neurons from the Nkx2.1-positive territory. At the most caudal positive part, we detected a thin stream of positive neurons migrating caudally into the mes-diencephalic tegmental region using time-lapse experiments on open neural tube explants. Late in development, we found a second migratory stream into the prethalamic territory. All these tangentially migrated neurons developed a gabaergic phenotype. In summary, we have described the contribution of interneurons from the Nkx2.1-positive hypothalamic territory into two different rostrocaudal territories: the mes-diencephalic reticular formation through a caudal tangential migration and the prethalamic zona incerta complex through a dorsocaudal tangential migration.


Assuntos
Movimento Celular , Hipotálamo/crescimento & desenvolvimento , Neurônios/fisiologia , Fator Nuclear 1 de Tireoide/fisiologia , Animais , Feminino , Interneurônios/fisiologia , Masculino , Camundongos Transgênicos , Vias Neurais/fisiologia , Neurogênese , Zona Incerta/crescimento & desenvolvimento
6.
Nat Commun ; 11(1): 4360, 2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32868762

RESUMO

The hypothalamus is a central regulator of many innate behaviors essential for survival, but the molecular mechanisms controlling hypothalamic patterning and cell fate specification are poorly understood. To identify genes that control hypothalamic development, we have used single-cell RNA sequencing (scRNA-Seq) to profile mouse hypothalamic gene expression across 12 developmental time points between embryonic day 10 and postnatal day 45. This identified genes that delineated clear developmental trajectories for all major hypothalamic cell types, and readily distinguished major regional subdivisions of the developing hypothalamus. By using our developmental dataset, we were able to rapidly annotate previously unidentified clusters from existing scRNA-Seq datasets collected during development and to identify the developmental origins of major neuronal populations of the ventromedial hypothalamus. We further show that our approach can rapidly and comprehensively characterize mutants that have altered hypothalamic patterning, identifying Nkx2.1 as a negative regulator of prethalamic identity. These data serve as a resource for further studies of hypothalamic development, physiology, and dysfunction.


Assuntos
Diferenciação Celular , Hipotálamo , Neurônios/metabolismo , Fator Nuclear 1 de Tireoide/metabolismo , Animais , Sequência de Bases , Padronização Corporal , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/citologia , Hipotálamo/embriologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Camundongos , Mutação , Análise de Célula Única , Fator Nuclear 1 de Tireoide/genética
7.
Medicine (Baltimore) ; 99(12): e19650, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32195974

RESUMO

RATIONALE: Mutations of the NKX2-1 gene are associated with brain-lung-thyroid syndrome, which is characterized by benign hereditary chorea, hypothyroidism, and pulmonary disease with variable presentation. Surfactant protein C (SFTPC) gene mutations result in chronic interstitial lung disease in adults or severe neonatal respiratory distress syndrome. PATIENT CONCERNS: Recurrent hypoxemia was observed shortly after birth in a baby at a gestational age of 40 weeks and birth weight of 3150 g. The need for respiratory support gradually increased. He had hypothyroidism and experienced feeding difficulties and irritability. DIAGNOSIS: Genetic examination of the peripheral blood revealed combined mutations of the NKX2-1 and SFTPC genes. INTERVENTIONS: The patient was administered respiratory support, antibiotics, low-dose dexamethasone, supplementary thyroxine, venous nutrition, and other supportive measures. OUTCOMES: The patient's guardian stopped treatment 3 months after commencement of treatment, due to the seriousness of his condition and the patient died. LESSONS: Combined mutations of NKX2-1 and SFTPC genes are very rare. Thus, idiopathic interstitial pneumonia with hypothyroidism and neurological disorders require special attention.


Assuntos
Atetose/genética , Coreia/genética , Hipotireoidismo Congênito/genética , Proteína C/metabolismo , Surfactantes Pulmonares/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Fator Nuclear 1 de Tireoide/genética , Atetose/sangue , Atetose/diagnóstico , Atetose/terapia , Coreia/sangue , Coreia/diagnóstico , Coreia/terapia , Hipotireoidismo Congênito/sangue , Hipotireoidismo Congênito/diagnóstico , Hipotireoidismo Congênito/terapia , Evolução Fatal , Transtornos da Alimentação e da Ingestão de Alimentos/diagnóstico , Transtornos da Alimentação e da Ingestão de Alimentos/etiologia , Humanos , Hipotireoidismo/diagnóstico , Hipotireoidismo/etiologia , Hipóxia/diagnóstico , Hipóxia/etiologia , Recém-Nascido , Cariotipagem , Doenças Pulmonares Intersticiais/diagnóstico , Doenças Pulmonares Intersticiais/etiologia , Masculino , Mutação , Cuidados Paliativos/métodos , Recidiva , Síndrome do Desconforto Respiratório do Recém-Nascido/sangue , Síndrome do Desconforto Respiratório do Recém-Nascido/diagnóstico , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia
8.
J Neurosci ; 39(21): 4023-4035, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-30886014

RESUMO

Food intake is tightly regulated by a group of neurons present in the arcuate nucleus of the hypothalamus, which release Pomc-encoded melanocortins, the absence of which induces marked hyperphagia and early-onset obesity. Although the relevance of hypothalamic POMC neurons in the regulation of body weight and energy balance is well appreciated, little is known about the transcription factors that establish the melanocortin neuron identity during brain development and its phenotypic maintenance in postnatal life. Here, we report that the transcription factor NKX2.1 is present in mouse hypothalamic POMC neurons from early development to adulthood. Electromobility shift assays showed that NKX2.1 binds in vitro to NKX binding motifs present in the neuronal Pomc enhancers nPE1 and nPE2 and chromatin immunoprecipitation assays detected in vivo binding of NKX2.1 to nPE1 and nPE2 in mouse hypothalamic extracts. Transgenic and mutant studies performed in mouse embryos of either sex and adult males showed that the NKX motifs present in nPE1 and nPE2 are essential for their transcriptional enhancer activity. The conditional early inactivation of Nkx2.1 in the ventral hypothalamus prevented the onset of Pomc expression. Selective Nkx2.1 ablation from POMC neurons decreased Pomc expression in adult males and mildly increased their body weight and adiposity. Our results demonstrate that NKX2.1 is necessary to activate Pomc expression by binding to conserved canonical NKX motifs present in nPE1 and nPE2. Therefore, NKX2.1 plays a critical role in the early establishment of hypothalamic melanocortin neuron identity and participates in the maintenance of Pomc expression levels during adulthood.SIGNIFICANCE STATEMENT Food intake and body weight regulation depend on hypothalamic neurons that release satiety-inducing neuropeptides, known as melanocortins. Central melanocortins are encoded byPomc, and Pomc mutations may lead to hyperphagia and severe obesity. Although the importance of central melanocortins is well appreciated, the genetic program that establishes and maintains fully functional POMC neurons remains to be explored. Here, we combined molecular, genetic, developmental, and functional studies that led to the discovery of NKX2.1, a transcription factor that participates in the early morphogenesis of the developing hypothalamus, as a key player in establishing the early identity of melanocortin neurons by activating Pomc expression. Thus, Nkx2.1 adds to the growing list of genes that participate in body weight regulation and adiposity.


Assuntos
Melanocortinas/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Fator Nuclear 1 de Tireoide/metabolismo , Animais , Peso Corporal/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipotálamo/embriologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
9.
Biochem Biophys Res Commun ; 496(1): 147-152, 2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29305861

RESUMO

Here, we report thyroid transcription factor 1 (TTF-1) as an important transcription factor for the expression of heme oxygenase-1 (HO-1). HO-1 is a well-known cytoprotective enzyme against inflammation. We observed that HO-1 co-expressed with TTF-1 in mouse hypothalamic cells. Results from luciferase and chromatin immunoprecipitation assays revealed that TTF-1 directly activated HO-1 transcription by binding to binding domains in the 5'-flanking region of the HO-1 gene. A proinflammatory cytokine, tumor necrosis factor-alpha (TNF-α), induced nuclear translocation of TTF-1 and increased binding affinity of TTF-1 to its binding sites on the HO-1 gene. HO-1 mRNA increased with TTF-1 overexpression but decreased with RNA interference of TTF-1 expression in rat astroglial C6 cells. Together with results showing involvement of TTF-1 in the TNF-α-induced increase in interleukin 1 beta and monocyte chemotactic protein 1 production, this study suggests that TTF-1 plays an important role in the mouse hypothalamus TNF-α-induced inflammatory response for regulating HO-1 gene expression.


Assuntos
Regulação da Expressão Gênica/fisiologia , Heme Oxigenase-1/metabolismo , Hipotálamo/metabolismo , Proteínas de Membrana/metabolismo , Fator Nuclear 1 de Tireoide/metabolismo , Ativação Transcricional/fisiologia , Animais , Linhagem Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos
10.
J Comp Neurol ; 526(3): 397-411, 2018 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-28921616

RESUMO

In mammals, thalamic axons are guided internally toward their neocortical target by corridor (Co) neurons that act as axonal guideposts. The existence of Co-like neurons in non-mammalian species, in which thalamic axons do not grow internally, raised the possibility that Co cells might have an ancestral role. Here, we investigated the contribution of corridor (Co) cells to mature brain circuits using a combination of genetic fate-mapping and assays in mice. We unexpectedly found that Co neurons contribute to striatal-like projection neurons in the central extended amygdala. In particular, Co-like neurons participate in specific nuclei of the bed nucleus of the stria terminalis, which plays essential roles in anxiety circuits. Our study shows that Co neurons possess an evolutionary conserved role in anxiety circuits independently from an acquired guidepost function. It furthermore highlights that neurons can have multiple sequential functions during brain wiring and supports a general role of tangential migration in the building of subpallial circuits.


Assuntos
Vias Aferentes/fisiologia , Orientação de Axônios/genética , Movimento Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Tegmento Pontino , Tálamo , Animais , Animais Recém-Nascidos , Toxina da Cólera/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Embrião de Mamíferos , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tegmento Pontino/citologia , Tegmento Pontino/embriologia , Tegmento Pontino/crescimento & desenvolvimento , Gravidez , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Tálamo/citologia , Tálamo/embriologia , Tálamo/crescimento & desenvolvimento , Fator Nuclear 1 de Tireoide/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Environ Toxicol ; 33(2): 209-219, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29139221

RESUMO

Perchlorate is a widespread endocrine disruptor that was previously correlated with increased serum TSH levels and decreased thyroid hormones production both in animals and humans. Even so, the regulation of gene/protein expression in the hypothalamus, pituitary and thyroid by chronic perchlorate exposure was not completely elucidated. Therefore, this study aimed to investigate the underlying mechanisms involved in the disruption of hypothalamus-pituitary-thyroid axis by chronic perchlorate exposure. Male Wistar rats were treated or not with NaClO4 in the drinking water (35 mg/Kg/day) for 60 days. Thereafter, hormone/cytokines serum levels were measured through multiplex assays; genes/proteins expression were investigated by qPCR/Western Blotting and thyroid morphology was evaluated through histological analysis. Serum TSH levels were increased and serum T4 /T3 levels were decreased in perchlorate-treated animals. This treatment also altered the thyrotropin-releasing hormone mRNA/protein content in the hypothalamus. Additionally, the expression of both subunits of TSH were increased in the pituitary of perchlorate-treated rats, which also presented significant alterations in the thyroid morphology/gene expression. Furthermore, perchlorate exposure reduced liver Dio1 mRNA expression and increased the content of pro-inflammatory cytokines in the thyroid and the serum. In conclusion, our study adds novel findings about the perchlorate-induced disruption of the hypothalamus-pituitary-thyroid axis gene/protein expression in male rats. The data presented herein also suggest that perchlorate induces thyroid and systemic inflammation through the increased production of cytokines. Taken together, our results suggest that perchlorate contamination should be monitored, especially in the individuals most susceptible to the deleterious effects of reduced levels of thyroid hormones.


Assuntos
Disruptores Endócrinos/toxicidade , Hipotálamo/efeitos dos fármacos , Percloratos/toxicidade , Hipófise/efeitos dos fármacos , Compostos de Sódio/toxicidade , Glândula Tireoide/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Fator de Transcrição PAX8/metabolismo , Hipófise/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia , Hormônios Tireóideos/sangue , Fator Nuclear 1 de Tireoide/metabolismo , Hormônio Liberador de Tireotropina/genética , Hormônio Liberador de Tireotropina/metabolismo , Tiroxina/sangue , Tri-Iodotironina/sangue
12.
Respir Med ; 129: 16-23, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28732825

RESUMO

We retrospectively studied the clinical presentation, treatment modalities and outcome in 16 patients with heterozygous NKX2-1 mutation associated with chronic lung disease. Twelve different NKX2-1 mutations, including 4 novel mutations, were identified in the 16 patients. Nine patients presented with brain-lung-thyroid syndrome, 3 had neurological and lung symptoms and 4 had only pulmonary symptoms. Ten patients had neonatal respiratory distress, and 6 of them developed infiltrative lung disease (ILD). The other patients were diagnosed with ILD in childhood (n = 3) or in adulthood (n = 3). The median age at diagnosis was 36 months (IQ 3.5-95). Patient testing included HRCT (n = 13), BALF analysis (n = 6), lung biopsies (n = 3) and lung function tests (n = 6). Six patients required supplemental oxygen support with a median duration of 18 months (IQ 2.5-29). All symptomatic ILD patients (n = 12) benefited from a treatment consisting of steroids, azithromycin (n = 9), and/or hydroxychloroquine (n = 4). The median follow-up was 36 months (IQ 24-71.5). One patient died of respiratory failure at 18 months and another is waiting for lung transplantation. In summary, the initial diagnosis was based on clinical presentation and radiological features, but the presentation was heterogeneous. Definitive diagnosis required genetic analysis, which should be performed, even in absence of neurological or thyroid symptoms.


Assuntos
Doenças Pulmonares Intersticiais/genética , Pneumopatias/genética , Pneumopatias/patologia , Proteinose Alveolar Pulmonar/genética , Proteína B Associada a Surfactante Pulmonar/deficiência , Fator Nuclear 1 de Tireoide/genética , Adolescente , Adulto , Atetose/complicações , Atetose/genética , Atetose/patologia , Líquido da Lavagem Broncoalveolar/química , Criança , Coreia/complicações , Coreia/genética , Coreia/patologia , Hipotireoidismo Congênito/complicações , Hipotireoidismo Congênito/genética , Hipotireoidismo Congênito/patologia , Feminino , França/epidemiologia , Genes Homeobox , Humanos , Pneumopatias/complicações , Pneumopatias/terapia , Doenças Pulmonares Intersticiais/complicações , Doenças Pulmonares Intersticiais/diagnóstico por imagem , Doenças Pulmonares Intersticiais/fisiopatologia , Doenças Pulmonares Intersticiais/terapia , Masculino , Mutação , Prognóstico , Proteinose Alveolar Pulmonar/complicações , Proteína B Associada a Surfactante Pulmonar/genética , Surfactantes Pulmonares/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/complicações , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Testes de Função Respiratória/métodos , Estudos Retrospectivos , Tomografia Computadorizada por Raios X , Resultado do Tratamento
13.
Sci Rep ; 7: 44444, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28294152

RESUMO

Melanocortin-3 receptors (MC3R) have a contextual role in appetite control that is amplified with hypocaloric conditioning. C57BL/6J (B6) mice subjected to hypocaloric feeding schedules (HFS) exhibit compulsive behavioral responses involving food anticipatory activity (FAA) and caloric loading following food access. These homeostatic responses to calorie-poor environs are attenuated in B6 mice in which Mc3r transcription is suppressed by a lox-stop-lox sequence in the 5'UTR (Mc3rTB/TB). Here, we report that optimization of caloric loading in B6 mice subject to HFS, characterized by increased meal size and duration, is not observed in Mc3rTB/TB mice. Analysis of hypothalamic and neuroendocrine responses to HFS throughout the light-dark cycle suggests uncoupling of hypothalamic responses involving appetite-stimulating fasting-responsive hypothalamic neurons expressing agouti-related peptide (AgRP) and neuropeptide Y (Npy). Rescuing Mc3rs expression in Nkx2.1(+ve) neurons is sufficient to restore normal hypothalamic responses to negative energy balance. In addition, Mc3rs expressed in Nkx2.1(+ve) neurons are also sufficient to restore FAA and caloric loading of B6 mice subjected to HFS. In summary, MC3Rs expressed in Nkx2.1(+ve) neurons are sufficient to coordinate hypothalamic response and expression of compulsive behavioral responses involving meal anticipation and consumption of large meals during situations of prolonged negative energy balance.


Assuntos
Proteína Relacionada com Agouti/genética , Metabolismo Energético/genética , Neuropeptídeo Y/genética , Receptor Tipo 3 de Melanocortina/genética , Animais , Apetite/genética , Ingestão de Energia/genética , Homeostase , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo , Fotoperíodo , Fator Nuclear 1 de Tireoide/genética
14.
Artigo em Inglês | MEDLINE | ID: mdl-28054989

RESUMO

Di-(2-ethylhexyl)phthalate (DEHP) was a widely used chemical with human toxicity. Recent in vivo and in vitro studies suggested that DEHP-exposure may be associated with altered serum thyroid hormones (THs) levels, but the underlying molecular mechanisms were largely unknown. To explore the possible molecular mechanisms, 128 Wistar rats were dosed with DEHP by gavage at 0, 150, 300, and 600 mg/kg/day for 3 months (M) and 6 M, respectively. After exposure, expression of genes and proteins in the thyroid, pituitary, and hypothalamus tissues of rats were analyzed by Q-PCR and western blot, while the sera and urine samples were assayed by radioimmunoassay and ELISA. Results showed that serum THs levels were suppressed by DEHP on the whole. DEHP treatment influenced the levels of rats' thyrotropin releasing hormone receptor (TRHr), Deiodinases 1 (D1), thyroid stimulating hormone beta (TSHß), sodium iodide symporter (NIS), thyroid stimulating hormone receptor (TSHr), thyroperoxidase (TPO), thyroid transcription factor 1 (TTF-1), and thyroglobulin (TG) mRNA/protein expression in the hypothalamus-pituitary-thyroid (HPT) axis and decreased urine iodine. Taken together, observed findings indicate that DEHP could reduce thyroid hormones via disturbing the HPT axis, and the activated TSH/TSHR pathway is required to regulate thyroid function via altering TRHr, TSHß, NIS, TSHr, TPO, TTF-1 and TG mRNA/protein expression of the HPT axis.


Assuntos
Dietilexilftalato/farmacologia , Hipotálamo/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Glândula Tireoide/efeitos dos fármacos , Hormônios Tireóideos/metabolismo , Animais , Autoantígenos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Iodeto Peroxidase/efeitos dos fármacos , Proteínas de Ligação ao Ferro/efeitos dos fármacos , Proteínas Nucleares/efeitos dos fármacos , Hormônios Hipofisários/metabolismo , Ratos , Ratos Wistar , Fator Nuclear 1 de Tireoide , Tireotropina/metabolismo , Fatores de Transcrição/efeitos dos fármacos
15.
PLoS One ; 11(5): e0155762, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27223697

RESUMO

Di-(2-ethylhexyl) phthalate (DEHP) has the potential to disrupt the thyroid endocrine system, but the underlying mechanism is unknown. In this study, zebrafish (Danio rerio) embryos were exposed to different concentrations of DEHP (0, 40, 100, 200, 400 µg/L) from 2 to 168 hours post fertilization (hpf). Thyroid hormones (THs) levels and transcriptional profiling of key genes related to hypothalamus-pituitary-thyroid (HPT) axis were examined. The result of whole-body thyroxine (T4) and triiodothyronine (T3) indicated that the thyroid hormone homeostasis was disrupted by DEHP in the zebrafish larvae. After exposure to DEHP, the mRNA expressions of thyroid stimulating hormone (tshß) and corticotrophin releasing hormone (crh) genes were increased in a concentration dependent manner, respectively. The expression level of genes involved in thyroid development (nkx2.1 and pax8) and thyroid synthesis (sodium/iodide symporter, nis, thyroglobulin, tg) were also measured. The transcripts of nkx2.1 and tg were significantly increased after DEHP exposure, while those of nis and pax8 had no significant change. Down-regulation of uridinediphosphate-glucuronosyl-transferase (ugt1ab) and up-regulation of thyronine deiodinase (dio2) might change the THs levels. In addition, the transcript of transthyretin (ttr) was up-regulated, while the mRNA levels of thyroid hormone receptors (trα and trß) remained unchanged. All the results demonstrated that exposure to DEHP altered the whole-body thyroid hormones in the zebrafish larvae and changed the expression profiling of key genes related to HPT axis, proving that DEHP induced the thyroid endocrine toxicity and potentially affected the synthesis, regulation and action of thyroid hormones.


Assuntos
Dietilexilftalato/toxicidade , Hipotálamo/embriologia , Hipófise/embriologia , Glândula Tireoide/embriologia , Peixe-Zebra/embriologia , Animais , Hormônio Liberador da Corticotropina/sangue , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas Nucleares/biossíntese , Fator de Transcrição PAX8/biossíntese , Simportadores/biossíntese , Tireoglobulina/biossíntese , Fator Nuclear 1 de Tireoide , Tireotropina/sangue , Tiroxina/sangue , Fatores de Transcrição/biossíntese , Tri-Iodotironina/sangue , Proteínas de Peixe-Zebra/biossíntese
16.
Mol Endocrinol ; 30(5): 494-503, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27003443

RESUMO

Brain-derived neurotrophic factor (BDNF) expressed in the paraventricular hypothalamus (PVH) has been shown to play a key role in regulating energy intake and energy expenditure. BDNF is also expressed in other hypothalamic nuclei; however, the role in the control of energy balance for BDNF produced in these structures remains largely unknown. We found that deleting the Bdnf gene in the ventromedial hypothalamus (VMH) during embryogenesis using the Sf1-Cre transgene had no effect on body weight in mice. In contrast, deleting the Bdnf gene in the adult VMH using Cre-expressing virus led to significant hyperphagia and obesity. These observations indicate that the lack of a hyperphagia phenotype in the Sf1-Cre/Bdnf mutant mice is likely due to developmental compensation. To investigate the role of BDNF expressed in other hypothalamic areas, we employed the hypothalamus-specific Nkx2.1-Cre transgene to delete the Bdnf gene. We found that the Nkx2.1-Cre transgene could abolish BDNF expression in many hypothalamic nuclei, but not in the PVH, and that the resulting mutant mice developed modest obesity due to reduced energy expenditure. Thus, BDNF produced in the VMH plays a role in regulating energy intake. Furthermore, BDNF expressed in hypothalamic areas other than PVH and VMH is also involved in the control of energy expenditure.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Peso Corporal/fisiologia , Ingestão de Energia/fisiologia , Hiperfagia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Fatores de Processamento de RNA/metabolismo , Fator Nuclear 1 de Tireoide/metabolismo , Transgenes/fisiologia
17.
Endocrinology ; 157(3): 1211-21, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26779746

RESUMO

The hypothalamic-pituitary-thyroid axis is governed by hypophysiotropic TRH-synthesizing neurons located in the hypothalamic paraventricular nucleus under control of the negative feedback of thyroid hormones. The mechanisms underlying the ontogeny of this phenomenon are poorly understood. We aimed to determine the onset of thyroid hormone-mediated hypothalamic-negative feedback and studied how local hypothalamic metabolism of thyroid hormones could contribute to this process in developing chicken. In situ hybridization revealed that whereas exogenous T4 did not induce a statistically significant inhibition of TRH expression in the paraventricular nucleus at embryonic day (E)19, T4 treatment was effective at 2 days after hatching (P2). In contrast, TRH expression responded to T3 treatment in both age groups. TSHß mRNA expression in the pituitary responded to T4 in a similar age-dependent manner. Type 2 deiodinase (D2) was expressed from E13 in tanycytes of the mediobasal hypothalamus, and its activity increased between E15 and P2 both in the mediobasal hypothalamus and in tanycyte-lacking hypothalamic regions. Nkx2.1 was coexpressed with D2 in E13 and P2 tanycytes and transcription of the cdio2 gene responded to Nkx2.1 in U87 glioma cells, indicating its potential role in the developmental regulation of D2 activity. The T3-degrading D3 enzyme was also detected in tanycytes, but its level was not markedly changed before and after the period of negative feedback acquisition. These findings suggest that increasing the D2-mediated T3 generation during E18-P2 could provide the sufficient local T3 concentration required for the onset of T3-dependent negative feedback in the developing chicken hypothalamus.


Assuntos
Retroalimentação Fisiológica/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Sistema Hipotálamo-Hipofisário/metabolismo , Iodeto Peroxidase/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , RNA Mensageiro/metabolismo , Glândula Tireoide/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Tiroxina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/metabolismo , Linhagem Celular Tumoral , Embrião de Galinha , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Sistema Hipotálamo-Hipofisário/embriologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/embriologia , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Iodeto Peroxidase/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/embriologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , RNA Mensageiro/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator Nuclear 1 de Tireoide , Tireotropina Subunidade beta/genética , Tiroxina/farmacologia , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Tri-Iodotironina/efeitos dos fármacos , Tri-Iodotironina/metabolismo , Iodotironina Desiodinase Tipo II
18.
PLoS One ; 10(12): e0144459, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26640945

RESUMO

Paternally Expressed Gene 3 (Peg3) is an imprinted gene that controls milk letdown and maternal-caring behaviors. In this study, a conditional knockout allele has been developed in Mus musculus to further characterize these known functions of Peg3 in a tissue-specific manner. The mutant line was first crossed with a germline Cre. The progeny of this cross displayed growth retardation phenotypes. This is consistent with those seen in the previous mutant lines of Peg3, confirming the usefulness of the new mutant allele. The mutant line was subsequently crossed individually with MMTV- and Nkx2.1-Cre lines to test Peg3's roles in the mammary gland and hypothalamus, respectively. According to the results, the milk letdown process was impaired in the nursing females with the Peg3 mutation in the mammary gland, but not in the hypothalamus. This suggests that Peg3's roles in the milk letdown process are more critical in the mammary gland than in the hypothalamus. In contrast, one of the maternal-caring behaviors, nest-building, was interrupted in the females with the mutation in both MMTV- and Nkx2.1-driven lines. Overall, this is the first study to introduce a conditional knockout allele of Peg3 and to further dissect its contribution to mammalian reproduction in a tissue-specific manner.


Assuntos
Comportamento Animal/fisiologia , Fatores de Transcrição Kruppel-Like/genética , Lactação/genética , Glândulas Mamárias Animais/citologia , Comportamento Materno/fisiologia , Animais , Feminino , Técnicas de Inativação de Genes , Hipotálamo/citologia , Hipotálamo/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Lactação/fisiologia , Masculino , Glândulas Mamárias Animais/fisiologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Proteínas Nucleares/genética , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética
19.
J Clin Invest ; 125(2): 796-808, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25555215

RESUMO

The hypothalamus is the central regulator of systemic energy homeostasis, and its dysfunction can result in extreme body weight alterations. Insights into the complex cellular physiology of this region are critical to the understanding of obesity pathogenesis; however, human hypothalamic cells are largely inaccessible for direct study. Here, we developed a protocol for efficient generation of hypothalamic neurons from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) obtained from patients with monogenetic forms of obesity. Combined early activation of sonic hedgehog signaling followed by timed NOTCH inhibition in human ESCs/iPSCs resulted in efficient conversion into hypothalamic NKX2.1+ precursors. Application of a NOTCH inhibitor and brain-derived neurotrophic factor (BDNF) further directed the cells into arcuate nucleus hypothalamic-like neurons that express hypothalamic neuron markers proopiomelanocortin (POMC), neuropeptide Y (NPY), agouti-related peptide (AGRP), somatostatin, and dopamine. These hypothalamic-like neurons accounted for over 90% of differentiated cells and exhibited transcriptional profiles defined by a hypothalamic-specific gene expression signature that lacked pituitary markers. Importantly, these cells displayed hypothalamic neuron characteristics, including production and secretion of neuropeptides and increased p-AKT and p-STAT3 in response to insulin and leptin. Our results suggest that these hypothalamic-like neurons have potential for further investigation of the neurophysiology of body weight regulation and evaluation of therapeutic targets for obesity.


Assuntos
Diferenciação Celular , Hipotálamo/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios , Obesidade/metabolismo , Antígenos de Diferenciação/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Proteínas Hedgehog/metabolismo , Humanos , Hipotálamo/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Proteínas Nucleares/metabolismo , Obesidade/patologia , Pró-Opiomelanocortina/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/metabolismo
20.
Diabetes ; 63(5): 1572-82, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24487022

RESUMO

Transcription factor forkhead box O1 (FoxO1) regulates energy expenditure (EE), food intake, and hepatic glucose production. These activities have been mapped to specific hypothalamic neuronal populations using cell type-specific knockout experiments in mice. To parse out the integrated output of FoxO1-dependent transcription from different neuronal populations and multiple hypothalamic regions, we used transgenic mice expressing Cre recombinase from the Nkx2.1 promoter to ablate loxP-flanked Foxo1 alleles from a majority of hypothalamic neurons (Foxo1KO(Nkx2.1) mice). This strategy resulted in the expected inhibition of FoxO1 expression, but only produced a transient reduction of body weight as well as a decreased body length. The transient decrease of body weight in male mice was accompanied by decreased fat mass. Male Foxo1KO(Nkx2.1) mice show food intake similar to that in wild-type controls, and, although female knockout mice eat less, they do so in proportion to a reduced body size. EE is unaffected in Foxo1KO(Nkx2.1) mice, although small increases in body temperature are present. Unlike other neuron-specific Foxo1 knockout mice, Foxo1KO(Nkx2.1) mice are not protected from diet-induced obesity. These studies indicate that, unlike the metabolic effects of highly restricted neuronal subsets (proopiomelanocortin, neuropeptide Y/agouti-related peptide, and steroidogenic factor 1), those of neurons derived from the Nkx2.1 lineage either occur in a FoxO1-independent fashion or are compensated for through developmental plasticity.


Assuntos
Metabolismo Energético/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Peso Corporal/fisiologia , Linhagem da Célula/fisiologia , Ingestão de Alimentos/fisiologia , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Masculino , Camundongos , Camundongos Transgênicos , Neuropeptídeo Y/metabolismo , Proteínas Nucleares/genética , Obesidade/genética , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais/fisiologia , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA