Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Neuroendocrinol ; 34(12): e13217, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36458331

RESUMO

Prader-Willi syndrome (PWS) is a neurodevelopmental disorder characterized by hyperphagia, obesity, developmental delay and intellectual disability. Studies suggest dysfunctional signaling of the neuropeptide oxytocin as one of the key mechanisms in PWS, and administration of oxytocin via intranasal or systemic routes yielded promising results in both humans and mouse models. However, a detailed assessment of the oxytocin system in mouse models of PWS such as the Magel2-deficient Magel2tm1.Stw mouse, is lacking. In the present study, we performed an automated counting of oxytocin cells in the entire paraventricular nucleus of the hypothalamus of Magel2tm1.Stw and wild-type control mice and found a significant reduction in the caudal part, which represents the parvocellular subdivision. In addition, based on the recent discovery that some astrocytes express the oxytocin receptor (OTR), we performed detailed analysis of astrocyte numbers and morphology in various brain regions, and assessed expression levels of the astrocyte marker glial fibrillary acidic protein, which was significantly decreased in the hypothalamus, but not other brain regions in Magel2tm1.Stw mice. Finally, we analyzed the number of OTR-expressing astrocytes in various brain regions and found a significant reduction in the nucleus accumbens of Magel2tm1.Stw mice, as well as a sex-specific difference in the lateral septum. This study suggests a role for caudal paraventricular nucleus oxytocin neurons as well as OTR-expressing astrocytes in a mouse model of PWS, provides novel information about sex-specific expression of astrocytic OTRs, and presents several new brain regions containing OTR-expressing astrocytes in the mouse brain.


Assuntos
Astrócitos , Hipotálamo , Neuropeptídeos , Ocitocina , Síndrome de Prader-Willi , Animais , Feminino , Masculino , Camundongos , Astrócitos/metabolismo , Modelos Animais de Doenças , Hipotálamo/metabolismo , Neuropeptídeos/metabolismo , Ocitocina/metabolismo , Síndrome de Prader-Willi/metabolismo , Receptores de Ocitocina/metabolismo
2.
J Neuroendocrinol ; 33(7): e12994, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34156126

RESUMO

Prader-Willi Syndrome (PWS) is a rare and incurable congenital neurodevelopmental disorder, resulting from the absence of expression of a group of genes on the paternally acquired chromosome 15q11-q13. Phenotypical characteristics of PWS include infantile hypotonia, short stature, incomplete pubertal development, hyperphagia and morbid obesity. Hypothalamic dysfunction in controlling body weight and food intake is a hallmark of PWS. Neuroimaging studies have demonstrated that PWS subjects have abnormal neurocircuitry engaged in the hedonic and physiological control of feeding behavior. This is translated into diminished production of hypothalamic effector peptides which are responsible for the coordination of energy homeostasis and satiety. So far, studies with animal models for PWS and with human post-mortem hypothalamic specimens demonstrated changes particularly in the infundibular and the paraventricular nuclei of the hypothalamus, both in orexigenic and anorexigenic neural populations. Moreover, many PWS patients have a severe endocrine dysfunction, e.g. central hypogonadism and/or growth hormone deficiency, which may contribute to the development of increased fat mass, especially if left untreated. Additionally, the role of non-neuronal cells, such as astrocytes and microglia in the hypothalamic dysregulation in PWS is yet to be determined. Notably, microglial activation is persistently present in non-genetic obesity. To what extent microglia, and other glial cells, are affected in PWS is poorly understood. The elucidation of the hypothalamic dysfunction in PWS could prove to be a key feature of rational therapeutic management in this syndrome. This review aims to examine the evidence for hypothalamic dysfunction, both at the neuropeptidergic and circuitry levels, and its correlation with the pathophysiology of PWS.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Rede Nervosa/fisiopatologia , Síndrome de Prader-Willi , Animais , Humanos , Hiperfagia/etiologia , Hiperfagia/metabolismo , Hiperfagia/psicologia , Hipogonadismo/etiologia , Hipogonadismo/metabolismo , Hipogonadismo/psicologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Rede Nervosa/metabolismo , Rede Nervosa/patologia , Neuropeptídeos/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/psicologia , Síndrome de Prader-Willi/complicações , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Síndrome de Prader-Willi/psicologia
3.
Hum Mol Genet ; 30(12): 1101-1110, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-33856031

RESUMO

The smallest genomic region causing Prader-Willi Syndrome (PWS) deletes the non-coding RNA SNORD116 cluster; however, the function of SNORD116 remains a mystery. Previous work in the field revealed the tantalizing possibility that expression of NHLH2, a gene previously implicated in both obesity and hypogonadism, was downregulated in PWS patients and differentiated stem cells. In silico RNA: RNA modeling identified several potential interaction domains between SNORD116 and NHLH2 mRNA. One of these interaction domains was highly conserved in most vertebrate NHLH2 mRNAs examined. A construct containing the Nhlh2 mRNA, including its 3'-UTR, linked to a c-myc tag was transfected into a hypothalamic neuron cell line in the presence and absence of exogenously-expressed Snord116. Nhlh2 mRNA expression was upregulated in the presence of Snord116 dependent on the length and type of 3'UTR used on the construct. Furthermore, use of actinomycin D to stop new transcription in N29/2 cells demonstrated that the upregulation occurred through increased stability of the Nhlh2 mRNA in the 45 minutes immediately following transcription. In silico modeling also revealed that a single nucleotide variant (SNV) in the NHLH2 mRNA could reduce the predicted interaction strength of the NHLH2:SNORD116 diad. Indeed, use of an Nhlh2 mRNA construct containing this SNV significantly reduces the ability of Snord116 to increase Nhlh2 mRNA levels. For the first time, these data identify a motif and mechanism for SNORD116-mediated regulation of NHLH2, clarifying the mechanism by which deletion of the SNORD116 snoRNAs locus leads to PWS phenotypes.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Síndrome de Prader-Willi/genética , Proteínas Proto-Oncogênicas c-myc/genética , RNA Nucleolar Pequeno/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Processamento Pós-Transcricional do RNA/genética , Estabilidade de RNA/genética
4.
Nutrients ; 12(10)2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-33066107

RESUMO

Prader-Willi syndrome (PWS) is a rare genetic disorder characterized by a wide range of clinical manifestations, including obesity, hyperphagia, and behavioral problems. Bifidobacterium animalis subsp. lactis strain BPL1 has been shown to improve central adiposity in adults with simple obesity. To evaluate BPL1's effects in children with PWS, we performed a randomized crossover trial among 39 patients (mean age 10.4 years). Participants were randomized to placebo-BPL1 (n = 19) or BPL1-placebo (n = 20) sequences and underwent a 12-week period with placebo/BPL1 treatments, a 12-week washout period, and a 12-week period with the crossover treatment. Thirty-five subjects completed the study. The main outcome was changes in adiposity, measured by dual-energy X-ray absorptiometry. Secondary outcomes included lipid and glucose metabolism, hyperphagia, and mental health symptoms. Generalized linear modeling was applied to assess differences between treatments. While BPL1 did not modify total fat mass compared to placebo, BPL1 decreased abdominal adiposity in a subgroup of patients older than 4.5 years (n = 28). BPL1 improved fasting insulin concentration and insulin sensitivity. Furthermore, we observed modest improvements in some mental health symptoms. A follow-up trial with a longer treatment period is warranted to determine whether BPL1 supplementation can provide a long-term therapeutic approach for children with PWS (ClinicalTrials.gov NCT03548480).


Assuntos
Adiposidade , Bifidobacterium animalis , Fenômenos Fisiológicos da Nutrição Infantil/fisiologia , Suplementos Nutricionais , Síndrome de Prader-Willi/dietoterapia , Síndrome de Prader-Willi/metabolismo , Probióticos/administração & dosagem , Adolescente , Criança , Comportamento Infantil , Pré-Escolar , Estudos Cross-Over , Feminino , Glucose/metabolismo , Humanos , Resistência à Insulina , Metabolismo dos Lipídeos , Masculino , Síndrome de Prader-Willi/psicologia , Resultado do Tratamento
5.
JCI Insight ; 5(12)2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32365348

RESUMO

Imprinted genes are highly expressed in the hypothalamus; however, whether specific imprinted genes affect hypothalamic neuromodulators and their functions is unknown. It has been suggested that Prader-Willi syndrome (PWS), a neurodevelopmental disorder caused by lack of paternal expression at chromosome 15q11-q13, is characterized by hypothalamic insufficiency. Here, we investigate the role of the paternally expressed Snord116 gene within the context of sleep and metabolic abnormalities of PWS, and we report a significant role of this imprinted gene in the function and organization of the 2 main neuromodulatory systems of the lateral hypothalamus (LH) - namely, the orexin (OX) and melanin concentrating hormone (MCH) - systems. We observed that the dynamics between neuronal discharge in the LH and the sleep-wake states of mice with paternal deletion of Snord116 (PWScrm+/p-) are compromised. This abnormal state-dependent neuronal activity is paralleled by a significant reduction in OX neurons in the LH of mutant mice. Therefore, we propose that an imbalance between OX- and MCH-expressing neurons in the LH of mutant mice reflects a series of deficits manifested in the PWS, such as dysregulation of rapid eye movement (REM) sleep, food intake, and temperature control.


Assuntos
Comportamento Animal/fisiologia , Região Hipotalâmica Lateral/metabolismo , Hipotálamo/metabolismo , Orexinas/metabolismo , RNA Nucleolar Pequeno/genética , Sono/fisiologia , Animais , Modelos Animais de Doenças , Comportamento Alimentar , Região Hipotalâmica Lateral/fisiopatologia , Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Camundongos , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatologia
6.
Nutrients ; 12(4)2020 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-32316673

RESUMO

Prader-Willi syndrome (PWS) is a genetic disorder characterized by hyperphagia with progressive, severe obesity, and an increased risk of obesity-related comorbidities in adult life. Although low dietary vitamin D intake and low 25-hydroxy vitamin D (25OHD) levels are commonly reported in PWS in the context of bone metabolism, the association of low 25OHD levels with fat mass has not been extensively evaluated in PWS adults. The aims of this study were to investigate the following in PWS adults: (1) 25OHD levels and the dietary vitamin D intake; (2) associations among 25OHD levels with anthropometric measurements and fat mass; (3) specific cut-off values for body mass index (BMI) and fat mass predictive of the 25OHD levels. In this cross-sectional, single-center study we enrolled 30 participants, 15 PWS adults (age 19-41 years and 40% males) and 15 control subjects matched by age, sex, and BMI from the same geographical area (latitude 40° 49' N; elevation 17 m). Fat mass was assessed using a bioelectrical impedance analysis (BIA) phase-sensitive system. The 25OHD levels were determined by a direct competitive chemiluminescence immunoassay. Dietary vitamin D intake data was collected by three-day food records. The 25OHD levels in the PWS adults were constantly lower across all categories of BMI and fat mass compared with their obese counterpart. The 25OHD levels were negatively associated with BMI (p = 0.04), waist circumference (p = 0.03), fat mass (p = 0.04), and dietary vitamin D intake (p < 0.001). During multiple regression analysis, dietary vitamin D intake was entered at the first step (p < 0.001), thus explaining 84% of 25OHD level variability. The threshold values of BMI and fat mass predicting the lowest decrease in the 25OHD levels were found at BMI ≥ 42 kg/m2 (p = 0.01) and fat mass ≥ 42 Kg (p = 0.003). In conclusion, our data indicate that: (i) 25OHD levels and dietary vitamin D intake were lower in PWS adults than in the control, independent of body fat differences; (ii) 25OHD levels were inversely associated with BMI, waist circumference, and fat mass, but low dietary vitamin D intake was the major determinant of low vitamin D status in these patients; (iii) sample-specific cut-off values of BMI and fat mass might help to predict risks of the lowest 25OHD level decreases in PWS adults. The presence of trained nutritionists in the integrated care teams of PWS adults is strongly suggested in order to provide an accurate nutritional assessment and tailored vitamin D supplementations.


Assuntos
Tecido Adiposo/metabolismo , Suplementos Nutricionais , Ingestão de Alimentos , Fenômenos Fisiológicos da Nutrição/fisiologia , Síndrome de Prader-Willi/sangue , Síndrome de Prader-Willi/metabolismo , Deficiência de Vitamina D , Vitamina D/análogos & derivados , Vitamina D/administração & dosagem , Adulto , Índice de Massa Corporal , Estudos Transversais , Feminino , Humanos , Masculino , Avaliação Nutricional , Risco , Vitamina D/sangue , Deficiência de Vitamina D/etiologia , Deficiência de Vitamina D/terapia , Circunferência da Cintura , Adulto Jovem
7.
Nat Commun ; 9(1): 1616, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29691382

RESUMO

Rhythmic oscillations of physiological processes depend on integrating the circadian clock and diurnal environment. DNA methylation is epigenetically responsive to daily rhythms, as a subset of CpG dinucleotides in brain exhibit diurnal rhythmic methylation. Here, we show a major genetic effect on rhythmic methylation in a mouse Snord116 deletion model of the imprinted disorder Prader-Willi syndrome (PWS). More than 23,000 diurnally rhythmic CpGs are identified in wild-type cortex, with nearly all lost or phase-shifted in PWS. Circadian dysregulation of a second imprinted Snord cluster at the Temple/Kagami-Ogata syndrome locus is observed at the level of methylation, transcription, and chromatin, providing mechanistic evidence of cross-talk. Genes identified by diurnal epigenetic changes in PWS mice overlapped rhythmic and PWS-specific genes in human brain and are enriched for PWS-relevant phenotypes and pathways. These results support the proposed evolutionary relationship between imprinting and sleep, and suggest possible chronotherapy in the treatment of PWS and related disorders.


Assuntos
Encéfalo/fisiologia , Córtex Cerebral/metabolismo , Ritmo Circadiano , Síndrome de Prader-Willi/genética , RNA Nucleolar Pequeno/genética , RNA Nucleolar Pequeno/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Metilação de DNA , Feminino , Deleção de Genes , Humanos , Masculino , Camundongos , Síndrome de Prader-Willi/metabolismo
8.
Cell Rep ; 22(13): 3401-3408, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590610

RESUMO

Transcriptional analysis of brain tissue from people with molecularly defined causes of obesity may highlight disease mechanisms and therapeutic targets. We performed RNA sequencing of hypothalamus from individuals with Prader-Willi syndrome (PWS), a genetic obesity syndrome characterized by severe hyperphagia. We found that upregulated genes overlap with the transcriptome of mouse Agrp neurons that signal hunger, while downregulated genes overlap with the expression profile of Pomc neurons activated by feeding. Downregulated genes are expressed mainly in neuronal cells and contribute to neurogenesis, neurotransmitter release, and synaptic plasticity, while upregulated, predominantly microglial genes are involved in inflammatory responses. This transcriptional signature may be mediated by reduced brain-derived neurotrophic factor expression. Additionally, we implicate disruption of alternative splicing as a potential molecular mechanism underlying neuronal dysfunction in PWS. Transcriptomic analysis of the human hypothalamus may identify neural mechanisms involved in energy homeostasis and potential therapeutic targets for weight loss.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Jejum/fisiologia , Hipotálamo/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Camundongos , Obesidade/metabolismo , Síndrome de Prader-Willi/patologia , Transcriptoma
9.
J Clin Invest ; 128(3): 960-969, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29376887

RESUMO

Profound hyperphagia is a major disabling feature of Prader-Willi syndrome (PWS). Characterization of the mechanisms that underlie PWS-associated hyperphagia has been slowed by the paucity of animal models with increased food intake or obesity. Mice with a microdeletion encompassing the Snord116 cluster of noncoding RNAs encoded within the Prader-Willi minimal deletion critical region have previously been reported to show growth retardation and hyperphagia. Here, consistent with previous reports, we observed growth retardation in Snord116+/-P mice with a congenital paternal Snord116 deletion. However, these mice neither displayed increased food intake nor had reduced hypothalamic expression of the proprotein convertase 1 gene PCSK1 or its upstream regulator NHLH2, which have recently been suggested to be key mediators of PWS pathogenesis. Specifically, we disrupted Snord116 expression in the mediobasal hypothalamus in Snord116fl mice via bilateral stereotaxic injections of a Cre-expressing adeno-associated virus (AAV). While the Cre-injected mice had no change in measured energy expenditure, they became hyperphagic between 9 and 10 weeks after injection, with a subset of animals developing marked obesity. In conclusion, we show that selective disruption of Snord116 expression in the mediobasal hypothalamus models the hyperphagia of PWS.


Assuntos
Hiperfagia/metabolismo , Hipotálamo/metabolismo , Síndrome de Prader-Willi/genética , RNA Nucleolar Pequeno/genética , Animais , Composição Corporal , Dependovirus , Modelos Animais de Doenças , Deleção de Genes , Genótipo , Hiperfagia/genética , Masculino , Camundongos , Camundongos Transgênicos , Obesidade/metabolismo , Síndrome de Prader-Willi/metabolismo
10.
Hum Mol Genet ; 26(21): 4215-4230, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973533

RESUMO

In Prader-Willi syndrome (PWS), obesity is caused by the disruption of appetite-controlling pathways in the brain. Two PWS candidate genes encode MAGEL2 and necdin, related melanoma antigen proteins that assemble into ubiquitination complexes. Mice lacking Magel2 are obese and lack leptin sensitivity in hypothalamic pro-opiomelanocortin neurons, suggesting dysregulation of leptin receptor (LepR) activity. Hypothalamus from Magel2-null mice had less LepR and altered levels of ubiquitin pathway proteins that regulate LepR processing (Rnf41, Usp8, and Stam1). MAGEL2 increased the cell surface abundance of LepR and decreased their degradation. LepR interacts with necdin, which interacts with MAGEL2, which complexes with RNF41 and USP8. Mutations in the MAGE homology domain of MAGEL2 suppress RNF41 stabilization and prevent the MAGEL2-mediated increase of cell surface LepR. Thus, MAGEL2 and necdin together control LepR sorting and degradation through a dynamic ubiquitin-dependent pathway. Loss of MAGEL2 and necdin may uncouple LepR from ubiquitination pathways, providing a cellular mechanism for obesity in PWS.


Assuntos
Antígenos de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Síndrome de Prader-Willi/metabolismo , Proteínas/metabolismo , Receptores para Leptina/metabolismo , Animais , Antígenos de Neoplasias/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Endopeptidases/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Células HEK293 , Humanos , Hipotálamo/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Leptina/genética , Leptina/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Obesidade/genética , Obesidade/metabolismo , Síndrome de Prader-Willi/genética , Transporte Proteico , Proteínas/genética , Receptores para Leptina/genética , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação
11.
J Neuroendocrinol ; 29(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28094877

RESUMO

The Snord116 gene cluster has been recognised as a critical contributor to the Prader-Willi syndrome (PWS), with mice lacking Snord116 displaying many classical PWS phenotypes, including low postnatal body weight, reduced bone mass and increased food intake. However, these mice do not develop obesity as a result of increased energy expenditure. To understand the physiological function of SNORD116 better and potentially rescue the altered metabolism of Snord116-/- mice, we used an adeno-associated viral (AAV) approach to reintroduce the product of the Snord116 gene into the hypothalamus in Snord116-/- mice at different ages. The results obtained show that mid-hypothalamic re-introduction of SNORD116 in 6-week-old Snord116-/- mice leads to significantly reduced body weight and weight gain, which is associated with elevated energy expenditure. Importantly, when the intervention targets other areas such as the anterior region of the hypothalamus or the reintroduction occurs in older mice, the positive effects on energy expenditure are diminished. These data indicate that the metabolic symptoms of PWS develop gradually and the Snord116 gene plays a critical role during this process. Furthermore, when we investigated the consequences of SNORD116 re-introduction under conditions of thermoneutrality where the mild cold stress influences are avoided, we also observed a significant increase in energy expenditure. In conclusion, the rescue of mid-hypothalamic Snord116 deficiency in young Snord116 germline deletion mice increases energy expenditure, providing fundamental information contributing to potential virus-mediated genetic therapy in PWS.


Assuntos
Metabolismo Energético , Hipotálamo/metabolismo , Síndrome de Prader-Willi/metabolismo , RNA Nucleolar Pequeno/metabolismo , Animais , Peso Corporal , Masculino , Camundongos , Camundongos Knockout , RNA Nucleolar Pequeno/genética
12.
Neuropeptides ; 61: 87-93, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27823858

RESUMO

Germline deletion of the Prader-Willi syndrome (PWS) candidate gene Snord116 in mice leads to some classical symptoms of human PWS, notably reductions in body weight, linear growth and bone mass. However, Snord116 deficient mice (Snord116-/-) do not develop an obese phenotype despite their increased food intake and the underlying mechanism for that is unknown. We tested the phenotypes of germline Snord116-/- as well as neuropeptide Y (NPY) neuron specific Snord116lox/lox/NPYcre/+ mice at 30°C, the thermoneutral temperature of mice, and compared these to previous reports studies conducted at normal room temperature. Snord116-/- mice at 30°C still weighed less than wild type but had increased body weight gain. Importantly, food intake and energy expenditure were no longer different at 30°C, and the reduced bone mass and nasal-anal length observed in Snord116-/- mice at room temperature were also normalized. Mechanistically, the thermoneutral condition led to the correction of the mRNA expression of NPY and pro-opiomelanocortin (POMC), which were both previously observed to be significantly up-regulated at room temperature. Importantly, almost identical phenotypes and NPY/POMC mRNA expression alterations were also observed in Snord116lox/lox/NPYcre/+ mice, which lack the Snord116 gene only in NPY neurons. These data illustrate that mild cold stress is a critical factor preventing the development of obesity in Snord116-/- mice via the NPY system. Our study highlights that the function of Snord116 in the hypothalamus may be to enhance energy expenditure, likely via the NPY system, and also indicates that Snord116 function in mice is strongly dependent on environmental conditions such as cold exposure.


Assuntos
Metabolismo Energético/genética , Homeostase/genética , Neurônios/metabolismo , Síndrome de Prader-Willi/genética , RNA Nucleolar Pequeno/genética , Animais , Peso Corporal/genética , Ingestão de Alimentos/genética , Hipotálamo/metabolismo , Camundongos , Camundongos Knockout , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Síndrome de Prader-Willi/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , RNA Nucleolar Pequeno/metabolismo , Temperatura
13.
Congenit Anom (Kyoto) ; 57(4): 96-103, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28004416

RESUMO

Prader-Will syndrome (PWS) is characterized by hyperphagia, growth hormone deficiency and central hypogonadism caused by the dysfunction of the hypothalamus. Patients with PWS present with methylation abnormalities of the PWS-imprinting control region in chromosome 15q11.2, subject to parent-of-origin-specific methylation and controlling the parent-of-origin-specific expression of other paternally expressed genes flanking the region. In theory, the reversal of hypermethylation in the hypothalamic cells could be a promising strategy for the treatment of PWS patients, since cardinal symptoms of PWS patients are correlated with dysfunction of the hypothalamus. The genome-wide methylation status dramatically changes during the reprograming of somatic cells into induced pluripotent stem cells (iPSCs) and during the in vitro culture of iPSCs. Here, we tested the methylation status of the chromosome 15q11.2 region in iPSCs from a PWS patient using pyrosequencing and a more detailed method of genome-wide DNA methylation profiling to reveal whether iPSCs with a partially unmethylated status for the chromosome 15q11.2 region exhibit global methylation aberrations. As a result, we were able to show that a fully methylated status for chromosome 15q11.2 in a PWS patient could be reversed to a partially unmethylated status in at least some of the PWS-iPSC lines. Genome-wide DNA methylation profiling revealed that the partial unmethylation occurred at differentially methylated regions located in chromosome 15q11.2, but not at other differentially methylated regions associated with genome imprinting. The present data potentially opens a door to cell-based therapy for PWS patients and, possibly, patients with other disorders associated with genomic imprinting.


Assuntos
Sequência de Bases , Epigênese Genética , Genoma Humano , Células-Tronco Pluripotentes Induzidas/metabolismo , Síndrome de Prader-Willi/genética , Deleção de Sequência , Reprogramação Celular , Criança , Cromossomos Humanos Par 15 , Metilação de DNA , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Estudo de Associação Genômica Ampla , Impressão Genômica , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Cultura Primária de Células
14.
Mol Metab ; 5(12): 1187-1199, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27900261

RESUMO

OBJECTIVE: Extreme obesity is a core phenotypic feature of Prader-Willi syndrome (PWS). Among numerous metabolic regulators, the endocannabinoid (eCB) system is critically involved in controlling feeding, body weight, and energy metabolism, and a globally acting cannabinoid-1 receptor (CB1R) blockade reverses obesity both in animals and humans. The first-in-class CB1R antagonist rimonabant proved effective in inducing weight loss in adults with PWS. However, it is no longer available for clinical use because of its centrally mediated, neuropsychiatric, adverse effects. METHODS: We studied eCB 'tone' in individuals with PWS and in the Magel2-null mouse model that recapitulates the major metabolic phenotypes of PWS and determined the efficacy of a peripherally restricted CB1R antagonist, JD5037 in treating obesity in these mice. RESULTS: Individuals with PWS had elevated circulating levels of 2-arachidonoylglycerol and its endogenous precursor and breakdown ligand, arachidonic acid. Increased hypothalamic eCB 'tone', manifested by increased eCBs and upregulated CB1R, was associated with increased fat mass, reduced energy expenditure, and decreased voluntary activity in Magel2-null mice. Daily chronic treatment of obese Magel2-null mice and their littermate wild-type controls with JD5037 (3 mg/kg/d for 28 days) reduced body weight, reversed hyperphagia, and improved metabolic parameters related to their obese phenotype. CONCLUSIONS: Dysregulation of the eCB/CB1R system may contribute to hyperphagia and obesity in Magel2-null mice and in individuals with PWS. Our results demonstrate that treatment with peripherally restricted CB1R antagonists may be an effective strategy for the management of severe obesity in PWS.


Assuntos
Síndrome de Prader-Willi/tratamento farmacológico , Síndrome de Prader-Willi/metabolismo , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Sulfonamidas/farmacologia , Adulto , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Ácidos Araquidônicos/sangue , Peso Corporal/efeitos dos fármacos , Estudos de Casos e Controles , Modelos Animais de Doenças , Endocanabinoides/sangue , Endocanabinoides/metabolismo , Feminino , Glicerídeos/sangue , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Prader-Willi/sangue , Proteínas/genética , Proteínas/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Redução de Peso/efeitos dos fármacos
15.
Gene ; 572(2): 266-73, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26220404

RESUMO

The loss of two gene clusters encoding small nucleolar RNAs, SNORD115 and SNORD116 contribute to Prader-Willi syndrome (PWS), the most common syndromic form of obesity in humans. SNORD115 and SNORD116 are considered to be orphan C/D box snoRNAs (SNORDs) as they do not target rRNAs or snRNAs. SNORD115 exhibits sequence complementarity towards the serotonin receptor 2C, but SNORD116 shows no extended complementarities to known RNAs. To identify molecular targets, we performed genome-wide array analysis after overexpressing SNORD115 and SNORD116 in HEK 293T cells, either alone or together. We found that SNORD116 changes the expression of over 200 genes. SNORD116 mainly changed mRNA expression levels. Surprisingly, we found that SNORD115 changes SNORD116's influence on gene expression. In similar experiments, we compared gene expression in post-mortem hypothalamus between individuals with PWS and aged-matched controls. The synopsis of these experiments resulted in 23 genes whose expression levels were influenced by SNORD116. Together our results indicate that SNORD115 and SNORD116 influence expression levels of multiple genes and modify each other activity.


Assuntos
Regulação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA Nucleolar Pequeno/genética , RNA Nucleolar Pequeno/metabolismo , Células HEK293 , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Família Multigênica , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , RNA Mensageiro/genética
16.
J Mol Endocrinol ; 54(3): R131-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25957321

RESUMO

Pubertal timing is influenced by complex interactions among genetic, nutritional, environmental, and socioeconomic factors. The role of MKRN3, an imprinted gene located in the Prader-Willi syndrome critical region (chromosome 15q11-13), in pubertal initiation was first described in 2013 after the identification of deleterious MKRN3 mutations in five families with central precocious puberty (CPP) using whole-exome sequencing analysis. Since then, additional loss-of-function mutations of MKRN3 have been associated with the inherited premature sexual development phenotype in girls and boys from different ethnic groups. In all of these families, segregation analysis clearly demonstrated autosomal dominant inheritance with complete penetrance, but with exclusive paternal transmission, consistent with the monoallelic expression of MKRN3 (a maternally imprinted gene). Interestingly, the hypothalamic Mkrn3 mRNA expression pattern in mice correlated with a putative inhibitory input on puberty initiation. Indeed, the initiation of puberty depends on a decrease in factors that inhibit the release of GnRH combined with an increase in stimulatory factors. These recent human and animal findings suggest that MKRN3 plays an inhibitory role in the reproductive axis to represent a new pathway in pubertal regulation.


Assuntos
Puberdade/genética , Ribonucleoproteínas/genética , Idade de Início , Sequência de Aminoácidos , Animais , Expressão Gênica , Estudos de Associação Genética , Humanos , Hipotálamo/metabolismo , Dados de Sequência Molecular , Fenótipo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Puberdade Precoce/genética , Puberdade Precoce/metabolismo , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases
17.
Hum Mol Genet ; 24(15): 4276-83, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25926624

RESUMO

Prader-Willi syndrome (PWS) is a multigene disorder associated with neonatal failure to thrive, developmental delay and endocrine abnormalities suggestive of hypothalamic dysfunction. Children with PWS typically develop overt hyperphagia and obesity ∼8 years of age, later than children with other genetic forms of obesity. This suggests a postnatal developmental or degenerative component to PWS-associated obesity. De novo inactivating mutations in one PWS candidate gene, MAGEL2, have been identified in children with features of PWS. Adult mice lacking Magel2 are insensitive to the anorexic effect of leptin treatment, and their hypothalamic pro-opiomelanocortin (POMC) neurons fail to depolarize in response to leptin. However, it is unclear whether this leptin insensitivity is congenital, or whether normal leptin sensitivity in neonatal Magel2-null mice is lost postnatally. We used in vitro cytosolic calcium imaging to follow the postnatal development of leptin responses in POMC neurons in these mice. Leptin caused an activation of POMC neurons in wild-type acute hypothalamic slice preparations at all ages, reflecting their normal leptin-invoked depolarization. Normal leptin responses were found in Magel2-null mice up to 4 weeks of age, but the proportion of leptin-responsive POMC neurons was reduced in 6-week-old Magel2-null mice. The number of α-melanocyte-stimulating hormone immunoreactive fibers in the paraventricular hypothalamic nucleus was also reduced in mutant mice at 6 weeks of age. A similar progressive loss of leptin sensitivity caused by loss of MAGEL2 in children with PWS could explain the delayed onset of increased appetite and weight gain in this complex disorder.


Assuntos
Antígenos de Neoplasias/genética , Leptina/metabolismo , Neurônios/metabolismo , Síndrome de Prader-Willi/genética , Proteínas/genética , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Modelos Animais de Doenças , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Hipotálamo/patologia , Leptina/administração & dosagem , Camundongos , Neurônios/patologia , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Pró-Opiomelanocortina/metabolismo , Aumento de Peso/genética
19.
PLoS One ; 8(2): e52390, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23390487

RESUMO

The Angelman/Prader-Willi syndrome (AS/PWS) domain contains at least 8 imprinted genes regulated by a bipartite imprinting center (IC) associated with the SNRPN gene. One component of the IC, the PWS-IC, governs the paternal epigenotype and expression of paternal genes. The mechanisms by which imprinting and expression of paternal genes within the AS/PWS domain - such as MKRN3 and NDN - are regulated by the PWS-IC are unclear. The syntenic region in the mouse is organized and imprinted similarly to the human domain with the murine PWS-IC defined by a 6 kb interval within the Snrpn locus that includes the promoter. To identify regulatory elements that may mediate PWS-IC function, we mapped the location and allele-specificity of DNase I hypersensitive (DH) sites within the PWS-IC in brain cells, then identified transcription factor binding sites within a subset of these DH sites. Six major paternal-specific DH sites were detected in the Snrpn gene, five of which map within the 6 kb PWS-IC. We postulate these five DH sites represent functional components of the murine PWS-IC. Analysis of transcription factor binding within multiple DH sites detected nuclear respiratory factors (NRF's) and YY1 specifically on the paternal allele. NRF's and YY1 were also detected in the paternal promoter region of the murine Mrkn3 and Ndn genes. These results suggest that NRF's and YY1 may facilitate PWS-IC function and coordinately regulate expression of paternal genes. The presence of NRF's also suggests a link between transcriptional regulation within the AS/PWS domain and regulation of respiration. 3C analyses indicated Mkrn3 lies in close proximity to the PWS-IC on the paternal chromosome, evidence that the PWS-IC functions by allele-specific interaction with its distal target genes. This could occur by allele-specific co-localization of the PWS-IC and its target genes to transcription factories containing NRF's and YY1.


Assuntos
Síndrome de Angelman/genética , Regulação da Expressão Gênica , Fatores Nucleares Respiratórios/genética , Síndrome de Prader-Willi/genética , Elementos Reguladores de Transcrição , Fator de Transcrição YY1/genética , Proteínas Centrais de snRNP/genética , Alelos , Síndrome de Angelman/metabolismo , Síndrome de Angelman/patologia , Animais , Sequência de Bases , Sítios de Ligação , Desoxirribonuclease I/metabolismo , Loci Gênicos , Impressão Genômica , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Fatores Nucleares Respiratórios/metabolismo , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Ligação Proteica , Sintenia , Transcrição Gênica , Fator de Transcrição YY1/metabolismo , Proteínas Centrais de snRNP/metabolismo
20.
Hum Mol Genet ; 21(21): 4703-17, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22865874

RESUMO

MAGED1, NECDIN and MAGEL2 are members of the MAGE gene family. The latter two of these genes have been involved in Prader-Willi syndrome (PWS), which includes hyperphagia, repetitive and compulsive behaviors, and cognitive impairment. Here, we show that Maged1-deficient mice develop progressive obesity associated with hyperphagia and reduced motor activity. Loss of Maged1 also results in a complex behavioral syndrome that includes reduced social interactions and memory, deficient sexual behavior, as well as increased anxiety and self-grooming. Oxytocin (OT), which is produced in the hypothalamus, can act as a neurotransmitter that reduces anxiety, promotes social behaviors and regulates food intake. Growing evidences indicate that OT is involved in autism. We found that Maged1 mutants showed a severe reduction in the levels of mature OT, but not of its precursors, in the hypothalamus. Moreover, the administration of OT rescued the deficit in social memory of these mice. We conclude that Maged1 is required for OT processing or stability. A decrease in mature OT levels in Maged1 mutants affects social interactions and possibly other behavioral processes. Our observations suggest that, in human, MAGED1 could play a role in autism or cause a neurodevelopmental condition that is reminiscent of the PWS.


Assuntos
Proteínas de Neoplasias , Ocitocina , Síndrome de Prader-Willi , Comportamento Sexual Animal , Animais , Ansiedade/genética , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Transtorno Autístico/metabolismo , Transtorno Autístico/fisiopatologia , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Relações Interpessoais , Camundongos , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Obesidade/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Ocitocina/administração & dosagem , Ocitocina/biossíntese , Ocitocina/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatologia , Comportamento Sexual Animal/efeitos dos fármacos , Comportamento Sexual Animal/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA