Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
JCI Insight ; 8(10)2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37014702

RESUMO

The prevalence of obesity and type 2 diabetes is growing at an alarming rate, including among pregnant women. Low-calorie sweeteners (LCSs) have increasingly been used as an alternative to sugar to deliver a sweet taste without the excessive caloric load. However, there is little evidence regarding their biological effects, particularly during development. Here, we used a mouse model of maternal LCS consumption to explore the impact of perinatal LCS exposure on the development of neural systems involved in metabolic regulation. We report that adult male, but not female, offspring from both aspartame- and rebaudioside A-exposed dams displayed increased adiposity and developed glucose intolerance. Moreover, maternal LCS consumption reorganized hypothalamic melanocortin circuits and disrupted parasympathetic innervation of pancreatic islets in male offspring. We then identified phenylacetylglycine (PAG) as a unique metabolite that was upregulated in the milk of LCS-fed dams and the serum of their pups. Furthermore, maternal PAG treatment recapitulated some of the key metabolic and neurodevelopmental abnormalities associated with maternal LCS consumption. Together, our data indicate that maternal LCS consumption has enduring consequences on the offspring's metabolism and neural development and that these effects are likely to be mediated through the gut microbial co-metabolite PAG.


Assuntos
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Animais , Camundongos , Masculino , Feminino , Humanos , Gravidez , Edulcorantes , Ingestão de Energia , Obesidade/metabolismo
2.
Mol Metab ; 71: 101707, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36933618

RESUMO

BACKGROUND/PURPOSE: Litter size is a biological variable that strongly influences adult physiology in rodents. Despite evidence from previous decades and recent studies highlighting its major impact on metabolism, information about litter size is currently underreported in the scientific literature. Here, we urge that this important biological variable should be explicitly stated in research articles. RESULTS/CONCLUSION: Below, we briefly describe the scientific evidence supporting the impact of litter size on adult physiology and outline a series of recommendations and guidelines to be implemented by investigators, funding agencies, editors in scientific journals, and animal suppliers to fill this important gap.


Assuntos
Roedores , Gravidez , Animais , Feminino , Tamanho da Ninhada de Vivíparos/fisiologia
3.
J Chem Neuroanat ; 123: 102117, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35680104

RESUMO

The hypothalamus is a large brain region made of nuclei and areas involved in the control of behaviors and physiological regulations. Among them, the arcuate nucleus (ARH) and the lateral hypothalamic area (LHA) contain key neuronal populations expressing the pro-opiomelanocortin (POMC), the agouti-related peptide (AgRP), and the melanin-concentrating hormone (MCH), respectively, that are involved in goal-oriented behaviors (such as feeding behavior) and glucose homeostasis. These neuronal populations are generated from distinct parts of the germinative neuroepithelium during embryonic life, and acquire their cell fate under the influence of morphogen proteins, specific transcription factors, and epigenetic modulators. POMC and MCH neuronal development continues by sending long descending axonal projections before birth under the control of axon guidance molecules such as Netrin1 and Slit2. Later, during the postnatal period, POMC and AgRP neurons develop intra-hypothalamic projections notably to the paraventricular nucleus of the hypothalamus through the influence of other axon guidance cues such as the class3 Semaphorins. Other cellular processes, such as autophagy and primary cilia function, and hormonal cues also appear critical for the proper development of POMC neurons.


Assuntos
Hipotálamo , Pró-Opiomelanocortina , Proteína Relacionada com Agouti/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo
4.
Exp Mol Med ; 54(4): 403-413, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35474338

RESUMO

The melanocortin system plays a critical role in the central regulation of food intake and energy balance. This system consists of neurons producing pro-opiomelanocortin (POMC), melanocortin receptors (MC4Rs), and the endogenous antagonist agouti-related peptide (AgRP). Pomc and Mc4r deficiency in rodents and humans causes early onset of obesity, whereas a loss of Agrp function is associated with leanness. Accumulating evidence shows that many chronic diseases, including obesity, might originate during early life. The melanocortin system develops during a relatively long period beginning during embryonic life with the birth of POMC and AgRP neurons and continuing postnatally with the assembly of their neuronal circuitry. The development of the melanocortin system requires the tight temporal regulation of molecular factors, such as transcription factors and axon guidance molecules, and cellular mechanisms, such as autophagy. It also involves a complex interplay of endocrine and nutritional factors. The disruption of one or more of these developmental factors can lead to abnormal maturation and function of the melanocortin system and has profound metabolic consequences later in life.


Assuntos
Melanocortinas , Pró-Opiomelanocortina , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Humanos , Hipotálamo/metabolismo , Melanocortinas/metabolismo , Obesidade/metabolismo , Peptídeos/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo
5.
Int J Obes (Lond) ; 46(6): 1138-1144, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35173277

RESUMO

BACKGROUND/OBJECTIVES: Alteration of the perinatal nutritional environment is an important risk factor for the development of metabolic diseases in later life. The hormone leptin plays a critical role in growth and development. Previous studies reported that postnatal overnutrition increases leptin secretion during the pre-weaning period. However, a direct link between leptin, neonatal overnutrition, and lifelong metabolic regulation has not been investigated. METHODS: We used the small litter mouse model combined with neonatal leptin antagonist injections to examine whether attenuating leptin during early life improves lifelong metabolic regulation in postnatally overnourished mice. RESULTS: Postnatally overnourished mice displayed rapid weight gain during lactation and remained overweight as adults. These mice also showed increased adiposity and perturbations in glucose homeostasis in adulthood. Neonatal administration of a leptin antagonist normalized fat mass and insulin sensitivity in postnatally overnourished mice. These metabolic improvements were associated with enhanced sensitivity of hypothalamic neurons to leptin. CONCLUSIONS: Early postnatal overnutrition causes metabolic alterations that can be permanently attenuated with the administration of a leptin antagonist during a restricted developmental window.


Assuntos
Leptina , Hipernutrição , Animais , Feminino , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos , Obesidade/metabolismo , Hipernutrição/metabolismo , Gravidez , Aumento de Peso
6.
Med Sci (Paris) ; 37(4): 366-371, 2021 Apr.
Artigo em Francês | MEDLINE | ID: mdl-33908854

RESUMO

The survival of the species depends on two closely interlinked processes: the correct functioning of the reproductive system, and the balance between the energy needs of an individual and the supply of energy sources through feeding. These two processes are regulated in the hypothalamus, which produces neurohormones that control various physiological functions. Among these neurohormones, GnRH controls not only the maturation and function of the reproductive organs, including the ovaries and the testes, during puberty and in adulthood, but also sexual attraction. Recent evidence suggest that neuropilin-1-mediated signaling in GnRH-synthesizing neurons could be a linchpin that holds together various neuroanatomical, physiological and behavioral adaptations involved in triggering puberty and achieving reproductive function.


TITLE: Signalisation impliquant la neuropiline dans les neurones sécrétant la GnRH - Son rôle dans le déclenchement de la puberté. ABSTRACT: La survie d'une espèce dépend de deux processus intimement liés : la reproduction, d'une part, et l'équilibre entre les besoins énergétiques et l'approvisionnement en sources d'énergie par l'alimentation, d'autre part. Ces deux processus sont contrôlés dans le cerveau par l'hypothalamus, qui produit des neurohormones agissant sur l'hypophyse pour piloter diverses fonctions physiologiques. L'une de ces neurohormones, la GnRH, contrôle non seulement la maturation et le fonctionnement des organes reproducteurs, incluant les ovaires et les testicules, lors de la puberté et à l'âge adulte, mais aussi l'attirance sexuelle. De récentes découvertes suggèrent que la signalisation impliquant la neuropiline-1 dans les neurones sécrétant la GnRH jouerait un rôle charnière dans la coordination du neurodéveloppement et des adaptations physiologiques et comportementales nécessaires au déclenchement de la puberté et à l'acquisition de la fonction de reproduction. Dans cet article de synthèse, nous replaçons ces découvertes dans le contexte de récents travaux montrant que les voies de signalisation des sémaphorines de classe 3 sont impliquées dans la physiopathologie non seulement de l'infertilité, mais aussi de l'obésité. Nous discutons également l'implication potentielle des neurones produisant la GnRH dans la perception des odeurs sociales et dans la précocité de la maturation sexuelle. L'hypothèse selon laquelle l'activité de ces neurones au cours du développement postnatal constituerait le chaînon manquant entre la prise de poids, le déclenchement de la puberté et le comportement sexuel, ouvre la voie à une meilleure compréhension de l'implication de l'homéostasie énergétique dans la maturation sexuelle, et pourrait aussi avoir des implications thérapeutiques pour la puberté précoce.


Assuntos
Hormônio Liberador de Gonadotropina/biossíntese , Neurônios/metabolismo , Neuropilina-1/metabolismo , Puberdade Precoce/etiologia , Puberdade/fisiologia , Animais , Ingestão de Energia , Metabolismo Energético/fisiologia , Feminino , Genitália/fisiologia , Humanos , Hipotálamo/fisiologia , Masculino , Camundongos , Reprodução/fisiologia , Caracteres Sexuais , Excitação Sexual
7.
Mol Metab ; 47: 101186, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33571700

RESUMO

OBJECTIVE: The ventromedial nucleus of the hypothalamus (VMH) is a critical component of the forebrain pathways that regulate energy homeostasis. It also plays an important role in the metabolic response to fasting. However, the mechanisms contributing to these physiological processes remain elusive. Autophagy is an evolutionarily conserved mechanism that maintains cellular homeostasis by turning over cellular components and providing nutrients to the cells during starvation. Here, we investigated the importance of the autophagy-related gene Atg7 in Sf1-expressing neurons of the VMH in control and fasted conditions. METHODS: We generated Sf1-Cre; Atg7loxP/loxP mice and examined their metabolic and cellular response to fasting. RESULTS: Fasting induces autophagy in the VMH, and mice lacking Atg7 in Sf1-expressing neurons display altered leptin sensitivity and impaired energy expenditure regulation in response to fasting. Moreover, loss of Atg7 in Sf1 neurons causes alterations in the central response to fasting. Furthermore, alterations in mitochondria morphology and activity are observed in mutant mice. CONCLUSION: Together, these data show that autophagy is nutritionally regulated in VMH neurons and that VMH autophagy participates in the control of energy homeostasis during fasting.


Assuntos
Autofagia , Jejum , Mitocôndrias/metabolismo , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Animais , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Metabolismo Energético , Feminino , Homeostase , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Transcriptoma
8.
J Neuroendocrinol ; 33(4): e12933, 2021 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-33438814

RESUMO

The discovery of insulin in 1921 was a major breakthrough in medicine and for therapy in patients with diabetes. The dramatic rise in the prevalence of overweight and obesity has been tightly linked to an increased prevalence of gestational diabetes mellitus (GDM), which poses major health concerns. Babies born to GDM mothers are more likely to develop obesity, type 2 diabetes and cardiovascular disease later in life. Evidence accumulated during the past two decades has revealed that high levels insulin, such as those observed during GDM, can have a widespread effect on the development and function of a variety of organs. This review summarises our current knowledge on the role of insulin in the placenta, cardiovascular system and brain during critical periods of development, as well as how it can contribute to lifelong metabolic regulation. We also discuss possible intervention strategies to ameliorate and hopefully reverse the developmental defects associated with obesity and GDM.

9.
EMBO J ; 39(19): e104633, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32761635

RESUMO

Hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH), the "master molecule" regulating reproduction and fertility, migrate from their birthplace in the nose to their destination using a system of guidance cues, which include the semaphorins and their receptors, the neuropilins and plexins, among others. Here, we show that selectively deleting neuropilin-1 in new GnRH neurons enhances their survival and migration, resulting in excess neurons in the hypothalamus and in their unusual accumulation in the accessory olfactory bulb, as well as an acceleration of mature patterns of activity. In female mice, these alterations result in early prepubertal weight gain, premature attraction to male odors, and precocious puberty. Our findings suggest that rather than being influenced by peripheral energy state, GnRH neurons themselves, through neuropilin-semaphorin signaling, might engineer the timing of puberty by regulating peripheral adiposity and behavioral switches, thus acting as a bridge between the reproductive and metabolic axes.


Assuntos
Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Neuropilina-1/biossíntese , Comportamento Sexual Animal , Maturidade Sexual , Aumento de Peso , Animais , Feminino , Hormônio Liberador de Gonadotropina/genética , Masculino , Camundongos , Camundongos Transgênicos , Neuropilina-1/genética
10.
Nat Commun ; 11(1): 1914, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32313051

RESUMO

Obesity is associated with the activation of cellular responses, such as endoplasmic reticulum (ER) stress. Here, we show that leptin-deficient ob/ob mice display elevated hypothalamic ER stress as early as postnatal day 10, i.e., prior to the development of obesity in this mouse model. Neonatal treatment of ob/ob mice with the ER stress-relieving drug tauroursodeoxycholic acid (TUDCA) causes long-term amelioration of body weight, food intake, glucose homeostasis, and pro-opiomelanocortin (POMC) projections. Cells exposed to ER stress often activate autophagy. Accordingly, we report that in vitro induction of ER stress and neonatal leptin deficiency in vivo activate hypothalamic autophagy-related genes. Furthermore, genetic deletion of autophagy in pro-opiomelanocortin neurons of ob/ob mice worsens their glucose homeostasis, adiposity, hyperphagia, and POMC neuronal projections, all of which are ameliorated with neonatal TUDCA treatment. Together, our data highlight the importance of early life ER stress-autophagy pathway in influencing hypothalamic circuits and metabolic regulation.


Assuntos
Autofagia/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Leptina/metabolismo , Neurogênese/fisiologia , Adiposidade , Animais , Antivirais/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína 7 Relacionada à Autofagia/genética , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Colagogos e Coleréticos/farmacologia , Modelos Animais de Doenças , Ingestão de Alimentos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Comportamento Alimentar , Homeostase , Hiperfagia/metabolismo , Leptina/genética , Masculino , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Neuroendocrinologia , Neurogênese/efeitos dos fármacos , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Ácido Tauroquenodesoxicólico
11.
PLoS Biol ; 18(3): e3000296, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32163401

RESUMO

The steady increase in the prevalence of obesity and associated type II diabetes mellitus is a major health concern, particularly among children. Maternal obesity represents a risk factor that contributes to metabolic perturbations in the offspring. Endoplasmic reticulum (ER) stress has emerged as a critical mechanism involved in leptin resistance and type 2 diabetes in adult individuals. Here, we used a mouse model of maternal obesity to investigate the importance of early life ER stress in the nutritional programming of this metabolic disease. Offspring of obese dams developed glucose intolerance and displayed increased body weight, adiposity, and food intake. Moreover, maternal obesity disrupted the development of melanocortin circuits associated with neonatal hyperleptinemia and leptin resistance. ER stress-related genes were up-regulated in the hypothalamus of neonates born to obese mothers. Neonatal treatment with the ER stress-relieving drug tauroursodeoxycholic acid improved metabolic and neurodevelopmental deficits and reversed leptin resistance in the offspring of obese dams.


Assuntos
Estresse do Retículo Endoplasmático , Hipotálamo/crescimento & desenvolvimento , Obesidade Materna/metabolismo , Animais , Animais Recém-Nascidos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Composição Corporal , Peso Corporal , Dieta/efeitos adversos , Estresse do Retículo Endoplasmático/genética , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/embriologia , Hipotálamo/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Pâncreas/crescimento & desenvolvimento , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Pró-Opiomelanocortina/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , alfa-MSH/metabolismo
12.
Neuroendocrinology ; 110(6): 552-562, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31484186

RESUMO

The hypothalamus contains integrative systems that support life, including physiological processes such as food intake, energy expenditure, and reproduction. Here, we show that anorexia nervosa (AN) patients, contrary to normal weight and constitutionally lean individuals, respond with a paradoxical reduction in hypothalamic levels of glutamate/glutamine (Glx) upon feeding. This reversal of the Glx response is associated with decreased wiring in the arcuate nucleus and increased connectivity in the lateral hypothalamic area, which are involved in the regulation on a variety of physiological and behavioral functions including the control of food intake and energy balance. The identification of distinct hypothalamic neurochemical dysfunctions and associated structural variations in AN paves the way for the development of new diagnostic and treatment strategies in conditions associated with abnormal body mass index and a maladaptive response to negative energy balance.


Assuntos
Anorexia Nervosa , Núcleo Arqueado do Hipotálamo , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Região Hipotalâmica Lateral , Adulto , Anorexia Nervosa/diagnóstico por imagem , Anorexia Nervosa/metabolismo , Anorexia Nervosa/patologia , Anorexia Nervosa/fisiopatologia , Núcleo Arqueado do Hipotálamo/diagnóstico por imagem , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Núcleo Arqueado do Hipotálamo/fisiopatologia , Feminino , Humanos , Região Hipotalâmica Lateral/diagnóstico por imagem , Região Hipotalâmica Lateral/metabolismo , Região Hipotalâmica Lateral/patologia , Região Hipotalâmica Lateral/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Espectroscopia de Prótons por Ressonância Magnética , Adulto Jovem
13.
Sci Rep ; 9(1): 1816, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755631

RESUMO

Emerging evidence from epidemiological and animal studies suggests that exposure to traffic-related air pollutants and particulate matter less than 2.5 µm in diameter (PM2.5) contributes to development of obesity and related metabolic abnormalities. However, it is not known whether nanoscale particulate matter (nPM) with aerodynamic diameter ≤200 nm have similar adverse metabolic effects. The goal of the present study was to determine the effects of prenatal and early life exposure to nPM on metabolic homeostasis in mice. C57BL/6 J mice were exposed to nPM or filtered air from gestation until 17 weeks of age and characterized for metabolic and behavioral parameters. In male mice, nPM exposure increased food intake, body weight, fat mass, adiposity, and whole-body glucose intolerance (p < 0.05). Consistent with these effects, male mice exposed to nPM displayed alterations in the expression of metabolically-relevant neuropeptides in the hypothalamus and decreased expression of insulin receptor signaling genes in adipose (p < 0.05). There were no differences in exploratory behavior or motor function, fasting lipid levels, or the inflammatory profile of adipose tissue. Our results provide evidence that chronic nPM exposure from gestation to early adulthood in male mice promotes metabolic dysregulation in part through modulation of feeding behavior and in the absence of an obesogenic diet.


Assuntos
Homeostase/efeitos dos fármacos , Material Particulado/toxicidade , Adiposidade/efeitos dos fármacos , Animais , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Feminino , Citometria de Fluxo , Intolerância à Glucose , Teste de Tolerância a Glucose , Resistência à Insulina , Locomoção/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
14.
Cell ; 176(4): 729-742.e18, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30661757

RESUMO

Hypothalamic melanocortin neurons play a pivotal role in weight regulation. Here, we examined the contribution of Semaphorin 3 (SEMA3) signaling to the development of these circuits. In genetic studies, we found 40 rare variants in SEMA3A-G and their receptors (PLXNA1-4; NRP1-2) in 573 severely obese individuals; variants disrupted secretion and/or signaling through multiple molecular mechanisms. Rare variants in this set of genes were significantly enriched in 982 severely obese cases compared to 4,449 controls. In a zebrafish mutagenesis screen, deletion of 7 genes in this pathway led to increased somatic growth and/or adiposity demonstrating that disruption of Semaphorin 3 signaling perturbs energy homeostasis. In mice, deletion of the Neuropilin-2 receptor in Pro-opiomelanocortin neurons disrupted their projections from the arcuate to the paraventricular nucleus, reduced energy expenditure, and caused weight gain. Cumulatively, these studies demonstrate that SEMA3-mediated signaling drives the development of hypothalamic melanocortin circuits involved in energy homeostasis.


Assuntos
Metabolismo Energético/genética , Melanocortinas/metabolismo , Semaforinas/genética , Adolescente , Adulto , Animais , Peso Corporal , Linhagem Celular , Criança , Pré-Escolar , Modelos Animais de Doenças , Ingestão de Alimentos , Feminino , Variação Genética/genética , Homeostase , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Obesidade/genética , Obesidade/metabolismo , Receptores de Superfície Celular/metabolismo , Semaforinas/metabolismo , Adulto Jovem , Peixe-Zebra
15.
Artigo em Inglês | MEDLINE | ID: mdl-31920985

RESUMO

With the prevalence of obesity, non-nutritive sweeteners (NNS) have been widely used as sugar substitutes as they deliver a sweet taste without excessive caloric load. However, it is increasingly recognized that NNS are not inert compounds and may cause long-term metabolic perturbations. Endoplasmic reticulum (ER) stress has emerged as a critical link in the development of obesity and type 2 diabetes. In this study, we investigated the effects of NNS found in common diet beverages (i.e., sucralose, aspartame, acesulfame potassium) and a natural sweetener (i.e., rebaudioside A) on ER stress in the hypothalamic cell line mHypoE-N43/5 in vivo and on axonal outgrowth ex vivo. Sucralose, aspartame, and acesulfame potassium caused elevated ER stress gene expression in mHypoE-N43/5 cells, with sucralose and acesulfame potassium having the most potent effect. Moreover, acesulfame potassium treatment reduced axon outgrowth from arcuate nucleus explants and this effect was attenuated with the ER stress-relieving drug tauroursodeoxycholic acid. Furthermore, sucralose induced cytotoxicity and acesulfame potassium increases caspase3/7 activity at high concentrations in mHypoE-N43/5 cells. In contrast, rebaudioside A only had moderate effects on hypothalamic ER stress and no adverse effects on axon outgrowth, cytotoxicity, or caspase3/7 activity. Together, our data reveal that commonly consumed NNS cause cellular stress in hypothalamic cells disrupting axon outgrowth and that these biological alterations are not seen with rebaudioside A. These data provide biological plausibility for some NNS to adversely impact metabolic health and identifies rebaudioside A as a sweetener with lower detrimental biological impact on hypothalamic cells.

16.
Elife ; 72018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30311908

RESUMO

Proopiomelanocortin (POMC) neurons are major negative regulators of energy balance. A distinct developmental property of POMC neurons is that they can adopt an orexigenic neuropeptide Y (NPY) phenotype. However, the mechanisms underlying the differentiation of Pomc progenitors remain unknown. Here, we show that the loss of the microRNA (miRNA)-processing enzyme Dicer in POMC neurons causes metabolic defects, an age-dependent decline in the number of PomcmRNA-expressing cells, and an increased proportion of Pomc progenitors acquiring a NPY phenotype. miRNome microarray screening further identified miR-103/107 as candidates that may be involved in the maturation of Pomc progenitors. In vitro inhibition of miR-103/107 causes a reduction in the number of Pomc-expressing cells and increases the proportion of Pomc progenitors differentiating into NPY neurons. Moreover, in utero silencing of miR-103/107 causes perturbations in glucose homeostasis. Together, these data suggest a role for prenatal miR-103/107 in the maturation of Pomc progenitors and glucose homeostasis.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/metabolismo , Neurônios/fisiologia , Neuropeptídeo Y/biossíntese , Pró-Opiomelanocortina/biossíntese , Animais , Glucose/metabolismo , Homeostase , Camundongos
17.
Mol Metab ; 15: 8-19, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29773464

RESUMO

BACKGROUND: The early life environment experienced by an individual in utero and during the neonatal period is a major factor in shaping later life disease risk-including susceptibility to develop obesity, diabetes, and cardiovascular disease. The incidence of metabolic disease is different between males and females. How the early life environment may underlie these sex differences is an area of active investigation. SCOPE OF REVIEW: The purpose of this review is to summarize our current understanding of how the early life environment influences metabolic disease risk in a sex specific manner. We also discuss the possible mechanisms responsible for mediating these sexually dimorphic effects and highlight the results of recent intervention studies in animal models. MAJOR CONCLUSIONS: Exposure to states of both under- and over-nutrition during early life predisposes both sexes to develop metabolic disease. Females seem particularly susceptible to develop increased adiposity and disrupted glucose homeostasis as a result of exposure to in utero undernutrition or high sugar environments, respectively. The male placenta is particularly vulnerable to damage by adverse nutritional states and this may underlie some of the metabolic phenotypes observed in adulthood. More studies investigating both sexes are needed to understand how changes to the early life environment impact differently on the long-term health of male and female individuals.


Assuntos
Desenvolvimento Embrionário , Metabolismo Energético , Doenças Metabólicas/etiologia , Caracteres Sexuais , Feminino , Humanos , Masculino , Doenças Metabólicas/epidemiologia , Fatores Sexuais
18.
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
19.
Diabetes ; 67(5): 805-817, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29467172

RESUMO

Amylin phosphorylates ERK (p-ERK) in the area postrema to reduce eating and synergizes with leptin to phosphorylate STAT3 in the arcuate (ARC) and ventromedial (VMN) hypothalamic nuclei to reduce food intake and body weight. The current studies assessed potential amylin and amylin-leptin ARC/VMN interactions on ERK signaling and their roles in postnatal hypothalamic pathway development. In amylin knockout mice, the density of agouti-related protein (AgRP)-immunoreactive (IR) fibers in the hypothalamic paraventricular nucleus (PVN) was increased, while the density of α-melanocyte-stimulating hormone (αMSH) fibers was decreased. In mice deficient of the amylin receptor components RAMP1/3, both AgRP and αMSH-IR fiber densities were decreased, while only αMSH-IR fiber density was decreased in rats injected neonatally in the ARC/VMN with an adeno-associated virus short hairpin RNA against the amylin core receptor. Amylin induced p-ERK in ARC neurons, 60% of which was present in POMC-expressing neurons, with none in NPY neurons. An amylin-leptin interaction was shown by an additive effect on ARC ERK signaling in neonatal rats and a 44% decrease in amylin-induced p-ERK in the ARC of leptin receptor-deficient and of ob/ob mice. Together, these results suggest that amylin directly acts, through a p-ERK-mediated process, on POMC neurons to enhance ARC-PVN αMSH pathway development.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Leptina/metabolismo , Sistema de Sinalização das MAP Quinases , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Pró-Opiomelanocortina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Alimentar , Feminino , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Obesos , Neuropeptídeo Y/metabolismo , Ratos Sprague-Dawley , Proteína 1 Modificadora da Atividade de Receptores/genética , Proteína 1 Modificadora da Atividade de Receptores/metabolismo , Proteína 3 Modificadora da Atividade de Receptores/genética , Proteína 3 Modificadora da Atividade de Receptores/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , alfa-MSH/metabolismo
20.
Artigo em Inglês | MEDLINE | ID: mdl-29250032

RESUMO

The area postrema (AP) and the nucleus of the solitary tract (NTS) are important hindbrain centers involved in the control of energy homeostasis. The AP mediates the anorectic action and the inhibitory effect on gastric emptying induced by the pancreatic hormone amylin. Amylin's target cells in the AP project to the NTS, an integrative relay center for enteroceptive signals. Perinatal hormonal and metabolic factors influence brain development. A postnatal surge of the adipocyte-derived hormone leptin represents a developmental signal for the maturation of projections between hypothalamic nuclei controlling energy balance. Amylin appears to promote neurogenesis in the AP in adult rats. Here, we examined whether amylin and leptin are required for the development of projections from the AP to the NTS in postnatal and adult mice by conducting neuronal tracing studies with DiI in amylin- (IAPP-/-) and leptin-deficient (ob/ob) mice. Compared to wild-type littermates, postnatal (P10) and adult (P60) IAPP-/- mice showed a significantly reduced density of AP-NTS projections. While AP projections were also reduced in postnatal (P14) ob/ob mice, AP-NTS fiber density did not differ between adult ob/ob and wild-type animals. Our findings suggest a crucial function of amylin for the maturation of neuronal brainstem pathways controlling energy balance and gastrointestinal function. The impaired postnatal development of neuronal AP-NTS projections in ob/ob mice appears to be compensated in this experimental model during later brain maturation. It remains to be elucidated whether an amylin- and leptin-dependent modulation in neuronal development translates into altered AP/NTS-mediated functions.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA