Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
1.
PLoS One ; 17(7): e0255201, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35853004

RESUMO

The underlying molecular pathophysiology of feeding disorders, particularly in peripheral organs, is still largely unknown. A range of molecular factors encoded by appetite-regulating genes are already described to control feeding behaviour in the brain. However, the important role of the gastrointestinal tract in the regulation of appetite and feeding in connection to the brain has gained more attention in the recent years. An example of such inter-organ connection can be the signals mediated by leptin, a key regulator of body weight, food intake and metabolism, with conserved anorexigenic effects in vertebrates. Leptin signals functions through its receptor (lepr) in multiple organs, including the brain and the gastrointestinal tract. So far, the regulatory connections between leptin signal and other appetite-regulating genes remain unclear, particularly in the gastrointestinal system. In this study, we used a zebrafish mutant with impaired function of leptin receptor to explore gut expression patterns of appetite-regulating genes, under different feeding conditions (normal feeding, 7-day fasting, 2 and 6-hours refeeding). We provide evidence that most appetite-regulating genes are expressed in the zebrafish gut. On one hand, we did not observed significant differences in the expression of orexigenic genes (except for hcrt) after changes in the feeding condition. On the other hand, we found 8 anorexigenic genes in wild-types (cart2, cart3, dbi, oxt, nmu, nucb2a, pacap and pomc), as well as 4 genes in lepr mutants (cart3, kiss1, kiss1r and nucb2a), to be differentially expressed in the zebrafish gut after changes in feeding conditions. Most of these genes also showed significant differences in their expression between wild-type and lepr mutant. Finally, we observed that impaired leptin signalling influences potential regulatory connections between anorexigenic genes in zebrafish gut. Altogether, these transcriptional changes propose a potential role of leptin signal in the regulation of feeding through changes in expression of certain anorexigenic genes in the gastrointestinal tract of zebrafish.


Assuntos
Apetite , Leptina , Animais , Apetite/genética , Regulação do Apetite/genética , Expressão Gênica , Leptina/genética , Leptina/metabolismo , Pró-Opiomelanocortina/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-34534676

RESUMO

Amylin is a 37-amino acid polypeptide that has been found to be involved in feeding regulation in some mammals, birds, and goldfish. We cloned amylin of Siberian sturgeon and detected its distribution pattern in 15 tissues. The expression levels in the periprandial period (pre-and post-feeding), the changes in the food intake, and the expression levels of related appetite factors after the intraperitoneal injection of amylin were detected. The expression of amylin was found to be the highest in the hypothalamus. Compared with 1 h pre-feeding, the expression levels of amylin in the hypothalamus and duodenum were increased significantly 1 h post-feeding. Compared with the control group (saline), intraperitoneal injection of 50 ng/g, 100 ng/g, and 200 ng/g of amylin significantly inhibited food intake at 1 h post injection, but not at 3 h and 6 h. The injection of 50 ng/g, 100 ng/g, and 200 ng/g amylin significantly inhibited the cumulative feed. After 1 h of 50 ng/g amylin injection, the levels of MC4R and somatostatin in the hypothalamus increased significantly, while the levels of amylin and NPY decreased significantly. The levels of CCK in the valvular intestine were increased significantly. Insulin in the duodenum was also increased significantly, but there was no significant change in ghrelin in the duodenum. These results show that amylin inhibits feeding in Siberian sturgeon by down-regulating the appetite-stimulating factor NPY and up-regulating the appetite-suppressing factors somatostatin, MC4R, CCK, and insulin. This study provides a theoretical basis for studying the feeding function and action mechanisms of amylin in Siberian sturgeon.


Assuntos
Proteínas de Peixes/metabolismo , Peixes/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Sequência de Aminoácidos , Animais , Depressores do Apetite/administração & dosagem , Depressores do Apetite/metabolismo , Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/genética , Regulação do Apetite/fisiologia , Sequência de Bases , Clonagem Molecular , Duodeno/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Proteínas de Peixes/administração & dosagem , Proteínas de Peixes/genética , Peixes/genética , Peixes/fisiologia , Expressão Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , Injeções Intraperitoneais , Polipeptídeo Amiloide das Ilhotas Pancreáticas/administração & dosagem , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Filogenia , Homologia de Sequência de Aminoácidos , Distribuição Tecidual
3.
Front Endocrinol (Lausanne) ; 12: 777997, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899611

RESUMO

Appetite is the basis for obtaining food and maintaining normal metabolism. Toll-like receptor 4 (TLR4) is an important receptor expressed in the brain that induces inflammatory signaling after activation. Inflammation is considered to affect the homeostatic and non-homeostatic systems of appetite, which are dominated by hypothalamic and mesolimbic dopamine signaling. Although the pathological features of many types of inflammation are known, their physiological functions in appetite are largely unknown. This review mainly addresses several key issues, including the structures of the homeostatic and non-homeostatic systems. In addition, the mechanism by which TLR4-induced inflammatory signaling contributes to these two systems to regulate appetite is also discussed. This review will provide potential opportunities to develop new therapeutic interventions that control appetite under inflammatory conditions.


Assuntos
Regulação do Apetite/fisiologia , Inflamação/fisiopatologia , Receptor 4 Toll-Like/fisiologia , Animais , Regulação do Apetite/genética , Ingestão de Alimentos/fisiologia , Homeostase/fisiologia , Humanos , Hipotálamo/fisiologia , Inflamação/genética , Inflamação/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
4.
Nature ; 600(7888): 269-273, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34789878

RESUMO

The brain is the seat of body weight homeostasis. However, our inability to control the increasing prevalence of obesity highlights a need to look beyond canonical feeding pathways to broaden our understanding of body weight control1-3. Here we used a reverse-translational approach to identify and anatomically, molecularly and functionally characterize a neural ensemble that promotes satiation. Unbiased, task-based functional magnetic resonance imaging revealed marked differences in cerebellar responses to food in people with a genetic disorder characterized by insatiable appetite. Transcriptomic analyses in mice revealed molecularly and topographically -distinct neurons in the anterior deep cerebellar nuclei (aDCN) that are activated by feeding or nutrient infusion in the gut. Selective activation of aDCN neurons substantially decreased food intake by reducing meal size without compensatory changes to metabolic rate. We found that aDCN activity terminates food intake by increasing striatal dopamine levels and attenuating the phasic dopamine response to subsequent food consumption. Our study defines a conserved satiation centre that may represent a novel therapeutic target for the management of excessive eating, and underscores the utility of a 'bedside-to-bench' approach for the identification of neural circuits that influence behaviour.


Assuntos
Manutenção do Peso Corporal/genética , Manutenção do Peso Corporal/fisiologia , Cerebelo/fisiologia , Alimentos , Biossíntese de Proteínas , Genética Reversa , Resposta de Saciedade/fisiologia , Adulto , Animais , Regulação do Apetite/genética , Regulação do Apetite/fisiologia , Núcleos Cerebelares/citologia , Núcleos Cerebelares/fisiologia , Cerebelo/citologia , Sinais (Psicologia) , Dopamina/metabolismo , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Feminino , Homeostase , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/metabolismo , Neurônios/fisiologia , Obesidade/genética , Filosofia , Adulto Jovem
5.
Front Endocrinol (Lausanne) ; 12: 727915, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34526970

RESUMO

In obesity and type 2 diabetes, numerous genes are differentially expressed, and microRNAs are involved in transcriptional regulation of target mRNAs, but miRNAs critically involved in the appetite control are not known. Here, we identified upregulation of miR-342-3p and its host gene Evl in brain and adipose tissues in C57BL/6 mice fed with high fat-high sucrose (HFHS) chow by RNA sequencing. Mir342 (-/-) mice fed with HFHS chow were protected from obesity and diabetes. The hypothalamic arcuate nucleus neurons co-express Mir342 and EVL. The percentage of activated NPY+pSTAT3+ neurons were reduced, while POMC+pSTAT3+ neurons increased in Mir342 (-/-) mice, and they demonstrated the reduction of food intake and amelioration of metabolic phenotypes. Snap25 was identified as a major target gene of miR-342-3p and the reduced expression of Snap25 may link to functional impairment hypothalamic neurons and excess of food intake. The inhibition of miR-342-3p may be a potential candidate for miRNA-based therapy.


Assuntos
Regulação do Apetite/genética , MicroRNAs/genética , Obesidade , Células 3T3-L1 , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Células Cultivadas , Dieta Hiperlipídica , Regulação da Expressão Gênica , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , MicroRNAs/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Obesidade/etiologia , Obesidade/genética , Obesidade/metabolismo , Regulação para Cima/genética
6.
Eur J Endocrinol ; 185(4): R93-R101, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34370694

RESUMO

In 2008, the first evidence of a new hormone called neuronostatin was published. The hormone was discovered using a bioinformatic method and found to originate from the same preprohormone as somatostatin. This small peptide hormone of 13 amino acids and a C-terminal amidation was soon found to exert pleiotropic physiological effects. In animal studies, neuronostatin has been shown to reduce food intake and delay gastric emptying and gastrointestinal transit. Furthermore, neuronostatin has been shown to affect glucose metabolism by increasing glucagon secretion during situations when glucose concentrations are low. Additionally, neuronostatin has been shown to affect neural tissue and cardiomyocytes by suppressing cardiac contractility. The effects of neuronostatin have not yet been delineated in humans, but if the effects found in animal studies translate to humans it could position neuronostatin as a promising target in the treatment of obesity, hypertension and diabetes. In this review, we describe the discovery of neuronostatin and the current understanding of its physiological role and potential therapeutic applicability.


Assuntos
Hormônios Peptídicos/fisiologia , Animais , Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/genética , Diabetes Mellitus/genética , Diabetes Mellitus/terapia , Esvaziamento Gástrico/efeitos dos fármacos , Esvaziamento Gástrico/genética , Humanos , Hipertensão/genética , Hipertensão/terapia , Contração Muscular/efeitos dos fármacos , Contração Muscular/genética , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Obesidade/genética , Obesidade/terapia , Hormônios Peptídicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Somatostatina/química , Somatostatina/farmacologia , Somatostatina/fisiologia
8.
Cell Metab ; 33(7): 1433-1448.e5, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34129812

RESUMO

Knowledge of how leptin receptor (LepR) neurons of the mediobasal hypothalamus (MBH) access circulating leptin is still rudimentary. Employing intravital microscopy, we found that almost half of the blood-vessel-enwrapping pericytes in the MBH express LepR. Selective disruption of pericytic LepR led to increased food intake, increased fat mass, and loss of leptin-dependent signaling in nearby LepR neurons. When delivered intravenously, fluorescently tagged leptin accumulated at hypothalamic LepR pericytes, which was attenuated upon pericyte-specific LepR loss. Because a paracellular tracer was also preferentially retained at LepR pericytes, we pharmacologically targeted regulators of inter-endothelial junction tightness and found that they affect LepR neuronal signaling and food intake. Optical imaging in MBH slices revealed a long-lasting, tonic calcium increase in LepR pericytes in response to leptin, suggesting pericytic contraction and vessel constriction. Together, our data indicate that LepR pericytes facilitate localized, paracellular blood-brain barrier leaks, enabling MBH LepR neurons to access circulating leptin.


Assuntos
Regulação do Apetite/genética , Hipotálamo/metabolismo , Leptina/farmacologia , Pericitos/fisiologia , Receptores para Leptina/fisiologia , Animais , Comportamento Alimentar/fisiologia , Feminino , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Leptina/sangue , Leptina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
9.
Cell Metab ; 33(7): 1466-1482.e7, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34043943

RESUMO

Sensory neurons relay gut-derived signals to the brain, yet the molecular and functional organization of distinct populations remains unclear. Here, we employed intersectional genetic manipulations to probe the feeding and glucoregulatory function of distinct sensory neurons. We reconstruct the gut innervation patterns of numerous molecularly defined vagal and spinal afferents and identify their downstream brain targets. Bidirectional chemogenetic manipulations, coupled with behavioral and circuit mapping analysis, demonstrated that gut-innervating, glucagon-like peptide 1 receptor (GLP1R)-expressing vagal afferents relay anorexigenic signals to parabrachial nucleus neurons that control meal termination. Moreover, GLP1R vagal afferent activation improves glucose tolerance, and their inhibition elevates blood glucose levels independent of food intake. In contrast, gut-innervating, GPR65-expressing vagal afferent stimulation increases hepatic glucose production and activates parabrachial neurons that control normoglycemia, but they are dispensable for feeding regulation. Thus, distinct gut-innervating sensory neurons differentially control feeding and glucoregulatory neurocircuits and may provide specific targets for metabolic control.


Assuntos
Regulação do Apetite , Eixo Encéfalo-Intestino/fisiologia , Glucose/metabolismo , Células Receptoras Sensoriais/fisiologia , Vias Aferentes/metabolismo , Animais , Apetite/fisiologia , Regulação do Apetite/genética , Comunicação Celular/genética , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos Transgênicos , Gânglio Nodoso/metabolismo , Gânglio Nodoso/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Nervo Vago/metabolismo , Nervo Vago/fisiologia , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
10.
Peptides ; 138: 170507, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33577839

RESUMO

Hypoxia controls metabolism at several levels, e.g., via mitochondrial ATP production, glucose uptake and glycolysis. Hence it is likely that hypoxia also affects the action and/or production of many peptide hormones linked to food intake and appetite control. Many of those are produced in the gastrointestinal tract, endocrine pancreas, adipose tissue, and selective areas in the brain which modulate and concert their actions. However, the complexity of the hypoxia response and the links to peptides/hormones involved in food intake and appetite control in the different organs are not well known. This review summarizes the role of the hypoxia response and its effects on major peptides linked to appetite regulation, nutrition and metabolism.


Assuntos
Apetite/genética , Hormônios Gastrointestinais/genética , Leptina/genética , Hormônios Peptídicos/genética , Tecido Adiposo/metabolismo , Apetite/fisiologia , Regulação do Apetite/genética , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Ingestão de Alimentos/genética , Metabolismo Energético/genética , Hormônios Gastrointestinais/metabolismo , Glucose/metabolismo , Glicólise/genética , Humanos , Leptina/metabolismo , Hormônios Peptídicos/metabolismo
11.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33468645

RESUMO

Mutations in the TrkB neurotrophin receptor lead to profound obesity in humans, and expression of TrkB in the dorsomedial hypothalamus (DMH) is critical for maintaining energy homeostasis. However, the functional implications of TrkB-fexpressing neurons in the DMH (DMHTrkB) on energy expenditure are unclear. Additionally, the neurocircuitry underlying the effect of DMHTrkB neurons on energy homeostasis has not been explored. In this study, we show that activation of DMHTrkB neurons leads to a robust increase in adaptive thermogenesis and energy expenditure without altering heart rate or blood pressure, while silencing DMHTrkB neurons impairs thermogenesis. Furthermore, we reveal neuroanatomically and functionally distinct populations of DMHTrkB neurons that regulate food intake or thermogenesis. Activation of DMHTrkB neurons projecting to the raphe pallidus (RPa) stimulates thermogenesis and increased energy expenditure, whereas DMHTrkB neurons that send collaterals to the paraventricular hypothalamus (PVH) and preoptic area (POA) inhibit feeding. Together, our findings provide evidence that DMHTrkB neuronal activity plays an important role in regulating energy expenditure and delineate distinct neurocircuits that underly the separate effects of DMHTrkB neuronal activity on food intake and thermogenesis.


Assuntos
Regulação do Apetite/genética , Metabolismo Energético/genética , Glicoproteínas de Membrana/genética , Núcleo Hipotalâmico Paraventricular/metabolismo , Área Pré-Óptica/metabolismo , Proteínas Tirosina Quinases/genética , Termogênese/genética , Animais , Ingestão de Alimentos/genética , Feminino , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Homeostase/genética , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Núcleo Pálido da Rafe/citologia , Núcleo Pálido da Rafe/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Área Pré-Óptica/citologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais
13.
Sci Rep ; 10(1): 22093, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33328492

RESUMO

Food intake is a complex behavior regulated by discrete brain nuclei that integrate homeostatic nutritional requirements with the hedonic properties of food. Homeostatic feeding (i.e. titration of caloric intake), is typically associated with hypothalamic brain nuclei, including the paraventricular nucleus of the hypothalamus (PVN). Hedonic feeding is driven, in part, by the reinforcing properties of highly palatable food (HPF), which is mediated by the nucleus accumbens (NAc). Dysregulation of homeostatic and hedonic brain nuclei can lead to pathological feeding behaviors, namely overconsumption of highly palatable food (HPF), that may drive obesity. Both homeostatic and hedonic mechanisms of food intake have been attributed to several brain regions, but the integration of homeostatic and hedonic signaling to drive food intake is less clear, therefore we aimed to identify the neuroanatomical, functional, and behavioral features of a novel PVN → NAc circuit. Using viral tracing techniques, we determined that PVN → NAc has origins in the parvocellular PVN, and that PVN → NAc neurons express VGLUT1, a marker of glutamatergic signaling. Next, we pharmacogenetically stimulated PVN → NAc neurons and quantified both gamma-aminobutyric acid (GABA) and glutamate release and phospho-cFos expression in the NAc and observed a robust and significant increase in extracellular glutamate and phospho-cFos expression. Finally, we pharmacogenetically stimulated PVN → NAc which decreased intake of highly palatable food, demonstrating that this glutamatergic circuitry regulates aspects of feeding.


Assuntos
Regulação do Apetite/genética , Encéfalo/fisiologia , Núcleo Accumbens/fisiologia , Obesidade/metabolismo , Animais , Encéfalo/metabolismo , Comportamento Alimentar , Ácido Glutâmico/metabolismo , Humanos , Hipotálamo/metabolismo , Vias Neurais/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Núcleo Accumbens/metabolismo , Obesidade/genética , Obesidade/patologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Filosofia , Ratos
14.
Int J Mol Sci ; 21(21)2020 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-33171667

RESUMO

Phoenixin (PNX) neuropeptide is a cleaved product of the Smim20 protein. Its most common isoforms are the 14- and 20-amino acid peptides. The biological functions of PNX are mediated via the activation of the GPR173 receptor. PNX plays an important role in the central nervous system (CNS) and in the female reproductive system where it potentiates LH secretion and controls the estrus cycle. Moreover, it stimulates oocyte maturation and increases the number of ovulated oocytes. Nevertheless, PNX not only regulates the reproduction system but also exerts anxiolytic, anti-inflammatory, and cell-protective effects. Furthermore, it is involved in behavior, food intake, sensory perception, memory, and energy metabolism. Outside the CNS, PNX exerts its effects on the heart, ovaries, adipose tissue, and pancreatic islets. This review presents all the currently available studies demonstrating the pleiotropic effects of PNX.


Assuntos
Neuropeptídeos/fisiologia , Hormônios Peptídicos/fisiologia , Reprodução/fisiologia , Sequência de Aminoácidos , Animais , Ansiedade/fisiopatologia , Regulação do Apetite/genética , Regulação do Apetite/fisiologia , Sistema Nervoso Central/fisiologia , Feminino , Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/fisiologia , Masculino , Memória/fisiologia , Neuropeptídeos/genética , Fármacos Neuroprotetores/metabolismo , Hormônios Peptídicos/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Reprodução/genética , Sede/fisiologia , Distribuição Tecidual
15.
Sci Rep ; 10(1): 19618, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33184349

RESUMO

Gestational complications, including preeclampsia and gestational diabetes, have long-term adverse consequences for offspring's metabolic and cardiovascular health. A low-grade systemic inflammatory response is likely mediating this. Here, we examine the consequences of LPS-induced gestational inflammation on offspring's health in adulthood. LPS was administered to pregnant C57Bl/6J mice on gestational day 10.5. Maternal plasma metabolomics showed oxidative stress, remaining for at least 5 days after LPS administration, likely mediating the consequences for the offspring. From weaning on, all offspring was fed a control diet; from 12 to 24 weeks of age, half of the offspring received a western-style diet (WSD). The combination of LPS-exposure and WSD resulted in hyperphagia and increased body weight and body fat mass in the female offspring. This was accompanied by changes in glucose tolerance, leptin and insulin levels and gene expression in liver and adipose tissue. In the hypothalamus, expression of genes involved in food intake regulation was slightly changed. We speculate that altered food intake behaviour is a result of dysregulation of hypothalamic signalling. Our results add to understanding of how maternal inflammation can mediate long-term health consequences for the offspring. This is relevant to many gestational complications with a pro-inflammatory reaction in place.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hiperfagia/etiologia , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/efeitos adversos , Troca Materno-Fetal/fisiologia , Caracteres Sexuais , Aumento de Peso , Tecido Adiposo/metabolismo , Animais , Regulação do Apetite/genética , Feminino , Hipotálamo/fisiopatologia , Insulina/metabolismo , Leptina/metabolismo , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Gravidez
16.
Int J Mol Sci ; 21(22)2020 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-33238373

RESUMO

Physical exercise is known to influence hormonal mediators of appetite, but the effect of short-term maximal intensity exercise on plasma levels of appetite hormones and cytokines has been little studied. We investigated the effect of a 30 s Wingate Test, followed by a postprandial period, on appetite sensations, food intake, and appetite hormones. Twenty-six physically active young males rated their subjective feelings of hunger, prospective food consumption, and fatigue on visual analogue scales at baseline, after exercise was completed, and during the postprandial period. Blood samples were obtained for the measurement of nesfatin-1, ghrelin, leptin, insulin, pancreatic polypeptide (PP), human growth factor (hGH) and cytokine interleukin-6 (IL-6), irisin and plasma lactate concentrations, at 30 min before exercise, immediately (210 s) after exercise, and 30 min following a meal and at corresponding times in control sedentary males without ad libitum meal intake, respectively. Appetite perceptions and food intake were decreased in response to exercise. Plasma levels of irisin, IL-6, lactate, nesfatin-1 and ghrelin was increased after exercise and then it was returned to postprandial/control period in both groups. A significant rise in plasma insulin, hGH and PP levels after exercise was observed while meal intake potentiated this response. In conclusion, an acute short-term fatiguing exercise can transiently suppress hunger sensations and food intake in humans. We postulate that this physiological response involves exercise-induced alterations in plasma hormones and the release of myokines such as irisin and IL-6, and supports the notion of existence of the skeletal muscle-brain-gut axis. Nevertheless, the detailed relationship between acute exercise releasing myokines, appetite sensations and impairment of this axis leading to several diseases should be further examined.


Assuntos
Regulação do Apetite/genética , Apetite/fisiologia , Exercício Físico , Fadiga/terapia , Adulto , Apetite/genética , Regulação do Apetite/fisiologia , Índice de Massa Corporal , Ingestão de Alimentos/fisiologia , Fadiga/sangue , Fadiga/fisiopatologia , Fibronectinas/sangue , Grelina/sangue , Humanos , Fome/fisiologia , Interleucina-6/sangue , Ácido Láctico/sangue , Masculino , Nucleobindinas/sangue , Polipeptídeo Pancreático/sangue , Período Pós-Prandial/fisiologia
17.
Med Sci (Paris) ; 36(10): 859-865, 2020 Oct.
Artigo em Francês | MEDLINE | ID: mdl-33026327

RESUMO

Obesity is a complex, multifactorial disorder. About 5% of obese patients actually present with a monogenic form of obesity where only one mutation is sufficient to cause the disease. So far, the genes that have been found to be mutated in these monogenic forms play a key role in the leptin/melanocortin pathway which is mainly active in the hypothalamus and which regulates food intake and energy expenditure. Our laboratory has recently reported a novel monogenic form of obesity due to MRAP2 deficiency where, contrary to previously described monogenic forms of obesity, the carriers presented with hyperglycemia and hypertension in addition to obesity, suggesting that MRAP2 might play a pleiotropic role in metabolic tissues, in addition to its role in brain control of food intake and energy expenditure.


TITLE: Du nouveau dans la génétique des formes monogéniques d'obésité et son impact pour mieux en comprendre la physiopathologie. ABSTRACT: L'obésité est une maladie complexe multifactorielle. Chez environ 5 % des individus obèses, il existe des formes monogéniques d'obésité pour laquelle une seule mutation est suffisante pour entraîner la maladie. Jusqu'à présent, les gènes mutés identifiés dans ces formes monogéniques sont impliqués dans la voie leptine/mélanocortine, principalement active au niveau de l'hypothalamus et régissant la satiété. Récemment, notre laboratoire a décrit une nouvelle forme monogénique d'obésité. Elle est due à une déficience du gène MRAP2 (melanocortin-2 receptor accessory protein 2) pour laquelle, contrairement aux formes précédemment décrites, on retrouve chez les porteurs une hyperglycémie et une hypertension, suggérant que MRAP2 joue un rôle biologique général dans les tissus métaboliques en plus de son rôle dans l'hypothalamus.


Assuntos
Regulação do Apetite/genética , Obesidade/genética , Animais , Predisposição Genética para Doença , Humanos , Leptina/genética , Leptina/metabolismo , Mutação/fisiologia , Obesidade/patologia , Obesidade/terapia , Medicina de Precisão/métodos , Medicina de Precisão/tendências , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo
18.
Psychoneuroendocrinology ; 119: 104718, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32535402

RESUMO

Growth hormone secretagogue receptor (GHSR), the receptor for ghrelin, is expressed in key brain nuclei that regulate food intake. The dopamine (DA) pathways have long been recognized to play key roles mediating GHSR effects on feeding behaviors. Here, we aimed to determine the role of GHSR in DA neurons controlling appetitive and consummatory behaviors towards high fat (HF) diet. For this purpose, we crossed reactivable GHSR-deficient mice with DA transporter (DAT)-Cre mice, which express Cre recombinase under the DAT promoter that is active exclusively in DA neurons, to generate mice with GHSR expression limited to DA neurons (DAT-GHSR mice). We found that DAT-GHSR mice show an increase of c-Fos levels in brain areas containing DA neurons after ghrelin treatment, in a similar fashion as seen in wild-type mice; however, they did not increase food intake or locomotor activity in response to systemically- or centrally-administered ghrelin. In addition, we found that satiated DAT-GHSR mice displayed both anticipatory activity to scheduled HF diet exposure and HF intake in a binge-like eating protocol similar to those in wild-type mice, whereas GHSR-deficient mice displayed impaired responses. We conclude that GHSR expression in DA neurons is sufficient to both mediate increased anticipatory activity to a scheduled HF diet exposure and fully orchestrate binge-like HF intake, but it is insufficient to restore the acute orexigenic or locomotor effects of ghrelin treatment. Thus, GHSR in DA neurons affects appetitive and consummatory behaviors towards HF diet that take place in the absence of caloric needs.


Assuntos
Comportamento Consumatório/fisiologia , Dieta Hiperlipídica , Comportamento Alimentar/fisiologia , Receptores de Grelina/fisiologia , Animais , Regulação do Apetite/genética , Comportamento Animal/fisiologia , Neurônios Dopaminérgicos/metabolismo , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Preferências Alimentares/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Grelina/genética , Receptores de Grelina/metabolismo
19.
Nutrients ; 12(4)2020 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-32325849

RESUMO

The aim of this study was to evaluate the distribution of energy intake and macronutrients consumption throughout the day, and how its effect on nutritional status can be modulated by the presence of the rs3749474 polymorphism of the CLOCK gene in the Cantoblanco Platform for Nutritional Genomics ("GENYAL Platform"). This cross-sectional study was carried out on 898 volunteers between 18 and 69 years old (65.5% women). Anthropometric measurements, social issues and health, dietary, biochemical, genetic, and physical activity data were collected. Subsequently, 21 statistical interaction models were designed to predict the body mass index (BMI) considering seven dietary variables analyzed by three genetic models (adjusted by age, sex, and physical activity). The average BMI was 26.9 ± 4.65 kg/m2, 62.14% presented an excess weight (BMI > 25 kg/m2). A significant interaction was observed between the presence of the rs3749474 polymorphism and the evening carbohydrate intake (% of the total daily energy intake [%TEI]) (adjusted p = 0.046), when predicting the BMI. Participants carrying TT/CT genotype showed a positive association between the evening carbohydrate intake (%TEI) and BMI (ß = 0.3379, 95% CI = (0.1689,0.5080)) and (ß = 0.1529, 95% CI = (-0.0164,0.3227)), respectively, whereas the wild type allele (CC) showed a negative association (ß = -0.0321, 95% CI = (-0.1505,0.0862)). No significant interaction with the remaining model variables was identified. New dietary strategies may be implemented to schedule the circadian distribution of macronutrients according to the genotype. Clinical Trial number: NCT04067921.


Assuntos
Regulação do Apetite/genética , Regulação do Apetite/fisiologia , Proteínas CLOCK/genética , Relógios Circadianos/genética , Relógios Circadianos/fisiologia , Carboidratos da Dieta/administração & dosagem , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Fenômenos Fisiológicos da Nutrição/genética , Fenômenos Fisiológicos da Nutrição/fisiologia , Estado Nutricional/genética , Estado Nutricional/fisiologia , Polimorfismo Genético , Adolescente , Adulto , Idoso , Alelos , Índice de Massa Corporal , Estudos Transversais , Ingestão de Energia/genética , Ingestão de Energia/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
20.
Artigo em Inglês | MEDLINE | ID: mdl-32153512

RESUMO

Context: The hypothalamic circuit has an essential role in the regulation of appetite and energy expenditure. Pathogenic variants in genes involved in the hypothalamic leptin-melanocortin pathway, including melanocortin-4-receptor (MC4R), have been associated with monogenic obesity. Objective: To determine the rate and spectrum of rare variants in genes involved in melanocortin pathway or hypothalamic development in patients with severe early-onset obesity (height-adjusted weight >60% before age 10 years). Methods: We used a custom-made targeted exome sequencing panel to assess peripheral blood DNA samples for rare (minor allele frequency <0.5%), pathogenic/likely pathogenic variants in 24 genes related to the hypothalamic circuit in 92 subjects (51% males, median age 13.7 years) with early-onset severe obesity (median body mass index (BMI) Z-score + 4.0). Results: We identified a novel frameshift deletion in MC4R (p.V103Afs5*) in two unrelated patients and a previously reported MC4R variant (p.T112M) in one patient. In addition, we identified rare heterozygous missense variants in ADCY3 (p.G1110R), MYT1L (p.R807Q), ISL1 (p.I347F), LRP2 (p.R2479I, and p.N3315S) and a hemizygous missense variant in GRPR (p.L87M) (each in one patient), possibly contributing to the obesity phenotype in these patients. Altogether 8 % (7/92) of the subjects had rare pathogenic/likely pathogenic variants in the studied genes. Conclusions: Rare genetic variants within the hypothalamic circuit are prevalent and contribute to the development of severe early-onset obesity. Targeted exome sequencing is useful in identifying affected subjects. Further studies are needed to evaluate the variants' clinical significance and to define optimal treatment.


Assuntos
Regulação do Apetite/genética , Biomarcadores/análise , Hipotálamo/patologia , Obesidade Mórbida/diagnóstico , Obesidade Pediátrica/diagnóstico , Polimorfismo de Nucleotídeo Único , Adolescente , Idade de Início , Criança , Feminino , Finlândia/epidemiologia , Seguimentos , Humanos , Hipotálamo/metabolismo , Masculino , Obesidade Mórbida/epidemiologia , Obesidade Mórbida/genética , Obesidade Pediátrica/epidemiologia , Obesidade Pediátrica/genética , Linhagem , Fenótipo , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...