Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 115
1.
PLoS One ; 17(1): e0262577, 2022.
Article En | MEDLINE | ID: mdl-35020776

cAMP responsive element binding protein (CREB)-regulated transcription coactivators (CRTCs) regulate gene transcription in response to an increase in intracellular cAMP or Ca2+ levels. To date, three isoforms of CRTC have been identified in mammals. All CRTCs are widely expressed in various regions of the brain. Numerous studies have shown the importance of CREB and CRTC in energy homeostasis. In the brain, the paraventricular nucleus of the hypothalamus (PVH) plays a critical role in energy metabolism, and CRTC1 and CRTC2 are highly expressed in PVH neuronal cells. The single-minded homolog 1 gene (Sim1) is densely expressed in PVH neurons and in some areas of the amygdala neurons. To determine the role of CRTCs in PVH on energy metabolism, we generated mice that lacked CRTC1 and CRTC2 in Sim1 cells using Sim-1 cre mice. We found that Sim1 cell-specific CRTC1 and CRTC2 double-knockout mice were sensitive to high-fat diet (HFD)-induced obesity. Sim1 cell-specific CRTC1 and CRTC2 double knockout mice showed hyperphagia specifically for the HFD, but not for the normal chow diet, increased fat mass, and no change in energy expenditure. Interestingly, these phenotypes were stronger in female mice than in male mice, and a weak phenotype was observed in the normal chow diet. The lack of CRTC1 and CRTC2 in Sim1 cells changed the mRNA levels of some neuropeptides that regulate energy metabolism in female mice fed an HFD. Taken together, our findings suggest that CRTCs in Sim1 cells regulate gene expression and suppress excessive fat intake, especially in female mice.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Diet, High-Fat , Energy Metabolism , Hyperphagia/pathology , Obesity/pathology , Repressor Proteins/metabolism , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Feeding Behavior , Female , Hyperphagia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Repressor Proteins/genetics
2.
Cell Rep ; 36(5): 109481, 2021 08 03.
Article En | MEDLINE | ID: mdl-34348145

Preadipocytes dynamically produce sensory cilia. However, the role of primary cilia in preadipocyte differentiation and adipose homeostasis remains poorly understood. We previously identified transition fiber component FBF1 as an essential player in controlling selective cilia import. Here, we establish Fbf1tm1a/tm1a mice and discover that Fbf1tm1a/tm1a mice develop severe obesity, but surprisingly, are not predisposed to adverse metabolic complications. Obese Fbf1tm1a/tm1a mice possess unexpectedly healthy white fat tissue characterized by spontaneous upregulated beiging, hyperplasia but not hypertrophy, and low inflammation along the lifetime. Mechanistically, FBF1 governs preadipocyte differentiation by constraining the beiging program through an AKAP9-dependent, cilia-regulated PKA signaling, while recruiting the BBS chaperonin to transition fibers to suppress the hedgehog signaling-dependent adipogenic program. Remarkably, obese Fbf1tm1a/tm1a mice further fed a high-fat diet are protected from diabetes and premature death. We reveal a central role for primary cilia in the fate determination of preadipocytes and the generation of metabolically healthy fat tissue.


Adaptor Proteins, Signal Transducing/deficiency , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , 3T3-L1 Cells , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/metabolism , Adipogenesis , Animals , Cell Respiration , Cilia/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Fibroblasts/metabolism , Hedgehog Proteins/metabolism , Homozygote , Humans , Hyperphagia/complications , Hyperphagia/pathology , Hyperplasia , Inflammation/pathology , Male , Metabolic Syndrome/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Obesity/complications , Signal Transduction , Transcription Factors/metabolism
3.
J Pediatr Endocrinol Metab ; 34(8): 971-978, 2021 Aug 26.
Article En | MEDLINE | ID: mdl-34147046

OBJECTIVES: Obesity is often the result of a high-calorie and unbalanced diet for a long time and can sometimes be associated with hyperphagia and eating disorders. Neurotensin (NT) is an anorexigenic peptide, which is secreted from the central nervous system and intestines, and increases intestinal fat absorption. In the literature, conflicting results regarding serum NT level in obesity and the relation of NT with metabolic parameters were reported. Besides, there is no data regarding the relation of NT with eating disorders or food preference in obese individuals. We aimed to evaluate the relation of serum NT level with metabolic parameters, hyperphagia, binge eating disorder (BED) and food preference in obese adolescents. METHODS: The study included 65 obese adolescents and 65 healthy controls. Anthropometric measurements, biochemical analyzes and body fat analyzes were performed in all cases. Hyperphagia score, presence of BED and three-day food intake records were also evaluated. RESULTS: NT level was significantly higher in obese adolescents than in controls and it was not associated with metabolic parameters, hyperphagia or food preference. In the obese group, NT level was not significantly different according to the presence of BED. CONCLUSIONS: Serum NT level is high in obese adolescents; however, it is not associated with metabolic parameters, hyperphagia, BED or food preference.


Biomarkers/blood , Energy Intake , Food Preferences/physiology , Hyperphagia/pathology , Neurotensin/blood , Pediatric Obesity/physiopathology , Adolescent , Case-Control Studies , Child , Female , Follow-Up Studies , Humans , Hyperphagia/blood , Male , Prognosis
4.
Poult Sci ; 100(2): 973-981, 2021 Feb.
Article En | MEDLINE | ID: mdl-33518151

It is known that nutrition and immunity are connected, but the mechanism is not very clear. Endogenous retroviruses (ERV) account for 8 to 10% of the human and mouse genomes and play an important role in some biological processes of animals. Recent studies indicate that the activation of ERV can affect the expression of the immunity- or inflammation-related genes, and the activities of ERV are subjected to regulation of many factors including nutritional factors. Therefore, we hypothesize that nutritional status can affect the expression of the immunity- or inflammation-related genes via ERV. To verify this hypothesis, the nutritional status of animals was altered by fasting or overfeeding, and the expression of intact ERV (ERVK18P, ERVK25P) and immunity- or inflammation-related genes (DDX41, IFIH1, IFNG, IRF7, STAT3) in the liver was determined by quantitative PCR, followed by overexpressing ERVK25P in goose primary hepatocytes and determining the expression of the immunity- or inflammation-related genes. The data showed that compared with the control group (no fasting), the expression of ERV and the immunity- or inflammation-related genes was increased in the liver of the fasted chickens but decreased in the liver of the fasted geese. Moreover, compared with the control group (routinely fed), the expression of ERV and the immunity- or inflammation-related genes was increased in the liver of the overfed geese. In addition, overexpression of ERVK25P in goose primary hepatocytes can induce the expression of the immunity- or inflammation-related genes. In conclusion, these findings suggest that ERV mediate the effects of fasting and overfeeding on the expression of the immunity- or inflammation-related genes, the mediation varied with poultry species, and ERV and the immunity- or inflammation-related genes may be involved in the development of goose fatty liver. This study provides a potential mechanism for the connection between nutrition and immunity.


Endogenous Retroviruses/physiology , Fasting/physiology , Hyperphagia/genetics , Poultry/genetics , Animals , Chickens , Hyperphagia/immunology , Hyperphagia/pathology , Immunity/physiology , Inflammation/genetics , Inflammation/veterinary , Liver/immunology , Liver/pathology , Mice , Poultry/immunology
5.
Pediatr Res ; 89(1): 143-149, 2021 01.
Article En | MEDLINE | ID: mdl-32305038

BACKGROUND: Increasing evidence suggests that postnatal overfeeding induces childhood obesity, which is strongly associated with metabolic syndrome. Insulin resistance is a risk factor for metabolic syndrome. MicroRNA-221 (miR-221) is involved in the development of obesity and has been reported to negatively regulate insulin sensitivity. However, the underlying mechanism remains unclear. METHODS: Rats raised in small litters (SLs, three pups/dam, n = 10) and normal litters (NLs, 10 pups/dam, n = 10) were used to model early postnatal overfeeding and act as controls, respectively. miR-221 and proteins related to the phosphoinositide 3-kinases (PI3K)/protein kinase B (AKT) pathway were assessed in the liver. RESULTS: Early postnatal overfeeding significantly increased body weight, visceral fat index, blood glucose, serum triglycerides, and the homeostasis model assessment of insulin resistance at 9 weeks. Real-time polymerase chain reaction (PCR) and western blot analysis revealed that postnatal overfeeding induced insulin receptor and insulin receptor substrate 2 expression, but decreased PI3K and AKT phosphorylation in the liver. Quantitative real-time PCR showed that hepatic miR-221 was significantly overexpressed in the SL group. CONCLUSIONS: These results indicate that postnatal overfeeding induces hepatic miR-221 overexpression and impairs the PI3K/AKT signal pathway, which may cause insulin resistance. IMPACT: We first report postnatal overfeeding induces hepatic miR-221 expression. Postnatal overfeeding impairs PI3K/AKT pathway in the liver of adult rats. Postnatal overfeeding induces obesity and high blood glucose. Avoidance of overfeeding during early postnatal life may prevent obesity and T2DM.


Hyperphagia/enzymology , Insulin Resistance , Liver/enzymology , MicroRNAs/metabolism , Obesity/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adiposity , Animals , Disease Models, Animal , Hyperphagia/genetics , Hyperphagia/pathology , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/genetics , Liver/pathology , Male , MicroRNAs/genetics , Obesity/genetics , Obesity/pathology , Phosphatidylinositol 3-Kinase/genetics , Phosphorylation , Rats, Sprague-Dawley , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction , Weight Gain
6.
Sci Rep ; 10(1): 18581, 2020 10 29.
Article En | MEDLINE | ID: mdl-33122657

Antipsychotic drugs (AP) are used to treat a multitude of psychiatric conditions including schizophrenia and bipolar disorder. However, APs also have metabolic side effects including increased food intake and body weight, but the underlying mechanisms remain unknown. We previously reported that minocycline (MINO) co-treatment abrogates olanzapine (OLZ)-induced hyperphagia and weight gain in mice. Using this model, we investigated the changes in the pharmacometabolome in the plasma and hypothalamus associated with OLZ-induced hyperphagia and weight gain. Female C57BL/6 mice were divided into groups and fed either i) control, CON (45% fat diet) ii) CON + MINO, iii) OLZ (45% fat diet with OLZ), iv) OLZ + MINO. We identified one hypothalamic metabolite indoxylsulfuric acid and 389 plasma metabolites (including 19 known metabolites) that were specifically associated with AP-induced hyperphagia and weight gain in mice. We found that plasma citrulline, tricosenoic acid, docosadienoic acid and palmitoleic acid were increased while serine, asparagine and arachidonic acid and its derivatives were decreased in response to OLZ. These changes were specifically blocked by co-treatment with MINO. These pharmacometabolomic profiles associated with AP-induced hyperphagia and weight gain provide candidate biomarkers and mechanistic insights related to the metabolic side effects of these widely used drugs.


Eating/drug effects , Hyperphagia/metabolism , Metabolome/drug effects , Minocycline/pharmacology , Olanzapine/toxicity , Weight Gain , Animals , Anti-Bacterial Agents/pharmacology , Antipsychotic Agents/toxicity , Female , Hyperphagia/chemically induced , Hyperphagia/drug therapy , Hyperphagia/pathology , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice , Mice, Inbred C57BL
7.
Genes (Basel) ; 11(4)2020 04 21.
Article En | MEDLINE | ID: mdl-32326226

To evaluate the potential role of ATP-sensitive potassium (KATP) channel activation in the treatment of hyperphagic obesity, a PubMed search was conducted focused on the expression of genes encoding the KATP channel, the response to activating the KATP channel in tissues regulating appetite and the establishment and maintenance of obesity, the evaluation of KATP activators in obese hyperphagic animal models, and clinical studies on syndromic obesity. KATP channel activation is mechanistically involved in the regulation of appetite in the arcuate nucleus; the regulation of hyperinsulinemia, glycemic control, appetite and satiety in the dorsal motor nucleus of vagus; insulin secretion by ß-cells; and the synthesis and ß-oxidation of fatty acids in adipocytes. KATP channel activators have been evaluated in hyperphagic obese animal models and were shown to reduce hyperphagia, induce fat loss and weight loss in older animals, reduce the accumulation of excess body fat in growing animals, reduce circulating and hepatic lipids, and improve glycemic control. Recent experience with a KATP channel activator in Prader-Willi syndrome is consistent with the therapeutic responses observed in animal models. KATP channel activation, given the breadth of impact and animal model and clinical results, is a viable target in hyperphagic obesity.


Anti-Obesity Agents/pharmacology , Hyperphagia/drug therapy , Insulin Secretion/drug effects , KATP Channels/agonists , Obesity/drug therapy , Animals , Humans , Hyperphagia/metabolism , Hyperphagia/pathology , Obesity/metabolism , Obesity/pathology
8.
Horm Behav ; 120: 104690, 2020 04.
Article En | MEDLINE | ID: mdl-31954709

Changes to neonatal nutrition result in long-lasting impairments in energy balance, which may be described as metabolic programing. Astrocytes, which are interconnected by gap junctions, have emerged as important players in the hypothalamic control of food intake. In order to study the effects of nutritional programming on glial morphology and protein expression, cross-fostered male Wistar rats at postnatal day 3 were assigned to three groups based on litter size: small litter (3 pups per dam, SL), normal litter (10 pups per dam, NL), and large litter (16 pups per dam, LL). Rats from the SL group exhibited higher body weight throughout the study and hyperphagia after weaning. LL animals exhibited hyperphagia, high energy efficiency and catch-up of body weight after weaning. Both the SL and LL groups at postnatal day 60 (PN60) exhibited increased levels of plasma leptin, the Lee index (as an index of obesity), adiposity content, immunoreactivity toward T-cell protein tyrosine phosphatase (TCPTP), and glial fibrillary acidic protein (GFAP) in the arcuate nucleus (ARC) of the hypothalamus. Astrocyte morphology was altered in the ARC of SL and LL animals, and this effect occurred in parallel with a reduction in immunoreactivity toward connexin 30 (CX30). The data obtained demonstrate that both neonatal over- and underfeeding promote not only alterations in the metabolic status but also morphological changes in glial cells in parallel with increasing TCPTP and changes in connexin expression.


Animal Nutritional Physiological Phenomena , Connexins/genetics , Gliosis/etiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Adiposity/physiology , Animals , Animals, Newborn , Connexins/metabolism , Female , Gene Expression Regulation, Developmental , Gliosis/genetics , Gliosis/metabolism , Hyperphagia/complications , Hyperphagia/genetics , Hyperphagia/metabolism , Hyperphagia/pathology , Hypothalamus/metabolism , Litter Size/physiology , Male , Obesity/complications , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Pregnancy , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Rats , Rats, Wistar , Sex Factors , Time Factors
9.
Acta Physiol (Oxf) ; 228(2): e13345, 2020 02.
Article En | MEDLINE | ID: mdl-31310704

AIM: Since foods with high hedonic value are often consumed in excess of energetic needs, this study was designed to identify the mechanisms that may counter anorexigenic signalling in the presence of hedonic foods in lean animals. METHODS: Mice, in different states of satiety (fed/fasted, or fed/fasted and treated with ghrelin or leptin, respectively), were allowed to choose between high-fat/high-sucrose and standard foods. Intake of each food type and the activity of hypothalamic neuropetidergic neurons that regulate appetite were monitored. In some cases, food choice was monitored in leptin-injected fasted mice that received microinjections of galanin receptor agonists into the lateral hypothalamus. RESULTS: Appetite-stimulating orexin neurons in the lateral hypothalamus are rapidly activated when lean, satiated mice consume a highly palatable food (PF); such activation (upregulated c-Fos expression) occurred even after administration of the anorexigenic hormone leptin and despite intact leptin signalling in the hypothalamus. The ability of leptin to restrain PF eating is restored when a galanin receptor 2 (Gal2R) agonist is injected into the lateral hypothalamus. CONCLUSION: Hedonically-loaded foods interrupt the inhibitory actions of leptin on orexin neurons and interfere with the homeostatic control of feeding. Overeating of palatable foods can be curtailed in lean animals by activating Gal2R in the lateral hypothalamus.


Eating/physiology , Hyperphagia/prevention & control , Hypothalamic Area, Lateral/drug effects , Leptin/pharmacology , Neurons/metabolism , Receptor, Galanin, Type 2/agonists , Animals , Disease Models, Animal , Eating/drug effects , Galanin/pharmacology , Ghrelin/metabolism , Hyperphagia/metabolism , Hyperphagia/pathology , Hypothalamic Area, Lateral/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Orexins/metabolism , Receptor, Galanin, Type 2/metabolism
10.
Am J Med Genet A ; 182(1): 53-63, 2020 01.
Article En | MEDLINE | ID: mdl-31729827

This study presents a broad overview of health issues and psychomotor development of 100 children with Angelman syndrome (AS), seen at the ENCORE Expertise Center for AS in Rotterdam, the Netherlands. We aimed to further delineate the phenotype of AS, to evaluate the association of the phenotype with genotype and other determinants such as epilepsy and to get insight in possible targets for intervention. We confirmed the presence of a more severe phenotype in the 15q11.2-q13 deletion subtype. Novel findings were an association of (early onset of) epilepsy with a negative effect on development, a high occurrence of nonconvulsive status epilepticus, a high rate of crouch gait in the older children with risk of deterioration of mobility, a relatively low occurrence of microcephaly, a higher mean weight for height in all genetic subtypes with a significant higher mean in the nondeletion children, and a high occurrence of hyperphagia across all genetic subtypes. Natural history data are needed to design future trials. With this large clinical cohort with structured prospective and multidisciplinary follow-up, we provide unbiased data on AS to support further intervention studies to optimize outcome and quality of life of children with AS and their family.


Angelman Syndrome/genetics , Epilepsy/genetics , Genetic Predisposition to Disease , Ubiquitin-Protein Ligases/genetics , Adolescent , Angelman Syndrome/epidemiology , Angelman Syndrome/physiopathology , Child , Child, Preschool , Chromosomes, Human, Pair 15/genetics , Cohort Studies , Epilepsy/physiopathology , Female , Genetic Association Studies , Genotype , Humans , Hyperphagia/genetics , Hyperphagia/pathology , Male , Microcephaly/genetics , Microcephaly/pathology , Netherlands/epidemiology , Phenotype , Psychomotor Performance/physiology
11.
Nat Med ; 25(11): 1733-1738, 2019 11.
Article En | MEDLINE | ID: mdl-31700171

The G-protein-coupled receptor accessory protein MRAP2 is implicated in energy control in rodents, notably via the melanocortin-4 receptor1. Although some MRAP2 mutations have been described in people with obesity1-3, their functional consequences on adiposity remain elusive. Using large-scale sequencing of MRAP2 in 9,418 people, we identified 23 rare heterozygous variants associated with increased obesity risk in both adults and children. Functional assessment of each variant shows that loss-of-function MRAP2 variants are pathogenic for monogenic hyperphagic obesity, hyperglycemia and hypertension. This contrasts with other monogenic forms of obesity characterized by excessive hunger, including melanocortin-4 receptor deficiency, that present with low blood pressure and normal glucose tolerance4. The pleiotropic metabolic effect of loss-of-function mutations in MRAP2 might be due to the failure of different MRAP2-regulated G-protein-coupled receptors in various tissues including pancreatic islets.


Adaptor Proteins, Signal Transducing/genetics , Genetic Predisposition to Disease , Hyperphagia/genetics , Obesity/genetics , Adolescent , Adult , Child , Energy Metabolism/genetics , Female , Humans , Hyperglycemia/complications , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperglycemia/pathology , Hyperphagia/complications , Hyperphagia/metabolism , Hyperphagia/pathology , Hypertension/complications , Hypertension/genetics , Hypertension/metabolism , Hypertension/pathology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Loss of Function Mutation/genetics , Male , Middle Aged , Obesity/complications , Obesity/metabolism , Obesity/pathology , Receptor, Melanocortin, Type 4/genetics , Risk Factors , Young Adult
12.
Mol Cell Endocrinol ; 498: 110574, 2019 12 01.
Article En | MEDLINE | ID: mdl-31494175

Several hypothalamic neuronal populations are directly responsive to growth hormone (GH) and central GH action regulates glucose and energy homeostasis. However, the potential role of GH signaling in proopiomelanocortin (POMC) neurons has not been studied yet. Thus, we investigated whether POMC neurons are responsive to GH and if ablation of GH receptor (GHR) or STAT5 in POMC cells leads to metabolic imbalances. Approximately 60% of POMC neurons of the arcuate nucleus exhibited STAT5 phosphorylation after intracerebroventricular GH injection. Ablation of GHR or STAT5 in POMC cells did not affect energy or glucose homeostasis. However, glucoprivic hyperphagia was blunted in male and female GHR knockout mice, and in male POMC-specific STAT5 knockout mice. Additionally, the absence of GHR in POMC neurons decreased glycemia during prolonged food restriction in male mice. Thus, GH action in POMC neurons regulates glucoprivic hyperphagia as well as blood glucose levels during prolonged food restriction.


Carrier Proteins/physiology , Glucose/metabolism , Hyperphagia/pathology , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , STAT5 Transcription Factor/physiology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Female , Hyperphagia/metabolism , Male , Mice , Mice, Knockout
13.
C R Biol ; 342(5-6): 209-219, 2019.
Article En | MEDLINE | ID: mdl-31151779

The aim of this study was to show, for the first time, the effect of a hypercaloric diet on the mitochondrial reshuffle of hepatocytes during the progression from steatosis to steatohepatitis to cirrhosis in Psammomys obesus, a typical animal model of the metabolic syndrome. Metabolic and oxidative stresses were induced by feeding the animal through a standard laboratory diet (SD) for nine months. Metabolic parameters, liver malondialdehyde (MDA) and glutathione (GSH), were evaluated. The pathological evolution was examined by histopathology and immunohistochemistry, using CD3 and CD20 antibodies. The dynamics of the mitochondrial structure was followed by transmission electron microscopy. SD induced a steatosis in this animal that evolved under the effect of oxidative and metabolic stress by the appearance of adaptive inflammation and fibrosis leading the animal to the cirrhosis stage with serious hepatocyte damage by the triggering, at first the mitochondrial fusion-fission cycles, which attempted to maintain the mitochondria intact and functional, but the hepatocellular oxidative damage was increased inducing a vicious circle of mitochondrial alteration and dysfunction and their elimination by mitophagy. P. obesus is an excellent animal model of therapeutic research that targets mitochondrial dysfunction in the progression of steatosis.


Gerbillinae , Hyperphagia/metabolism , Insulin Resistance , Liver/metabolism , Mitochondria, Liver/metabolism , Neutrophil Infiltration , Oxidative Stress , Animals , Diet , Disease Models, Animal , Disease Progression , Fatty Liver/metabolism , Fatty Liver/pathology , Glutathione/metabolism , Hyperphagia/pathology , Inflammation/metabolism , Inflammation/pathology , Liver/pathology , Male , Malondialdehyde/metabolism , Mitochondria, Liver/pathology
14.
Rev Endocr Metab Disord ; 20(2): 239-250, 2019 06.
Article En | MEDLINE | ID: mdl-31065942

Prader-Willi syndrome (PWS) is a genetic disorder characterized by short stature, low lean body mass, muscular hypotonia, mental retardation, behavioral abnormalities, dysmorphic features, and excessive appetite with progressive obesity. It is caused by lack of expression of genes on the paternally inherited chromosome 15q11.2-q13. This genetic disorder has an estimated prevalence that ranges between 1/10,000-1/30,000. Hypothalamic dysfunction is a common finding in PWS and it has been implicated in several manifestations of this syndrome such as hyperphagia, temperature instability, high pain threshold, sleep disordered breathing, and multiple endocrine abnormalities. These include growth hormone deficiency, central adrenal insufficiency, hypogonadism, hypothyroidism, and obesity often complicated by type 2 diabetes. The aim of this manuscript is to overview the current literature on metabolic and endocrine complications of PWS, focusing on human studies and providing insights on the physio pathological mechanisms. A careful management of metabolic and endocrine complications can contribute to improve quality of life, prevent complications, and prolong life expectancy of PW patients.


Prader-Willi Syndrome/metabolism , Prader-Willi Syndrome/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Hyperphagia/metabolism , Hyperphagia/pathology , Quality of Life
15.
Sci Rep ; 9(1): 6989, 2019 05 06.
Article En | MEDLINE | ID: mdl-31061470

Obesity is a progressive, chronic disease, which can be caused by long-term miscommunication between organs. It remains challenging to understand how chronic dysfunction in a particular tissue remotely impairs other organs to eventually imbalance organismal energy homeostasis. Here we introduce RNAi Pulse Induction (RiPI) mediated by short hairpin RNA (shRiPI) or double-stranded RNA (dsRiPI) to generate chronic, organ-specific gene knockdown in the adult Drosophila fat tissue. We show that organ-restricted RiPI targeting Stromal interaction molecule (Stim), an essential factor of store-operated calcium entry (SOCE), results in progressive fat accumulation in fly adipose tissue. Chronic SOCE-dependent adipose tissue dysfunction manifests in considerable changes of the fat cell transcriptome profile, and in resistance to the glucagon-like Adipokinetic hormone (Akh) signaling. Remotely, the adipose tissue dysfunction promotes hyperphagia likely via increased secretion of Akh from the neuroendocrine system. Collectively, our study presents a novel in vivo paradigm in the fly, which is widely applicable to model and functionally analyze inter-organ communication processes in chronic diseases.


Adipose Tissue/metabolism , Calcium/metabolism , Drosophila Proteins/genetics , Hyperphagia/genetics , Insect Hormones/genetics , Obesity/genetics , Oligopeptides/genetics , Pyrrolidonecarboxylic Acid/analogs & derivatives , Stromal Interaction Molecule 1/genetics , Adipose Tissue/pathology , Animals , Aspartate Aminotransferase, Cytoplasmic/genetics , Aspartate Aminotransferase, Cytoplasmic/metabolism , Calcium Signaling , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila melanogaster , Energy Metabolism/genetics , Female , Gene Expression Regulation , Homeostasis/genetics , Humans , Hyperphagia/metabolism , Hyperphagia/pathology , Insect Hormones/metabolism , Ion Transport , Isoenzymes/genetics , Isoenzymes/metabolism , Lipid Metabolism/genetics , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , Male , Obesity/metabolism , Obesity/pathology , Oligopeptides/metabolism , Pyrrolidonecarboxylic Acid/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stromal Interaction Molecule 1/antagonists & inhibitors , Stromal Interaction Molecule 1/metabolism
16.
J Clin Invest ; 129(6): 2417-2430, 2019 04 02.
Article En | MEDLINE | ID: mdl-30938715

Glial cells have emerged as key players in the central control of energy balance and etiology of obesity. Astrocytes play a central role in neural communication via the release of gliotransmitters. Acyl-CoA binding protein (ACBP)-derived endozepines are secreted peptides that modulate the GABAA receptor. In the hypothalamus, ACBP is enriched in arcuate nucleus (ARC) astrocytes, ependymocytes and tanycytes. Central administration of the endozepine octadecaneuropeptide (ODN) reduces feeding and improves glucose tolerance, yet the contribution of endogenous ACBP in energy homeostasis is unknown. We demonstrated that ACBP deletion in GFAP+ astrocytes, but not in Nkx2.1-lineage neural cells, promoted diet-induced hyperphagia and obesity in both male and female mice, an effect prevented by viral rescue of ACBP in ARC astrocytes. ACBP-astrocytes were observed in apposition with proopiomelanocortin (POMC) neurons and ODN selectively activated POMC neurons through the ODN-GPCR but not GABAA, and supressed feeding while increasing carbohydrate utilization via the melanocortin system. Similarly, ACBP overexpression in ARC astrocytes reduced feeding and weight gain. Finally, the ODN-GPCR agonist decreased feeding and promoted weight loss in ob/ob mice. These findings uncover ACBP as an ARC gliopeptide playing a key role in energy balance control and exerting strong anorectic effects via the central melanocortin system.


Astrocytes/metabolism , Diazepam Binding Inhibitor/metabolism , Eating , Energy Metabolism , Hyperphagia/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Astrocytes/pathology , Cell Line , Diazepam Binding Inhibitor/genetics , Female , Hyperphagia/genetics , Hyperphagia/pathology , Male , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Obesity/genetics , Obesity/pathology , Pro-Opiomelanocortin/genetics
17.
J Neurosci ; 39(15): 2810-2822, 2019 04 10.
Article En | MEDLINE | ID: mdl-30737307

Neurogenesis in the adult brain, a powerful mechanism for neuronal plasticity and brain repair, is altered by aging and pathological conditions, including metabolic disorders. The search for mechanisms and therapeutic solutions to alter neurogenesis requires understanding of cell kinetics within neurogenic niches using a high-throughput quantitative approach. The challenge is in the dynamic nature of the process and multiple cell types involved, each having several potential modes of division or cell fate. Here we show that cell kinetics can be revealed through a combination of the BrdU/EdU pulse-chase, based on the circadian pattern of DNA replication, and a differential equations model that describes time-dependent cell densities. The model is validated through the analysis of cell kinetics in the cerebellar neurogenic niche of normal young adult male zebrafish, with cells quantified in 2D (sections), and with neuronal fate and reactivation of stem cells confirmed in 3D whole-brain images (CLARITY). We then reveal complex alterations in cell kinetics associated with accelerated aging due to chronic high caloric intake. Low activity of neuronal stem cells in this condition persists 2 months after reverting to normal diet, and is accompanied by overproduction of transient amplifying cells, their accelerated cell death, and slow migration of postmitotic progeny. This combined experimental and mathematical approach should allow for relatively high-throughput analysis of early signs of pathological and age-related changes in neurogenesis, evaluation of specific therapeutic targets, and drug efficacy.SIGNIFICANCE STATEMENT Understanding normal cell kinetics of adult neurogenesis and the type of cells affected by a pathological process is needed to develop effective prophylactic and therapeutic measures directed at specific cell targets. Complex time-dependent mechanisms involved in the kinetics of multiple cell types require a combination of experimental and mathematical modeling approaches. This study demonstrates such a combined approach by comparing normal neurogenesis with that altered by diet-induced accelerated aging in adult zebrafish.


Aging, Premature/pathology , Diet/adverse effects , Energy Intake , Neurogenesis/physiology , Stem Cell Niche/physiology , Zebrafish/physiology , Animals , Brain/diagnostic imaging , Cell Division , Circadian Rhythm , DNA Replication , Hyperphagia/pathology , Kinetics , Magnetic Resonance Imaging , Male , Mitosis , Models, Theoretical , Neural Stem Cells
18.
Nat Commun ; 9(1): 5103, 2018 11 30.
Article En | MEDLINE | ID: mdl-30504766

Palatable foods (fat and sweet) induce hyperphagia, and facilitate the development of obesity. Whether and how overnutrition increases appetite through the adipose-to-brain axis is unclear. O-linked beta-D-N-acetylglucosamine (O-GlcNAc) transferase (OGT) couples nutrient cues to O-GlcNAcylation of intracellular proteins at serine/threonine residues. Chronic dysregulation of O-GlcNAc signaling contributes to metabolic diseases. Here we show that adipocyte OGT is essential for high fat diet-induced hyperphagia, but is dispensable for baseline food intake. Adipocyte OGT stimulates hyperphagia by transcriptional activation of de novo lipid desaturation and accumulation of N-arachidonyl ethanolamine (AEA), an endogenous appetite-inducing cannabinoid (CB). Pharmacological manipulation of peripheral CB1 signaling regulates hyperphagia in an adipocyte OGT-dependent manner. These findings define adipocyte OGT as a fat sensor that regulates peripheral lipid signals, and uncover an unexpected adipose-to-brain axis to induce hyperphagia and obesity.


Adipocytes/metabolism , Adipose Tissue/metabolism , Hyperphagia/metabolism , Hyperphagia/pathology , Obesity/metabolism , Obesity/pathology , Acetylglucosamine/metabolism , Adipose Tissue/pathology , Animals , Blotting, Western , Body Weight/genetics , Body Weight/physiology , Cannabinoids/metabolism , Cell Line , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
19.
Int J Mol Sci ; 19(8)2018 Aug 08.
Article En | MEDLINE | ID: mdl-30096853

Alzheimer's disease (AD), a progressive neurodegenerative disease is highly associated with metabolic syndromes. We previously demonstrated that glycemic dysregulation and obesity are augmented in high fat diet (HFD)-treated APPswe/PS1dE9 (APP/PS1) transgenic mice. In the current study, the underlying mechanism mediating exacerbated metabolic stresses in HFD APP/PS1 transgenic mice was further examined. APP/PS1 mice developed insulin resistance and, consequently, impaired glucose homeostasis after 10 weeks on HFD. [18F]-2-fluoro-2-deoxy-d-glucose ([18F]-FDG) positron emission tomography showed that interscapular brown adipose tissue is vulnerable to HFD and AD-related pathology. Chronic HFD induced hyperphagia, with limited effects on basal metabolic rates in APP/PS1 transgenic mice. Excessive food intake may be caused by impairment of leptin signaling in the hypothalamus because leptin failed to suppress the food intake of HFD APP/PS1 transgenic mice. Leptin-induced pSTAT3 signaling in the arcuate nucleus was attenuated. Dysregulated energy homeostasis including hyperphagia and exacerbated obesity was elicited prior to the presence of the amyloid pathology in the hypothalamus of HFD APP/PS1 transgenic mice; nevertheless, cortical neuroinflammation and the level of serum Aß and IL-6 were significantly elevated. Our study demonstrates the pivotal role of AD-related pathology in augmenting HFD-induced insulin and leptin resistance and impairing hypothalamic regulation of energy homeostasis.


Alzheimer Disease/genetics , Hyperphagia/drug therapy , Insulin Resistance/genetics , Obesity/genetics , Adipose Tissue, Brown/drug effects , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Blood Glucose , Diet, High-Fat/adverse effects , Disease Models, Animal , Eating/genetics , Homeostasis , Humans , Hyperphagia/genetics , Hyperphagia/pathology , Insulin/metabolism , Insulin/therapeutic use , Leptin/metabolism , Leptin/therapeutic use , Metabolic Syndrome/drug therapy , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , Mice , Mice, Transgenic , Obesity/complications , Obesity/pathology
20.
Eur J Med Genet ; 61(8): 465-467, 2018 Aug.
Article En | MEDLINE | ID: mdl-29545012

Congenital Leptin receptor (LEPR) deficiency is a rare genetic cause of early-onset morbid obesity characterised by severe early onset obesity, major hyperphagia, hypogonadotropic hypogonadism and immune and neuroendocrine/metabolic dysfunction. We identified a homozygous loss-of-function mutation, NM_002303.5:c.464 T > G; p.(Tyr155*), in the LEPR in an extended consanguineous family with multiple individuals affected by early-onset severe obesity and hyperphagia. Interestingly, the LEPR-deficient adult females have extremely high body mass index (BMI) with hypogonadal infertility, the BMI of the affected males began to decline around the onset of puberty (13-15 years) with fertility being preserved. These findings lead to the speculation that LEPR deficiency may have a gender-specific effect on the regulation of body weight. In order to elucidate gender-specific effects of LEPR deficiency on reproduction further investigations are needed. The limitations of this study are that our conclusion is based on observations of two males and two females. Further LEPR deficient males and females are required for comparison in order to support this finding more confidently.


Hyperphagia/genetics , Loss of Function Mutation , Obesity/genetics , Receptors, Leptin/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Hyperphagia/pathology , Male , Obesity/pathology , Pedigree , Sex Factors
...