Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.347
Filter
1.
J Agric Food Chem ; 72(28): 15765-15777, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38970495

ABSTRACT

Konjac glucomannan (KGM), high-viscosity dietary fiber, is utilized in weight management. Previous investigations on the appetite-suppressing effects of KGM have centered on intestinal responses to nutrients and gastric emptying rates, with less focus on downstream hypothalamic neurons of satiety hormones. In our studies, the molecular mechanisms through which KGM and its degradation products influence energy homeostasis via the adipocyte-hypothalamic axis have been examined. It was found that high-viscosity KGM more effectively stimulates enteroendocrine cells to release glucagon-like peptide-1 (GLP-1) and reduces ghrelin production, thereby activating hypothalamic neurons and moderating short-term satiety. Conversely, low-viscosity DKGM has been shown to exhibit stronger anti-inflammatory properties in the hypothalamus, enhancing hormone sensitivity and lowering the satiety threshold. Notably, both KGM and DKGM significantly reduced leptin signaling and fatty acid signaling in adipose tissue and activated brown adipose tissue thermogenesis to suppress pro-opiomelanocortin (POMC) expression and activate agouti-related protein (AgRP) expression, thereby reducing food intake and increasing energy expenditure. Additionally, high-viscosity KGM has been found to activate the adipocyte-hypothalamus axis more effectively than DKGM, thereby promoting greater daily energy expenditure. These findings provide novel insights into the adipocyte-hypothalamic axis for KGM to suppress appetite and reduce weight.


Subject(s)
Adipocytes , Appetite Regulation , Diet, High-Fat , Energy Metabolism , Hypothalamus , Mice, Inbred C57BL , Animals , Mice , Energy Metabolism/drug effects , Hypothalamus/metabolism , Hypothalamus/drug effects , Diet, High-Fat/adverse effects , Male , Appetite Regulation/drug effects , Adipocytes/metabolism , Adipocytes/drug effects , Humans , Glucagon-Like Peptide 1/metabolism , Ghrelin/metabolism , Leptin/metabolism , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Thermogenesis/drug effects , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Obesity/metabolism , Obesity/physiopathology , Obesity/diet therapy , Mannans
2.
J Neurosci ; 44(32)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38977301

ABSTRACT

Overexpression of the agouti-signaling protein (asip1), an endogenous melanocortin antagonist, under the control of a constitutive promoter in zebrafish [Tg(Xla.Eef1a1:Cau.Asip1]iim4] (asip1-Tg) increases food intake by reducing sensitivity of the central satiety systems and abolish circadian activity rhythms. The phenotype also shows increased linear growth and body weight, yet no enhanced aggressiveness in dyadic fights is observed. In fact, asip1-Tg animals choose to flee to safer areas rather than face a potential threat, thus suggesting a potential anxiety-like behavior (ALB). Standard behavioral tests, i.e., the open field test (OFT), the novel object test (NOT), and the novel tank dive test (NTDT), were used to investigate thigmotaxis and ALB in male and female zebrafish. Results showed that the asip1-Tg strain exhibited severe ALB in every test, mainly characterized by pronounced freezing behavior and increased linear and angular swimming velocities. asip1-Tg animals exhibited low central serotonin (5-HT) and dopamine (DA) levels and high turnover rates, thus suggesting that central monoaminergic pathways might mediate melanocortin antagonist-induced ALB. Accordingly, the treatment of asip1-Tg animals with fluoxetine, a selective serotonin reuptake inhibitor (SSRI), reversed the ALB phenotype in NTDT as well as 5-HT turnover. Genomic and anatomical data further supported neuronal interaction between melanocortinergic and serotonergic systems. These results suggest that inhibition of the melanocortin system by ubiquitous overexpression of endogenous antagonist has an anxiogenic effect mediated by serotonergic transmission.


Subject(s)
Anxiety , Serotonin , Zebrafish , Animals , Anxiety/metabolism , Anxiety/psychology , Male , Female , Serotonin/metabolism , Animals, Genetically Modified , Behavior, Animal/drug effects , Behavior, Animal/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Fluoxetine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Dopamine/metabolism , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics
3.
Science ; 385(6707): 438-446, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38935778

ABSTRACT

Glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RAs) are effective antiobesity drugs. However, the precise central mechanisms of GLP-1RAs remain elusive. We administered GLP-1RAs to patients with obesity and observed a heightened sense of preingestive satiation. Analysis of human and mouse brain samples pinpointed GLP-1 receptor (GLP-1R) neurons in the dorsomedial hypothalamus (DMH) as candidates for encoding preingestive satiation. Optogenetic manipulation of DMHGLP-1R neurons caused satiation. Calcium imaging demonstrated that these neurons are actively involved in encoding preingestive satiation. GLP-1RA administration increased the activity of DMHGLP-1R neurons selectively during eating behavior. We further identified that an intricate interplay between DMHGLP-1R neurons and neuropeptide Y/agouti-related peptide neurons of the arcuate nucleus (ARCNPY/AgRP neurons) occurs to regulate food intake. Our findings reveal a hypothalamic mechanism through which GLP-1RAs control preingestive satiation, offering previously unexplored neural targets for obesity and metabolic diseases.


Subject(s)
Arcuate Nucleus of Hypothalamus , Dorsomedial Hypothalamic Nucleus , Glucagon-Like Peptide-1 Receptor Agonists , Obesity , Satiation , Animals , Female , Humans , Male , Mice , Agouti-Related Protein/metabolism , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Dorsomedial Hypothalamic Nucleus/drug effects , Dorsomedial Hypothalamic Nucleus/metabolism , Eating/drug effects , Feeding Behavior/drug effects , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Neuropeptide Y/metabolism , Obesity/drug therapy , Obesity/psychology , Optogenetics , Satiation/drug effects , Glucagon-Like Peptide-1 Receptor Agonists/administration & dosage , Glucagon-Like Peptide-1 Receptor Agonists/pharmacology
4.
Mol Metab ; 86: 101975, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38925247

ABSTRACT

OBJECTIVE: The lactational period is associated with profound hyperphagia to accommodate the energy demands of nursing. These changes are important for the long-term metabolic health of the mother and children as altered feeding during lactation increases the risk of mothers and offspring developing metabolic disorders later in life. However, the specific behavioral mechanisms and neural circuitry mediating the hyperphagia of lactation are incompletely understood. METHODS: Here, we utilized home cage feeding devices to characterize the dynamics of feeding behavior in lactating mice. A combination of pharmacological and behavioral assays were utilized to determine how lactation alters meal structure, circadian aspects of feeding, hedonic feeding, and sensitivity to hunger and satiety signals in lactating mice. Finally, we utilized chemogenetic, immunohistochemical, and in vivo imaging approaches to characterize the role of hypothalamic agouti-related peptide (AgRP) neurons in lactational-hyperphagia. RESULTS: The lactational period is associated with increased meal size, altered circadian patterns of feeding, reduced sensitivity to gut-brain satiety signals, and enhanced sensitivity to negative energy balance. Hypothalamic AgRP neurons display increased sensitivity to negative energy balance and altered in vivo activity during the lactational state. Further, using in vivo imaging approaches we demonstrate that AgRP neurons are directly activated by lactation. Chemogenetic inhibition of AgRP neurons acutely reduces feeding in lactating mice, demonstrating an important role for these neurons in lactational-hyperphagia. CONCLUSIONS: Together, these results show that lactation collectively alters multiple components of feeding behavior and position AgRP neurons as an important cellular substrate mediating the hyperphagia of lactation.


Subject(s)
Agouti-Related Protein , Feeding Behavior , Hyperphagia , Hypothalamus , Lactation , Neurons , Animals , Agouti-Related Protein/metabolism , Lactation/metabolism , Hyperphagia/metabolism , Female , Mice , Neurons/metabolism , Hypothalamus/metabolism , Feeding Behavior/physiology , Energy Metabolism , Mice, Inbred C57BL
5.
Metabolism ; 158: 155959, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38942170

ABSTRACT

OBJECTIVE: The molecular control of feeding after fasting is essential for maintaining energy homeostasis, while overfeeding usually leads to obesity. Identifying non-coding microRNAs (miRNAs) that control food intake could reveal new oligonucleotide-based therapeutic targets for treating obesity and its associated diseases. This study aims to identify a miRNA modulating food intake and its mechanism in neuronal regulation of food intake and energy homeostasis. METHODS: A comprehensive genome-wide miRNA screening in the arcuate nucleus of the hypothalamus (ARC) of fasted mice and ad libitum mice was performed. Through stereotactic virus injections, intracerebroventricular injections, and miRNA sponge technology, miR-7a-5p was inhibited specifically in AgRP neurons and the central nervous system, and metabolic phenotypes were monitored. Quantitative real-time PCR, Western blotting, immunofluorescence, whole-cell patch-clamp recording, and luciferase reporter assay were used to investigate the mechanisms underlying miR-7a-5p's regulation of food intake. RESULTS: We found a significant increase in miR-7a-5p levels after fasting. miR-7a-5p was highly expressed in the ARC, and inhibition of miR-7a-5p specifically in AgRP neurons reduced food intake and body weight gain. miR-7a-5p inhibited S6K1 gene expression by binding to its 3'-UTR. Furthermore, the knockdown of ribosomal S6 kinase 1 (S6K1) in AgRP neurons can partially reverse the effects caused by miR-7a-5p inhibition. Importantly, intracerebroventricular administration of the miR-7a-5p inhibitor could also reduce food intake and body weight gain. CONCLUSION: Our findings suggest that miR-7a-5p responds to energy deficit and regulates food intake by fine-tuning mTOR1/S6K1 signaling in the AgRP neurons, which could be a promising oligonucleotide-based therapeutic target for treating obesity and its associated diseases.


Subject(s)
Agouti-Related Protein , Eating , Fasting , MicroRNAs , Neurons , Animals , MicroRNAs/genetics , Neurons/metabolism , Mice , Fasting/physiology , Eating/physiology , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Male , Arcuate Nucleus of Hypothalamus/metabolism , Mice, Inbred C57BL , TOR Serine-Threonine Kinases/metabolism
6.
Nutr Diabetes ; 14(1): 40, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38844453

ABSTRACT

BACKGROUND: High-protein diets are often enriched with branched-chain amino acids (BCAAs) known to enhance protein synthesis and provide numerous physiological benefits, but recent studies reveal their association with obesity and diabetes. In support of this, protein or BCAA supplementation is shown to disrupt glucose metabolism while restriction improves it. However, it is not clear if these are primary, direct effects of BCAAs or secondary to other physiological changes during chronic manipulation of dietary BCAAs. METHODS: Three-month-old C57Bl/6 mice were acutely treated with either vehicle/BCAAs or BT2, a BCAA-lowering compound, and detailed in vivo metabolic phenotyping, including frequent sampling and pancreatic clamps, were conducted. RESULTS: Using a catheter-guided frequent sampling method in mice, here we show that a single infusion of BCAAs was sufficient to acutely elevate blood glucose and plasma insulin. While pre-treatment with BCAAs did not affect glucose tolerance, a constant infusion of BCAAs during hyperinsulinemic-euglycemic clamps impaired whole-body insulin sensitivity. Similarly, a single injection of BT2 was sufficient to prevent BCAA rise during fasting and markedly improve glucose tolerance in high-fat-fed mice, suggesting that abnormal glycemic control in obesity may be causally linked to high circulating BCAAs. We further show that chemogenetic over-activation of AgRP neurons in the hypothalamus, as present in obesity, significantly impairs glucose tolerance that is completely normalized by acute BCAA reduction. Interestingly, most of these effects were demonstrated only in male, but not in female mice. CONCLUSION: These findings suggest that BCAAs per se can acutely impair glucose homeostasis and insulin sensitivity, thus offering an explanation for how they may disrupt glucose metabolism in the long-term as observed in obesity and diabetes. Our findings also reveal that AgRP neuronal regulation of blood glucose is mediated through BCAAs, further elucidating a novel mechanism by which brain controls glucose homeostasis.


Subject(s)
Agouti-Related Protein , Amino Acids, Branched-Chain , Blood Glucose , Insulin Resistance , Mice, Inbred C57BL , Neurons , Animals , Insulin Resistance/physiology , Agouti-Related Protein/metabolism , Neurons/metabolism , Neurons/drug effects , Male , Mice , Blood Glucose/metabolism , Female , Amino Acids, Branched-Chain/metabolism , Insulin/blood , Insulin/metabolism , Glucose Clamp Technique , Diet, High-Fat , Obesity/metabolism
7.
Nat Commun ; 15(1): 5439, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937485

ABSTRACT

Efficient control of feeding behavior requires the coordinated adjustment of complex motivational and affective neurocircuits. Neuropeptides from energy-sensing hypothalamic neurons are potent feeding modulators, but how these endogenous signals shape relevant circuits remains unclear. Here, we examine how the orexigenic neuropeptide Y (NPY) adapts GABAergic inputs to the bed nucleus of the stria terminalis (BNST). We find that fasting increases synaptic connectivity between agouti-related peptide (AgRP)-expressing 'hunger' and BNST neurons, a circuit that promotes feeding. In contrast, GABAergic input from the central amygdala (CeA), an extended amygdala circuit that decreases feeding, is reduced. Activating NPY-expressing AgRP neurons evokes these synaptic adaptations, which are absent in NPY-deficient mice. Moreover, fasting diminishes the ability of CeA projections in the BNST to suppress food intake, and NPY-deficient mice fail to decrease anxiety in order to promote feeding. Thus, AgRP neurons drive input-specific synaptic plasticity, enabling a selective shift in hunger and anxiety signaling during starvation through NPY.


Subject(s)
Agouti-Related Protein , Feeding Behavior , Neuronal Plasticity , Neuropeptide Y , Septal Nuclei , Starvation , Animals , Neuropeptide Y/metabolism , Neuropeptide Y/genetics , Neuronal Plasticity/physiology , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Feeding Behavior/physiology , Septal Nuclei/metabolism , Septal Nuclei/physiology , Mice , Starvation/metabolism , Male , Amygdala/metabolism , Amygdala/physiology , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/physiology , GABAergic Neurons/metabolism , Eating/physiology , Fasting/physiology , Anxiety/metabolism , Anxiety/physiopathology , Hunger/physiology
8.
Neuromolecular Med ; 26(1): 18, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38691185

ABSTRACT

Seipin is a key regulator of lipid metabolism, the deficiency of which leads to severe lipodystrophy. Hypothalamus is the pivotal center of brain that modulates appetite and energy homeostasis, where Seipin is abundantly expressed. Whether and how Seipin deficiency leads to systemic metabolic disorders via hypothalamus-involved energy metabolism dysregulation remains to be elucidated. In the present study, we demonstrated that Seipin-deficiency induced hypothalamic inflammation, reduction of anorexigenic pro-opiomelanocortin (POMC), and elevation of orexigenic agonist-related peptide (AgRP). Importantly, administration of rosiglitazone, a thiazolidinedione antidiabetic agent, rescued POMC and AgRP expression, suppressed hypothalamic inflammation, and restored energy homeostasis in Seipin knockout mice. Our findings offer crucial insights into the mechanism of Seipin deficiency-associated energy imbalance and indicates that rosiglitazone could serve as potential intervening agent towards metabolic disorders linked to Seipin.


Subject(s)
Agouti-Related Protein , Energy Metabolism , GTP-Binding Protein gamma Subunits , Homeostasis , Hypothalamus , Mice, Knockout , Pro-Opiomelanocortin , Rosiglitazone , Animals , Mice , Hypothalamus/metabolism , Energy Metabolism/drug effects , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/biosynthesis , Agouti-Related Protein/genetics , GTP-Binding Protein gamma Subunits/genetics , Rosiglitazone/pharmacology , Male , Neuroinflammatory Diseases/etiology , Mice, Inbred C57BL , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Neuropeptides/genetics , Neuropeptides/deficiency , Gene Expression Regulation/drug effects
9.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38815068

ABSTRACT

The growth hormone secretagogue receptor (GHSR), primarily known as the receptor for the hunger hormone ghrelin, potently controls food intake, yet the specific Ghsr-expressing cells mediating the orexigenic effects of this receptor remain incompletely characterized. Since Ghsr is expressed in gamma-aminobutyric acid (GABA)-producing neurons, we sought to investigate whether the selective expression of Ghsr in a subset of GABA neurons is sufficient to mediate GHSR's effects on feeding. First, we crossed mice that express a tamoxifen-dependent Cre recombinase in the subset of GABA neurons that express glutamic acid decarboxylase 2 (Gad2) enzyme (Gad2-CreER mice) with reporter mice, and found that ghrelin mainly targets a subset of Gad2-expressing neurons located in the hypothalamic arcuate nucleus (ARH) and that is predominantly segregated from Agouti-related protein (AgRP)-expressing neurons. Analysis of various single-cell RNA-sequencing datasets further corroborated that the primary subset of cells coexpressing Gad2 and Ghsr in the mouse brain are non-AgRP ARH neurons. Next, we crossed Gad2-CreER mice with reactivable GHSR-deficient mice to generate mice expressing Ghsr only in Gad2-expressing neurons (Gad2-GHSR mice). We found that ghrelin treatment induced the expression of the marker of transcriptional activation c-Fos in the ARH of Gad2-GHSR mice, yet failed to induce food intake. In contrast, food deprivation-induced refeeding was higher in Gad2-GHSR mice than in GHSR-deficient mice and similar to wild-type mice, suggesting that ghrelin-independent roles of GHSR in a subset of GABA neurons is sufficient for eliciting full compensatory hyperphagia in mice.


Subject(s)
Arcuate Nucleus of Hypothalamus , Food Deprivation , GABAergic Neurons , Ghrelin , Glutamate Decarboxylase , Hyperphagia , Receptors, Ghrelin , Animals , Male , Mice , GABAergic Neurons/metabolism , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Hyperphagia/metabolism , Ghrelin/metabolism , Ghrelin/pharmacology , Arcuate Nucleus of Hypothalamus/metabolism , Food Deprivation/physiology , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/genetics , Mice, Transgenic , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Mice, Inbred C57BL
10.
Nat Commun ; 15(1): 4646, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821928

ABSTRACT

AgRP neurons in the arcuate nucleus of the hypothalamus (ARC) coordinate homeostatic changes in appetite associated with fluctuations in food availability and leptin signaling. Identifying the relevant transcriptional regulatory pathways in these neurons has been a priority, yet such attempts have been stymied due to their low abundance and the rich cellular diversity of the ARC. Here we generated AgRP neuron-specific transcriptomic and chromatin accessibility profiles from male mice during three distinct hunger states of satiety, fasting-induced hunger, and leptin-induced hunger suppression. Cis-regulatory analysis of these integrated datasets enabled the identification of 18 putative hunger-promoting and 29 putative hunger-suppressing transcriptional regulators in AgRP neurons, 16 of which were predicted to be transcriptional effectors of leptin. Within our dataset, Interferon regulatory factor 3 (IRF3) emerged as a leading candidate mediator of leptin-induced hunger-suppression. Measures of IRF3 activation in vitro and in vivo reveal an increase in IRF3 nuclear occupancy following leptin administration. Finally, gain- and loss-of-function experiments in vivo confirm the role of IRF3 in mediating the acute satiety-evoking effects of leptin in AgRP neurons. Thus, our findings identify IRF3 as a key mediator of the acute hunger-suppressing effects of leptin in AgRP neurons.


Subject(s)
Hunger , Interferon Regulatory Factor-3 , Leptin , Neurons , Animals , Male , Mice , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Arcuate Nucleus of Hypothalamus/metabolism , Chromatin , Epigenesis, Genetic , Fasting , Gene Expression Regulation , Hunger/physiology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Leptin/metabolism , Mice, Inbred C57BL , Neurons/metabolism , Signal Transduction , Transcriptome
11.
Neuron ; 112(13): 2130-2141.e7, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38642553

ABSTRACT

Elevated anxiety often precedes anorexia nervosa and persists after weight restoration. Patients with anorexia nervosa often describe self-starvation as pleasant, potentially because food restriction can be anxiolytic. Here, we tested whether repeated stress can cause animals to prefer a starvation-like state. We developed a virtual reality place preference paradigm in which head-fixed mice can voluntarily seek a starvation-like state induced by optogenetic stimulation of hypothalamic agouti-related peptide (AgRP) neurons. Prior to stress exposure, males but not females showed a mild aversion to AgRP stimulation. Strikingly, following multiple days of stress, a subset of females developed a strong preference for AgRP stimulation that was predicted by high baseline anxiety. Such stress-induced changes in preference were reflected in changes in facial expressions during AgRP stimulation. Our study suggests that stress may cause females predisposed to anxiety to seek a starvation state and provides a powerful experimental framework for investigating the underlying neural mechanisms.


Subject(s)
Agouti-Related Protein , Anxiety , Starvation , Stress, Psychological , Animals , Female , Mice , Agouti-Related Protein/metabolism , Male , Optogenetics , Neurons/metabolism , Mice, Inbred C57BL , Hypothalamus/metabolism
12.
Mol Autism ; 15(1): 14, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38570876

ABSTRACT

BACKGROUND: SH3 and multiple ankyrin repeat domains protein 3 (SHANK3) monogenic mutations or deficiency leads to excessive stereotypic behavior and impaired sociability, which frequently occur in autism cases. To date, the underlying mechanisms by which Shank3 mutation or deletion causes autism and the part of the brain in which Shank3 mutation leads to the autistic phenotypes are understudied. The hypothalamus is associated with stereotypic behavior and sociability. p38α, a mediator of inflammatory responses in the brain, has been postulated as a potential gene for certain cases of autism occurrence. However, it is unclear whether hypothalamus and p38α are involved in the development of autism caused by Shank3 mutations or deficiency. METHODS: Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and immunoblotting were used to assess alternated signaling pathways in the hypothalamus of Shank3 knockout (Shank3-/-) mice. Home-Cage real-time monitoring test was performed to record stereotypic behavior and three-chamber test was used to monitor the sociability of mice. Adeno-associated viruses 9 (AAV9) were used to express p38α in the arcuate nucleus (ARC) or agouti-related peptide (AgRP) neurons. D176A and F327S mutations expressed constitutively active p38α. T180A and Y182F mutations expressed inactive p38α. RESULTS: We found that Shank3 controls stereotypic behavior and sociability by regulating p38α activity in AgRP neurons. Phosphorylated p38 level in hypothalamus is significantly enhanced in Shank3-/- mice. Consistently, overexpression of p38α in ARC or AgRP neurons elicits excessive stereotypic behavior and impairs sociability in wild-type (WT) mice. Notably, activated p38α in AgRP neurons increases stereotypic behavior and impairs sociability. Conversely, inactivated p38α in AgRP neurons significantly ameliorates autistic behaviors of Shank3-/- mice. In contrast, activated p38α in pro-opiomelanocortin (POMC) neurons does not affect stereotypic behavior and sociability in mice. LIMITATIONS: We demonstrated that SHANK3 regulates the phosphorylated p38 level in the hypothalamus and inactivated p38α in AgRP neurons significantly ameliorates autistic behaviors of Shank3-/- mice. However, we did not clarify the biochemical mechanism of SHANK3 inhibiting p38α in AgRP neurons. CONCLUSIONS: These results demonstrate that the Shank3 deficiency caused autistic-like behaviors by activating p38α signaling in AgRP neurons, suggesting that p38α signaling in AgRP neurons is a potential therapeutic target for Shank3 mutant-related autism.


Subject(s)
Autistic Disorder , Animals , Mice , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Hypothalamus/metabolism , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Mitogen-Activated Protein Kinase 14/metabolism
13.
Sci Rep ; 14(1): 7844, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38570726

ABSTRACT

Obesity, a worldwide epidemic, leads to various metabolic disorders threatening human health. In response to stress or fasting, glucocorticoid (GC) levels are elevated to promote food intake. This involves GC-induced expression of the orexigenic neuropeptides in agouti-related protein (AgRP) neurons of the hypothalamic arcuate nucleus (ARC) via the GC receptor (GR). Here, we report a selective GR modulator (SGRM) that suppresses GR-induced transcription of genes with non-classical glucocorticoid response elements (GREs) such as Agrp-GRE, but not with classical GREs, and via this way may serve as a novel anti-obesity agent. We have identified a novel SGRM, 2-O-trans-p-coumaroylalphitolic acid (Zj7), a triterpenoid extracted from the Ziziphus jujube plant, that selectively suppresses GR transcriptional activity in Agrp-GRE without affecting classical GREs. Zj7 reduces the expression of orexigenic genes in the ARC and exerts a significant anorexigenic effect with weight loss in both high fat diet-induced obese and genetically obese db/db mouse models. Transcriptome analysis showed that Zj7 represses the expression of a group of orexigenic genes including Agrp and Npy induced by the synthetic GR ligand dexamethasone (Dex) in the hypothalamus. Taken together, Zj7, as a selective GR modulator, showed beneficial metabolic activities, in part by suppressing GR activity in non-classical GREs in orexigenic genes. This study demonstrates that a potential anorexigenic molecule may allow GRE-specific inhibition of GR transcriptional activity, which is a promising approach for the treatment of metabolic disorders.


Subject(s)
Metabolic Diseases , Receptors, Glucocorticoid , Mice , Animals , Humans , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Glucocorticoids/pharmacology , Agouti-Related Protein/metabolism , Obesity/drug therapy , Obesity/metabolism
14.
J Neuroendocrinol ; 36(5): e13389, 2024 05.
Article in English | MEDLINE | ID: mdl-38599683

ABSTRACT

Hunger increases the motivation for calorie consumption, often at the expense of low-taste appeal. However, the neural mechanisms integrating calorie-sensing with increased motivation for calorie consumption remain unknown. Agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus sense hunger, and the ingestion of caloric solutions promotes dopamine release in the absence of sweet taste perception. Therefore, we hypothesised that metabolic-sensing of hunger by AgRP neurons would be essential to promote dopamine release in the nucleus accumbens in response to caloric, but not non-caloric solutions. Moreover, we examined whether metabolic sensing in AgRP neurons affected taste preference for bitter solutions under conditions of energy need. Here we show that impaired metabolic sensing in AgRP neurons attenuated nucleus accumbens dopamine release in response to sucrose, but not saccharin, consumption. Furthermore, metabolic sensing in AgRP neurons was essential to distinguish nucleus accumbens dopamine response to sucrose consumption when compared with saccharin. Under conditions of hunger, metabolic sensing in AgRP neurons increased the preference for sucrose solutions laced with the bitter tastant, quinine, to ensure calorie consumption, whereas mice with impaired metabolic sensing in AgRP neurons maintained a strong aversion to sucrose/quinine solutions despite ongoing hunger. In conclusion, we demonstrate normal metabolic sensing in AgRP neurons drives the preference for calorie consumption, primarily when needed, by engaging dopamine release in the nucleus accumbens.


Subject(s)
Agouti-Related Protein , Dopamine , Nucleus Accumbens , Sucrose , Nucleus Accumbens/metabolism , Animals , Dopamine/metabolism , Agouti-Related Protein/metabolism , Mice , Male , Food Preferences/physiology , Mice, Inbred C57BL , Neurons/metabolism , Hunger/physiology , Taste Perception/physiology
15.
Trends Endocrinol Metab ; 35(6): 456-458, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599900

ABSTRACT

Obesity is associated with dysfunctions in hypothalamic neurons that regulate metabolism, including agouti-related protein (AgRP)-expressing neurons. In a recent article, Zhang et al. demonstrated that either diet- or genetically induced obesity promoted iron accumulation specifically in AgRP neurons. Preventing iron overload in AgRP neurons mitigated diet-induced obesity and related comorbidities in male mice.


Subject(s)
Agouti-Related Protein , Iron , Obesity , Obesity/metabolism , Animals , Humans , Iron/metabolism , Agouti-Related Protein/metabolism , Mice , Neurons/metabolism , Male , Hypothalamus/metabolism , Iron Overload/metabolism
16.
Nature ; 628(8009): 826-834, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538787

ABSTRACT

Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.


Subject(s)
Brain Stem , Ependymoglial Cells , Feeding Behavior , Hot Temperature , Hypothalamus , Neural Pathways , Neurons , Animals , Female , Male , Mice , Agouti-Related Protein/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/cytology , Brain Stem/cytology , Brain Stem/physiology , Dopamine/metabolism , Eating/physiology , Ependymoglial Cells/cytology , Ependymoglial Cells/physiology , Feeding Behavior/physiology , Glutamic Acid/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Neural Pathways/metabolism , Neurons/metabolism , Parabrachial Nucleus/cytology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiology , Thermosensing/physiology , Time Factors , Vascular Endothelial Growth Factor A/cerebrospinal fluid , Vascular Endothelial Growth Factor A/metabolism
17.
Cell Rep ; 43(3): 113900, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38460132

ABSTRACT

Iron overload is closely associated with metabolic dysfunction. However, the role of iron in the hypothalamus remains unclear. Here, we find that hypothalamic iron levels are increased, particularly in agouti-related peptide (AgRP)-expressing neurons in high-fat-diet-fed mice. Using pharmacological or genetic approaches, we reduce iron overload in AgRP neurons by central deferoxamine administration or transferrin receptor 1 (Tfrc) deletion, ameliorating diet-induced obesity and related metabolic dysfunction. Conversely, Tfrc-mediated iron overload in AgRP neurons leads to overeating and adiposity. Mechanistically, the reduction of iron overload in AgRP neurons inhibits AgRP neuron activity; improves insulin and leptin sensitivity; and inhibits iron-induced oxidative stress, endoplasmic reticulum stress, nuclear factor κB signaling, and suppression of cytokine signaling 3 expression. These results highlight the critical role of hypothalamic iron in obesity development and suggest targets for treating obesity and related metabolic disorders.


Subject(s)
Iron Overload , Metabolic Diseases , Mice , Animals , Agouti-Related Protein/metabolism , Obesity/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Neurons/metabolism , Diet, High-Fat/adverse effects , Metabolic Diseases/metabolism , Iron/metabolism , Mice, Inbred C57BL
18.
Nat Commun ; 15(1): 2131, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38459068

ABSTRACT

AgRP neurons drive hunger, and excessive nutrient intake is the primary driver of obesity and associated metabolic disorders. While many factors impacting central regulation of feeding behavior have been established, the role of microRNAs in this process is poorly understood. Utilizing unique mouse models, we demonstrate that miR-33 plays a critical role in the regulation of AgRP neurons, and that loss of miR-33 leads to increased feeding, obesity, and metabolic dysfunction in mice. These effects include the regulation of multiple miR-33 target genes involved in mitochondrial biogenesis and fatty acid metabolism. Our findings elucidate a key regulatory pathway regulated by a non-coding RNA that impacts hunger by controlling multiple bioenergetic processes associated with the activation of AgRP neurons, providing alternative therapeutic approaches to modulate feeding behavior and associated metabolic diseases.


Subject(s)
Hunger , MicroRNAs , Animals , Mice , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Hunger/physiology , Hypothalamus/metabolism , MicroRNAs/metabolism , Neurons/metabolism , Obesity/metabolism
19.
Phytomedicine ; 126: 155297, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38342019

ABSTRACT

BACKGROUND: Research on the imbalance of proopiomelanocortin (POMC)/agouti-related protein (AgRP) neurons in the hypothalamus holds potential insights into the pathophysiology of diabetes. Jinkui Shenqi pills (JSP), a prevalent traditional Chinese medicine, regulate hypothalamic function and treat diabetes. PURPOSE: To investigate the hypoglycemic effect of JSP and explore the probable mechanism in treating diabetes. METHODS: A type 2 diabetes mouse model was used to investigate the pharmacodynamics of JSP. The glucose-lowering efficacy of JSP was assessed through various metrics including body weight, food consumption, fasting blood glucose (FBG), serum insulin levels, and an oral glucose tolerance test (OGTT). To elucidate the modulatory effects of JSP on hypothalamic mechanisms, we quantified the expression and activity of POMC and AgRP and assessed the insulin-mediated phosphoinositide 3-kinase (PI3K)/protein kinase A (AKT)/forkhead box O1 (FOXO1) pathway in diabetic mice via western blotting and immunohistochemistry. Additionally, primary hypothalamic neurons were exposed to high glucose and palmitic acid levels to induce insulin resistance, and the influence of JSP on POMC/AgRP protein expression and activation was evaluated by PI3K protein inhibition using western blotting and immunofluorescence. RESULTS: Medium- and high-dose JSP treatment effectively inhibited appetite, resulting in a steady declining trend in body weight, FBG, and OGTT results in diabetic mice (p < 0.05). These JSP groups also had significantly increased insulin levels (p < 0.05). Importantly, the medium-dose group exhibited notable protection of hypothalamic neuronal and synaptic structures, leading to augmentation of dendritic length and branching (p < 0.05). Furthermore, low-, medium-, and high-dose JSP groups exhibited increased phosphorylated (p) INSR, PI3K, pPI3K, AKT, and pAKT expression, as well as decreased FOXO1 and increased pFOXO1 expression, indicating improved hypothalamic insulin resistance in diabetic mice (p < 0.05). Treatment with 10% JSP-enriched serum produced a marked elevation of both expression and activation of POMC (p < 0.05), with a concurrent reduction in AgRP expression and activation within primary hypothalamic neurons (p < 0.05). Intriguingly, these effects could be attributed to the regulatory dynamics of PI3K activity. CONCLUSION: Our findings suggest that JSP can ameliorate diabetes by regulating POMC/AgRP expression and activity. The insulin-mediated PI3K/AKT/FOXO1 pathway plays an important regulatory role in this intricate process.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Drugs, Chinese Herbal , Insulin Resistance , Mice , Animals , Agouti-Related Protein/metabolism , Agouti-Related Protein/pharmacology , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Glucose/metabolism , Body Weight
20.
J Neuroendocrinol ; 36(3): e13370, 2024 03.
Article in English | MEDLINE | ID: mdl-38344844

ABSTRACT

Excess levels of circulating androgens during prenatal or peripubertal development are an important cause of polycystic ovary syndrome (PCOS), with the brain being a key target. Approximately half of the women diagnosed with PCOS also experience metabolic syndrome; common features including obesity, insulin resistance and hyperinsulinemia. Although a large amount of clinical and preclinical evidence has confirmed this relationship between androgens and the reproductive and metabolic features of PCOS, the mechanisms by which androgens cause this dysregulation are unknown. Neuron-specific androgen receptor knockout alleviates some PCOS-like features in a peripubertal dihydrotestosterone (DHT) mouse model, but the specific neuronal populations mediating these effects are undefined. A candidate population is the agouti-related peptide (AgRP)-expressing neurons, which are important for both reproductive and metabolic function. We used a well-characterised peripubertal androgenized mouse model and Cre-loxP transgenics to investigate whether deleting androgen receptors specifically from AgRP neurons can alleviate the induced reproductive and metabolic dysregulation. Androgen receptors were co-expressed in 66% of AgRP neurons in control mice, but only in <2% of AgRP neurons in knockout mice. The number of AgRP neurons was not altered by the treatments. Only 20% of androgen receptor knockout mice showed rescue of DHT-induced androgen-induced anovulation and acyclicity. Furthermore, androgen receptor knockout did not rescue metabolic dysfunction (body weight, adiposity or glucose and insulin tolerance). While we cannot rule out developmental compensation in our model, these results suggest peripubertal androgen excess does not markedly influence Agrp expression and does not dysregulate reproductive and metabolic function through direct actions of androgens onto AgRP neurons.


Subject(s)
Androgens , Polycystic Ovary Syndrome , Animals , Female , Humans , Mice , Pregnancy , Agouti-Related Protein/metabolism , Androgens/metabolism , Dihydrotestosterone/pharmacology , Mice, Knockout , Neurons/metabolism , Obesity/metabolism , Peptides/pharmacology , Receptors, Androgen/metabolism , Virilism/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL