Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38659859

ABSTRACT

Previous studies have been focused on lipid metabolism in peripheral tissues such as adipose tissues, while little or nothing is known about that in the brain. It is well recognized that cold acclimations enhance adipocyte functions, including white adipose tissue (WAT) lipid lipolysis and beiging, and brown adipose tissue (BAT) thermogenesis in mammals. However, it remains unclear whether and how the genes responsible for lipid metabolism in the brain are also under the control of cold acclimations. Here, we show that cold exposure predominantly increases the expressions of the genes encoding lipid lipolysis in the paraventricular nucleus of the hypothalamus (PVH). Mechanistically, we find that inactivation of neurons in the PVH blunts the cold-induced lipid peroxidation and lipolysis. Together, these findings indicate that lipid metabolism in the PVH is cold sensitive, potentially participating in cold regulations of energy metabolism.

2.
J Clin Invest ; 133(7)2023 04 03.
Article in English | MEDLINE | ID: mdl-36787185

ABSTRACT

Glucose homeostasis can be improved after bariatric surgery, which alters bile flow and stimulates gut hormone secretion, particularly FGF15/19. FGFR1 expression in AGRP-expressing cells is required for bile acids' ability to improve glucose control. We show that the mouse Agrp gene has 3 promoter/enhancer regions that direct transcription of each of their own AGRP transcripts. One of these Agrp promoters/enhancers, Agrp-B, is regulated by bile acids. We generated an Agrp-B knockin FLP/knockout allele. AGRP-B-expressing cells are found in endocrine cells of the pars tuberalis and coexpress diacylglycerol lipase B - an endocannabinoid biosynthetic enzyme - distinct from pars tuberalis thyrotropes. AGRP-B expression is also found in the folliculostellate cells of the pituitary's anterior lobe. Mice without AGRP-B were protected from glucose intolerance induced by high-fat feeding but not from excess weight gain. Chemogenetic inhibition of AGRP-B cells improved glucose tolerance by enhancing glucose-stimulated insulin secretion. Inhibition of the AGRP-B cells also caused weight loss. The improved glucose tolerance and reduced body weight persisted up to 6 weeks after cessation of the DREADD-mediated inhibition, suggesting the presence of a biological switch for glucose homeostasis that is regulated by long-term stability of food availability.


Subject(s)
Hypothalamus , Neurons , Mice , Animals , Agouti-Related Protein/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Homeostasis , Glucose/metabolism , Bile Acids and Salts/metabolism , Eating
3.
Int J Obes (Lond) ; 47(4): 325-334, 2023 04.
Article in English | MEDLINE | ID: mdl-36782056

ABSTRACT

BACKGROUND: It is established that pulmonary disorders are comorbid with metabolic disorders such as obesity. Previous studies show that the stimulator of interferon genes (STING) signaling plays crucial roles in obesity-induced chronic inflammation via TANK-binding kinase 1 (TBK1) pathways. However, it remains unknown whether and how the STING signaling is implicated in the inflammatory processes in the lung in obesity. METHODS: Human lung tissues were obtained from obese patients (n = 3) and controls (n = 3). Mice were fed with the high-fat diet or regular control diet to establish the diet-induced obese (DIO) and lean mice, and were treated with C-176 (a specific STING inhibitor) or vehicle respectively. The lung macrophages were exposed to palmitic acid (PA) in vitro. The levels of STING singaling and metabolic inflammation factors were detected and anlyzed. RESULTS: We find that STING+/CD68+ macrophages are increased in lung tissues in patients with obesity. Our data also show that the expressions of STING and the levels of proinflammatory cytokines are increased both in lung tissues and bronchoalveolar lavage fluid (BALF) in obesity compared to controls, and inhibition of the STING blunted the obesity-induced lung inflammation. Mechanistically, our data demonstrate that the STING signaling pathway is involved in the PA-induced inflammation through the STING-TBK1-IRF3 (interferon regulatory factor 3)/NF-κB (nuclear factor kappa B) pathways in the lung macrophages. CONCLUSIONS: Our results collectively suggest that the STING signaling contributes to obesity-associated inflammation by stimulating proinflammatory processes in lung macrophages, one that may serve as a therapeutic target in ameliorating obesity-related lung dysfunctions.


Subject(s)
Pneumonia , Signal Transduction , Animals , Humans , Mice , Inflammation/metabolism , NF-kappa B/metabolism , Obesity/complications
4.
Nat Commun ; 13(1): 7536, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36477150

ABSTRACT

The role of non-neuronal glial cells in the regulation of adipose sympathetic nerve activity and adipocyte functions such as white adipose tissue lipid lipolysis is poorly understood. Here, we combine chemo/optogenetic manipulations of medio-basal hypothalamic astrocytes, real-time fiber photometry monitoring of white adipose tissue norepinephrine (NE) contents and nerve activities, electrophysiological recordings of local sympathetic inputs to inguinal white adipose tissue (iWAT), and adipose tissue lipid lipolytic assays to define the functional roles of hypothalamic astrocytes in the regulation of iWAT sympathetic outflow and lipolysis. Our results show that astrocyte stimulation elevates iWAT NE contents, excites sympathetic neural inputs and promotes lipolysis. Mechanistically, we find that sympathetic paravertebral ganglia (PG) partake in those astrocyte effects. We also find that astrocyte stimulation excites pro-opiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARH), and chemogenetic inhibition of POMC neurons blunts the effects induced by astrocyte stimulation. While we cannot exclude potential roles played by other cell populations such as microglia, our findings in this study reveal a central astrocyte-peripheral adipocyte axis modulating sympathetic drive to adipose tissues and adipocyte functions, one that might serve as a target for therapeutic intervention in the treatment of obesity.


Subject(s)
Adipocytes, White , Nerve Tissue , Mice , Animals , Lipids
6.
Mol Psychiatry ; 26(12): 7270-7279, 2021 12.
Article in English | MEDLINE | ID: mdl-34489531

ABSTRACT

The neuropeptide oxytocin (OXT) is well recognized for eliciting anxiolytic effects and promoting social reward. However, emerging evidence shows that OXT increases aversive events. These seemingly inconsistent results may be attributable to the broad OXT receptor (OXTr) expression in the central nervous system. This study selectively activated septal neurons expressing OXTr using chemogenetics. We found that chemogenetic activation of septal OXTr neurons induced anxiety- but not depressive-like behavior. In addition, septal OXTr neurons projected dense fibers to the horizontal diagonal band of Broca (HDB), and selective stimulation of those HDB projections also elicited anxiety-like behaviors. We also found that septal OXTr neurons express the vesicular GABA transporter (vGAT) protein and optogenetic stimulation of septal OXTr projections to the HDB inactivated HDB neurons. Our data collectively reveal that septal OXTr neurons increase anxiety by projecting inhibitory GABAergic inputs to the HDB.


Subject(s)
Oxytocin , Receptors, Oxytocin , Anxiety , Neurons/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Social Behavior
7.
Mol Psychiatry ; 26(7): 2805-2819, 2021 07.
Article in English | MEDLINE | ID: mdl-33067580

ABSTRACT

It is essential to elucidate brain-adipocyte interactions in order to tackle obesity and its comorbidities, as the precise control of brain-adipose tissue cross-talk is crucial for energy and glucose homeostasis. Recent studies show that in the peripheral adipose tissue, adenosine induces adipogenesis through peripheral adenosine A1 receptor (pADORA1) signaling; however, it remains unclear whether systemic and adipose tissue metabolism would also be under the control of central (c) ADORA1 signaling. Here, we use tissue-specific pharmacology and metabolic tools to clarify the roles of cADORA1 signaling in energy and adipocyte physiology. We found that cADORA1 signaling reduces body weight while also inducing adipose tissue lipolysis. cADORA1 signaling also increases adipose tissue sympathetic norepinephrine content. In contrast, pADORA1 signaling facilitates a high-fat diet-induced obesity (DIO). We propose here a novel mechanism in which cADORA1 and pADORA1 signaling hinder and aggravate DIO, respectively.


Subject(s)
Adipose Tissue , Lipid Metabolism , Adipocytes , Adipose Tissue/metabolism , Body Weight , Brain , Diet, High-Fat , Energy Metabolism , Humans
8.
Nat Commun ; 11(1): 6326, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33303759

ABSTRACT

It is well recognized that ventromedial hypothalamus (VMH) serves as a satiety center in the brain. However, the feeding circuit for the VMH regulation of food intake remains to be defined. Here, we combine fiber photometry, chemo/optogenetics, virus-assisted retrograde tracing, ChR2-assisted circuit mapping and behavioral assays to show that selective activation of VMH neurons expressing steroidogenic factor 1 (SF1) rapidly inhibits food intake, VMH SF1 neurons project dense fibers to the paraventricular thalamus (PVT), selective chemo/optogenetic stimulation of the PVT-projecting SF1 neurons or their projections to the PVT inhibits food intake, and chemical genetic inactivation of PVT neurons diminishes SF1 neural inhibition of feeding. We also find that activation of SF1 neurons or their projections to the PVT elicits a flavor aversive effect, and selective optogenetic stimulation of ChR2-expressing SF1 projections to the PVT elicits direct excitatory postsynaptic currents. Together, our data reveal a neural circuit from VMH to PVT that inhibits food intake.


Subject(s)
Feeding Behavior/physiology , Neural Pathways/physiology , Thalamus/physiology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Designer Drugs/pharmacology , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Glucose Tolerance Test , Integrases/metabolism , Leptin/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Neural Pathways/drug effects , Neurons/drug effects , Neurons/physiology , Thalamus/drug effects , Ventromedial Hypothalamic Nucleus/drug effects
9.
Sci Rep ; 10(1): 18368, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110180

ABSTRACT

Ample evidence indicates that obesity causes dysfunctions in the lung. Previous studies also show that cholinergic anti-inflammatory pathways play crucial roles in obesity-induced chronic inflammation via α7 nicotinic acetylcholine receptor (α7nAChR) signaling. However, it remains unclear whether and how obesity affects the expressions of α7nAChR in myeloid cells in the lung. To address this question, we treated regular chow diet-fed mice or high-fat diet induced obese mice with lipopolysaccharide (LPS) or vehicle via endotracheal injections. By using a multicolor flow cytometry approach to analyze and characterize differential cell subpopulations and α7nAChR expressions, we find no detectable α7nAChR in granulocytes, monocytes and alveolar macrophages, and low expression levels of α7nAChR were detected in interstitial macrophages. Interestingly, we find that a challenge with LPS treatment significantly increased expression levels of α7nAChR in monocytes, alveolar and interstitial macrophages. Meanwhile, we observed that the expression levels of α7nAChR in alveolar and interstitial macrophages in high-fat diet induced obese mice were lower than regular chow diet-fed mice challenged by the LPS. Together, our findings indicate that obesity alters the expressions of α7nAChR in differential lung myeloid cells.


Subject(s)
Diet, High-Fat , Lung/metabolism , Myeloid Cells/metabolism , Obesity/etiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Immunophenotyping , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Obesity/immunology , Obesity/metabolism , Obesity/pathology
10.
Stem Cell Res Ther ; 10(1): 352, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31779687

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a common neurotrauma leading to brain dysfunction and death. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) hold promise in the treatment of TBI. However, their efficacy is modest due to low survival and differentiation under the harsh microenvironment of the injured brain. MG53, a member of TRIM family protein, plays a vital role in cell and tissue damage repair. The present study aims to test whether MG53 preserves hUC-MSCs against oxidative stress and enhances stem cell survival and efficacy in TBI treatment. METHODS: In this study, we performed a series of in vitro and in vivo experiments in hUC-MSCs and mice to define the function of MG53 enhancing survival, neurogenesis, and therapeutic efficacy of stem cells in murine traumatic brain injury. RESULTS: We found that recombinant human MG53 (rhMG53) protein protected hUC-MSCs against H2O2-induced oxidative damage and stimulated hUC-MSC proliferation and migration. In a mouse model of contusion-induced TBI, intravenous administration of MG53 protein preserved the survival of transplanted hUC-MSCs, mitigated brain edema, reduced neurological deficits, and relieved anxiety and depressive-like behaviors. Co-treatment of MG53 and hUC-MSCs enhanced neurogenesis by reducing apoptosis and improving PI3K/Akt-GSK3ß signaling. CONCLUSION: MG53 enhances the efficacy of hUC-MSCs in the recovery of TBI, indicating that such adjunctive therapy may provide a novel strategy to lessen damage and optimize recovery for brain injury.


Subject(s)
Brain Injuries, Traumatic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Oxidative Stress , Signal Transduction , Tripartite Motif Proteins/metabolism , Umbilical Cord , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/therapy , Cell Survival , Disease Models, Animal , Heterografts , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Umbilical Cord/metabolism , Umbilical Cord/pathology
11.
Transl Psychiatry ; 9(1): 111, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850579

ABSTRACT

Recent studies indicate that activation of hypothalamic Agouti-related protein (Agrp) neurons can increase forage-related/repetitive behavior and decrease anxiety levels. However, the impact of physiological hunger states and food deprivation on anxiety-related behaviors have not been clarified. In the present study, we evaluated changes in anxiety levels induced by physiological hunger states and food deprivation, and identified the neuron population involved. Ad libitum fed and fasted mice were tested in the open field and elevated plus-maze behavioral tests. The DREADD approach was applied to selectively inhibit and stimulate neurons expressing Agrp in hypothalamic arcuate nucleus in Agrp-Cre transgenic mice. We found that anxiety levels were significantly reduced in the late light period when mice have increased need for food and increased Agrp neurons firing, in contrast to the levels in the early light period. Consistently, we also found that anxiety was potently reduced in 24-h fasted mice, relative to 12-h fasted mice or fed ad libitum. Mechanistically, we found that chemogenetic activation of Agrp neurons reduced anxiety in fed mice, and inactivation of Agrp neurons reduced fasting-induced anxiolytic effects. Our results suggest that anxiety levels may vary physiologically with the increasing need for food, and are influenced by acute fasting in a time-dependent manner. Agrp neurons contribute to fasting-induced anxiolytic effects, supporting the notion that Agrp neuron may serve as an entry point for the treatment of energy states-related anxiety disorders.


Subject(s)
Agouti-Related Protein/metabolism , Anxiety , Circadian Rhythm , Fasting/physiology , Neural Pathways/physiology , Neurons/physiology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/physiology , Eating/physiology , Female , Food Deprivation , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity
12.
Neurosci Lett ; 662: 91-97, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29030220

ABSTRACT

This study aims to investigate the relationship between DNA methylation and expression of human dopamine transporter (hDAT). We examined methylation status of hDAT in cells with various hDAT expression levels, including two dopaminergic neural cell lines (SK-N-AS and SH-SY-5Y) and one non-dopaminergic cell line (HEK293) by bisulfite sequencing PCR(BSP). The effects of DNA methyltransferase inhibitor 5-aza-dC or/and histone deacetylase inhibitor (HDACi, sodium butyrate, NaB) on the DNA methylation status and mRNA expression levels of hDAT were examined. The results revealed marked hypomethylation of the two promoter regions (-1214 to -856bp and -48 to 439bp, the first base of exon 1 was taken as +1 bp)of hDAT in SK-N-AS (4.7%±2.0mC and 3.5%±1.0mC, respectively) compared with SH-SY-5Y (88.0%±4.4%mC and 81.1%±8.8%mC) and HEK293 (90.7%±2.4mC and 84.4%±8.6% mC) cell lines, indicating a cell-specific methylation regulation of hDAT. 5-aza-dC and NaB decreased hypermethylation,while increase hDAT expression in SH-SY-5Y cells and recovered hDAT mRNA expression in HEK293 cells. DNA methylation enabled the cell-specific differential expression of the hDAT gene. hDAT silencing was reversed by the introduction of DNA hypomethylation via 5-aza-dC or/and NaB.


Subject(s)
DNA Methylation , Dopamine Plasma Membrane Transport Proteins/metabolism , Cell Line , Cell Line, Tumor , Dopamine Plasma Membrane Transport Proteins/genetics , Epigenesis, Genetic , Gene Expression , Humans , Promoter Regions, Genetic , RNA/metabolism , Transcription, Genetic
13.
Proc Natl Acad Sci U S A ; 114(52): 13816-13821, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229861

ABSTRACT

Feeding behavior is controlled by diverse neurons and neural circuits primarily concentrated in the hypothalamus and hindbrain in mammals. In this study, by using chemo/optogenetic techniques along with feeding assays, we investigate how neurons within the medial septal complex (MSc), a brain area implicated in emotion and cognition, contribute to food intake. We find that chemo/optogenetic activation of MSc glutamatergic neurons profoundly reduces food intake during both light and dark periods of the rodent light cycle. Furthermore, we find that selective activation of MSc glutamatergic projections in paraventricular hypothalamus (PVH) reduces food intake, suggesting that MSc glutamatergic neurons suppress feeding by activating downstream neurons in the PVH. Open-field behavioral assays reveal that these neurons do not overtly affect anxiety levels and locomotion. Collectively, our findings demonstrate that septal glutamatergic neurons exert anorexigenic effects by projecting to the PVH without affecting anxiety and physical activities.


Subject(s)
Appetite Regulation/physiology , Eating/physiology , Locomotion/physiology , Neurons/physiology , Septal Nuclei/physiology , Animals , Female , Male , Mice , Mice, Transgenic
14.
Trends Endocrinol Metab ; 28(6): 437-448, 2017 06.
Article in English | MEDLINE | ID: mdl-28279562

ABSTRACT

The neural circuits controlling feeding and emotional behaviors are intricately and reciprocally connected. Recent technological developments, including cell type-specific optogenetic and chemogenetic approaches, allow functional characterization of genetically defined cell populations and neural circuits in feeding and emotional processes. Here we review recent studies that have utilized circuit-based manipulations to decipher the functional interactions between neural circuits controlling feeding and those controlling emotional processes. Specifically, we highlight newly described neural circuit interactions between classical emotion-related brain regions, such as the hippocampus and amygdala, and homeostatic feeding circuitry in the arcuate nucleus and lateral hypothalamus (LH). Together these circuits will provide a template for future studies to examine functional interactions between feeding and emotion.


Subject(s)
Brain/physiology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Brain/metabolism , Emotions/physiology , Feeding Behavior/physiology , Feeding Behavior/psychology , Humans , Hypothalamic Area, Lateral/metabolism , Hypothalamic Area, Lateral/physiology
15.
Schizophr Res ; 179: 41-49, 2017 01.
Article in English | MEDLINE | ID: mdl-27613506

ABSTRACT

OBJECTIVE: It is well established that schizophrenia patients have high cardiovascular morbidity and mortality. However, the underlying risk factors in the earliest stages of both schizophrenia illness and antipsychotics treatment are less clear. This study aimed to characterize the metabolic features of those patients. METHODS: We performed a retrospective cohort study in a naturalistic setting, which included antipsychotic-naïve, first-episode schizophrenia (FES) inpatients with the baseline metabolic measurements and changes following a short term treatment with antipsychotic drugs. RESULTS: Although prevalence of hypertriglyceridemia, hypercholesterolemia, higher-LDL-C and hyperglycaemia in patients with FES were much lower than those of the general population (7.5% v.s. 14.9%, 9.2% v.s. 18.4%, 8.1% v.s. 14.9%, 8.6% v.s.18.3%, respectively), lower-HDL-C in patients with FES were much more prevalent than that of the general population (19.9% v.s. 6.4%). Despite significant metabolic risk profiles (as such lipid abnormalities and insulin resistance) increase, mean fasting glucose and glucosylated serum protein (GSP) were significantly decreased after the short term (median of 23days) antipsychotics exposure, compared to baseline. There is no significant difference of the metabolic profile change between monopharmacy and polypharmacy. CONCLUSION: These results indicated an early-onset nature of HDL-C abnormalities in drug-naïve FES patients. Lipids metabolism risk may develop early and quickly after antipsychotic exposure. Early monitoring is required for the purpose of early detection and hence prevention of the initial metabolic risk which may lead to diabetes mellitus and cardiovascular disease.


Subject(s)
Antipsychotic Agents/adverse effects , Cardiovascular Diseases/blood , Lipid Metabolism Disorders/blood , Schizophrenia/blood , Adult , Cardiovascular Diseases/chemically induced , Female , Humans , Lipid Metabolism Disorders/chemically induced , Male , Retrospective Studies , Schizophrenia/drug therapy , Young Adult
16.
J Neurosci ; 36(44): 11185-11195, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27807162

ABSTRACT

Feeding behavior is orchestrated by neural circuits primarily residing in the hypothalamus and hindbrain. However, the relative influence of cognitive and emotional brain circuits to the feeding circuitry in the hypothalamus and hindbrain remains unclear. Here, using the cell-type selectivity of genetic methods, circuit mapping, and behavior assays, we sought to decipher neural circuits emanating from the septal nucleus to the lateral hypothalamus (LH) that contribute to neural regulation of food intake in mice. We found that chemogenetic and optogenetic activation of septal vesicular GABA transporter (vGAT)-containing neurons or their projections in the LH reduced food intake in mice. Consistently, chemogenetic inhibition of septal vGAT neurons increased food intake. Furthermore, we investigated a previously unknown neural circuit originating from septal vGAT neurons to a subset of vGAT neurons in the LH, an area involved in homeostatic and hedonic control of energy states. Collectively, our data reveal an inhibitory septohypothalamic feeding circuit that might serve as a therapeutic target for the treatment of eating disorders such as anorexia nervosa. SIGNIFICANCE STATEMENT: Our results demonstrate that top-down projections from the septum to the hypothalamus control food intake negatively. Given the known role for both of these brain regions in the control of feeding and emotion-related behaviors, these findings reveal previously unknown neural circuitry that is likely implicated in emotional aspects of food intake and provide new insights into the development of therapeutic targets for the treatment of eating disorders.


Subject(s)
Appetite/physiology , Feeding Behavior/physiology , GABAergic Neurons/physiology , Hypothalamic Area, Lateral/physiology , Neural Inhibition/physiology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Nerve Net/physiology , Neural Pathways/physiology
17.
Glia ; 64(12): 2263-2273, 2016 12.
Article in English | MEDLINE | ID: mdl-27658520

ABSTRACT

Emerging evidence shows that hypothalamic astrocytes react to and counteract energy surfeit produced by high-fat diet (HFD) feeding. However, the functional role of astrocytes in the control of energy states and the underlying molecular mechanism(s) during physiological conditions remain largely underexplored. In the present study, by taking advantage of spatiotemporally precise optogenetic approaches, real-time measurements of extracellular adenosine, and behavioral assays, we find that optogenetic stimulation of astrocytes localized in the medial basal hypothalamus (MBH) suppresses food intake in a frequency dependent manner with high frequency, but not low frequency, stimulation of astrocytes reducing food intake. Furthermore, stimulation of MBH astrocytes diminishes orexigenic ghrelin or fasting-induced hyperphagia without effecting anxiety-related behavior. Consistent with a frequency dependent role for MBH astrocytes in feeding behavior, optogenetic stimulation of MBH astrocytes increases extracellular levels of adenosine in a frequency dependent manner. Collectively, our results provide new insights into the role of astrocytes in physiological functions during naturally occurring behaviors, such as feeding. GLIA 2016;64:2263-2273.


Subject(s)
Astrocytes/metabolism , Feeding Behavior/physiology , Hypothalamus/cytology , Adenosine/metabolism , Animals , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Diet, High-Fat , Emotions/physiology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Optogenetics
18.
Appetite ; 101: 171-7, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26972351

ABSTRACT

The objective of this study was to determine the effects of different doses of caffeine on appetite and anxiety-related behavior. Additionally, we sought to determine if withdrawal from chronic caffeine administration promotes anxiety. In this study, we utilized rodent open field testing and feeding behavior assays to determine the effects of caffeine on feeding and anxiety-related behavior (n = 8 mice; 4-8 weeks old). We also measured 2 h and 24 h food intake and body-weight during daily administration of caffeine (n = 12 mice; 4-8 weeks old). To test for caffeine withdrawal induced anxiety, anxiety-related behavior in rodents was quantified following withdrawal from four consecutive days of caffeine administration (n = 12 mice; 4-8 weeks old). We find that acute caffeine administration increases food intake in a dose-dependent manner with lower doses of caffeine more significantly increasing food intake than higher doses. Acute caffeine administration also reduced anxiety-related behaviors in mice without significantly altering locomotor activity. However, we did not observe any differences in 24 h food intake or body weight following chronic caffeine administration and there were no observable differences in anxiety-related behaviors during caffeine withdrawal. In conclusion, we find that caffeine can both increase appetite and decrease anxiety-related behaviors in a dose dependent fashion. Given the complex relationship between appetite and anxiety, the present study provides additional insights into potential caffeine-based pharmacological mechanisms governing appetite and anxiety disorders, such as bulimia nervosa.


Subject(s)
Anxiety/drug therapy , Appetite/drug effects , Caffeine/administration & dosage , Animals , Behavior, Animal , Body Weight , Caffeine/pharmacology , Dose-Response Relationship, Drug , Eating , Female , Male , Mice , Mice, Transgenic
19.
Nat Commun ; 6: 10188, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26666960

ABSTRACT

Previous research has focused on feeding circuits residing in the hindbrain and midbrain that govern homeostatic or hedonic control of food intake. However, the feeding circuits controlling emotional or cognitive aspects of food intake are largely unknown. Here we use chemical genetics and optogenetic techniques to dissect appetite control circuits originating from ventral hippocampus (vHPC), a brain region implicated in emotion and cognition. We find that the vHPC projects functional glutamatergic synaptic inputs to the lateral septum (LS) and optogenetic activation of vHPC projections in LS reduces food intake. Consistently, food intake is suppressed by chemogenetic activation of glutamatergic neurons in the vHPC that project to the LS and inactivation of LS neurons blunts vHPC-induced suppression of feeding. Collectively, our results identify an anorexigenic neural circuit originating from vHPC to LS in the brain, revealing a potential therapeutic target for the treatment of anorexia or other appetite disorders.


Subject(s)
Eating/physiology , Hippocampus/anatomy & histology , Hippocampus/physiology , Septal Nuclei/anatomy & histology , Septal Nuclei/physiology , Animals , Anxiety , Brain Mapping , Locomotion , Mice , Neurons/physiology , Random Allocation
20.
Elife ; 42015 Sep 11.
Article in English | MEDLINE | ID: mdl-26359633

ABSTRACT

N-Methyl-D-aspartate receptors (NMDA-Rs) are ion channels that are important for synaptic plasticity, which is involved in learning and drug addiction. We show enzymatic targeting of an NMDA-R antagonist, MK801, to a molecularly defined neuronal population with the cell-type-selectivity of genetic methods and the temporal control of pharmacology. We find that NMDA-Rs on dopamine neurons are necessary for cocaine-induced synaptic potentiation, demonstrating that cell type-specific pharmacology can be used to dissect signaling pathways within complex brain circuits.


Subject(s)
Dizocilpine Maleate/pharmacology , Dopaminergic Neurons/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Cocaine/pharmacokinetics , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...