Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R929-R939, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32130027

ABSTRACT

Women in low- and middle-income countries frequently consume a protein-deficient diet during pregnancy and breastfeeding. The effects of gestational malnutrition on fetal and early postnatal development can have lasting adverse effects on offspring metabolism. Expanding on previous studies in rodent models, we utilized a nonhuman primate model of gestational and early-life protein restriction (PR) to evaluate effects on the organ development and glucose metabolism of juvenile offspring. Offspring were born to dams that had consumed a control diet containing 26% protein or a PR diet containing 13% protein. Offspring were maintained on the PR diet and studied [body and serum measurements, intravenous glucose tolerance tests (ivGTTs), and dual-energy X-ray absorptiometry scans] up to 7 mo of age, at which time tissues were collected for analysis. PR offspring had age-appropriate body weight and were euglycemic but exhibited elevated fasting insulin and reduced initial, but increased total, insulin secretion during an ivGTT at 6 mo of age. No changes were detected in pancreatic islets of PR juveniles; however, PR did induce changes, including reduced kidney size, and changes in liver, adipose tissue, and muscle gene expression in other peripheral organs. Serum osteocalcin was elevated and bone mineral content and density were reduced in PR juveniles, indicating a significant impact of PR on early postnatal bone development.


Subject(s)
Animal Nutritional Physiological Phenomena , Diet, Protein-Restricted , Energy Metabolism , Fetal Growth Retardation/metabolism , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects , Age Factors , Animals , Blood Glucose/metabolism , Body Composition , Bone Development , Disease Models, Animal , Energy Metabolism/genetics , Female , Fetal Growth Retardation/etiology , Fetal Growth Retardation/genetics , Fetal Growth Retardation/physiopathology , Gene Expression Regulation, Developmental , Insulin Resistance , Macaca mulatta , Male , Nutritional Status , Pregnancy
2.
Am J Physiol Regul Integr Comp Physiol ; 313(2): R169-R179, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28404581

ABSTRACT

Maternal high-fat-diet (HFD) consumption during pregnancy decreased fetal body weight and impacted development of hypothalamic melanocortin neural circuitry in nonhuman primate offspring. We investigated whether these impairments during gestation persisted in juvenile offspring and examined the interaction between maternal and early postnatal HFD consumption. Adult dams consumed either a control diet (CTR; 15% calories from fat) or a high-saturated-fat diet (HFD; 37% calories from fat) during pregnancy. Offspring were weaned onto a CTR or HFD at ~8 mo of age. Offspring from HFD-fed dams displayed early catch-up growth and elevated body weight at 6 and 13 mo of age. Maternal and postnatal HFD exposure reduced the amount of agouti-related peptide fibers in the paraventricular nucleus of the hypothalamus. Postnatal HFD consumption also decreased the amount of agouti-related peptide fibers in the arcuate nucleus of the hypothalamus. Postnatal HFD was associated with decreased food intake and increased activity. These results support and extend our previous findings of maternal diet effects on fetal development and reveal, for the first time in a nonhuman primate model, that maternal HFD-induced disturbances in offspring body weight regulation extended past gestation into the juvenile period. Maternal HFD consumption increases the risk for offspring developing obesity, with the developmental timing of HFD exposure differentially impacting the melanocortin system and energy balance regulation. The present findings provide translational insight into human clinical populations, suggesting that profound health consequences may await individuals later in life following intrauterine and postnatal HFD exposure.


Subject(s)
Diet, High-Fat/adverse effects , Eating , Hypothalamus/physiopathology , Melanocortins/metabolism , Obesity/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Energy Metabolism , Feeding Behavior , Female , Fetal Development , Humans , Macaca , Male , Obesity/etiology , Pregnancy , Pregnancy, Animal , Signal Transduction
3.
J Neurosci ; 35(22): 8558-69, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26041922

ABSTRACT

Neurons coexpressing neuropeptide Y, agouti-related peptide, and GABA (NAG) play an important role in ingestive behavior and are located in the arcuate nucleus of the hypothalamus. NAG neurons receive both GABAergic and glutamatergic synaptic inputs, however, the developmental time course of synaptic input organization of NAG neurons in mice is unknown. In this study, we show that these neurons have low numbers of GABAergic synapses and that GABA is inhibitory to NAG neurons during early postnatal period. In contrast, glutamatergic inputs onto NAG neurons are relatively abundant by P13 and are comparatively similar to the levels observed in the adult. As mice reach adulthood (9-10 weeks), GABAergic tone onto NAG neurons increases. At this age, NAG neurons received similar numbers of inhibitory and EPSCs. To further differentiate age-associated changes in synaptic distribution, 17- to 18-week-old lean and diet-induced obesity (DIO) mice were studied. Surprisingly, NAG neurons from lean adult mice exhibit a reduction in the GABAergic synapses compared with younger adults. Conversely, DIO mice display reductions in the number of GABAergic and glutamatergic inputs onto NAG neurons. Based on these experiments, we propose that synaptic distribution in NAG neurons is continuously restructuring throughout development to accommodate the animals' energy requirements.


Subject(s)
Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/growth & development , Neurons/physiology , Synapses/physiology , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Age Factors , Animals , Animals, Newborn , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Lysine/analogs & derivatives , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Sodium Channel Blockers/pharmacology , Synapses/drug effects , Synapses/genetics , Tetrodotoxin/pharmacology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/pharmacology
4.
J Neurosci ; 34(30): 9982-94, 2014 Jul 23.
Article in English | MEDLINE | ID: mdl-25057200

ABSTRACT

Leptin is well known for its role in the regulation of energy homeostasis in adults, a mechanism that at least partially results from the inhibition of the activity of NPY/AgRP/GABA neurons (NAG) in the arcuate nucleus of the hypothalamus (ARH). During early postnatal development in the rodent, leptin promotes axonal outgrowth from ARH neurons, and preautonomic NAG neurons are particularly responsive to leptin's trophic effects. To begin to understand how leptin could simultaneously promote axonal outgrowth from and inhibit the activity of NAG neurons, we characterized the electrochemical effects of leptin on NAG neurons in mice during early development. Here, we show that NAG neurons do indeed express a functional leptin receptor throughout the early postnatal period in the mouse; however, at postnatal days 13-15, leptin causes membrane depolarization in NAG neurons, rather than the expected hyperpolarization. Leptin action on NAG neurons transitions from stimulatory to inhibitory in the periweaning period, in parallel with the acquisition of functional ATP-sensitive potassium channels. These findings are consistent with the idea that leptin provides an orexigenic drive through the NAG system to help rapidly growing pups meet their energy requirements.


Subject(s)
Arcuate Nucleus of Hypothalamus/growth & development , Leptin/physiology , Neurons/physiology , Receptors, Leptin/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , Male , Mice , Mice, Transgenic , Receptors, Leptin/biosynthesis
5.
Circulation ; 129(4): 471-8, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24163066

ABSTRACT

BACKGROUND: Inflammation and insulin resistance (IR) are associated processes that potentiate risk for cardiovascular disease in obesity. The temporal relation between IR and inflammation is not completely characterized. We hypothesized that endothelial cell adhesion molecule expression in large arteries is an early event that coincides with diet-induced obesity and IR in primates. METHODS AND RESULTS: Ten adult male rhesus macaques were studied at baseline and every 4 to 6 months on a high-fat diet for 2 years. Truncal fat, carotid intima-media thickness, plasma inflammatory biomarkers, and carotid P-selectin and vascular cell adhesion molecule-1 expression by contrast-enhanced ultrasound molecular imaging were assessed. Intravenous glucose tolerance test was performed at baseline and at 4 and 18 months. A high-fat diet produced a rapid increase (P<0.01) in weight, truncal fat, and degree of IR indicated by the insulin area under the curve and glucose disappearance rate on intravenous glucose tolerance test, all of which worsened minimally thereafter. Molecular imaging detected a progressive increase in endothelial cell adhesion molecule expression over time (5- to 7-fold greater than control agent signal at 2 years; P<0.01). Changes in intima-media thickness were not detected until 2 years and, although there was a trend toward an increase in plasma markers of inflammation (monocyte chemotactic protein-1, C-reactive protein), the pattern of increase varied considerably over time. CONCLUSIONS: In primates with diet-induced obesity, endothelial inflammatory activation is an early event that occurs coincident with the development of IR and long before any measurable change in carotid intima-media thickness. Endothelial activation is related more to the duration rather than to the severity of IR and is not mirrored by changes in plasma biomarkers.


Subject(s)
Carotid Arteries/physiopathology , Disease Progression , Endothelium, Vascular/physiopathology , Insulin Resistance/physiology , Macaca mulatta/physiology , Obesity/physiopathology , Vasculitis/physiopathology , Animals , Biomarkers/metabolism , C-Reactive Protein/metabolism , Carotid Arteries/diagnostic imaging , Carotid Arteries/metabolism , Carotid Intima-Media Thickness , Chemokine CCL2/metabolism , Disease Models, Animal , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/metabolism , Male , Microbubbles , Molecular Diagnostic Techniques , Obesity/metabolism , P-Selectin/metabolism , Time Factors , Ultrasonography, Interventional , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/metabolism
6.
J Neurosci ; 33(38): 15306-17, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048859

ABSTRACT

Neuropeptide Y (NPY) neurons in both the arcuate nucleus of the hypothalamus (ARH) and the dorsomedial hypothalamus (DMH) have been implicated in food intake and obesity. However, while ARH NPY is highly expressed in the lean animal, DMH NPY mRNA expression is observed only after diet-induced obesity (DIO). Furthermore, while ARH NPY neurons are inhibited by leptin, the effect of this adipokine on DMH NPY neurons is unknown. In this study we show that in contrast to the consistent expression in the ARH, DMH NPY mRNA expression was undetectable until after 10 weeks in mice fed a high-fat diet, and peaked at 20 weeks. Surprisingly, electrophysiological experiments demonstrated that leptin directly depolarized and increased the firing rate of DMH NPY neurons in DIO mice. To further differentiate the regulation of DMH and ARH NPY populations, fasting decreased expression of DMH NPY expression, while it increased ARH NPY expression. However, treatment with a leptin receptor antagonist failed to alter DMH NPY expression, indicating that leptin may not be the critical factor regulating mRNA expression. Importantly, we also demonstrated that DMH NPY neurons coexpress cocaine amphetamine-regulated transcript (CART); however, CART mRNA expression in the DMH peaked earlier in the progression of DIO. This study demonstrates novel and important findings. First, NPY and CART are coexpressed in the same neurons within the DMH, and second, leptin stimulates DMH NPY neurons. These studies suggest that during the progression of DIO, there is an unknown signal that drives the expression of the orexigenic NPY signal within the DMH, and that the chronic hyperleptinemia increases the activity of these NPY/CART neurons.


Subject(s)
Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Leptin/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Obesity/pathology , Action Potentials/drug effects , Action Potentials/genetics , Analysis of Variance , Animals , Diet/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hypothalamus/pathology , In Vitro Techniques , Insulin/blood , Leptin/antagonists & inhibitors , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neuropeptide Y/genetics , Obesity/blood , Obesity/etiology , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Radioimmunoassay , STAT3 Transcription Factor/metabolism , Time Factors
7.
Neuroendocrinology ; 99(3-4): 190-203, 2014.
Article in English | MEDLINE | ID: mdl-25011649

ABSTRACT

BACKGROUND/AIMS: Kisspeptin is the major excitatory regulator of gonadotropin-releasing hormone (GnRH) neurons and is responsible for basal GnRH/LH release and the GnRH/LH surge. Although it is widely assumed, based on mutations in kisspeptin and Kiss1R, that kisspeptin acts to sustain basal GnRH neuronal activity, there have been no studies to investigate whether endogenous basal kisspeptin tone plays a direct role in basal spontaneous GnRH neuronal excitability. It is also of interest to examine possible interactions between endogenous kisspeptin tone and other neuropeptides that have direct effects on GnRH neurons, such as neuropeptide Y (NPY) or cocaine- and amphetamine-regulated transcript (CART), since the activity of all these neuropeptides changes during states of negative energy balance. METHODS: Loose cell-attached and whole-cell current patch-clamp recordings were made from GnRH-GFP neurons in hypothalamic slices from female and male rats. RESULTS: Kisspeptin activated GnRH neurons in a concentration-dependent manner with an EC50 of 3.32 ± 0.02 nM. Surprisingly, a kisspeptin antagonist, Peptide 347, suppressed spontaneous activity in GnRH neurons, demonstrating the essential nature of the endogenous kisspeptin tone. Furthermore, inhibition of endogenous kisspeptin tone blocked the direct activation of GnRH cells that occurs in response to antagonism of NPY Y5 receptor or by CART. CONCLUSIONS: Our electrophysiology studies suggest that basal endogenous kisspeptin tone is not only essential for spontaneous GnRH neuronal firing, but it is also required for the net excitatory effects of other neuropeptides, such as CART or NPY antagonism, on GnRH neurons. Therefore, endogenous kisspeptin tone could serve as the linchpin in GnRH activation or inhibition.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Kisspeptins/metabolism , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Neuropeptide Y/pharmacology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Estradiol/pharmacology , Female , Gonadotropin-Releasing Hormone/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Kisspeptins/antagonists & inhibitors , Kisspeptins/pharmacology , Male , Neurons/physiology , Ovariectomy , Patch-Clamp Techniques , Preoptic Area/cytology , Rats , Rats, Transgenic , Rats, Wistar , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
8.
J Clin Invest ; 133(19)2023 10 02.
Article in English | MEDLINE | ID: mdl-37581939

ABSTRACT

The adipose-derived hormone leptin acts via its receptor (LepRb) in the brain to control energy balance. A potentially unidentified population of GABAergic hypothalamic LepRb neurons plays key roles in the restraint of food intake and body weight by leptin. To identify markers for candidate populations of LepRb neurons in an unbiased manner, we performed single-nucleus RNA-Seq of enriched mouse hypothalamic LepRb cells, identifying several previously unrecognized populations of hypothalamic LepRb neurons. Many of these populations displayed strong conservation across species, including GABAergic Glp1r-expressing LepRb (LepRbGlp1r) neurons, which expressed more Lepr than other LepRb cell populations. Ablating Lepr from LepRbGlp1r cells provoked hyperphagic obesity without impairing energy expenditure. Similarly, improvements in energy balance caused by Lepr reactivation in GABA neurons of otherwise Lepr-null mice required Lepr expression in GABAergic Glp1r-expressing neurons. Furthermore, restoration of Glp1r expression in LepRbGlp1r neurons in otherwise Glp1r-null mice enabled food intake suppression by the GLP1R agonist, liraglutide. Thus, the conserved GABAergic LepRbGlp1r neuron population plays crucial roles in the suppression of food intake by leptin and GLP1R agonists.


Subject(s)
Leptin , Obesity , Mice , Animals , Leptin/genetics , Leptin/metabolism , Obesity/genetics , Obesity/prevention & control , Obesity/metabolism , Hypothalamus/metabolism , Mice, Knockout , GABAergic Neurons/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Eating/genetics
9.
Endocrinology ; 150(1): 333-40, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18719019

ABSTRACT

Increased neuropeptide Y (NPY) activity drives the chronic hyperphagia of lactation and may contribute to the suppression of GnRH activity. The majority of GnRH neurons are contacted by NPY fibers, and GnRH cells express NPY Y5 receptor (Y5R). Therefore, NPY provides a neurocircuitry for information about food intake/energy balance to be directly transmitted to GnRH neurons. To investigate the effects of lactation on GnRH neuronal activity, hypothalamic slices were prepared from green fluorescent protein-GnRH transgenic rats. Extracellular loose-patch recordings determined basal GnRH neuronal activity from slices of ovariectomized control and lactating rats. Compared with controls, hypothalamic slices from lactating rats had double the number of quiescent GnRH neurons (14.51 +/- 2.86 vs. 7.04 +/- 2.84%) and significantly lower firing rates of active GnRH neurons (0.25 +/- 0.02 vs. 0.37 +/- 0.03 Hz). To study the NPY-postsynaptic Y5R system, whole-cell current-clamp recordings were performed in hypothalamic slices from control rats to examine NPY/Y5R antagonist effects on GnRH neuronal resting membrane potential. Under tetrodotoxin treatment, NPY hyperpolarized GnRH neurons from -56.7 +/- 1.94 to -62.1 +/- 1.83 mV; NPY's effects were blocked by Y5R antagonist. To determine whether increased endogenous NPY tone contributes to GnRH neuronal suppression during lactation, hypothalamic slices were treated with Y5R antagonist. A significantly greater percentage of GnRH cells were activated in slices from lactating rats (52%) compared with controls (28%). These results suggest that: 1) basal GnRH neuronal activity is suppressed during lactation; 2) NPY can hyperpolarize GnRH neurons via postsynaptic Y5R; and 3) increased inhibitory NPY tone during lactation is a component of the mechanisms responsible for suppression of GnRH neuronal activity.


Subject(s)
Brain/physiology , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Lactation/physiology , Neuropeptide Y/pharmacology , Animals , Animals, Genetically Modified , Brain/drug effects , Electrophysiology , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Models, Neurological , Neurons/drug effects , Neurons/physiology , Rats , Rats, Wistar
10.
Diabetes ; 68(7): 1462-1472, 2019 07.
Article in English | MEDLINE | ID: mdl-31048370

ABSTRACT

Fibroblast growth factor 1 (FGF1) has been shown to reverse hyperglycemia in diabetic rodent models through peripheral and central administration routes. Previous studies demonstrated that insulin is required for central and peripheral FGF1 metabolic improvements; however, it is unknown if FGF1 targets insulin secretion at the islet level. Here we show for the first time that FGF1 increases islet insulin secretion in diabetic mouse models. FGF1 was administered via a single intracerebroventricular or multiple subcutaneous injections to leptin receptor-deficient (db/db), diet-induced obese, and control mice; pancreatic islets were isolated 7 days later for analysis of insulin secretion. Central and peripheral FGF1 significantly lowered blood glucose in vivo and increased ex vivo islet insulin secretion from diabetic, but not control, mice. FGF1 injections to the cisterna magna mimicked intracerebroventricular outcomes, pointing to a novel therapeutic potential. Central effects of FGF1 appeared dependent on reductions in food intake, whereas peripheral FGF1 had acute actions on islet function prior to significant changes in food intake or blood glucose. Additionally, peripheral, but not central, FGF1 increased islet ß-cell density, suggesting that peripheral FGF1 may induce long-term changes in islet structure and function that are not present with central treatment.


Subject(s)
Fibroblast Growth Factor 1/therapeutic use , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Fibroblast Growth Factor 1/administration & dosage , Glucose Tolerance Test , Immunohistochemistry , Infusions, Intraventricular , Injections, Subcutaneous , Insulin Secretion/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fos/metabolism
11.
Mol Metab ; 26: 18-29, 2019 08.
Article in English | MEDLINE | ID: mdl-31230943

ABSTRACT

OBJECTIVE: Reelin (RELN) is a large glycoprotein involved in synapse maturation and neuronal organization throughout development. Deficits in RELN signaling contribute to multiple psychological disorders, such as autism spectrum disorder, schizophrenia, and bipolar disorder. Nutritional stress alters RELN expression in brain regions associated with these disorders; however, the involvement of RELN in the neural circuits involved in energy metabolism is unknown. The RELN receptors apolipoprotein E receptor 2 (ApoER2) and very low-density lipoprotein receptor (VLDLR) are involved in lipid metabolism and expressed in the hypothalamus. Here we explored the involvement of RELN in hypothalamic signaling and the impact of diet-induced obesity (DIO) on this system. METHODS: Adult male mice were fed a chow diet or maintained on a high-fat diet (HFD) for 12-16 weeks. HFD-fed DIO mice exhibited decreased ApoER2 and VLDLR expression and increased RELN protein in the hypothalamus. Electrophysiology was used to determine the mechanism by which the central fragment of RELN (CF-RELN) acts on arcuate nucleus (ARH) satiety-promoting proopiomelanocortin (POMC) neurons and the impact of DIO on this circuitry. RESULTS: CF-RELN exhibited heterogeneous presynaptic actions on inhibitory inputs onto ARH-POMC-EGFP neurons and consistent postsynaptic actions. Additionally, central administration of CF-RELN caused a significant increase in ARH c-Fos expression and an acute decrease in food intake and body weight. CONCLUSIONS: We conclude that RELN signaling is modulated by diet, that RELN is involved in synaptic signaling onto ARH-POMC neurons, and that altering central CF-RELN levels can impact food intake and body weight.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Serine Endopeptidases/metabolism , Animals , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/chemically induced , Reelin Protein
12.
eNeuro ; 4(1)2017.
Article in English | MEDLINE | ID: mdl-28144621

ABSTRACT

Kisspeptin (Kiss1) neurons in the hypothalamic arcuate nucleus (ARC) are key components of the hypothalamic-pituitary-gonadal axis, as they regulate the basal pulsatile release of gonadotropin releasing hormone (GnRH). ARC Kiss1 action is dependent on energy status, and unmasking metabolic factors responsible for modulating ARC Kiss1 neurons is of great importance. One possible factor is glucagon-like peptide 1 (GLP-1), an anorexigenic neuropeptide produced by brainstem preproglucagon neurons. Because GLP fiber projections and the GLP-1 receptor (GLP-1R) are abundant in the ARC, we hypothesized that GLP-1R signaling could modulate ARC Kiss1 action. Using ovariectomized mice, we found that GLP-producing fibers come in close apposition with ARC Kiss1 neurons; these neurons also contain Glp1r mRNA. Electrophysiological recordings revealed that liraglutide (a long-acting GLP-1R agonist) increased action potential firing and caused a direct membrane depolarization of ARC Kiss1 cells in brain slices. We determined that brainstem preproglucagon mRNA is decreased after a 48-h fast in mice, a negative energy state in which ARC Kiss1 expression and downstream GnRH/luteinizing hormone (LH) release are potently suppressed. However, activation of GLP-1R signaling in fasted mice with liraglutide was not sufficient to prevent LH inhibition. Furthermore, chronic central infusions of the GLP-1R antagonist, exendin(9-39), in ad libitum-fed mice did not alter ARC Kiss1 mRNA or plasma LH. As a whole, these data identify a novel interaction of the GLP-1 system with ARC Kiss1 neurons but indicate that CNS GLP-1R signaling alone is not critical for the maintenance of LH during fasting or normal feeding.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Fasting/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/blood , Neurons/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/metabolism , Drug Implants , Eating/physiology , Estradiol/administration & dosage , Estrogens/administration & dosage , Female , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptides/metabolism , Luteinizing Hormone/antagonists & inhibitors , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Ovariectomy , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tissue Culture Techniques
13.
Obesity (Silver Spring) ; 23(11): 2157-64, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26530932

ABSTRACT

OBJECTIVE: To utilize a nonhuman primate model to examine the impact of maternal high-fat diet (HFD) consumption and pre-pregnancy obesity on offspring intake of palatable food and to examine whether maternal HFD consumption impaired development of the dopamine system, critical for the regulation of hedonic feeding. METHODS: The impact of exposure to maternal HFD and obesity on offspring consumption of diets of varying composition was assessed after weaning. The influence of maternal HFD consumption on the development of the prefrontal cortex-dopaminergic system at 13 months of age was also examined. RESULTS: During a preference test, offspring exposed to maternal HFD consumption and obesity displayed increased intake of food high in fat and sugar content relative to offspring from lean control mothers. Maternal HFD consumption suppressed offspring dopamine signaling (as assessed by immunohistochemistry) relative to control offspring. Specifically, there was decreased abundance of dopamine fibers and of dopamine receptor 1 and 2 proteins. CONCLUSIONS: This study reveals that offspring exposed to both maternal HFD consumption and maternal obesity during early development are at increased risk for obesity due to overconsumption of palatable energy-dense food, a behavior that may be related to reduced central dopamine signaling.


Subject(s)
Diet, High-Fat/adverse effects , Dopamine/metabolism , Eating/physiology , Maternal Nutritional Physiological Phenomena , Obesity/metabolism , Pregnancy Complications/metabolism , Prenatal Exposure Delayed Effects , Animals , Feeding Behavior/physiology , Female , Male , Models, Animal , Obesity/etiology , Pregnancy , Pregnancy Complications/physiopathology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Primates , Signal Transduction , Taste/physiology
14.
Endocrinology ; 151(4): 1598-610, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20194730

ABSTRACT

Childhood obesity increases the risk of adult obesity and diabetes, suggesting that early overnutrition permanently programs altered energy and glucose homeostasis. In the present studies, we used a mouse model to investigate whether early overnutrition increases susceptibility to obesity and insulin resistance in response to a high-fat diet (HFD). Litters from Swiss Webster dams were culled to three [chronic postnatal overnutrition (CPO)] or 10 (control) pups and then weaned onto standard chow at postnatal day (P) 23. At 6 wk of age, a subset of mice was placed on HFD, and glucose and insulin tolerance were examined at 16-17 wk of age. Leptin sensitivity was determined by hypothalamic phosphorylated signal transducer and activator of transcription-3 immunoreactivity at P16 and adulthood after ip leptin. CPO mice exhibited accelerated body weight gain and hyperleptinemia during the preweaning period but only a slightly heavier body weight and normal glucose tolerance in adulthood on standard chow diet. Importantly, CPO mice exhibited significant leptin resistance in the arcuate nucleus, demonstrated by reduced activation of phospho-signal transducer and activator of transcription-3, as early as P16 and throughout life, despite normalized leptin levels. In response to HFD, CPO but not control mice displayed insulin resistance in response to an insulin tolerance test. In conclusion, CPO mice exhibited early and persistent leptin resistance in the arcuate nucleus and, in response to HFD, rapid development of obesity and insulin resistance. These studies suggest that early overnutrition can permanently alter energy homeostasis and significantly increase susceptibility to obesity and insulin resistance.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Dietary Fats/metabolism , Leptin/metabolism , Overnutrition/metabolism , STAT3 Transcription Factor/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/drug effects , Area Under Curve , Blood Glucose , Body Weight/genetics , Eating/physiology , Energy Metabolism/physiology , Glucose Tolerance Test , Immunohistochemistry , Insulin/metabolism , Insulin Resistance/genetics , Leptin/pharmacology , Mice , Motor Activity/physiology , Overnutrition/genetics , Phosphorylation/physiology , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics
15.
Endocrinology ; 150(9): 4231-40, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19470705

ABSTRACT

Negative energy balance during lactation is reflected by low levels of insulin and leptin and is associated with chronic hyperphagia and suppressed GnRH/LH activity. We studied whether restoration of insulin and/or leptin to physiological levels would reverse the lactation-associated hyperphagia, changes in hypothalamic neuropeptide expression [increased neuropeptide Y (NPY) and agouti-related protein (AGRP) and decreased proopiomelanocortin (POMC), kisspeptin (Kiss1), and neurokinin B (NKB)] and suppression of LH. Ovariectomized lactating rats (eight pups) were treated for 48 h with sc minipumps containing saline, human insulin, or rat leptin. The arcuate nucleus (ARH) was analyzed for NPY, AGRP, POMC, Kiss1, and NKB mRNA expression; the dorsal medial hypothalamus (DMH) was analyzed for NPY mRNA. Insulin replacement reversed the increase in ARH NPY/AGRP mRNAs, partially recovered POMC, but had no effect on recovering Kiss1/NKB. Leptin replacement only affected POMC, which was fully recovered. Insulin/leptin dual replacement had similar effects as insulin replacement alone but with a slight increase in Kiss1/NKB. The lactation-induced increase in DMH NPY was unchanged after treatments. Restoration of insulin and/or leptin had no effect on food intake, body weight, serum glucose or serum LH. These results suggest that the negative energy balance of lactation is not required for the hyperphagic drive, although it is involved in the orexigenic changes in the ARH. The chronic hyperphagia of lactation is most likely sustained by the induction of NPY in the DMH. The negative energy balance also does not appear to be a necessary prerequisite for the suppression of GnRH/LH activity.


Subject(s)
Insulin/physiology , Lactation/physiology , Leptin/physiology , Animals , Blood Glucose/metabolism , Eating , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , Insulin/blood , Lactation/drug effects , Leptin/blood , Luteinizing Hormone/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL