Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Neurosci ; 24(7): 913-929, 2021 07.
Article in English | MEDLINE | ID: mdl-34002087

ABSTRACT

Pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus represent key regulators of metabolic homeostasis. Electrophysiological and single-cell sequencing experiments have revealed a remarkable degree of heterogeneity of these neurons. However, the exact molecular basis and functional consequences of this heterogeneity have not yet been addressed. Here, we have developed new mouse models in which intersectional Cre/Dre-dependent recombination allowed for successful labeling, translational profiling and functional characterization of distinct POMC neurons expressing the leptin receptor (Lepr) and glucagon like peptide 1 receptor (Glp1r). Our experiments reveal that POMCLepr+ and POMCGlp1r+ neurons represent largely nonoverlapping subpopulations with distinct basic electrophysiological properties. They exhibit a specific anatomical distribution within the arcuate nucleus and differentially express receptors for energy-state communicating hormones and neurotransmitters. Finally, we identify a differential ability of these subpopulations to suppress feeding. Collectively, we reveal a notably distinct functional microarchitecture of critical metabolism-regulatory neurons.


Subject(s)
Feeding Behavior/physiology , Hypothalamus/physiology , Neurons/physiology , Pro-Opiomelanocortin/metabolism , Animals , Energy Metabolism/physiology , Homeostasis/physiology , Hypothalamus/cytology , Mice , Mice, Transgenic , Neurons/cytology
2.
Diabetes Obes Metab ; 22(8): 1328-1338, 2020 08.
Article in English | MEDLINE | ID: mdl-32196896

ABSTRACT

AIMS: To test specific mono-agonists to the glucagon-like peptide-1 receptor (GLP-1R), glucagon receptor (GCGR) and glucose-dependent insulinotropic peptide receptor (GIPR), individually and in combination, in a mouse model of diet-induced non-alcoholic steatohepatitis (NASH) and fibrosis in order to decipher the contribution of their activities and potential additive effects to improving systemic and hepatic metabolism. MATERIALS AND METHODS: We induced NASH by pre-feeding C57BL/6J mice a diet rich in fat, fructose and cholesterol for 36 weeks. This was followed by 8 weeks of treatment with the receptor-specific agonists 1-GCG (20 µg/kg twice daily), 2-GLP1 (3 µg/kg twice daily) or 3-GIP (30 µg/kg twice daily), or the dual (1 + 2) or triple (1 + 2 + 3) combinations thereof. A dual GLP-1R/GCGR agonistic peptide, 4-dual-GLP1/GCGR (30 µg/kg twice daily), and liraglutide (100 µg/kg twice daily) were included as references. RESULTS: Whereas low-dose 1-GCG or 3-GIP alone did not influence body weight, liver lipids and histology, their combination with 2-GLP1 provided additional weight loss, reduction in liver triglycerides and improvement in histological disease activity score. Notably, 4-dual-GLP-1R/GCGR and the triple combination of selective mono-agonists led to a significantly stronger reduction in the histological non-alcoholic fatty liver disease activity score compared to high-dose liraglutide, at the same extent of body weight loss. CONCLUSIONS: GCGR and GIPR agonism provide additional, body weight-independent improvements on top of GLP-1R agonism in a murine model of manifest NASH with fibrosis.


Subject(s)
Incretins , Non-alcoholic Fatty Liver Disease , Animals , Glucagon-Like Peptide-1 Receptor , Incretins/therapeutic use , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Glucagon
3.
J Cereb Blood Flow Metab ; 37(3): 1014-1029, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27193035

ABSTRACT

Aging causes major alterations of all components of the neurovascular unit and compromises brain blood supply. Here, we tested how aging affects vascular reactivity in basilar arteries from young (<10 weeks; y-BA), old (>22 months; o-BA) and old (>22 months) heterozygous MYPT1-T-696A/+ knock-in mice. In isometrically mounted o-BA, media thickness was increased by ∼10% while the passive length tension relations were not altered. Endothelial denudation or pan-NOS inhibition (100 µmol/L L-NAME) increased the basal tone by 11% in y-BA and 23% in o-BA, while inhibition of nNOS (1 µmol/L L-NPA) induced ∼10% increase in both ages. eNOS expression was ∼2-fold higher in o-BA. In o-BA, U46619-induced force was augmented (pEC50 ∼6.9 vs. pEC50 ∼6.5) while responsiveness to DEA-NONOate, electrical field stimulation or nicotine was decreased. Basal phosphorylation of MLC20-S19 and MYPT1-T-853 was higher in o-BA and was reversed by apocynin. Furthermore, permeabilized o-BA showed enhanced Ca2+-sensitivity. Old T-696A/+ BA displayed a reduced phosphorylation of MYPT1-T696 and MLC20, a lower basal tone in response to L-NAME and a reduced eNOS expression. The results indicate that the vascular hypercontractility found in o-BA is mediated by inhibition of MLCP and is partially compensated by an upregulation of endothelial NO release.


Subject(s)
Acetophenones/pharmacology , Aging , Basilar Artery/physiology , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Animals , Enzyme Inhibitors , Mice , Myosin-Light-Chain Phosphatase/metabolism , Nitric Oxide Synthase Type I/metabolism , Phosphorylation , Protein Subunits/metabolism , Vasoconstriction
4.
Nat Neurosci ; 14(7): 911-8, 2011 Jun 05.
Article in English | MEDLINE | ID: mdl-21642975

ABSTRACT

Steroidogenic factor 1 (SF-1)-expressing neurons of the ventromedial hypothalamus (VMH) control energy homeostasis, but the role of insulin action in these cells remains undefined. We show that insulin activates phosphatidylinositol-3-OH kinase (PI3K) signaling in SF-1 neurons and reduces firing frequency in these cells through activation of K(ATP) channels. These effects were abrogated in mice with insulin receptor deficiency restricted to SF-1 neurons (SF-1(ΔIR) mice). Whereas body weight and glucose homeostasis remained the same in SF-1(ΔIR) mice as in controls under a normal chow diet, they were protected from diet-induced leptin resistance, weight gain, adiposity and impaired glucose tolerance. High-fat feeding activated PI3K signaling in SF-1 neurons of control mice, and this response was attenuated in the VMH of SF-1(ΔIR) mice. Mimicking diet-induced overactivation of PI3K signaling by disruption of the phosphatidylinositol-3,4,5-trisphosphate phosphatase PTEN led to increased body weight and hyperphagia under a normal chow diet. Collectively, our experiments reveal that high-fat diet-induced, insulin-dependent PI3K activation in VMH neurons contributes to obesity development.


Subject(s)
Dietary Fats/adverse effects , Neurons/drug effects , Obesity/chemically induced , Obesity/pathology , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Insulin/metabolism , Ventromedial Hypothalamic Nucleus/pathology , Action Potentials/drug effects , Action Potentials/genetics , Age Factors , Animals , Animals, Newborn , Blood Glucose/drug effects , Body Weight/drug effects , Calorimetry/methods , Dose-Response Relationship, Drug , Eating/drug effects , Eating/physiology , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Female , Gene Expression Regulation/drug effects , Glucose Tolerance Test , Green Fluorescent Proteins/genetics , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Injections, Intraventricular/methods , Insulin/pharmacology , Leptin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Patch-Clamp Techniques , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Time Factors , Tolbutamide/pharmacology , Ventromedial Hypothalamic Nucleus/cytology
5.
Physiol Behav ; 97(5): 632-8, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19351541

ABSTRACT

As the obesity epidemic, diabetes mellitus type 2, and associated comorbidities show no signs of abating, large efforts have been put into a better understanding of the homeostatic control mechanisms involved in regulation of body weight and energy homeostasis. For decades, the hypothalamic arcuate nucleus (ARC), which integrates peripheral signals and modulates appetite and metabolism, has been the focus of investigation. Besides these basic homeostatic circuits, food palatability and reward are thought to be major factors involved in the regulation of food intake. Highly palatable food is easily available, and is ingested even when there is no metabolic need for it. Thus, overriding of the homeostatic control systems by the cognitive, rewarding, social, and emotional aspects of palatable food may contribute to the obesity epidemic. This review aims to provide an updated view, how insulin and leptin as signals originating from the periphery of the body and communicating energy availability to the CNS act not only on ARC neurons, but also directly control the activity of neuronal circuits in control of food-associated reward mechanisms.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Energy Metabolism/physiology , Homeostasis/physiology , Insulin/metabolism , Leptin/metabolism , Animals , Humans
6.
Cell Metab ; 7(4): 291-301, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18396135

ABSTRACT

Insulin- and leptin-stimulated phosphatidylinositol-3 kinase (PI3K) activation has been demonstrated to play a critical role in central control of energy homeostasis. To delineate the importance of pathways downstream of PI3K specifically in pro-opiomelanocortin (POMC) cell regulation, we have generated mice with selective inactivation of 3-phosphoinositide-dependent protein kinase 1 (PDK1) in POMC-expressing cells (PDK1(DeltaPOMC) mice). PDK1(DeltaPOMC) mice initially display hyperphagia, increased body weight, and impaired glucose metabolism caused by reduced hypothalamic POMC expression. On the other hand, PDK1(DeltaPOMC) mice exhibit progressive, severe hypocortisolism caused by loss of POMC-expressing corticotrophs in the pituitary. Expression of a dominant-negative mutant of FOXO1 specifically in POMC cells is sufficient to ameliorate positive energy balance in PDK1(DeltaPOMC) mice but cannot restore regular pituitary function. These results reveal important but differential roles for PDK1 signaling in hypothalamic and pituitary POMC cells in the control of energy homeostasis and stress response.


Subject(s)
Energy Metabolism , Forkhead Transcription Factors/metabolism , Pro-Opiomelanocortin/metabolism , Protein Serine-Threonine Kinases/deficiency , Stress, Physiological , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Body Weight/drug effects , Corticosterone/metabolism , Corticosterone/pharmacology , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Gene Deletion , Gene Expression Regulation , Hyperphagia/genetics , Hypothalamus/cytology , Hypothalamus/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pituitary Gland/metabolism , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...