Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
EMBO Rep ; 24(8): e57344, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37314252

ABSTRACT

The counterregulatory response to hypoglycemia (CRR), which ensures a sufficient glucose supply to the brain, is an essential survival function. It is orchestrated by incompletely characterized glucose-sensing neurons, which trigger a coordinated autonomous and hormonal response that restores normoglycemia. Here, we investigate the role of hypothalamic Tmem117, identified in a genetic screen as a regulator of CRR. We show that Tmem117 is expressed in vasopressin magnocellular neurons of the hypothalamus. Tmem117 inactivation in these neurons increases hypoglycemia-induced vasopressin secretion leading to higher glucagon secretion in male mice, and this effect is estrus cycle phase dependent in female mice. Ex vivo electrophysiological analysis, in situ hybridization, and in vivo calcium imaging reveal that Tmem117 inactivation does not affect the glucose-sensing properties of vasopressin neurons but increases ER stress, ROS production, and intracellular calcium levels accompanied by increased vasopressin production and secretion. Thus, Tmem117 in vasopressin neurons is a physiological regulator of glucagon secretion, which highlights the role of these neurons in the coordinated response to hypoglycemia.


Subject(s)
Glucagon , Hypoglycemia , Mice , Male , Female , Animals , Glucagon/adverse effects , Calcium , Hypoglycemia/genetics , Hypoglycemia/chemically induced , Vasopressins/adverse effects , Glucose , Neurons/physiology , Blood Glucose , Insulin
2.
Nat Commun ; 13(1): 5761, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36180454

ABSTRACT

The counterregulatory response to hypoglycemia that restores normal blood glucose levels is an essential physiological function. It is initiated, in large part, by incompletely characterized brain hypoglycemia sensing neurons that trigger the secretion of counterregulatory hormones, in particular glucagon, to stimulate hepatic glucose production. In a genetic screen of recombinant inbred BXD mice we previously identified Agpat5 as a candidate regulator of hypoglycemia-induced glucagon secretion. Here, using genetic mouse models, we demonstrate that Agpat5 expressed in agouti-related peptide neurons is required for their activation by hypoglycemia, for hypoglycemia-induced vagal nerve activity, and glucagon secretion. We find that inactivation of Agpat5 leads to increased fatty acid oxidation and ATP production and that suppressing Cpt1a-dependent fatty acid import into mitochondria restores hypoglycemia sensing. Collectively, our data show that AgRP neurons are involved in the control of glucagon secretion and that Agpat5, by partitioning fatty acyl-CoAs away from mitochondrial fatty acid oxidation and ATP generation, ensures that the fall in intracellular ATP, which triggers neuronal firing, faithfully reflects changes in glycemia.


Subject(s)
Glucagon , Hypoglycemia , Adenosine Triphosphate , Agouti-Related Protein/genetics , Animals , Blood Glucose , Fatty Acids , Glucose , Insulin , Lipids/adverse effects , Mice , Neurons
3.
iScience ; 24(10): 103122, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34622169

ABSTRACT

The paraventricular nucleus of the thalamus (PVT) controls goal-oriented behavior through its connections to the nucleus accumbens (NAc). We previously characterized Glut2aPVT neurons that are activated by hypoglycemia, and which increase sucrose seeking behavior through their glutamatergic projections to the NAc. Here, we identified glucokinase (Gck)-expressing neurons of the PVT (GckaPVT) and generated a mouse line expressing the Cre recombinase from the glucokinase locus (Gck Cre/+ mice). Ex vivo calcium imaging and whole-cell patch clamp recordings revealed that GckaPVT neurons that project to the NAc were mostly activated by hyperglycemia. Their chemogenetic inhibition or optogenetic stimulation, respectively, enhanced food intake or decreased sucrose-seeking behavior. Collectively, our results describe a neuronal population of Gck-expressing neurons in the PVT, which has opposite glucose sensing properties and control over feeding behavior than the previously characterized Glut2aPVT neurons. This study allows a better understanding of the complex regulation of feeding behavior by the PVT.

4.
Diabetes ; 70(7): 1443-1457, 2021 07.
Article in English | MEDLINE | ID: mdl-33883213

ABSTRACT

The counterregulatory response to hypoglycemia is an essential survival function. It is controlled by an integrated network of glucose-responsive neurons, which trigger endogenous glucose production to restore normoglycemia. The complexity of this glucoregulatory network is, however, only partly characterized. In a genetic screen of a panel of recombinant inbred mice we previously identified Fgf15, expressed in neurons of the dorsomedial hypothalamus (DMH), as a negative regulator of glucagon secretion. Here, we report on the generation of Fgf15CretdTomato mice and their use to further characterize these neurons. We show that they were glutamatergic and comprised glucose-inhibited and glucose-excited neurons. When activated by chemogenetics, Fgf15 neurons prevented the increase in vagal nerve firing and the secretion of glucagon normally triggered by insulin-induced hypoglycemia. On the other hand, they increased the activity of the sympathetic nerve in the basal state and prevented its silencing by glucose overload. Higher sympathetic tone increased hepatic Creb1 phosphorylation, Pck1 mRNA expression, and hepatic glucose production leading to glucose intolerance. Thus, Fgf15 neurons of the DMH participate in the counterregulatory response to hypoglycemia by a direct adrenergic stimulation of hepatic glucose production while suppressing vagally induced glucagon secretion. This study provides new insights into the complex neuronal network that prevents the development of hypoglycemia.


Subject(s)
Fibroblast Growth Factors/physiology , Glucagon/metabolism , Gluconeogenesis/physiology , Hypothalamus/metabolism , Liver/metabolism , Neurons/physiology , Animals , Cyclic AMP Response Element-Binding Protein/physiology , Female , Hypoglycemia/prevention & control , Male , Mice , Mice, Inbred C57BL , Sympathetic Nervous System/physiology
5.
PLoS Biol ; 18(11): e3000680, 2020 11.
Article in English | MEDLINE | ID: mdl-33253166

ABSTRACT

Proopiomelanocortin (POMC) neurons are major regulators of energy balance and glucose homeostasis. In addition to being regulated by hormones and nutrients, POMC neurons are controlled by glutamatergic input originating from multiple brain regions. However, the factors involved in the formation of glutamatergic inputs and how they contribute to bodily functions remain largely unknown. Here, we show that during the development of glutamatergic inputs, POMC neurons exhibit enriched expression of the Efnb1 (EphrinB1) and Efnb2 (EphrinB2) genes, which are known to control excitatory synapse formation. In vivo loss of Efnb1 in POMC-expressing progenitors decreases the amount of glutamatergic inputs, associated with a reduced number of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) receptor subunits and excitability of these cells. We found that mice lacking Efnb1 in POMC-expressing progenitors display impaired glucose tolerance due to blunted vagus nerve activity and decreased insulin secretion. However, despite reduced excitatory inputs, mice lacking Efnb2 in POMC-expressing progenitors showed no deregulation of insulin secretion and only mild alterations in feeding behavior and gluconeogenesis. Collectively, our data demonstrate the role of ephrins in controlling excitatory input amount into POMC-expressing progenitors and show an isotype-specific role of ephrins on the regulation of glucose homeostasis and feeding.


Subject(s)
Ephrin-B1/metabolism , Glucose/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Brain/metabolism , Energy Metabolism/physiology , Ephrin-B1/physiology , Ephrin-B2/metabolism , Ephrin-B2/physiology , Excitatory Amino Acid Agents/metabolism , Homeostasis/physiology , Male , Mice , Mice, Knockout , N-Methylaspartate/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
6.
Diabetes ; 69(11): 2253-2266, 2020 11.
Article in English | MEDLINE | ID: mdl-32839348

ABSTRACT

The ventromedial nucleus of the hypothalamus (VMN) is involved in the counterregulatory response to hypoglycemia. VMN neurons activated by hypoglycemia (glucose-inhibited [GI] neurons) have been assumed to play a critical although untested role in this response. Here, we show that expression of a dominant negative form of AMPK or inactivation of AMPK α1 and α2 subunit genes in Sf1 neurons of the VMN selectively suppressed GI neuron activity. We found that Txn2, encoding a mitochondrial redox enzyme, was strongly downregulated in the absence of AMPK activity and that reexpression of Txn2 in Sf1 neurons restored GI neuron activity. In cell lines, Txn2 was required to limit glucopenia-induced reactive oxygen species production. In physiological studies, absence of GI neuron activity after AMPK suppression in the VMN had no impact on the counterregulatory hormone response to hypoglycemia or on feeding. Thus, AMPK is required for GI neuron activity by controlling the expression of the antioxidant enzyme Txn2. However, the glucose-sensing capacity of VMN GI neurons is not required for the normal counterregulatory response to hypoglycemia. Instead, it may represent a fail-safe system in case of impaired hypoglycemia sensing by peripherally located glucose detection systems that are connected to the VMN.


Subject(s)
Glucose/metabolism , Hypoglycemia/blood , Neurons/physiology , Thioredoxins/metabolism , Ventromedial Hypothalamic Nucleus/cytology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Blood Glucose , Cells, Cultured , Humans , Patch-Clamp Techniques , Thioredoxins/genetics
7.
J Cereb Blood Flow Metab ; 39(9): 1725-1736, 2019 09.
Article in English | MEDLINE | ID: mdl-29561214

ABSTRACT

Glucose transporter 2 (Glut2)-positive cells are sparsely distributed in brain and play an important role in the stimulation of glucagon secretion in response to hypoglycemia. We aimed to determine if Glut2-positive cells can influence another response to hypoglycemia, i.e. increased cerebral blood flow (CBF). CBF of adult male mice devoid of Glut2, either globally (ripglut1:glut2-/-) or in the nervous system only (NG2KO), and their respective controls were studied under basal glycemia and insulin-induced hypoglycemia using quantitative perfusion magnetic resonance imaging at 9.4 T. The effect on CBF of optogenetic activation of hypoglycemia responsive Glut2-positive neurons of the paraventricular thalamic area was measured in mice expressing channelrhodopsin2 under the control of the Glut2 promoter. We found that in both ripglut1:glut2-/- mice and NG2KO mice, CBF in basal conditions was higher than in their respective controls and not further activated by hypoglycemia, as measured in the hippocampus, hypothalamus and whole brain. Conversely, optogenetic activation of Glut2-positive cells in the paraventricular thalamic nucleus induced a local increase in CBF similar to that induced by hypoglycemia. Thus, Glut2 expression in the nervous system is required for the control of CBF in response to changes in blood glucose concentrations.


Subject(s)
Blood Glucose/metabolism , Cerebrovascular Circulation , Glucose Transporter Type 2/metabolism , Hypoglycemia/metabolism , Animals , Brain/blood supply , Brain/metabolism , Hypoglycemia/blood , Male , Mice, Inbred C57BL
8.
Methods Mol Biol ; 1713: 255-267, 2018.
Article in English | MEDLINE | ID: mdl-29218531

ABSTRACT

Brain glucose sensing plays an essential role in the regulation of energy homeostasis. Recent publications report that neurons expressing glucose transporter GLUT2 act as glucose sensors in different regions of the brain and contribute to the control of glucose homeostasis and feeding behavior. In this chapter we describe the methods used to explore glucose sensing in genetically tagged GLUT2-expressing neurons with slice electrophysiology.


Subject(s)
Brain/physiology , Electrophysiological Phenomena , Gene Expression , Glucose Transporter Type 2/genetics , Glucose/metabolism , Neurons/physiology , Animals , Glucose Transporter Type 2/metabolism , Male , Mice , Molecular Imaging , Patch-Clamp Techniques
9.
J Neurosci ; 36(45): 11469-11481, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27911750

ABSTRACT

Signals of energy homeostasis interact closely with neural circuits of motivation to control food intake. An emerging hypothesis is that the transition to maladaptive feeding behavior seen in eating disorders or obesity may arise from dysregulation of these interactions. Focusing on key brain regions involved in the control of food intake (ventral tegmental area, striatum, hypothalamus, and thalamus), we describe how activity of specific cell types embedded within these regions can influence distinct components of motivated feeding behavior. We review how signals of energy homeostasis interact with these regions to influence motivated behavioral output and present evidence that experience-dependent neural adaptations in key feeding circuits may represent cellular correlates of impaired food intake control. Future research into mechanisms that restore the balance of control between signals of homeostasis and motivated feeding behavior may inspire new treatment options for eating disorders and obesity.


Subject(s)
Appetite Regulation/physiology , Brain/physiology , Eating/physiology , Energy Metabolism/physiology , Homeostasis/physiology , Motivation/physiology , Animals , Body Weight/physiology , Humans
10.
Diabetes ; 65(10): 2920-31, 2016 10.
Article in English | MEDLINE | ID: mdl-27422385

ABSTRACT

Glucokinase (Gck) is a critical regulator of glucose-induced insulin secretion by pancreatic ß-cells. It has been suggested to also play an important role in glucose signaling in neurons of the ventromedial hypothalamic nucleus (VMN), a brain nucleus involved in the control of glucose homeostasis and feeding. To test the role of Gck in VMN glucose sensing and physiological regulation, we studied mice with genetic inactivation of the Gck gene in Sf1 neurons of the VMN (Sf1Gck(-/-) mice). Compared with control littermates, Sf1Gck(-/-) mice displayed increased white fat mass and adipocyte size, reduced lean mass, impaired hypoglycemia-induced glucagon secretion, and a lack of parasympathetic and sympathetic nerve activation by neuroglucopenia. However, these phenotypes were observed only in female mice. To determine whether Gck was required for glucose sensing by Sf1 neurons, we performed whole-cell patch clamp analysis of brain slices from control and Sf1Gck(-/-) mice. Absence of Gck expression did not prevent the glucose responsiveness of glucose-excited or glucose-inhibited Sf1 neurons in either sex. Thus Gck in the VMN plays a sex-specific role in the glucose-dependent control of autonomic nervous activity; this is, however, unrelated to the control of the firing activity of classical glucose-responsive neurons.


Subject(s)
Glucokinase/metabolism , Hypothalamus/enzymology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Female , Glucagon/metabolism , Glucokinase/genetics , Glucose/pharmacology , Homeostasis/drug effects , Hypothalamus/cytology , Hypothalamus/metabolism , Male , Mice , Mice, Mutant Strains , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/enzymology , Ventromedial Hypothalamic Nucleus/metabolism
11.
Nat Neurosci ; 19(8): 999-1002, 2016 08.
Article in English | MEDLINE | ID: mdl-27322418

ABSTRACT

Feeding behavior is governed by homeostatic needs and motivational drive to obtain palatable foods. Here, we identify a population of glutamatergic neurons in the paraventricular thalamus of mice that express the glucose transporter Glut2 (encoded by Slc2a2) and project to the nucleus accumbens. These neurons are activated by hypoglycemia and, in freely moving mice, their activation by optogenetics or Slc2a2 inactivation increases motivated sucrose-seeking but not saccharin-seeking behavior. These neurons may control sugar overconsumption in obesity and diabetes.


Subject(s)
Behavior, Animal/physiology , Glucose/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Thalamus/metabolism , Animals , Feeding Behavior , Hypoglycemia/metabolism , Mice, Transgenic , Motivation/physiology , Nucleus Accumbens/metabolism , Obesity/metabolism , Self Administration/methods , Sucrose/metabolism
12.
Trends Endocrinol Metab ; 26(9): 455-66, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26163755

ABSTRACT

Glucose homeostasis as well as homeostatic and hedonic control of feeding is regulated by hormonal, neuronal, and nutrient-related cues. Glucose, besides its role as a source of metabolic energy, is an important signal controlling hormone secretion and neuronal activity, hence contributing to whole-body metabolic integration in coordination with feeding control. Brain glucose sensing plays a key, but insufficiently explored, role in these metabolic and behavioral controls, which when deregulated may contribute to the development of obesity and diabetes. The recent introduction of innovative transgenic, pharmacogenetic, and optogenetic techniques allows unprecedented analysis of the complexity of central glucose sensing at the molecular, cellular, and neuronal circuit levels, which will lead to a new understanding of the pathogenesis of metabolic diseases.


Subject(s)
Brain Chemistry/physiology , Food Preferences/physiology , Glucose/metabolism , Homeostasis/physiology , Animals , Energy Metabolism , Humans , Neurons/physiology
13.
Cell Metab ; 19(3): 527-38, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24606905

ABSTRACT

Glucose-sensing neurons in the brainstem participate in the regulation of energy homeostasis but have been poorly characterized because of the lack of specific markers to identify them. Here we show that GLUT2-expressing neurons of the nucleus of the tractus solitarius form a distinct population of hypoglycemia-activated neurons. Their response to low glucose is mediated by reduced intracellular glucose metabolism, increased AMP-activated protein kinase activity, and closure of leak K(+) channels. These are GABAergic neurons that send projections to the vagal motor nucleus. Light-induced stimulation of channelrhodospin-expressing GLUT2 neurons in vivo led to increased parasympathetic nerve firing and glucagon secretion. Thus GLUT2 neurons of the nucleus tractus solitarius link hypoglycemia detection to counterregulatory response. These results may help identify the cause of hypoglycemia-associated autonomic failure, a major threat in the insulin treatment of diabetes.


Subject(s)
GABAergic Neurons/physiology , Glucagon/metabolism , Glucose Transporter Type 2/metabolism , Solitary Nucleus/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Channelrhodopsins , Deoxyglucose/pharmacology , GABAergic Neurons/drug effects , Glucosamine/pharmacology , Glucose/pharmacology , Hypoglycemia/metabolism , Hypoglycemia/pathology , In Vitro Techniques , Membrane Potentials/drug effects , Mice , Mice, Transgenic , Patch-Clamp Techniques , Potassium Channels/metabolism
14.
Nat Neurosci ; 16(3): 300-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23354329

ABSTRACT

The prevalence of obesity has markedly increased over the past few decades. Exploration of how hunger and satiety signals influence the reward system can help us understand non-homeostatic feeding. Insulin may act in the ventral tegmental area (VTA), a critical site for reward-seeking behavior, to suppress feeding. However, the neural mechanisms underlying insulin effects in the VTA remain unknown. We demonstrate that insulin, a circulating catabolic peptide that inhibits feeding, can induce long-term depression (LTD) of mouse excitatory synapses onto VTA dopamine neurons. This effect requires endocannabinoid-mediated presynaptic inhibition of glutamate release. Furthermore, after a sweetened high-fat meal, which elevates endogenous insulin, insulin-induced LTD is occluded. Finally, insulin in the VTA reduces food anticipatory behavior in mice and conditioned place preference for food in rats. Taken together, these results suggest that insulin in the VTA suppresses excitatory synaptic transmission and reduces anticipatory activity and preference for food-related cues.


Subject(s)
Dopaminergic Neurons/drug effects , Endocannabinoids/metabolism , Insulin/pharmacology , Long-Term Synaptic Depression/drug effects , Ventral Tegmental Area/drug effects , Animals , Association Learning/drug effects , Association Learning/physiology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Dietary Fats/pharmacology , Dopaminergic Neurons/cytology , Dopaminergic Neurons/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Glutamic Acid/metabolism , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , TOR Serine-Threonine Kinases/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology
15.
Neuron ; 73(6): 1173-83, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22445344

ABSTRACT

Salient but aversive stimuli inhibit the majority of dopamine (DA) neurons in the ventral tegmental area (VTA) and cause conditioned place aversion (CPA). The cellular mechanism underlying DA neuron inhibition has not been investigated and the causal link to behavior remains elusive. Here, we show that GABA neurons of the VTA inhibit DA neurons through neurotransmission at GABA(A) receptors. We also observe that GABA neurons increase their firing in response to a footshock and provide evidence that driving GABA neurons with optogenetic effectors is sufficient to affect behavior. Taken together, our data demonstrate that synaptic inhibition of DA neurons drives place aversion.


Subject(s)
Conditioning, Operant/physiology , Dopaminergic Neurons/physiology , Escape Reaction/physiology , GABAergic Neurons/physiology , Ventral Tegmental Area/cytology , Action Potentials/drug effects , Action Potentials/genetics , Analgesics, Opioid/pharmacology , Analysis of Variance , Animals , Apomorphine/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Channelrhodopsins , Conditioning, Operant/drug effects , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dopaminergic Neurons/drug effects , Electroshock/adverse effects , Escape Reaction/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/deficiency , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , GABAergic Neurons/drug effects , Glutamate Decarboxylase/genetics , Haloperidol/pharmacology , In Vitro Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphine/pharmacology , Optics and Photonics , Time Factors , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/drug effects
16.
Nature ; 463(7282): 769-74, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20148031

ABSTRACT

Benzodiazepines are widely used in clinics and for recreational purposes, but will lead to addiction in vulnerable individuals. Addictive drugs increase the levels of dopamine and also trigger long-lasting synaptic adaptations in the mesolimbic reward system that ultimately may induce the pathological behaviour. The neural basis for the addictive nature of benzodiazepines, however, remains elusive. Here we show that benzodiazepines increase firing of dopamine neurons of the ventral tegmental area through the positive modulation of GABA(A) (gamma-aminobutyric acid type A) receptors in nearby interneurons. Such disinhibition, which relies on alpha1-containing GABA(A) receptors expressed in these cells, triggers drug-evoked synaptic plasticity in excitatory afferents onto dopamine neurons and underlies drug reinforcement. Taken together, our data provide evidence that benzodiazepines share defining pharmacological features of addictive drugs through cell-type-specific expression of alpha1-containing GABA(A) receptors in the ventral tegmental area. The data also indicate that subunit-selective benzodiazepines sparing alpha1 may be devoid of addiction liability.


Subject(s)
Behavior, Addictive/chemically induced , Behavior, Addictive/physiopathology , Benzodiazepines/adverse effects , Benzodiazepines/pharmacology , Neurons/drug effects , Action Potentials/drug effects , Administration, Oral , Animals , Behavior, Addictive/pathology , Benzodiazepines/administration & dosage , Dopamine/metabolism , Electric Conductivity , Glutamic Acid/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Interneurons/drug effects , Interneurons/metabolism , Mice , Mice, Inbred C57BL , Midazolam/administration & dosage , Midazolam/adverse effects , Midazolam/pharmacology , Models, Biological , Morphine/pharmacology , Neuronal Plasticity/drug effects , Neurons/metabolism , Organ Specificity , Receptors, AMPA/metabolism , Receptors, GABA-A/deficiency , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Substrate Specificity , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , gamma-Aminobutyric Acid/metabolism
18.
PLoS One ; 5(12): e15870, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21209835

ABSTRACT

BACKGROUND: Addictive drugs have in common that they cause surges in dopamine (DA) concentration in the mesolimbic reward system and elicit synaptic plasticity in DA neurons of the ventral tegmental area (VTA). Cocaine for example drives insertion of GluA2-lacking AMPA receptors (AMPARs) at glutamatergic synapes in DA neurons. However it remains elusive which molecular target of cocaine drives such AMPAR redistribution and whether other addictive drugs (morphine and nicotine) cause similar changes through their effects on the mesolimbic DA system. METHODOLOGY/PRINCIPAL FINDINGS: We used in vitro electrophysiological techniques in wild-type and transgenic mice to observe the modulation of excitatory inputs onto DA neurons by addictive drugs. To observe AMPAR redistribution, post-embedding immunohistochemistry for GluA2 AMPAR subunit was combined with electron microscopy. We also used a double-floxed AAV virus expressing channelrhodopsin together with a DAT Cre mouse line to selectively express ChR2 in VTA DA neurons. We find that in mice where the effect of cocaine on the dopamine transporter (DAT) is specifically blocked, AMPAR redistribution was absent following administration of the drug. Furthermore, addictive drugs known to increase dopamine levels cause a similar AMPAR redistribution. Finally, activating DA VTA neurons optogenetically is sufficient to drive insertion of GluA2-lacking AMPARs, mimicking the changes observed after a single injection of morphine, nicotine or cocaine. CONCLUSIONS/SIGNIFICANCE: We propose the mesolimbic dopamine system as a point of convergence at which addictive drugs can alter neural circuits. We also show that direct activation of DA neurons is sufficient to drive AMPAR redistribution, which may be a mechanism associated with early steps of non-substance related addictions.


Subject(s)
Dopamine/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Animals , Cocaine/pharmacology , Dependovirus/metabolism , Electrophysiology/methods , Glutamine/metabolism , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Nicotine/pharmacology , Synapses , Ventral Tegmental Area/metabolism
19.
Nat Neurosci ; 10(12): 1559-68, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17965710

ABSTRACT

Agonists of GABA(B) receptors exert a bi-directional effect on the activity of dopamine (DA) neurons of the ventral tegmental area, which can be explained by the fact that coupling between GABA(B) receptors and G protein-gated inwardly rectifying potassium (GIRK) channels is significantly weaker in DA neurons than in GABA neurons. Thus, low concentrations of agonists preferentially inhibit GABA neurons and thereby disinhibit DA neurons. This disinhibition might confer reinforcing properties on addictive GABA(B) receptor agonists such as gamma-hydroxybutyrate (GHB) and its derivatives. Here we show that, in DA neurons of mice, the low coupling efficiency reflects the selective expression of heteromeric GIRK2/3 channels and is dynamically modulated by a member of the regulator of G protein signaling (RGS) protein family. Moreover, repetitive exposure to GHB increases the GABA(B) receptor-GIRK channel coupling efficiency through downregulation of RGS2. Finally, oral self-administration of GHB at a concentration that is normally rewarding becomes aversive after chronic exposure. On the basis of these results, we propose a mechanism that might underlie tolerance to GHB.


Subject(s)
Dopamine/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Neurons/physiology , RGS Proteins/metabolism , Receptors, GABA-A/physiology , Ventral Tegmental Area/cytology , Animals , Animals, Newborn , Baclofen/pharmacology , Barium Compounds/pharmacology , Behavior, Animal/drug effects , Chlorides/pharmacology , Dose-Response Relationship, Drug , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/ultrastructure , GABA Agonists/pharmacology , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Homeodomain Proteins/genetics , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Sodium Oxybate/pharmacology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL