Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Development ; 149(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35815619

ABSTRACT

The midbrain reticular formation (MRF) is a mosaic of diverse GABAergic and glutamatergic neurons that have been associated with a variety of functions, including sleep regulation. However, the molecular characteristics and development of MRF neurons are poorly understood. As the transcription factor, Gata2 is required for the development of all GABAergic neurons derived from the embryonic mouse midbrain, we hypothesized that the genes expressed downstream of Gata2 could contribute to the diversification of GABAergic neuron subtypes in this brain region. Here, we show that Gata2 is required for the expression of several GABAergic lineage-specific transcription factors, including Nkx2-2 and Skor2, which are co-expressed in a restricted group of post-mitotic GABAergic precursors in the MRF. Both Gata2 and Nkx2-2 function is required for Skor2 expression in GABAergic precursors. In the adult mouse and rat midbrain, Nkx2-2-and Skor2-expressing GABAergic neurons locate at the boundary of the ventrolateral periaqueductal gray and the MRF, an area containing REM-off neurons regulating REM sleep. In addition to the characteristic localization, Skor2+ cells increase their activity upon REM-sleep inhibition, send projections to the dorsolateral pons, a region associated with sleep control, and are responsive to orexins, consistent with the known properties of midbrain REM-off neurons.


Subject(s)
GABAergic Neurons , Sleep, REM , Animals , GABAergic Neurons/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Homeobox Protein Nkx-2.2/metabolism , Mesencephalon , Mice , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Rats , Sleep/physiology , Sleep, REM/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Transl Psychiatry ; 10(1): 357, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087695

ABSTRACT

The neural circuits regulating motivation and movement include midbrain dopaminergic neurons and associated inhibitory GABAergic and excitatory glutamatergic neurons in the anterior brainstem. Differentiation of specific subtypes of GABAergic and glutamatergic neurons in the mouse embryonic brainstem is controlled by a transcription factor Tal1. This study characterizes the behavioral and neurochemical changes caused by the absence of Tal1 function. The Tal1cko mutant mice are hyperactive, impulsive, hypersensitive to reward, have learning deficits and a habituation defect in a novel environment. Only minor changes in their dopaminergic system were detected. Amphetamine induced striatal dopamine release and amphetamine induced place preference were normal in Tal1cko mice. Increased dopamine signaling failed to stimulate the locomotor activity of the Tal1cko mice, but instead alleviated their hyperactivity. Altogether, the Tal1cko mice recapitulate many features of the attention and hyperactivity disorders, suggesting a role for Tal1 regulated developmental pathways and neural structures in the control of motivation and movement.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Brain Stem/cytology , Dopaminergic Neurons , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Amphetamine/pharmacology , Animals , Attention Deficit Disorder with Hyperactivity/genetics , Mesencephalon , Mice
3.
Pharmacol Biochem Behav ; 190: 172852, 2020 03.
Article in English | MEDLINE | ID: mdl-31952939

ABSTRACT

Acute lipopolysaccharide (LPS) administration induces innate inflammatory signalling and produces sickness reaction characterized by reduced drinking, eating and reduced locomotor exploration, as well as emotional changes indicating increased helplessness/despair. LPS administration has been used to model behavioral and emotional responses to inflammatory reactions. Our aim was to find out whether the lack of metabotropic glutamate receptor 3 (mGluR3) in the knockout (KO) mice affects behavioral effects of LPS in vivo, as mGluR3 may have a role in inflammatory signalling. After LPS (1 mg/kg, i.p.) administration, we compared wild-type (WT) and mGluR3-KO mice for differences in gross appearance and locomotion at 3- and 6-h time points, anxiety-like behavior in the light-dark test at 24-h, depression-like behavior in the tail-suspension test at 25-h, and in the forced-swim test at 48-h time points. Body weight and water consumption were monitored. Based on behavioral scorings at the 3-h and 6-h time points, the mGluR3-KO mice reacted to LPS in a similar way as the WT mice. LPS-induced reductions in the body weight or water consumption did not differ between genotypes. Interestingly, LPS-induced reductions in the body temperature were significantly enhanced in male and female mGluR3-KO mice at 6-h and 3-h time points, respectively. In the light-dark anxiety-test the saline-treated mGluR3-KOs showed increased anxiolytic-like behaviors compared to the saline-treated WT mice. LPS treatment significantly reduced the KO entries to the light compartment to the same level as WT mice given saline. Total locomotion was significantly reduced in both genotypes by LPS. In the despair models, no genotype difference was observed after saline or LPS, neither had LPS treatment any significant effect on immobility. Although changes in glutamatergic neurotransmission may partly mediate effects of systemic LPS administration, mGluR3 appears not to be crucial in behavioral responses to acute activation of innate immune system.


Subject(s)
Behavior, Animal/drug effects , Gene Knockout Techniques , Lipopolysaccharides/pharmacology , Receptors, Metabotropic Glutamate/genetics , Animals , Anxiety/chemically induced , Body Temperature/drug effects , Body Weight/drug effects , Depression/chemically induced , Drinking/drug effects , Genotype , Inflammation/chemically induced , Locomotion/drug effects , Locomotion/genetics , Male , Mice , Mice, Knockout
4.
Front Mol Neurosci ; 12: 130, 2019.
Article in English | MEDLINE | ID: mdl-31178693

ABSTRACT

THIP (gaboxadol), a superagonist of the δ subunit-containing extrasynaptic GABAA receptors, produces persistent neuroplasticity in dopamine (DA) neurons of the ventral tegmental area (VTA), similarly to rewarding drugs of abuse. However, unlike them THIP lacks abuse potential and induces conditioned place aversion in mice. The mechanism underlying the aversive effects of THIP remains elusive. Here, we show that mild aversive effects of THIP were detected 2 h after administration likely reflecting an anxiety-like state with increased corticosterone release and with central recruitment of corticotropin-releasing factor corticotropin-releasing factor receptor 1 (CRF1) receptors. A detailed immunohistochemical c-Fos expression mapping for THIP-activated brain areas revealed a correlation between the activation of CRF-expressing neurons in the oval nucleus of the bed nuclei of stria terminalis and THIP-induced aversive effects. In addition, the neuroplasticity of mesolimbic DA system (24 h after administration) and conditioned place aversion by THIP after four daily acute sessions were dependent on extrasynaptic GABAA receptors (abolished in δ-GABAA receptor knockout mice) and activation of the CRF1 receptors (abolished in wildtype mice by a CRF1 receptor antagonist). A selective THIP-induced activation of CRF-expressing neurons in the oval part of the bed nucleus of stria terminalis may constitute a novel mechanism for inducing plasticity in a population of VTA DA neurons and aversive behavioral states.

5.
J Psychopharmacol ; 33(7): 855-864, 2019 07.
Article in English | MEDLINE | ID: mdl-31070489

ABSTRACT

BACKGROUND: Alcohol use associates with environmental cues that can later reinstate drinking patterns without any alcohol exposure. Alcohol-induced reward, when combined with contextual signals of various sensory modalities in the central synapses of mesolimbic reward circuitries, can lead to the formation of conditioned responses. AIMS: As the activation of glutamatergic synapses is pivotal in such processes, we aimed to investigate whether the metabotropic glutamate receptor subtype 3 plays a role in alcohol-induced behaviours including place preference. METHODS: The metabotropic glutamate receptor subtype 3 knockout (mGluR3-KO) mouse line was used to study alcohol-induced place preference, locomotor activating and ataxic effects, limited access alcohol drinking, and preference for sucrose and saccharin. RESULTS: Alcohol-induced horizontal locomotor stimulation and reduced rearing behaviour remained unchanged in the mGluR3-KO mice. However, alcohol-induced place conditioning in an unbiased paradigm setup was lacking in the mGluR3-KO mice, but clearly present in wildtype mice. Locomotor activity was not different between the mGluR3-KO and wildtype mice during the acquisition and expression trials. Alcohol consumption, studied through the 'drinking in the dark' model, remained unchanged in the mGluR3-KO mice, although low consumption in both wildtype and knockout mice hampers interpretation. The mGluR3-KO mice also showed normal sucrose and saccharin preference. CONCLUSIONS: These studies indicate a role for metabotropic glutamate receptor subtype 3 in the conditioned contextual alcohol responses, but not in stimulatory, and ataxic alcohol effects.


Subject(s)
Alcohol Drinking/psychology , Conditioning, Psychological/drug effects , Ethanol/pharmacology , Receptors, Metabotropic Glutamate/genetics , Animals , Cues , Ethanol/administration & dosage , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reward
6.
Front Pharmacol ; 10: 309, 2019.
Article in English | MEDLINE | ID: mdl-30984001

ABSTRACT

Gene-targeted mice with deficient AMPA receptor GluA1 subunits (Gria1-/- mice) show robust hyperlocomotion in a novel environment, suggesting them to constitute a model for hyperactivity disorders such as mania, schizophrenia and attention deficit hyperactivity disorder. This behavioral alteration has been associated with increased neuronal activation in the hippocampus, and it can be attenuated by chronic treatment with antimanic drugs, such as lithium, valproic acid, and lamotrigine. Now we found that systemic cannabidiol strongly blunted the hyperactivity and the hippocampal c-Fos expression of the Gria1-/- mice, while not affecting the wild-type littermate controls. Acute bilateral intra-dorsal hippocampal infusion of cannabidiol partially blocked the hyperactivity of the Gria1-/- mice, but had no effect on wild-types. The activation of the inhibitory DREADD receptor hM4Gi in the dorsal hippocampus by clozapine-N-oxide robustly inhibited the hyperactivity of the Gria1-/- mice, but had no effect on the locomotion of wild-type mice. Our results show that enhanced neuronal excitability in the hippocampus is associated with pronounced novelty-induced hyperactivity of GluA1 subunit-deficient mice. When this enhanced response of hippocampal neurons to novel stimuli is specifically reduced in the hippocampus by pharmacological treatment or by chemogenetic inhibition, Gria1-/- mice recover from behavioral hyperactivity, suggesting a hippocampal dysfunction in hyperactive behaviors that can be treated with cannabidiol.

7.
Sci Rep ; 9(1): 3311, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824794

ABSTRACT

Animal studies remain an essential part of drug discovery since in vitro models are not capable of describing the complete living organism. We developed and qualified a microchip electrophoresis-electrochemical detection (MCE-EC) method for rapid analysis of morphine in mouse plasma using a commercial MCE-EC device. Following liquid-liquid extraction (LLE), we achieved within-run precision of 3.7 and 4.5% (coefficient of variation, CV, n = 6) and accuracy of 106.9% and 100.7% at biologically relevant morphine concentrations of 5 and 20 µM in plasma, respectively. The same method was further challenged by morphine detection in mouse brain homogenates with equally good within-run precision (7.8% CV, n = 5) at 1 µM concentration. The qualified method was applied to analyze a set of plasma and brain homogenate samples derived from a behavioral animal study. After intraperitoneal administration of 20 mg/kg morphine hydrochloride, the detected morphine concentrations in plasma were between 6.7 and 17 µM. As expected, the morphine concentrations in the brain were significantly lower, ca. 80-125 nM (280-410 pg morphine/mg dissected brain), and could only be detected after preconcentration achieved during LLE. In all, the microchip-based separation system is proven feasible for rapid analysis of morphine to provide supplementary chemical information to behavioral animal studies.


Subject(s)
Brain/metabolism , Electrophoresis, Microchip , Morphine , Plasma/metabolism , Animals , Injections, Intraperitoneal , Mice , Morphine/pharmacokinetics , Morphine/pharmacology
8.
Pharmacol Res ; 141: 384-391, 2019 03.
Article in English | MEDLINE | ID: mdl-30648615

ABSTRACT

The dopamine D3 receptor (D3R), in the nucleus accumbens (NAc), plays an important role in alcohol reward mechanisms. The major neuronal type within the NAc is the GABAergic medium spiny neuron (MSN), whose activity is regulated by dopaminergic inputs. We previously reported that genetic deletion or pharmacological blockade of D3R increases GABAA α6 subunit in the ventral striatum. Here we tested the hypothesis that D3R-dependent changes in GABAA α6 subunit in the NAc affect voluntary alcohol intake, by influencing the inhibitory transmission of MSNs. We performed in vivo and ex vivo experiments in D3R knockout (D3R -/-) mice and wild type littermates (D3R +/+). Ro 15-4513, a high affinity α6-GABAA ligand was used to study α6 activity. At baseline, NAc α6 expression was negligible in D3R+/+, whereas it was robust in D3R-/-; other relevant GABAA subunits were not changed. In situ hybridization and qPCR confirmed α6 subunit mRNA expression especially in the NAc. In the drinking-in-the-dark paradigm, systemic administration of Ro 15-4513 inhibited alcohol intake in D3R+/+, but increased it in D3R-/-; this was confirmed by intra-NAc administration of Ro 15-4513 and furosemide, a selective α6-GABAA antagonist. Whole-cell patch-clamp showed peak amplitudes of miniature inhibitory postsynaptic currents in NAc medium spiny neurons higher in D3R-/- compared to D3R+/+; Ro 15-4513 reduced the peak amplitude in the NAc of D3R-/-, but not in D3R+/+. We conclude that D3R-dependent enhanced expression of α6 GABAA subunit inhibits voluntary alcohol intake by increasing GABA inhibition in the NAc.


Subject(s)
Binge Drinking/genetics , GABAergic Neurons/pathology , Receptors, Dopamine D3/genetics , Receptors, GABA-A/genetics , Animals , Binge Drinking/pathology , GABAergic Neurons/metabolism , Gene Expression Regulation , Male , Mice , Mice, Knockout , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Protein Subunits/genetics , RNA, Messenger/genetics
9.
Behav Pharmacol ; 30(5): 405-411, 2019 08.
Article in English | MEDLINE | ID: mdl-30376459

ABSTRACT

Extinction and reinstatement of morphine-induced conditioned place preference were studied in glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-receptor GluA1 subunit-deficient mice (global GluA1-KO mice). In line with previous findings, both acquisition and expression of conditioned place preference to morphine (20 mg/kg, subcutaneously) were fully functional in GluA1 KO mice compared with wild-type littermate controls (GluA1-WT), thus enabling the study of extinction. With a 10-session extinction paradigm, the GluA1 KO mice showed complete extinction similar to that of the GluA1-WT mice. Morphine-induced reinstatement (10 mg/kg, subcutaneously) was detected in both mouse lines. GluA1 KO mice moved more during all the phases of the experiment, including the place conditioning trials, extinction sessions, and place preference tests. The results suggest that the GluA1 subunit may be dispensable or prone to compensation at the neural circuitries delineating extinction and reinstatement. The GluA1 KO mice show altered long-term between-session habituation, which extends longer than previously anticipated.


Subject(s)
Conditioning, Classical/drug effects , Extinction, Psychological/drug effects , Morphine/pharmacology , Animals , Conditioning, Psychological/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Narcotics/pharmacology , Receptors, AMPA/genetics
10.
Curr Biol ; 28(16): 2557-2569.e4, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30100338

ABSTRACT

The central cholinergic system and the amygdala are important for motivation and mnemonic processes. Different cholinergic populations innervate the amygdala, but it is unclear how these projections impact amygdala processes. Using optogenetic circuit-mapping strategies in choline acetyltransferase (ChAT)-cre mice, we demonstrate that amygdala-projecting basal forebrain and brainstem ChAT-containing neurons can differentially affect amygdala circuits and behavior. Photo-activating ChAT terminals in vitro revealed the underlying synaptic impact of brainstem inputs to the central lateral division to be excitatory, mediated via the synergistic glutamatergic activation of AMPA and NMDA receptors. In contrast, stimulating basal forebrain inputs to the basal nucleus resulted in endogenous acetylcholine (ACh) release, resulting in biphasic inhibition-excitation responses onto principal neurons. Such response profiles are physiological hallmarks of neural oscillations and could thus form the basis of ACh-mediated rhythmicity in amygdala networks. Consistent with this, in vivo basal forebrain ChAT+ activation strengthened amygdala basal nucleus theta and gamma frequency rhythmicity, both of which continued for seconds after stimulation and were dependent on local muscarinic and nicotinic receptor activation, respectively. Activation of brainstem ChAT-containing neurons, however, resulted in a transient increase in central lateral amygdala activity that was independent of cholinergic receptors. In addition, driving these respective inputs in behaving animals induced opposing appetitive and defensive learning-related behavioral changes. Because learning and memory are supported by both cellular and network-level processes in central cholinergic and amygdala networks, these results provide a route by which distinct cholinergic inputs can convey salient information to the amygdala and promote associative biophysical changes that underlie emotional memories.


Subject(s)
Amygdala/physiology , Basal Forebrain/physiology , Brain Stem/physiology , Cholinergic Neurons/physiology , Learning/physiology , Memory/physiology , Animals , Choline O-Acetyltransferase/metabolism , Male , Mice , Mice, Transgenic , Optogenetics
11.
Basic Clin Pharmacol Toxicol ; 123(5): 558-566, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29781560

ABSTRACT

Extrasynaptic δ subunit-containing γ-aminobutyric acid type A receptors (δ-GABAA Rs) are emerging as targets for a number of neuropsychopharmacological drugs, including the direct GABA site agonist gaboxadol and neuroactive steroids. Among other regions, these δ-GABAA Rs are functionally expressed in the ventral tegmental area (VTA), the cell body region of mesocorticolimbic dopamine (DA) system important for motivated behaviours, and in the target region, the nucleus accumbens. Gaboxadol and neurosteroids induce VTA DA neuron plasticity ex vivo, by inhibiting the VTA GABA neurons, and aversive place conditioning, which are absent in the δ-GABAA R knockout mice (δ-KO). It is not known whether δ-GABAA Rs are important for the effects of other drugs, such as opioids (that also inhibit GABA neurons) and stimulants (that primarily elevate monoamine levels). Here, we used δ-KO mice and conditioned place preference (CPP) test to study the rewarding effects of morphine (20 mg/kg), methamphetamine (1 mg/kg) and mephedrone (5 mg/kg). Morphine-induced nociception was also assessed using tail-flick and hot-plate tests. We found that the δ-KO mice failed to express morphine-induced CPP, but that they were more sensitive to morphine-induced analgesia in the tail-flick test. In contrast, stimulant-induced CPP in the δ-KO mice was similar to that in the wild-type controls. Thus, the conditioned rewarding effect by opioids, but not that of stimulants, was impaired in the absence of δ-GABAA Rs. Further studies are warranted to assess the potential of δ-GABAA R antagonists as possible targets for reducing morphine reward and potentiating morphine analgesia.


Subject(s)
Conditioning, Psychological/drug effects , Methamphetamine/analogs & derivatives , Methamphetamine/pharmacology , Morphine/pharmacology , Motivation , Receptors, GABA-A , Analgesics, Opioid/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , GABA-A Receptor Antagonists/pharmacology , Mice , Mice, Knockout , Motivation/drug effects , Motivation/physiology , Neuronal Plasticity/drug effects , Nociception/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Reward , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
12.
eNeuro ; 4(2)2017.
Article in English | MEDLINE | ID: mdl-28497110

ABSTRACT

Satiety, rather than all or none, can instead be viewed as a cumulative decrease in the drive to eat that develops over the course of a meal. The nucleus accumbens (NAc) is known to play a critical role in this type of value reappraisal, but the underlying circuits that influence such processes are unclear. Although NAc cholinergic interneurons (CINs) comprise only a small proportion of NAc neurons, their local impact on reward-based processes provides a candidate cell population for investigating the neural underpinnings of satiety. The present research therefore aimed to determine the role of NAc-CINs in motivation for food reinforcers in relation to satiety signaling. Through bidirectional control of CIN activity in mice, we show that when motivated by food restriction, increasing CIN activity led to a reduction in palatable food consumption while reducing CIN excitability enhanced food intake. These activity-dependent changes developed only late in the session and were unlikely to be driven by the innate reinforcer strength, suggesting that CIN modulation was instead impacting the cumulative change in motivation underlying satiety signaling. We propose that on a circuit level, an overall increase in inhibitory tone onto NAc output neurons played a role in the behavioral results, as activating NAc-CINs led to an inhibition of medium spiny neurons that was dependent on nicotinic receptor activation. Our results reveal an important role for NAc-CINs in controlling motivation for food intake and additionally provide a circuit-level framework for investigating the endogenous cholinergic circuits that signal satiety.


Subject(s)
Cholinergic Neurons/physiology , Interneurons/physiology , Motivation/physiology , Reward , Animals , Cholinergic Agents/pharmacology , Cholinergic Neurons/drug effects , Eating/physiology , Interneurons/drug effects , Mice, Transgenic , Nucleus Accumbens/physiology
13.
Neurobiol Learn Mem ; 136: 183-188, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27746379

ABSTRACT

The hypothalamic hypocretin/orexin (HO) system holds a central role in the regulation of several physiological functions critical for food-seeking behavior including mnemonic processes for effective foraging behavior. It is unclear however whether physiological increases in HO neuronal activity can support such processes. Using a designer rM3Ds receptor activation approach increasing HO neuronal activity resulted in improved short-term memory for novel locations. When tested on a non-spatial novelty object recognition task no significant difference was detected between groups indicating that hypothalamic HO neuronal activation can selectively facilitate short-term spatial memory for potentially supporting memory for locations during active exploration.


Subject(s)
Hypothalamus/physiology , Memory, Short-Term/physiology , Neurons/physiology , Orexins/physiology , Recognition, Psychology/physiology , Spatial Memory/physiology , Animals , Behavior, Animal/physiology , Female , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Orexins/metabolism
14.
PLoS One ; 10(8): e0133327, 2015.
Article in English | MEDLINE | ID: mdl-26247203

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) play an important role in regulating appetite and have been shown to do so by influencing neural activity in the hypothalamus. To shed light on the hypothalamic circuits governing acetylcholine's (ACh) regulation of appetite this study investigated the influence of hypothalamic nAChRs expressing the α4 subunit. We found that antagonizing the α4ß2 nAChR locally in the lateral hypothalamus with di-hydro-ß-erythroidine (DHßE), an α4 nAChR antagonist with moderate affinity, caused an increase in food intake following free access to food after a 12 hour fast, compared to saline-infused animals. Immunocytochemical analysis revealed that orexin/hypocretin (HO), oxytocin, and tyrosine hydroxylase (TH)-containing neurons in the A13 and A12 of the hypothalamus expressed the nAChR α4 subunit in varying amounts (34%, 42%, 50%, and 51%, respectively) whereas melanin concentrating hormone (MCH) neurons did not, suggesting that DHßE-mediated increases in food intake may be due to a direct activation of specific hypothalamic circuits. Systemic DHßE (2 mg/kg) administration similarly increased food intake following a 12 hour fast. In these animals a subpopulation of orexin/hypocretin neurons showed elevated activity compared to control animals and MCH neuronal activity was overall lower as measured by expression of the immediate early gene marker for neuronal activity cFos. However, oxytocin neurons in the paraventricular hypothalamus and TH-containing neurons in the A13 and A12 did not show differential activity patterns. These results indicate that various neurochemically distinct hypothalamic populations are under the influence of α4ß2 nAChRs and that cholinergic inputs to the lateral hypothalamus can affect satiety signals through activation of local α4ß2 nAChR-mediated transmission.


Subject(s)
Eating/physiology , Hypothalamus/metabolism , Motor Activity/physiology , Neurons/metabolism , Receptors, Nicotinic/metabolism , Animals , Dihydro-beta-Erythroidine/pharmacology , Eating/drug effects , Hypothalamus/drug effects , Male , Motor Activity/drug effects , Neural Pathways/drug effects , Neural Pathways/metabolism , Neurons/drug effects , Nicotinic Antagonists/pharmacology , Orexins/metabolism , Oxytocin/metabolism , Rats , Rats, Wistar , Tyrosine 3-Monooxygenase/metabolism
15.
J Neurosci ; 35(14): 5435-41, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25855162

ABSTRACT

The lateral hypothalamus (LH) is a key regulator of multiple vital behaviors. The firing of brain-wide-projecting LH neurons releases neuropeptides promoting wakefulness (orexin/hypocretin; OH), or sleep (melanin-concentrating hormone; MCH). OH neurons, which coexpress glutamate and dynorphin, have been proposed to excite their neighbors, including MCH neurons, suggesting that LH may sometimes coengage its antagonistic outputs. However, it remains unclear if, when, and how OH actions promote temporal separation of the sleep and wake signals, a process that fails in narcolepsy caused by OH loss. To explore this directly, we paired optogenetic stimulation of OH cells (at rates that promoted awakening in vivo) with electrical monitoring of MCH cells in mouse brain slices. Membrane potential recordings showed that OH cell firing inhibited action potential firing in most MCH neurons, an effect that required GABAA but not dynorphin receptors. Membrane current analysis showed that OH cell firing increased the frequency of fast GABAergic currents in MCH cells, an effect blocked by antagonists of OH but not dynorphin or glutamate receptors, and mimicked by bath-applied OH peptide. In turn, neural network imaging with a calcium indicator genetically targeted to MCH neurons showed that excitation by bath-applied OH peptides occurs in a minority of MCH cells. Collectively, our data provide functional microcircuit evidence that intra-LH feedforward loops may facilitate appropriate switching between sleep and wake signals, potentially preventing sleep disorders.


Subject(s)
Hypothalamic Hormones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Melanins/metabolism , Nerve Net/physiology , Neural Inhibition/physiology , Neurons/physiology , Neuropeptides/metabolism , Optogenetics , Pituitary Hormones/metabolism , Signal Transduction/physiology , Animals , Calcium/metabolism , Channelrhodopsins , Excitatory Amino Acid Antagonists/pharmacology , Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/genetics , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Melanins/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Neural Inhibition/drug effects , Neuropeptides/genetics , Neuropeptides/pharmacology , Orexins , Patch-Clamp Techniques , Pituitary Hormones/genetics , Transduction, Genetic , gamma-Aminobutyric Acid/metabolism , Red Fluorescent Protein
16.
Front Pharmacol ; 5: 256, 2014.
Article in English | MEDLINE | ID: mdl-25505414

ABSTRACT

GABAA receptors are the main fast inhibitory neurotransmitter receptors in the mammalian brain, and targets for many clinically important drugs widely used in the treatment of anxiety disorders, insomnia and in anesthesia. Nonetheless, there are significant risks associated with the long-term use of these drugs particularly related to development of tolerance and addiction. Addictive mechanisms of GABAA receptor drugs are poorly known, but recent findings suggest that those drugs may induce aberrant neuroadaptations in the brain reward circuitry. Recently, benzodiazepines, acting on synaptic GABAA receptors, and modulators of extrasynaptic GABAA receptors (THIP and neurosteroids) have been found to induce plasticity in the ventral tegmental area (VTA) dopamine neurons and their main target projections. Furthermore, depending whether synaptic or extrasynaptic GABAA receptor populations are activated, the behavioral outcome of repeated administration seems to correlate with rewarding or aversive behavioral responses, respectively. The VTA dopamine neurons project to forebrain centers such as the nucleus accumbens and medial prefrontal cortex, and receive afferent projections from these brain regions and especially from the extended amygdala and lateral habenula, forming the major part of the reward and aversion circuitry. Both synaptic and extrasynaptic GABAA drugs inhibit the VTA GABAergic interneurons, thus activating the VTA DA neurons by disinhibition and this way inducing glutamatergic synaptic plasticity. However, the GABAA drugs failed to alter synaptic spine numbers as studied from Golgi-Cox-stained VTA dendrites. Since the GABAergic drugs are known to depress the brain metabolism and gene expression, their likely way of inducing neuroplasticity in mature neurons is by disinhibiting the principal neurons, which remains to be rigorously tested for a number of clinically important anxiolytics, sedatives and anesthetics in different parts of the circuitry.

17.
Eur J Pharmacol ; 745: 36-45, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25446922

ABSTRACT

Malfunction of glutamate transmission is implicated in several neuropsychiatric disorders. Gria1-/- mouse line with knocked-out GluA1 subunits of ionotropic AMPA glutamate receptor displays several behavioural features of schizoaffective disorder. Typically, these mice show hyperactivity provoked by environmental novelty, which is attenuated after 4-week treatment with the standard mood-stabilisers lithium and valproate and the mood-stabilising anticonvulsants topiramate and lamotrigine (Maksimovic, M., Vekovischeva, O.Y., Aitta-Aho, T., Korpi, E.R., 2014. Chronic treatment with mood-stabilizers attenuates abnormal hyperlocomotion of GluA1-subunit deficient mice. PloS One. 9, e100188). Here, we complement our study by treating these mice chronically with perampanel, a novel non-competitive antagonist of AMPA receptors, for 4 weeks at the dose of 60 mg/kg diet, and found reduced locomotor hyperactivity in the Gria1-/- animals, while not affecting the wild-type littermates. To study the cellular mechanism by which chronic treatments with glutamate-modulating mood-stabilizing drugs alleviate this hyperactivity, we used the immediate early gene c-Fos protein expression as a marker of neuronal activity in the brain. Chronic lithium, valproate and topiramate blunted the c-Fos expression especially in the dorsal hippocampus of the Gria1-/- mice, with all of them reducing the number of c-Fos-positive cells in the CA3 region and valproate and topiramate also in the dentate gyrus (DG). Lamotrigine and perampanel treatments had the same effect in the all CA1, CA3 and DG subfields of the dorsal hippocampus of Gria1-/- mice. The results suggest that abnormal (hippocampal) glutamatergic transmission underlies the hyperactive phenotype of the Gria1-/- mice in a novel environment, and based on the efficacies of the present chronic drug treatments, this mouse model may serve as a predictive tool for studying novel mood-stabilisers.


Subject(s)
Exploratory Behavior/physiology , Hippocampus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, AMPA/deficiency , Animals , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , Exploratory Behavior/drug effects , Female , Fructose/administration & dosage , Fructose/analogs & derivatives , Glutamic Acid/metabolism , Hippocampus/drug effects , Lamotrigine , Lithium Compounds/administration & dosage , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Neuroprotective Agents/administration & dosage , Nitriles , Pyridones/administration & dosage , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synaptic Transmission , Topiramate , Triazines/administration & dosage , Valproic Acid/administration & dosage
18.
PLoS One ; 9(6): e100188, 2014.
Article in English | MEDLINE | ID: mdl-24932798

ABSTRACT

Abnormal excitatory glutamate neurotransmission and plasticity have been implicated in schizophrenia and affective disorders. Gria1-/- mice lacking GluA1 subunit (encoded by Gria1 gene) of AMPA-type glutamate receptor show robust novelty-induced hyperactivity, social deficits and heightened approach features, suggesting that they could be used to test for anti-manic activity of drugs. Here, we tested the efficacy of chronic treatment with established anti-manic drugs on behavioural properties of the Gria1-/- mice. The mice received standard mood stabilizers (lithium and valproate) and novel ones (topiramate and lamotrigine, used more as anticonvulsants) as supplements in rodent chow for at least 4 weeks. All drugs attenuated novelty-induced locomotor hyperactivity of the Gria1-/- mice, especially by promoting the habituation, while none of them attenuated 2-mg/kg amphetamine-induced hyperactivity as compared to control diet. Treatment with lithium and valproate reversed the elevated exploratory activity of Gria1-/- mice. Valproate treatment also reduced struggling behaviour in tail suspension test and restored reciprocally-initiated social contacts of Gria1-/- mice to the level shown by the wild-type Gria1+/+ mice. Gria1-/- mice consumed slightly more sucrose during intermittent sucrose exposure than the wild-types, but ran similar distances on running wheels. These behaviours were not consistently affected by lithium and valproate in the Gria1-/- mice. The efficacy of various anti-manic drug treatments on novelty-induced hyperactivity suggests that the Gria1-/- mouse line can be utilized in screening for new therapeutics.


Subject(s)
Anticonvulsants/pharmacology , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Hyperkinesis/prevention & control , Motor Activity/drug effects , Receptors, AMPA/physiology , Animals , Female , Lithium/pharmacology , Male , Maze Learning/drug effects , Mice , Mice, Knockout , Valproic Acid/pharmacology
19.
Neuropsychopharmacology ; 39(3): 727-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24077066

ABSTRACT

The main fast-acting inhibitory receptors in the mammalian brain are γ-aminobutyric acid type-A (GABAA) receptors for which neurosteroids, a subclass of steroids synthesized de novo in the brain, constitute a group of endogenous ligands with the most potent positive modulatory actions known. Neurosteroids can act on all subtypes of GABAA receptors, with a preference for δ-subunit-containing receptors that mediate extrasynaptic tonic inhibition. Pathological conditions characterized by emotional and motivational disturbances are often associated with perturbation in the levels of endogenous neurosteroids. We studied the effects of ganaxolone (GAN)-a synthetic analog of endogenous allopregnanolone that lacks activity on nuclear steroid receptors-on the mesolimbic dopamine (DA) system involved in emotions and motivation. A single dose of GAN in young mice induced a dose-dependent, long-lasting neuroplasticity of glutamate synapses of DA neurons ex vivo in the ventral tegmental area (VTA). Increased α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)/N-methyl-D-aspartate ratio and rectification of AMPA receptor responses even at 6 days after GAN administration suggested persistent synaptic targeting of GluA2-lacking AMPA receptors. This glutamate neuroplasticity was not observed in GABAA receptor δ-subunit-knockout (δ-KO) mice. GAN (500 nM) applied locally to VTA selectively increased tonic inhibition of GABA interneurons and triggered potentiation of DA neurons within 4 h in vitro. Place-conditioning experiments in adult wild-type C57BL/6J and δ-KO mice revealed aversive properties of repeated GAN administration that were dependent on the δ-subunits. Prolonged neuroadaptation to neurosteroids in the VTA might contribute to both the physiology and pathophysiology underlying processes and changes in motivation, mood, cognition, and drug addiction.


Subject(s)
Dopaminergic Neurons/drug effects , Neuronal Plasticity/drug effects , Neurotransmitter Agents/pharmacology , Receptors, GABA-A/metabolism , Ventral Tegmental Area/cytology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Dopamine/metabolism , Excitatory Amino Acid Agents/pharmacology , Female , GABA Agents/pharmacology , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Picrotoxin/pharmacology , Pregnanolone/analogs & derivatives , Pregnanolone/pharmacology , Receptors, GABA-A/genetics , gamma-Aminobutyric Acid/pharmacology
20.
PLoS One ; 7(5): e38325, 2012.
Article in English | MEDLINE | ID: mdl-22675452

ABSTRACT

In state-dependency, information retrieval is most efficient when the animal is in the same state as it was during the information acquisition. State-dependency has been implicated in a variety of learning and memory processes, but its mechanisms remain to be resolved. Here, mice deficient in AMPA-type glutamate receptor GluA1 subunits were first conditioned to morphine (10 or 20 mg/kg s.c. during eight sessions over four days) using an unbiased procedure, followed by testing for conditioned place preference at morphine states that were the same as or different from the one the mice were conditioned to. In GluA1 wildtype littermate mice the same-state morphine dose produced the greatest expression of place preference, while in the knockout mice no place preference was then detected. Both wildtype and knockout mice expressed moderate morphine-induced place preference when not at the morphine state (saline treatment at the test); in this case, place preference was weaker than that in the same-state test in wildtype mice. No correlation between place preference scores and locomotor activity during testing was found. Additionally, as compared to the controls, the knockout mice showed unchanged sensitization to morphine, morphine drug discrimination and brain regional µ-opioid receptor signal transduction at the G-protein level. However, the knockout mice failed to show increased AMPA/NMDA receptor current ratios in the ventral tegmental area dopamine neurons of midbrain slices after a single injection of morphine (10 mg/kg, s.c., sliced prepared 24 h afterwards), in contrast to the wildtype mice. The results indicate impaired drug-induced state-dependency in GluA1 knockout mice, correlating with impaired opioid-induced glutamate receptor neuroplasticity.


Subject(s)
Morphine Dependence/genetics , Receptors, AMPA/genetics , Analgesics, Opioid/pharmacology , Animals , Dopaminergic Neurons/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Morphine Dependence/metabolism , Motor Activity/drug effects , Motor Activity/genetics , Narcotics/pharmacology , Receptors, AMPA/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...