Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Eur J Neurosci ; 55(4): 939-951, 2022 02.
Article in English | MEDLINE | ID: mdl-33253450

ABSTRACT

Several studies support the notion that exploratory behaviour depends on the functionality of the cannabinoid type 1 (CB1) receptor in a cell type-specific manner. Mice lacking the CB1 receptor in forebrain GABAergic or dorsal telencephalic glutamatergic neurons have served as essential tools revealing the necessary CB1 receptor functions in these two neuronal populations. However, whether these specific CB1 receptor populations are also sufficient within the endocannabinoid system for wild-type-like exploratory behaviour has remained unknown. To evaluate cell-type-specific sufficiency of CB1 receptor signalling exclusively in dorsal telencephalic glutamatergic neurons (Glu-CB1-RS) or in forebrain GABAergic neurons (GABA-CB1-RS), we utilised a mouse model in which CB1 receptor expression can be reactivated conditionally at endogenous levels from a complete CB1-KO background. The two types of conditional CB1-rescue mice were compared with CB1 receptor-deficient [no reactivation (Stop-CB1)] and wild-type [ubiquitous reactivation of endogenous CB1 receptor (CB1-RS)] controls to investigate the behavioural consequences. We evaluated social and object exploratory behaviour in four different paradigms. Remarkably, the reduced exploration observed in Stop-CB1 animals was rescued in Glu-CB1-RS mice and sometimes even surpassed CB1-RS (wild-type) exploration. In contrast, GABA-CB1-RS animals showed the lowest exploratory drive in all paradigms, with an even stronger phenotype than Stop-CB1 mice. Interestingly, these effects weakened with increasing familiarity with the environment, suggesting a causal role for altered neophobia in the observed phenotypes. Taken together, using our genetic approach, we were able to substantiate the opposing role of the CB1 receptor in dorsal telencephalic glutamatergic versus forebrain GABAergic neurons regarding exploratory behaviour.


Subject(s)
Exploratory Behavior , Receptor, Cannabinoid, CB1 , Animals , Endocannabinoids , Exploratory Behavior/physiology , GABAergic Neurons/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Cannabinoid, CB1/genetics , gamma-Aminobutyric Acid
2.
Neuropharmacology ; 179: 108287, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32860777

ABSTRACT

Δ9-tetrahydrocannabinol (THC), the major psychoactive ingredient of Cannabis sativa, exerts its actions through the endocannabinoid system by stimulation of the cannabinoid type 1 (CB1) receptor. The widespread distribution of this receptor in different neuronal cell types and the plethora of functions that is modulated by the endocannabinoid system explain the versatility of the effects of THC. However, the cell types involved in the different THC effects are still not fully known. Conditional CB1 receptor knock-out mice were previously used to identify CB1 receptor subpopulations that are "necessary" for the tetrad effects of a high dose of THC: hypothermia, hypolocomotion, catalepsy and analgesia. Here, we used mouse models for conditional CB1 receptor "rescue" in dorsal telencephalic glutamatergic and forebrain GABAergic neurons to determine which CB1 receptor subpopulations are "sufficient" for these tetrad effects. Glutamatergic CB1 receptor was not only necessary but also sufficient for THC-induced hypothermia and hypolocomotion. Analgesic and cataleptic effects of THC are largely independent of glutamatergic and GABAergic CB1 receptors, since no sufficiency was found, in agreement with the previously reported lack of necessity. We also revealed a novel aspect of GABAergic CB1 receptor signaling. In animals with CB1 receptors exclusively in forebrain GABAergic neurons, THC stimulated rather than reduced locomotion. This cell-type selective and hitherto unsuspected hyperlocomotive effect may be occluded in wild-types and conditional knockouts and only be exposed when CB1 signaling is absent in all other cell types, thus underlining the importance of investigating both necessary and sufficient functions to unequivocally unravel cell-type specific actions.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Dronabinol/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptors, GABA , Receptors, Glutamate , Analgesia/methods , Animals , Cannabinoid Receptor Agonists/metabolism , Catalepsy/chemically induced , Catalepsy/metabolism , Dronabinol/metabolism , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/pharmacology , GABA Agonists/metabolism , GABA Agonists/pharmacology , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Knockout , Receptor, Cannabinoid, CB1/metabolism , Receptors, GABA/metabolism , Receptors, Glutamate/metabolism
3.
Nature ; 558(7711): 590-594, 2018 06.
Article in English | MEDLINE | ID: mdl-29925954

ABSTRACT

Escaping from imminent danger is an instinctive behaviour that is fundamental for survival, and requires the classification of sensory stimuli as harmless or threatening. The absence of threat enables animals to forage for essential resources, but as the level of threat and potential for harm increases, they have to decide whether or not to seek safety 1 . Despite previous work on instinctive defensive behaviours in rodents2-11, little is known about how the brain computes the threat level for initiating  escape. Here we show that the probability and vigour of escape in mice scale with the saliency of innate threats, and are well described by a model that computes the distance between the threat level and an escape threshold. Calcium imaging and optogenetics in the midbrain of freely behaving mice show that the activity of excitatory neurons in the deep layers of the medial superior colliculus (mSC) represents the saliency of the threat stimulus and is predictive of escape, whereas glutamatergic neurons of the dorsal periaqueductal grey (dPAG) encode exclusively the choice to escape and control escape vigour. We demonstrate a feed-forward monosynaptic excitatory connection from mSC to dPAG neurons, which is weak and unreliable-yet required for escape behaviour-and provides a synaptic threshold for dPAG activation and the initiation of escape. This threshold can be overcome by high mSC network activity because of short-term synaptic facilitation and recurrent excitation within the mSC, which amplifies and sustains synaptic drive to the dPAG. Therefore, dPAG glutamatergic neurons compute escape decisions and escape vigour using a synaptic mechanism to  threshold threat information received from the mSC, and provide a biophysical model of how the brain performs a critical behavioural computation.


Subject(s)
Decision Making , Escape Reaction/physiology , Models, Neurological , Synapses/metabolism , Animals , Calcium/analysis , Female , Male , Mice , Mice, Inbred C57BL , Neural Pathways , Optogenetics , Periaqueductal Gray/physiology , Superior Colliculi/physiology
4.
Glia ; 66(7): 1417-1431, 2018 07.
Article in English | MEDLINE | ID: mdl-29480581

ABSTRACT

Astroglial type-1 cannabinoid (CB1 ) receptors are involved in synaptic transmission, plasticity and behavior by interfering with the so-called tripartite synapse formed by pre- and post-synaptic neuronal elements and surrounding astrocyte processes. However, little is known concerning the subcellular distribution of astroglial CB1 receptors. In particular, brain CB1 receptors are mostly localized at cells' plasmalemma, but recent evidence indicates their functional presence in mitochondrial membranes. Whether CB1 receptors are present in astroglial mitochondria has remained unknown. To investigate this issue, we included conditional knock-out mice lacking astroglial CB1 receptor expression specifically in glial fibrillary acidic protein (GFAP)-containing astrocytes (GFAP-CB1 -KO mice) and also generated genetic rescue mice to re-express CB1 receptors exclusively in astrocytes (GFAP-CB1 -RS). To better identify astroglial structures by immunoelectron microscopy, global CB1 knock-out (CB1 -KO) mice and wild-type (CB1 -WT) littermates were intra-hippocampally injected with an adeno-associated virus expressing humanized renilla green fluorescent protein (hrGFP) under the control of human GFAP promoter to generate GFAPhrGFP-CB1 -KO and -WT mice, respectively. Furthermore, double immunogold (for CB1 ) and immunoperoxidase (for GFAP or hrGFP) revealed that CB1 receptors are present in astroglial mitochondria from different hippocampal regions of CB1 -WT, GFAP-CB1 -RS and GFAPhrGFP-CB1 -WT mice. Only non-specific gold particles were detected in mouse hippocampi lacking CB1 receptors. Altogether, we demonstrated the existence of a precise molecular architecture of the CB1 receptor in astrocytes that will have to be taken into account in evaluating the functional activity of cannabinergic signaling at the tripartite synapse.


Subject(s)
Astrocytes/metabolism , Astrocytes/ultrastructure , Hippocampus/metabolism , Hippocampus/ultrastructure , Receptor, Cannabinoid, CB1/metabolism , Animals , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Immunoenzyme Techniques , Mice, Knockout , Microscopy, Immunoelectron , Mitochondria/metabolism , Mitochondria/ultrastructure , Receptor, Cannabinoid, CB1/genetics
5.
J Neurochem ; 142(4): 521-533, 2017 08.
Article in English | MEDLINE | ID: mdl-28608535

ABSTRACT

Numerous studies have been carried out in the mouse model, investigating the role of the cannabinoid receptor type 1 (CB1). However, mouse CB1 (mCB1) receptor differs from human CB1 (hCB1) receptor in 13 amino acid residues. Two splice variants, hCB1a and hCB1b, diverging in their amino-termini, have been reported to be unique for hCB1 and, via different signaling properties, contribute to CB1 receptor physiology and pathophysiology. We hypothesized that splice variants also exist for the mCB1 receptor and have different signaling properties. On murine hippocampal cDNA, we identified two novel mCB1 receptor splice variants generated by splicing of introns with 117 bp and 186 bp in the N-terminal domain, corresponding to deletions of 39 or 62 amino acids, respectively. The mRNAs for the splice variants mCB1a and mCB1b are expressed at low levels in different brain regions. Western blot analysis of protein extracts from stably transfected HEK293 cells indicates a strongly reduced glycosylation because of the absence of two glycosylation sites in mCB1b. On-cell western analysis in these stable lines revealed increased internalization of mCB1a and mCB1b upon stimulation with the agonist WIN55,212-2 as compared to mCB1. Results also point toward an increased affinity to SR141716 for mCB1a, as well as slightly enhanced inhibition of neurotransmission compared to mCB1. In mCB1b, agonist-induced MAPK phosphorylation was decreased compared to mCB1 and mCB1a. Identification of mouse CB1 receptor splice variants may help to explain differences found between human and mouse endocannabinoid systems and improve the understanding of CB1 receptor signaling and trafficking in different species.


Subject(s)
Endocannabinoids/metabolism , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Benzoxazines/pharmacology , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mice, Knockout , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/metabolism , RNA Splicing/drug effects , RNA Splicing/genetics , RNA, Messenger/drug effects , Receptor, Cannabinoid, CB1/drug effects , Signal Transduction/drug effects
6.
Brain Struct Funct ; 222(8): 3431-3452, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28393261

ABSTRACT

Genetic inactivation of the cannabinoid CB1 receptor gene in different cell types in the brain has previously revealed necessary functions for distinct synaptic plasticity processes and behaviors. Here, we sought to identify CB1 receptor expression sites that are minimally required to reconstruct normal phenotypes. In a CB1-null background, we re-expressed endogenous CB1 receptors in forebrain GABAergic neurons, thereby assessing the sufficiency of CB1 receptors. Depolarization-induced suppression of inhibitory, but not excitatory, transmission was restored in hippocampal and amygdalar circuits. GABAergic CB1 receptors did not convey protection against chemically induced seizures, but prevented the spontaneous mortality observed in CB1 null mutants. Rescue of GABAergic CB1 receptors largely restored normal anxiety-like behavior but improved extinction of learned fear only marginally. This study illustrates that the approach of genetic reconstruction of complex behaviors is feasible. It also revealed distinct degrees of modulation for different emotional behaviors by the GABAergic population of CB1 receptors.


Subject(s)
Behavior, Animal , GABAergic Neurons/physiology , Prosencephalon/physiology , Receptor, Cannabinoid, CB1/physiology , Amygdala/metabolism , Amygdala/physiology , Animals , Anxiety , Extinction, Psychological , Fear , Hippocampus/metabolism , Hippocampus/physiology , Inhibitory Postsynaptic Potentials , Male , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Seizures/chemically induced
7.
J Comp Neurol ; 525(2): 302-318, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27339436

ABSTRACT

Type 1 cannabinoid (CB1 ) receptors are widely distributed in the brain. Their physiological roles depend on their distribution pattern, which differs remarkably among cell types. Hence, subcellular compartments with little but functionally relevant CB1 receptors can be overlooked, fostering an incomplete mapping. To overcome this, knockin mice with cell-type-specific rescue of CB1 receptors have emerged as excellent tools for investigating CB1 receptors' cell-type-specific localization and sufficient functional role with no bias. However, to know whether these rescue mice maintain endogenous CB1 receptor expression level, detailed anatomical studies are necessary. The subcellular distribution of hippocampal CB1 receptors of rescue mice that express the gene exclusively in dorsal telencephalic glutamatergic neurons (Glu-CB1 -RS) or GABAergic neurons (GABA-CB1 -RS) was studied by immunoelectron microscopy. Results were compared with conditional CB1 receptor knockout lines. As expected, CB1 immunoparticles appeared at presynaptic plasmalemma, making asymmetric and symmetric synapses. In the hippocampal CA1 stratum radiatum, the values of the CB1 receptor-immunopositive excitatory and inhibitory synapses were Glu-CB1 -RS, 21.89% (glutamatergic terminals); 2.38% (GABAergic terminals); GABA-CB1 -RS, 1.92% (glutamatergic terminals); 77.92% (GABAergic terminals). The proportion of CB1 receptor-immunopositive excitatory and inhibitory synapses in the inner one-third of the dentate molecular layer was Glu-CB1 -RS, 53.19% (glutamatergic terminals); 2.30% (GABAergic terminals); GABA-CB1 -RS, 3.19% (glutamatergic terminals); 85.07% (GABAergic terminals). Taken together, Glu-CB1 -RS and GABA-CB1 -RS mice show the usual CB1 receptor distribution and expression in hippocampal cell types with specific rescue of the receptor, thus being ideal for in-depth anatomical and functional investigations of the endocannabinoid system. J. Comp. Neurol. 525:302-318, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hippocampus/metabolism , Hippocampus/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Receptor, Cannabinoid, CB1/biosynthesis , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Immunoelectron
8.
Proc Natl Acad Sci U S A ; 113(35): 9904-9, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27528659

ABSTRACT

Stressful events can generate emotional memories linked to the traumatic incident, but they also can impair the formation of nonemotional memories. Although the impact of stress on emotional memories is well studied, much less is known about the influence of the emotional state on the formation of nonemotional memories. We used the novel object-recognition task as a model of nonemotional memory in mice to investigate the underlying mechanism of the deleterious effect of stress on memory consolidation. Systemic, hippocampal, and peripheral blockade of cannabinoid type-1 (CB1) receptors abolished the stress-induced memory impairment. Genetic deletion and rescue of CB1 receptors in specific cell types revealed that the CB1 receptor population specifically in dopamine ß-hydroxylase (DBH)-expressing cells is both necessary and sufficient for stress-induced impairment of memory consolidation, but CB1 receptors present in other neuronal populations are not involved. Strikingly, pharmacological manipulations in mice expressing CB1 receptors exclusively in DBH(+) cells revealed that both hippocampal and peripheral receptors mediate the impact of stress on memory consolidation. Thus, CB1 receptors on adrenergic and noradrenergic cells provide previously unrecognized cross-talk between central and peripheral mechanisms in the stress-dependent regulation of nonemotional memory consolidation, suggesting new potential avenues for the treatment of cognitive aspects on stress-related disorders.


Subject(s)
Memory Consolidation/physiology , Memory Disorders/physiopathology , Receptor, Cannabinoid, CB1/physiology , Stress, Psychological/physiopathology , Animals , Anisomycin/pharmacology , Dopamine beta-Hydroxylase/metabolism , Electroshock/adverse effects , Hindlimb Suspension/adverse effects , Indoles/pharmacology , Male , Memory Consolidation/drug effects , Memory Disorders/etiology , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Rimonabant , Stress, Psychological/etiology
9.
Neuron ; 90(2): 308-19, 2016 04 20.
Article in English | MEDLINE | ID: mdl-27068790

ABSTRACT

In daylight, the input to the retinal circuit is provided primarily by cone photoreceptors acting as band-pass filters, but the retinal output also contains neuronal populations transmitting sustained signals. Using in vivo imaging of genetically encoded calcium reporters, we investigated the circuits that generate these sustained channels within the inner retina of zebrafish. In OFF bipolar cells, sustained transmission was found to depend on crossover inhibition from the ON pathway through GABAergic amacrine cells. In ON bipolar cells, the amplitude of low-frequency signals was regulated by glycinergic amacrine cells, while GABAergic inhibition regulated the gain of band-pass signals. We also provide the first functional description of a subset of sustained ON bipolar cells in which synaptic activity was suppressed by fluctuations at frequencies above ∼0.2 Hz. These results map out the basic circuitry by which the inner retina generates sustained visual signals and describes a new function of crossover inhibition.


Subject(s)
Amacrine Cells/physiology , Neural Inhibition/physiology , Retina/physiology , Retinal Bipolar Cells/physiology , Vision, Ocular/physiology , Amacrine Cells/metabolism , Animals , GABAergic Neurons/physiology , Glycine/metabolism , Glycine/physiology , Photic Stimulation , Synaptic Transmission/physiology , Zebrafish
10.
Proc Natl Acad Sci U S A ; 111(36): E3805-14, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25157152

ABSTRACT

Genetically encoded fluorescent proteins and immunostaining are widely used to detect cellular and subcellular structures in fixed biological samples. However, for thick or whole-mount tissue, each approach suffers from limitations, including limited spectral flexibility and lower signal or slow speed, poor penetration, and high background labeling, respectively. We have overcome these limitations by using transgenically expressed chemical tags for rapid, even, high-signal and low-background labeling of thick biological tissues. We first construct a platform of widely applicable transgenic Drosophila reporter lines, demonstrating that chemical labeling can accelerate staining of whole-mount fly brains by a factor of 100. Using viral vectors to deliver chemical tags into the mouse brain, we then demonstrate that this labeling strategy works well in mice. Thus this tag-based approach drastically improves the speed and specificity of labeling genetically marked cells in intact and/or thick biological samples.


Subject(s)
Brain/metabolism , Fluorescent Dyes/metabolism , Staining and Labeling/methods , Animals , Drosophila , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism
11.
Nat Neurosci ; 17(3): 407-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24509429

ABSTRACT

Hunger arouses sensory perception, eventually leading to an increase in food intake, but the underlying mechanisms remain poorly understood. We found that cannabinoid type-1 (CB1) receptors promote food intake in fasted mice by increasing odor detection. CB1 receptors were abundantly expressed on axon terminals of centrifugal cortical glutamatergic neurons that project to inhibitory granule cells of the main olfactory bulb (MOB). Local pharmacological and genetic manipulations revealed that endocannabinoids and exogenous cannabinoids increased odor detection and food intake in fasted mice by decreasing excitatory drive from olfactory cortex areas to the MOB. Consistently, cannabinoid agonists dampened in vivo optogenetically stimulated excitatory transmission in the same circuit. Our data indicate that cortical feedback projections to the MOB crucially regulate food intake via CB1 receptor signaling, linking the feeling of hunger to stronger odor processing. Thus, CB1 receptor-dependent control of cortical feedback projections in olfactory circuits couples internal states to perception and behavior.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Eating/physiology , Endocannabinoids/physiology , Feeding Behavior/physiology , Olfactory Pathways/physiology , Olfactory Perception/physiology , Receptor, Cannabinoid, CB1/metabolism , Synaptic Transmission/physiology , Animals , Eating/drug effects , Endocannabinoids/metabolism , Feedback, Physiological/physiology , Feeding Behavior/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Olfactory Bulb/physiology , Olfactory Pathways/drug effects , Olfactory Pathways/metabolism , Olfactory Perception/drug effects , Receptor, Cannabinoid, CB1/genetics , Synaptic Transmission/drug effects
12.
J Neurosci ; 33(25): 10264-77, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785142

ABSTRACT

A major goal in current neuroscience is to understand the causal links connecting protein functions, neural activity, and behavior. The cannabinoid CB1 receptor is expressed in different neuronal subpopulations, and is engaged in fine-tuning excitatory and inhibitory neurotransmission. Studies using conditional knock-out mice revealed necessary roles of CB1 receptor expressed in dorsal telencephalic glutamatergic neurons in synaptic plasticity and behavior, but whether this expression is also sufficient for brain functions is still to be determined. We applied a genetic strategy to reconstitute full wild-type CB1 receptor functions exclusively in dorsal telencephalic glutamatergic neurons and investigated endocannabinoid-dependent synaptic processes and behavior. Using this approach, we partly restored the phenotype of global CB1 receptor deletion in anxiety-like behaviors and fully restored hippocampus-dependent neuroprotection from chemically induced epileptiform seizures. These features coincided with a rescued hippocampal depolarization-induced suppression of excitation (DSE), a CB1 receptor-dependent form of synaptic plasticity at glutamatergic neurons. By comparison, the rescue of the CB1 receptor on dorsal telencephalic glutamatergic neurons prolonged the time course of DSE in the amygdala, and impaired fear extinction in auditory fear conditioning. These data reveal that CB1 receptor in dorsal telencephalic glutamatergic neurons plays a sufficient role to control neuronal functions that are in large part hippocampus-dependent, while it is insufficient for proper amygdala functions, suggesting an unexpectedly complex circuit regulation by endocannabinoid signaling in the amygdala. Our data pave the way to a better understanding of neuronal networks in the context of behavior, by fine-tuned interference with synaptic transmission processes.


Subject(s)
Amygdala/physiology , Behavior, Animal/physiology , Glutamic Acid/physiology , Hippocampus/physiology , Neurons/physiology , Receptor, Cannabinoid, CB1/physiology , Synapses/physiology , Telencephalon/physiology , Animals , Anxiety/psychology , Blotting, Western , Electrophysiological Phenomena , Excitatory Amino Acid Agonists/toxicity , Immunohistochemistry , Kainic Acid/toxicity , Light , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/physiology , RNA/biosynthesis , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Telencephalon/cytology
13.
J Neurochem ; 124(6): 795-807, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23289830

ABSTRACT

Type 1 cannabinoid receptor (CB1) is expressed in different neuronal populations in the mammalian brain. In particular, CB1 on GABAergic or glutamatergic neurons exerts different functions and display different pharmacological properties in vivo. This suggests the existence of neuron-type specific signalling pathways activated by different subpopulations of CB1. In this study, we analysed CB1 expression, binding and signalling in the hippocampus of conditional mutant mice, bearing CB1 deletion in GABAergic (GABA-CB1-KO mice) or cortical glutamatergic neurons (Glu-CB1-KO mice). Compared to their wild-type littermates, Glu-CB1-KO displayed a small decrease of CB1 mRNA amount, immunoreactivity and [³H]CP55,940 binding. Conversely, GABA-CB1-KO mice showed a drastic reduction of these parameters, confirming that CB1 is present at much higher density on hippocampal GABAergic interneurons than glutamatergic neurons. Surprisingly, however, saturation analysis of HU210-stimulated [(35) S]GTPγS binding demonstrated that 'glutamatergic' CB1 is more efficiently coupled to G protein signalling than 'GABAergic' CB1. Thus, the minority of CB1 on glutamatergic neurons is paradoxically several fold more strongly coupled to G protein signalling than 'GABAergic' CB1. This selective signalling mechanism raises the possibility of designing novel cannabinoid ligands that differentially activate only a subset of physiological effects of CB1 stimulation, thereby optimizing therapeutic action.


Subject(s)
Cannabinoids/metabolism , GABAergic Neurons/physiology , GTP-Binding Proteins/physiology , Hippocampus/metabolism , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/physiology , Animals , Hippocampus/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/physiology , Receptor, Cannabinoid, CB1/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL