Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Development ; 147(24)2020 12 23.
Article in English | MEDLINE | ID: mdl-33168583

ABSTRACT

The endocannabinoid (eCB) system, via the cannabinoid CB1 receptor, regulates neurodevelopment by controlling neural progenitor proliferation and neurogenesis. CB1 receptor signalling in vivo drives corticofugal deep layer projection neuron development through the regulation of BCL11B and SATB2 transcription factors. Here, we investigated the role of eCB signalling in mouse pluripotent embryonic stem cell-derived neuronal differentiation. Characterization of the eCB system revealed increased expression of eCB-metabolizing enzymes, eCB ligands and CB1 receptors during neuronal differentiation. CB1 receptor knockdown inhibited neuronal differentiation of deep layer neurons and increased upper layer neuron generation, and this phenotype was rescued by CB1 re-expression. Pharmacological regulation with CB1 receptor agonists or elevation of eCB tone with a monoacylglycerol lipase inhibitor promoted neuronal differentiation of deep layer neurons at the expense of upper layer neurons. Patch-clamp analyses revealed that enhancing cannabinoid signalling facilitated neuronal differentiation and functionality. Noteworthy, incubation with CB1 receptor agonists during human iPSC-derived cerebral organoid formation also promoted the expansion of BCL11B+ neurons. These findings unveil a cell-autonomous role of eCB signalling that, via the CB1 receptor, promotes mouse and human deep layer cortical neuron development.


Subject(s)
Cell Differentiation/genetics , Matrix Attachment Region Binding Proteins/genetics , Neurons/metabolism , Receptor, Cannabinoid, CB1/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Proliferation/drug effects , Cerebellum/growth & development , Embryonic Development/genetics , Endocannabinoids/agonists , Endocannabinoids/genetics , Endocannabinoids/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Induced Pluripotent Stem Cells/drug effects , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Organoids/growth & development , Signal Transduction/genetics
2.
Nat Commun ; 11(1): 2217, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32371879

ABSTRACT

Theta oscillations play a major role in temporarily defining the hippocampal rate code by translating behavioral sequences into neuronal representations. However, mechanisms constraining phase timing and cell-type-specific phase preference are unknown. Here, we employ computational models tuned with evolutionary algorithms to evaluate phase preference of individual CA1 pyramidal cells recorded in mice and rats not engaged in any particular memory task. We applied unbiased and hypothesis-free approaches to identify effects of intrinsic and synaptic factors, as well as cell morphology, in determining phase preference. We found that perisomatic inhibition delivered by complementary populations of basket cells interacts with input pathways to shape phase-locked specificity of deep and superficial pyramidal cells. Somatodendritic integration of fluctuating glutamatergic inputs defined cycle-by-cycle by unsupervised methods demonstrated that firing selection is tuneable across sublayers. Our data identify different mechanisms of phase-locking selectivity that are instrumental for flexible dynamical representations of theta sequences.


Subject(s)
CA1 Region, Hippocampal/physiology , Neurons/physiology , Synapses/physiology , Theta Rhythm/physiology , Action Potentials/physiology , Algorithms , Animals , CA1 Region, Hippocampal/cytology , Computer Simulation , Female , Kinetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Patch-Clamp Techniques , Pyramidal Cells/physiology , Rats, Wistar
3.
Neuropsychopharmacology ; 45(5): 877-886, 2020 04.
Article in English | MEDLINE | ID: mdl-31982904

ABSTRACT

Prenatal exposure to Δ9-tetrahydrocannabinol (THC), the most prominent active constituent of cannabis, alters neurodevelopmental plasticity with a long-term functional impact on adult offspring. Specifically, THC affects the development of pyramidal neurons and GABAergic interneurons via cannabinoid CB1 receptors (CB1R). However, the particular contribution of these two neuronal lineages to the behavioral alterations and functional deficits induced by THC is still unclear. Here, by using conditional CB1R knockout mice, we investigated the neurodevelopmental consequences of prenatal THC exposure in adulthood, as well as their potential sex differences. Adult mice that had been exposed to THC during embryonic development showed altered hippocampal oscillations, brain hyperexcitability, and spatial memory impairment. Remarkably, we found a clear sexual dimorphism in these effects, with males being selectively affected. At the neuronal level, we found a striking interneuronopathy of CCK-containing interneurons in the hippocampus, which was restricted to male progeny. This THC-induced CCK-interneuron reduction was not evident in mice lacking CB1R selectively in GABAergic interneurons, thus pointing to a cell-autonomous THC action. In vivo electrophysiological recordings of hippocampal LFPs revealed alterations in hippocampal oscillations confined to the stratum pyramidale of CA1 in male offspring. In addition, sharp-wave ripples, a major high-frequency oscillation crucial for learning and memory consolidation, were also altered, pointing to aberrant circuitries caused by persistent reduction of CCK+ basket cells. Taken together, these findings provide a mechanistic explanation for the long-term interneuronopathy responsible for the sex-dimorphic cognitive impairment induced by prenatal THC.


Subject(s)
Cannabinoid Receptor Agonists/administration & dosage , Dronabinol/administration & dosage , Hippocampus/drug effects , Hippocampus/pathology , Interneurons/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Sex Characteristics , Spatial Memory/drug effects , Animals , Female , Hippocampus/physiology , Interneurons/pathology , Male , Mice, Knockout , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/psychology , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/genetics , Spatial Memory/physiology
4.
Front Pharmacol ; 9: 1508, 2018.
Article in English | MEDLINE | ID: mdl-30687088

ABSTRACT

Alterations of the PI3K/Akt/mammalian target of rapamycin complex 1 (mTORC1) signaling pathway are causally involved in a subset of malformations of cortical development (MCDs) ranging from focal cortical dysplasia (FCD) to hemimegalencephaly and megalencephaly. These MCDs represent a frequent cause of refractory pediatric epilepsy. The endocannabinoid system -especially cannabinoid CB1 receptor- exerts a neurodevelopmental regulatory role at least in part via activation of mTORC1 signaling. Therefore, we sought to characterize the possible contribution of endocannabinoid system signaling to FCD. Confocal microscopy characterization of the CB1 receptor expression and mTORC1 activation was conducted in FCD Type II resection samples. FCD samples were subjected to single nucleotide polymorphism screening for endocannabinoid system elements, as well as CB1 receptor gene sequencing. Cannabinoid CB1 receptor levels were increased in FCD with overactive mTORC1 signaling. CB1 receptors were enriched in phospho-S6-positive cells including balloon cells (BCs) that co-express aberrant markers of undifferentiated cells and dysplastic neurons. Pharmacological regulation of CB1 receptors and the mTORC1 pathway was performed in fresh FCD-derived organotypic cultures. HU-210-evoked activation of CB1 receptors was unable to further activate mTORC1 signaling, whereas CB1 receptor blockade with rimonabant attenuated mTORC1 overactivation. Alterations of the endocannabinoid system may thus contribute to FCD pathological features, and blockade of cannabinoid signaling might be a new therapeutic intervention in FCD.

5.
Cereb Cortex ; 27(11): 5303-5317, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28334226

ABSTRACT

Neuronal migration is a fundamental process of brain development, and its disruption underlies devastating neurodevelopmental disorders. The transcriptional programs governing this process are relatively well characterized. However, how environmental cues instruct neuronal migration remains poorly understood. Here, we demonstrate that the cannabinoid CB1 receptor is strictly required for appropriate pyramidal neuron migration in the developing cortex. Acute silencing of the CB1 receptor alters neuronal morphology and impairs radial migration. Consequently, CB1 siRNA-electroporated mice display cortical malformations mimicking subcortical band heterotopias and increased seizure susceptibility in adulthood. Importantly, rescuing the CB1 deficiency-induced radial migration arrest by knockdown of the GTPase protein RhoA restored the hyperexcitable neuronal network and seizure susceptibility. Our findings show that CB1 receptor/RhoA signaling regulates pyramidal neuron migration, and that deficient CB1 receptor signaling may contribute to cortical development malformations leading to refractory epilepsy independently of its canonical neuromodulatory role in the adult brain.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/abnormalities , Cerebral Cortex/metabolism , Pyramidal Cells/metabolism , Receptor, Cannabinoid, CB1/deficiency , Seizures/metabolism , Animals , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Disease Models, Animal , Disease Susceptibility/metabolism , Disease Susceptibility/pathology , Electroporation , Fluorescent Antibody Technique , Gene Knockdown Techniques , In Situ Hybridization , Mice, Transgenic , Microscopy, Confocal , Pentylenetetrazole , Pyramidal Cells/pathology , RNA, Small Interfering , Receptor, Cannabinoid, CB1/genetics , Seizures/pathology , Tissue Culture Techniques , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
6.
Proc Natl Acad Sci U S A ; 112(44): 13693-8, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26460022

ABSTRACT

The CB1 cannabinoid receptor, the main target of Δ(9)-tetrahydrocannabinol (THC), the most prominent psychoactive compound of marijuana, plays a crucial regulatory role in brain development as evidenced by the neurodevelopmental consequences of its manipulation in animal models. Likewise, recreational cannabis use during pregnancy affects brain structure and function of the progeny. However, the precise neurobiological substrates underlying the consequences of prenatal THC exposure remain unknown. As CB1 signaling is known to modulate long-range corticofugal connectivity, we analyzed the impact of THC exposure on cortical projection neuron development. THC administration to pregnant mice in a restricted time window interfered with subcerebral projection neuron generation, thereby altering corticospinal connectivity, and produced long-lasting alterations in the fine motor performance of the adult offspring. Consequences of THC exposure were reminiscent of those elicited by CB1 receptor genetic ablation, and CB1-null mice were resistant to THC-induced alterations. The identity of embryonic THC neuronal targets was determined by a Cre-mediated, lineage-specific, CB1 expression-rescue strategy in a CB1-null background. Early and selective CB1 reexpression in dorsal telencephalic glutamatergic neurons but not forebrain GABAergic neurons rescued the deficits in corticospinal motor neuron development of CB1-null mice and restored susceptibility to THC-induced motor alterations. In addition, THC administration induced an increase in seizure susceptibility that was mediated by its interference with CB1-dependent regulation of both glutamatergic and GABAergic neuron development. These findings demonstrate that prenatal exposure to THC has long-lasting deleterious consequences in the adult offspring solely mediated by its ability to disrupt the neurodevelopmental role of CB1 signaling.


Subject(s)
Cerebral Cortex/metabolism , Dronabinol/administration & dosage , Maternal Exposure , Neurons/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Cerebral Cortex/growth & development , Female , Mice , Pregnancy
7.
Cereb Cortex ; 25(9): 2395-408, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24610119

ABSTRACT

The CB1 cannabinoid receptor regulates cortical progenitor proliferation during embryonic development, but the molecular mechanism of this action remains unknown. Here, we report that CB1-deficient mouse embryos show premature cell cycle exit, decreased Pax6- and Tbr2-positive cell number, and reduced mammalian target of rapamycin complex 1 (mTORC1) activation in the ventricular and subventricular cortical zones. Pharmacological stimulation of the CB1 receptor in cortical slices and progenitor cell cultures activated the mTORC1 pathway and increased the number of Pax6- and Tbr2-expressing cells. Likewise, acute CB1 knockdown in utero reduced mTORC1 activation and cannabinoid-induced Tbr2-positive cell generation. Luciferase reporter and chromatin immunoprecipitation assays revealed that the CB1 receptor drives Tbr2 expression downstream of Pax6 induction in an mTORC1-dependent manner. Altogether, our results demonstrate that the CB1 receptor tunes dorsal telencephalic progenitor proliferation by sustaining the transcriptional activity of the Pax6-Tbr2 axis via the mTORC1 pathway, and suggest that alterations of CB1 receptor signaling, by producing the missexpression of progenitor identity determinants may contribute to neurodevelopmental alterations.


Subject(s)
Cerebral Cortex , Gene Expression Regulation, Developmental/genetics , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/genetics , Stem Cells/physiology , T-Box Domain Proteins/metabolism , Animals , Animals, Newborn , Cell Culture Techniques , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Embryo, Mammalian , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Ki-67 Antigen/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation/genetics , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Cannabinoid, CB1/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Box Domain Proteins/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...