Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Molecules ; 26(22)2021 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-34833992

RESUMO

Numerous studies have investigated the roles of the type 1 cannabinoid receptor (CB1) in glutamatergic and GABAergic neurons. Here, we used the cell-type-specific CB1 rescue model in mice to gain insight into the organizational principles of plasma membrane targeting and Gαi/o protein signalling of the CB1 receptor at excitatory and inhibitory terminals of the frontal cortex and hippocampus. By applying biochemical fractionation techniques and Western blot analyses to synaptosomal membranes, we explored the subsynaptic distribution (pre-, post-, and extra-synaptic) and CB1 receptor compartmentalization into lipid and non-lipid raft plasma membrane microdomains and the signalling properties. These data infer that the plasma membrane partitioning of the CB1 receptor and its functional coupling to Gαi/o proteins are not biased towards the cell type of CB1 receptor rescue. The extent of the canonical Gαi/o protein-dependent CB1 receptor signalling correlated with the abundance of CB1 receptor in the respective cell type (glutamatergic versus GABAergic neurons) both in frontal cortical and hippocampal synaptosomes. In summary, our results provide an updated view of the functional coupling of the CB1 receptor to Gαi/o proteins at excitatory and inhibitory terminals and substantiate the utility of the CB1 rescue model in studying endocannabinoid physiology at the subcellular level.


Assuntos
Lobo Frontal/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Hipocampo/metabolismo , Microdomínios da Membrana/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Sinaptossomos/metabolismo , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Microdomínios da Membrana/genética , Camundongos , Camundongos Knockout , Receptor CB1 de Canabinoide/genética , Sinapses/genética
2.
J Neurochem ; 142(4): 521-533, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28608535

RESUMO

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.


Assuntos
Endocanabinoides/metabolismo , RNA Mensageiro/metabolismo , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Animais , Benzoxazinas/farmacologia , Células HEK293 , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Camundongos Knockout , Morfolinas/farmacologia , Naftalenos/farmacologia , Neurônios/metabolismo , Splicing de RNA/efeitos dos fármacos , Splicing de RNA/genética , RNA Mensageiro/efeitos dos fármacos , Receptor CB1 de Canabinoide/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
3.
J Neurosci ; 35(9): 3842-50, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25740514

RESUMO

The endocannabinoid system negatively regulates the release of various neurotransmitters in an activity-dependent manner, thereby influencing the excitability of neuronal circuits. In the hippocampus, cannabinoid type 1 (CB1) receptor is present on both GABAergic and glutamatergic axon terminals. CB1 receptor-deficient mice were previously shown to have increased hippocampal long-term potentiation (LTP). In this study, we have investigated the consequences of cell-type-specific deletion of the CB1 receptor on the induction of hippocampal LTP and on CA1 pyramidal cell morphology. Deletion of CB1 receptor in GABAergic neurons in GABA-CB1-KO mice leads to a significantly decreased hippocampal LTP compared with WT controls. Concomitantly, CA1 pyramidal neurons have a significantly reduced dendritic branching both on the apical and on the basal dendrites. Moreover, the average spine density on the apical dendrites of CA1 pyramidal neurons is significantly diminished. In contrast, in mice lacking CB1 receptor in glutamatergic cells (Glu-CB1-KO), hippocampal LTP is significantly enhanced and CA1 pyramidal neurons show an increased branching and an increased spine density in the apical dendritic region. Together, these results indicate that the CB1 receptor signaling system both on inhibitory and excitatory neurons controls functional and structural synaptic plasticity of pyramidal neurons in the hippocampal CA1 region to maintain an appropriate homeostatic state upon neuronal activation. Consequently, if the CB1 receptor is lost in either neuronal population, an allostatic shift will occur leading to a long-term dysregulation of neuronal functions.


Assuntos
Hipocampo/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Sinapses/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal/fisiologia , Receptor CB1 de Canabinoide/genética , Transmissão Sináptica/fisiologia
4.
J Neurosci ; 35(41): 13975-88, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26468198

RESUMO

Adolescence is characterized by drastic behavioral adaptations and comprises a particularly vulnerable period for the emergence of various psychiatric disorders. Growing evidence reveals that the pathophysiology of these disorders might derive from aberrations of normal neurodevelopmental changes in the adolescent brain. Understanding the molecular underpinnings of adolescent behavior is therefore critical for understanding the origin of psychopathology, but the molecular mechanisms that trigger adolescent behavior are unknown. Here, we hypothesize that the cannabinoid type-1 receptor (CB1R) may play a critical role in mediating adolescent behavior because enhanced endocannabinoid (eCB) signaling has been suggested to occur transiently during adolescence. To study enhanced CB1R signaling, we introduced a missense mutation (F238L) into the rat Cnr1 gene that encodes for the CB1R. According to our hypothesis, rats with the F238L mutation (Cnr1(F238L)) should sustain features of adolescent behavior into adulthood. Gain of function of the mutated receptor was demonstrated by in silico modeling and was verified functionally in a series of biochemical and electrophysiological experiments. Mutant rats exhibit an adolescent-like phenotype during adulthood compared with wild-type littermates, with typical high risk/novelty seeking, increased peer interaction, enhanced impulsivity, and augmented reward sensitivity for drug and nondrug reward. Partial inhibition of CB1R activity in Cnr1(F238L) mutant rats normalized behavior and led to a wild-type phenotype. We conclude that the activity state and functionality of the CB1R is critical for mediating adolescent behavior. These findings implicate the eCB system as an important research target for the neuropathology of adolescent-onset mental health disorders. SIGNIFICANCE STATEMENT: We present the first rodent model with a gain-of-function mutation in the cannabinoid type-1 receptor (CB1R). Adult mutant rats exhibit an adolescent-like phenotype with typical high risk seeking, impulsivity, and augmented drug and nondrug reward sensitivity. Adolescence is a critical period for suboptimal behavioral choices and the emergence of neuropsychiatric disorders. Understanding the basis of these disorders therefore requires a comprehensive knowledge of how adolescent neurodevelopment triggers behavioral reactions. Our behavioral observations in adult mutant rats, together with reports on enhanced adolescent CB1R signaling, suggest a pivotal role for the CB1R in an adolescent brain as an important molecular mediator of adolescent behavior. These findings implicate the endocannabinoid system as a notable research target for adolescent-onset mental health disorders.


Assuntos
Comportamento do Adolescente/fisiologia , Comportamento Animal/fisiologia , Receptor CB1 de Canabinoide/metabolismo , Adolescente , Fatores Etários , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Antagonistas de Receptores de Canabinoides/farmacologia , Cocaína/administração & dosagem , Corpo Estriado/citologia , Endocanabinoides/metabolismo , Endocanabinoides/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Humanos , Técnicas In Vitro , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Modelos Animais , Mutação/genética , Cintilografia , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos , Receptor CB1 de Canabinoide/genética , Assunção de Riscos , Comportamento Social , Isótopos de Enxofre/farmacocinética
5.
Proc Natl Acad Sci U S A ; 110(12): 4786-91, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23487769

RESUMO

Complex interactions between periphery and the brain regulate food intake in mammals. Cannabinoid type-1 (CB1) receptor antagonists are potent hypophagic agents, but the sites where this acute action is exerted and the underlying mechanisms are not fully elucidated. To dissect the mechanisms underlying the hypophagic effect of CB1 receptor blockade, we combined the acute injection of the CB1 receptor antagonist rimonabant with the use of conditional CB1-knockout mice, as well as with pharmacological modulation of different central and peripheral circuits. Fasting/refeeding experiments revealed that CB1 receptor signaling in many specific brain neurons is dispensable for the acute hypophagic effects of rimonabant. CB1 receptor antagonist-induced hypophagia was fully abolished by peripheral blockade of ß-adrenergic transmission, suggesting that this effect is mediated by increased activity of the sympathetic nervous system. Consistently, we found that rimonabant increases gastrointestinal metabolism via increased peripheral ß-adrenergic receptor signaling in peripheral organs, including the gastrointestinal tract. Blockade of both visceral afferents and glutamatergic transmission in the nucleus tractus solitarii abolished rimonabant-induced hypophagia. Importantly, these mechanisms were specifically triggered by lipid-deprivation, revealing a nutrient-specific component acutely regulated by CB1 receptor blockade. Finally, peripheral blockade of sympathetic neurotransmission also blunted central effects of CB1 receptor blockade, such as fear responses and anxiety-like behaviors. These data demonstrate that, independently of their site of origin, important effects of CB1 receptor blockade are expressed via activation of peripheral sympathetic activity. Thus, CB1 receptors modulate bidirectional circuits between the periphery and the brain to regulate feeding and other behaviors.


Assuntos
Ansiedade/metabolismo , Regulação do Apetite , Encéfalo/metabolismo , Transtornos da Alimentação e da Ingestão de Alimentos/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Sistema Nervoso Simpático/metabolismo , Transmissão Sináptica , Animais , Ansiedade/genética , Ansiedade/patologia , Ansiedade/fisiopatologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Transtornos da Alimentação e da Ingestão de Alimentos/genética , Transtornos da Alimentação e da Ingestão de Alimentos/fisiopatologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Trato Gastrointestinal/fisiopatologia , Camundongos , Camundongos Knockout , Receptor CB1 de Canabinoide/genética , Sistema Nervoso Simpático/patologia , Sistema Nervoso Simpático/fisiopatologia
6.
Proc Natl Acad Sci U S A ; 109(51): 21134-9, 2012 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-23150578

RESUMO

Allosteric modulation of G-protein-coupled receptors represents a key goal of current pharmacology. In particular, endogenous allosteric modulators might represent important targets of interventions aimed at maximizing therapeutic efficacy and reducing side effects of drugs. Here we show that the anti-inflammatory lipid lipoxin A(4) is an endogenous allosteric enhancer of the CB(1) cannabinoid receptor. Lipoxin A(4) was detected in brain tissues, did not compete for the orthosteric binding site of the CB(1) receptor (vs. (3)H-SR141716A), and did not alter endocannabinoid metabolism (as opposed to URB597 and MAFP), but it enhanced affinity of anandamide at the CB1 receptor, thereby potentiating the effects of this endocannabinoid both in vitro and in vivo. In addition, lipoxin A(4) displayed a CB(1) receptor-dependent protective effect against ß-amyloid (1-40)-induced spatial memory impairment in mice. The discovery of lipoxins as a class of endogenous allosteric modulators of CB(1) receptors may foster the therapeutic exploitation of the endocannabinoid system, in particular for the treatment of neurodegenerative disorders.


Assuntos
Anti-Inflamatórios/metabolismo , Lipoxinas/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Sítio Alostérico , Proteínas Amiloidogênicas/metabolismo , Animais , Encéfalo/metabolismo , Endocanabinoides/metabolismo , Inflamação , Cinética , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fármacos Neuroprotetores/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Comportamento Espacial
7.
Proc Natl Acad Sci U S A ; 108(27): 11256-61, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690345

RESUMO

Brain aging is associated with cognitive decline that is accompanied by progressive neuroinflammatory changes. The endocannabinoid system (ECS) is involved in the regulation of glial activity and influences the progression of age-related learning and memory deficits. Mice lacking the Cnr1 gene (Cnr1(-/-)), which encodes the cannabinoid receptor 1 (CB1), showed an accelerated age-dependent deficit in spatial learning accompanied by a loss of principal neurons in the hippocampus. The age-dependent decrease in neuronal numbers in Cnr1(-/-) mice was not related to decreased neurogenesis or to epileptic seizures. However, enhanced neuroinflammation characterized by an increased density of astrocytes and activated microglia as well as an enhanced expression of the inflammatory cytokine IL-6 during aging was present in the hippocampus of Cnr1(-/-) mice. The ongoing process of pyramidal cell degeneration and neuroinflammation can exacerbate each other and both contribute to the cognitive deficits. Deletion of CB1 receptors from the forebrain GABAergic, but not from the glutamatergic neurons, led to a similar neuronal loss and increased neuroinflammation in the hippocampus as observed in animals lacking CB1 receptors in all cells. Our results suggest that CB1 receptor activity on hippocampal GABAergic neurons protects against age-dependent cognitive decline by reducing pyramidal cell degeneration and neuroinflammation.


Assuntos
Envelhecimento/fisiologia , Encéfalo/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Animais , Encéfalo/citologia , Contagem de Células , Feminino , Expressão Gênica , Hipocampo/citologia , Hipocampo/fisiologia , Interleucina-6/genética , Interleucina-6/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/fisiologia , Neuroglia/citologia , Neuroglia/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Receptor CB1 de Canabinoide/deficiência , Receptor CB1 de Canabinoide/genética , Convulsões/patologia , Convulsões/fisiopatologia , Ácido gama-Aminobutírico/fisiologia
8.
J Neurochem ; 124(6): 795-807, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23289830

RESUMO

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.


Assuntos
Canabinoides/metabolismo , Neurônios GABAérgicos/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Hipocampo/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais/fisiologia , Animais , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica/fisiologia , Receptor CB1 de Canabinoide/deficiência
9.
Brain ; 134(Pt 1): 119-36, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20929960

RESUMO

Endocannabinoids act as neuromodulatory and neuroprotective cues by engaging type 1 cannabinoid receptors. These receptors are highly abundant in the basal ganglia and play a pivotal role in the control of motor behaviour. An early downregulation of type 1 cannabinoid receptors has been documented in the basal ganglia of patients with Huntington's disease and animal models. However, the pathophysiological impact of this loss of receptors in Huntington's disease is as yet unknown. Here, we generated a double-mutant mouse model that expresses human mutant huntingtin exon 1 in a type 1 cannabinoid receptor-null background, and found that receptor deletion aggravates the symptoms, neuropathology and molecular pathology of the disease. Moreover, pharmacological administration of the cannabinoid Δ(9)-tetrahydrocannabinol to mice expressing human mutant huntingtin exon 1 exerted a therapeutic effect and ameliorated those parameters. Experiments conducted in striatal cells show that the mutant huntingtin-dependent downregulation of the receptors involves the control of the type 1 cannabinoid receptor gene promoter by repressor element 1 silencing transcription factor and sensitizes cells to excitotoxic damage. We also provide in vitro and in vivo evidence that supports type 1 cannabinoid receptor control of striatal brain-derived neurotrophic factor expression and the decrease in brain-derived neurotrophic factor levels concomitant with type 1 cannabinoid receptor loss, which may contribute significantly to striatal damage in Huntington's disease. Altogether, these results support the notion that downregulation of type 1 cannabinoid receptors is a key pathogenic event in Huntington's disease, and suggest that activation of these receptors in patients with Huntington's disease may attenuate disease progression.


Assuntos
Corpo Estriado/metabolismo , Doença de Huntington/genética , Neurônios/metabolismo , Receptor CB1 de Canabinoide/genética , Análise de Variância , Animais , Western Blotting , Sobrevivência Celular , Dronabinol/farmacologia , Hormônio Liberador de Hormônio do Crescimento/análogos & derivados , Doença de Huntington/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Receptor CB1 de Canabinoide/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Teste de Desempenho do Rota-Rod
10.
Cell Death Dis ; 13(7): 585, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35798697

RESUMO

Cannabinoids are known to modulate oligodendrogenesis and developmental CNS myelination. However, the cell-autonomous action of these compounds on oligodendroglial cells in vivo, and the molecular mechanisms underlying these effects have not yet been studied. Here, by using oligodendroglial precursor cell (OPC)-targeted genetic mouse models, we show that cannabinoid CB1 receptors exert an essential role in modulating OPC differentiation at the critical periods of postnatal myelination. We found that selective genetic inactivation of CB1 receptors in OPCs in vivo perturbs oligodendrogenesis and postnatal myelination by altering the RhoA/ROCK signaling pathway, leading to hypomyelination, and motor and cognitive alterations in young adult mice. Conversely, pharmacological CB1 receptor activation, by inducing E3 ubiquitin ligase-dependent RhoA proteasomal degradation, promotes oligodendrocyte development and CNS myelination in OPCs, an effect that was not evident in OPC-specific CB1 receptor-deficient mice. Moreover, pharmacological inactivation of ROCK in vivo overcomes the defects in oligodendrogenesis and CNS myelination, and behavioral alterations found in OPC-specific CB1 receptor-deficient mice. Overall, this study supports a cell-autonomous role for CB1 receptors in modulating oligodendrogenesis in vivo, which may have a profound impact on the scientific knowledge and therapeutic manipulation of CNS myelination by cannabinoids.


Assuntos
Canabinoides , Células Precursoras de Oligodendrócitos , Receptor CB1 de Canabinoide , Animais , Canabinoides/farmacologia , Diferenciação Celular/fisiologia , Inativação Gênica , Camundongos , Bainha de Mielina/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Receptor CB1 de Canabinoide/metabolismo
11.
Proc Natl Acad Sci U S A ; 105(25): 8760-5, 2008 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-18562289

RESUMO

Endocannabinoids (eCBs) have recently been identified as axon guidance cues shaping the connectivity of local GABAergic interneurons in the developing cerebrum. However, eCB functions during pyramidal cell specification and establishment of long-range axonal connections are unknown. Here, we show that eCB signaling is operational in subcortical proliferative zones from embryonic day 12 in the mouse telencephalon and controls the proliferation of pyramidal cell progenitors and radial migration of immature pyramidal cells. When layer patterning is accomplished, developing pyramidal cells rely on eCB signaling to initiate the elongation and fasciculation of their long-range axons. Accordingly, CB(1) cannabinoid receptor (CB(1)R) null and pyramidal cell-specific conditional mutant (CB(1)R(f/f,NEX-Cre)) mice develop deficits in neuronal progenitor proliferation and axon fasciculation. Likewise, axonal pathfinding becomes impaired after in utero pharmacological blockade of CB(1)Rs. Overall, eCBs are fundamental developmental cues controlling pyramidal cell development during corticogenesis.


Assuntos
Axônios/metabolismo , Moduladores de Receptores de Canabinoides/metabolismo , Endocanabinoides , Células Piramidais/metabolismo , Transdução de Sinais , Animais , Antagonistas de Receptores de Canabinoides , Diferenciação Celular , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Gravidez , Células Piramidais/citologia , Receptores de Canabinoides/genética , Receptores de Canabinoides/metabolismo
12.
Nat Neurosci ; 10(7): 870-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17558404

RESUMO

Although endocannabinoids constitute one of the first lines of defense against pain, the anatomical locus and the precise receptor mechanisms underlying cannabinergic modulation of pain are uncertain. Clinical exploitation of the system is severely hindered by the cognitive deficits, memory impairment, motor disturbances and psychotropic effects resulting from the central actions of cannabinoids. We deleted the type 1 cannabinoid receptor (CB1) specifically in nociceptive neurons localized in the peripheral nervous system of mice, preserving its expression in the CNS, and analyzed these genetically modified mice in preclinical models of inflammatory and neuropathic pain. The nociceptor-specific loss of CB1 substantially reduced the analgesia produced by local and systemic, but not intrathecal, delivery of cannabinoids. We conclude that the contribution of CB1-type receptors expressed on the peripheral terminals of nociceptors to cannabinoid-induced analgesia is paramount, which should enable the development of peripherally acting CB1 analgesic agonists without any central side effects.


Assuntos
Analgesia , Canabinoides/farmacologia , Nociceptores/efeitos dos fármacos , Sistema Nervoso Periférico/efeitos dos fármacos , Receptor CB1 de Canabinoide/efeitos dos fármacos , Alelos , Animais , Moduladores de Receptores de Canabinoides/fisiologia , Sistema Nervoso Central/efeitos dos fármacos , Primers do DNA , Eletrofisiologia , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Nervosas Amielínicas/efeitos dos fármacos , Fibras Nervosas Amielínicas/fisiologia , Neurônios Aferentes/fisiologia , Doenças do Sistema Nervoso Periférico/patologia , Receptor CB1 de Canabinoide/genética
13.
Neuron ; 51(4): 455-66, 2006 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-16908411

RESUMO

Balanced control of neuronal activity is central in maintaining function and viability of neuronal circuits. The endocannabinoid system tightly controls neuronal excitability. Here, we show that endocannabinoids directly target hippocampal glutamatergic neurons to provide protection against acute epileptiform seizures in mice. Functional CB1 cannabinoid receptors are present on glutamatergic terminals of the hippocampal formation, colocalizing with vesicular glutamate transporter 1 (VGluT1). Conditional deletion of the CB1 gene either in cortical glutamatergic neurons or in forebrain GABAergic neurons, as well as virally induced deletion of the CB1 gene in the hippocampus, demonstrate that the presence of CB1 receptors in glutamatergic hippocampal neurons is both necessary and sufficient to provide substantial endogenous protection against kainic acid (KA)-induced seizures. The direct endocannabinoid-mediated control of hippocampal glutamatergic neurotransmission may constitute a promising therapeutic target for the treatment of disorders associated with excessive excitatory neuronal activity.


Assuntos
Moduladores de Receptores de Canabinoides/fisiologia , Endocanabinoides , Epilepsia/patologia , Epilepsia/fisiopatologia , Hipocampo/patologia , Rede Nervosa/patologia , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Benzoxazinas , Bloqueadores dos Canais de Cálcio/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Epilepsia/induzido quimicamente , Epilepsia/genética , Expressão Gênica/fisiologia , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Ácido Caínico/toxicidade , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Potenciais da Membrana/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfolinas/farmacologia , Naftalenos/farmacologia , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiopatologia , Células Piramidais/fisiologia , Células Piramidais/efeitos da radiação , Receptor CB1 de Canabinoide/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Ácido gama-Aminobutírico/genética
14.
Eur J Neurosci ; 32(5): 693-706, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21050275

RESUMO

A role for endocannabinoid signaling in neuronal morphogenesis as the brain develops has recently been suggested. Here we used the developing somatosensory circuit as a model system to examine the role of endocannabinoid signaling in neural circuit formation. We first show that a deficiency in cannabinoid receptor type 1 (CB(1)R), but not G-protein-coupled receptor 55 (GPR55), leads to aberrant fasciculation and pathfinding in both corticothalamic and thalamocortical axons despite normal target recognition. Next, we localized CB(1)R expression to developing corticothalamic projections and found little if any expression in thalamocortical axons, using a newly established reporter mouse expressing GFP in thalamocortical projections. A similar thalamocortical projection phenotype was observed following removal of CB(1)R from cortical principal neurons, clearly demonstrating that CB(1)R in corticothalamic axons was required to instruct their complimentary connections, thalamocortical axons. When reciprocal thalamic and cortical connections meet, CB(1)R-containing corticothalamic axons are intimately associated with elongating thalamocortical projections containing DGLß, a 2-arachidonoyl glycerol (2-AG) synthesizing enzyme. Thus, 2-AG produced in thalamocortical axons and acting at CB(1)Rs on corticothalamic axons is likely to modulate axonal patterning. The presence of monoglyceride lipase, a 2-AG degrading enzyme, in both thalamocortical and corticothalamic tracts probably serves to restrict 2-AG availability. In summary, our study provides strong evidence that endocannabinoids are a modulator for the proposed 'handshake' interactions between corticothalamic and thalamocortical axons, especially for fasciculation. These findings are important in understanding the long-term consequences of alterations in CB(1)R activity during development, a potential etiology for the mental health disorders linked to prenatal cannabis use.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Morfogênese/fisiologia , Vias Neurais/crescimento & desenvolvimento , Células Piramidais/crescimento & desenvolvimento , Receptor CB1 de Canabinoide/fisiologia , Tálamo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Moduladores de Receptores de Canabinoides/metabolismo , Moduladores de Receptores de Canabinoides/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese/genética , Vias Neurais/embriologia , Técnicas de Rastreamento Neuroanatômico/métodos , Células Piramidais/embriologia , Receptor CB1 de Canabinoide/genética , Receptores de Canabinoides/genética , Receptores de Canabinoides/fisiologia , Tálamo/embriologia
15.
PLoS Biol ; 5(10): e269, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17927447

RESUMO

Marijuana and its main psychotropic ingredient Delta(9)-tetrahydrocannabinol (THC) exert a plethora of psychoactive effects through the activation of the neuronal cannabinoid receptor type 1 (CB1), which is expressed by different neuronal subpopulations in the central nervous system. The exact neuroanatomical substrates underlying each effect of THC are, however, not known. We tested locomotor, hypothermic, analgesic, and cataleptic effects of THC in conditional knockout mouse lines, which lack the expression of CB1 in different neuronal subpopulations, including principal brain neurons, GABAergic neurons (those that release gamma aminobutyric acid), cortical glutamatergic neurons, and neurons expressing the dopamine receptor D1, respectively. Surprisingly, mice lacking CB1 in GABAergic neurons responded to THC similarly as wild-type littermates did, whereas deletion of the receptor in all principal neurons abolished or strongly reduced the behavioural and autonomic responses to the drug. Moreover, locomotor and hypothermic effects of THC depend on cortical glutamatergic neurons, whereas the deletion of CB1 from the majority of striatal neurons and a subpopulation of cortical glutamatergic neurons blocked the cataleptic effect of the drug. These data show that several important pharmacological actions of THC do not depend on functional expression of CB1 on GABAergic interneurons, but on other neuronal populations, and pave the way to a refined interpretation of the pharmacological effects of cannabinoids on neuronal functions.


Assuntos
Sistema Nervoso Autônomo/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Dronabinol/farmacologia , Psicotrópicos/farmacologia , Receptor CB1 de Canabinoide/efeitos dos fármacos , Animais , Temperatura Corporal/efeitos dos fármacos , Catalepsia/induzido quimicamente , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Ácido Glutâmico/metabolismo , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Interneurônios/patologia , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Neocórtex/efeitos dos fármacos , Neocórtex/patologia , Neocórtex/fisiopatologia , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Limiar da Dor/efeitos dos fármacos , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Ácido gama-Aminobutírico/metabolismo
16.
Neuropharmacology ; 54(1): 141-50, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17709120

RESUMO

Anandamide and 2-arachidonoyl glycerol, referred to as endocannabinoids (eCBs), are the endogenous agonists for the cannabinoid receptor type 1 (CB1). Several pieces of evidence support a role for eCBs in the attenuation of anxiety-related behaviours, although the precise mechanism has remained uncertain. The fatty acid amid hydrolase (FAAH), an enzyme responsible for the degradation of eCBs, has emerged as a promising target for anxiety-related disorders, since FAAH inhibitors are able to increase the levels of anandamide and thereby induce anxiolytic-like effects in rodents. The present study adopted both genetic and pharmacological approaches and tested the hypothesis that FAAH-deficient (FAAH(-/-)) mice as well as C57BL/6N mice treated with an FAAH inhibitor (URB597) would express reduced anxiety-like responses. Furthermore, as it is known that anandamide can bind several other targets than CB1 receptors, we investigated whether FAAH inhibition reduces anxiety via CB1 receptors. FAAH(-/-) mice showed reduced anxiety both in the elevated plus maze and in the light-dark test. These genotype-related differences were prevented by the CB1 receptor antagonist rimonabant (3mg/kg). Moreover, URB597 (1mg/kg) induced an anxiolytic-like effect in C57BL/6N mice exposed to the elevated plus maze, which was prevented by rimonabant (3mg/kg). The present work provides genetic and pharmacological evidence supporting the inhibition of FAAH as an important mechanism for the alleviation of anxiety. In addition, it indicates an increased activation of CB1 receptors as a mechanism underlying the effects of FAAH inhibition in two models of anxiety.


Assuntos
Amidoidrolases/metabolismo , Ansiedade/genética , Ácidos Araquidônicos/metabolismo , Glicerídeos/metabolismo , Alcamidas Poli-Insaturadas/metabolismo , Receptor CB1 de Canabinoide/fisiologia , Amidoidrolases/antagonistas & inibidores , Amidoidrolases/deficiência , Análise de Variância , Animais , Ansiedade/tratamento farmacológico , Comportamento Animal , Benzamidas/uso terapêutico , Carbamatos/uso terapêutico , Modelos Animais de Doenças , Endocanabinoides , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Piperidinas/uso terapêutico , Pirazóis/uso terapêutico , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto
17.
Neuroscience ; 384: 397-405, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29885522

RESUMO

Endocannabinoids acting through cannabinoid receptor type 1 (CB1) are major modulators of peripheral somatic and visceral nociception. Although only partially studied, some evidence suggests a particular role of CB1 within the brain in nociceptive processes. As the endocannabinoid system regulates affect and emotional behaviors, we hypothesized that cerebral CB1 influences affective processing of visceral pain-related behaviors in laboratory animals. To study nocifensive responses modulated by supraspinal CB1, we used conditional knock-out mice lacking CB1 either in cortical glutamatergic neurons (Glu-CB1-KO), or in forebrain GABAergic neurons (GABA-CB1-KO), or in principal neurons of the forebrain (CaMK-CB1-KO). These mutant mice and mice treated with the CB1 antagonist SR141716 were tested for different pain-related behaviors. In an acetic acid-induced abdominal constriction test, supraspinal CB1 deletions did not affect nocifensive responses. In the cerulein-model of acute pancreatitis, mechanical allodynia or hyperalgesia were not changed, but Glu-CB1- and CaMK-CB1-KO mice showed significantly increased facial grimacing scores indicating increased affective responses to this noxious visceral stimulus. Similarly, these brain-specific CB1 KO mice also showed significantly changed thermal nociception in a hot-plate test. These results reveal a novel, and important role of CB1 expressed by cortical glutamatergic neurons in the affective component of visceral nociception.


Assuntos
Afeto/fisiologia , Encéfalo/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Dor Visceral/metabolismo , Afeto/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Encéfalo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Receptor CB1 de Canabinoide/genética , Rimonabanto/farmacologia , Dor Visceral/psicologia
18.
Front Mol Neurosci ; 11: 230, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30026687

RESUMO

Defining functional domains and amino acid residues in G protein coupled receptors (GPCRs) represent an important way to improve rational drug design for this major class of drug targets. The cannabinoid type 1 (CB1) receptor is one of the most abundant GPCRs in the central nervous system and is involved in many physiological and pathophysiological processes. Interestingly, cannabinoid type 1 receptor with a phenylalanine 238 to leucine mutation (CB1F238L) has been already linked to a number of both in vitro and in vivo alterations. While CB1F238L causes significantly reduced presynaptic neurotransmitter release at the cellular level, behaviorally this mutation induces increased risk taking, social play behavior and reward sensitivity in rats. However, the molecular mechanisms underlying these changes are not fully understood. In this study, we tested whether the F238L mutation affects trafficking and axonal/presynaptic polarization of the CB1 receptor in vitro. Steady state or ligand modulated surface expression and lipid raft association was analyzed in human embryonic kidney 293 (HEK293) cells stably expressing either wild-type cannabinoid type 1 receptor (CB1wt) or CB1F238L receptor. Axonal/presynaptic polarization of the CB1F238L receptor was assessed in transfected primary hippocampal neurons. We show that in vitro the CB1F238L receptor displays increased association with lipid rafts, which coincides with increased lipid raft mediated constitutive endocytosis, leading to a reduction in steady state surface expression of the CB1F238L receptor. Furthermore, the CB1F238L receptor showed increased axonal polarization in primary hippocampal neurons. These data demonstrate that endocytosis of the CB1 receptor is an important mediator of axonal/presynaptic polarization and that phenylalanine 238 plays a key role in CB1 receptor trafficking and axonal polarization.

19.
Neuropsychopharmacology ; 43(5): 964-977, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28102227

RESUMO

The dorsal striatum is a key node for many neurobiological processes such as motor activity, cognitive functions, and affective processes. The proper functioning of striatal neurons relies critically on metabotropic receptors. Specifically, the main adenosine and endocannabinoid receptors present in the striatum, ie, adenosine A2A receptor (A2AR) and cannabinoid CB1 receptor (CB1R), are of pivotal importance in the control of neuronal excitability. Facilitatory and inhibitory functional interactions between striatal A2AR and CB1R have been reported, and evidence supports that this cross-talk may rely, at least in part, on the formation of A2AR-CB1R heteromeric complexes. However, the specific location and properties of these heteromers have remained largely unknown. Here, by using techniques that allowed a precise visualization of the heteromers in situ in combination with sophisticated genetically modified animal models, together with biochemical and pharmacological approaches, we provide a high-resolution expression map and a detailed functional characterization of A2AR-CB1R heteromers in the dorsal striatum. Specifically, our data unveil that the A2AR-CB1R heteromer (i) is essentially absent from corticostriatal projections and striatonigral neurons, and, instead, is largely present in striatopallidal neurons, (ii) displays a striking G protein-coupled signaling profile, where co-stimulation of both receptors leads to strongly reduced downstream signaling, and (iii) undergoes an unprecedented dysfunction in Huntington's disease, an archetypal disease that affects striatal neurons. Altogether, our findings may open a new conceptual framework to understand the role of coordinated adenosine-endocannabinoid signaling in the indirect striatal pathway, which may be relevant in motor function and neurodegenerative diseases.


Assuntos
Corpo Estriado/metabolismo , Estrutura Quaternária de Proteína , Receptor A2A de Adenosina/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais , Animais , Humanos , Doença de Huntington/metabolismo , Camundongos , Vias Neurais/metabolismo , Subunidades Proteicas/biossíntese
20.
J Neurosci ; 26(25): 6677-86, 2006 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-16793875

RESUMO

The interplay between fear expression and fear extinction provides an important prerequisite for adequate coping with aversive encounters. Current models propose that extinction of conditioned fear is mediated by associative safety learning. Here, we demonstrate that the cannabinoid CB1 receptor, which is crucially involved in fear extinction, is dispensable for associative safety learning. In fact, our results indicate that CB1 mediates fear extinction primarily via habituation-like processes. CB1 null-mutant mice were severely impaired not only in extinction of the fear response to a tone after fear conditioning but also in habituation of the fear response to a tone after sensitization with an inescapable footshock. Surprisingly, long-term habituation was generally affected even in situations with proper short-term adaptation, suggesting the existence of two separated CB1-dependent effector systems for short- and long-term fear adaptation. Our findings underscore the importance of habituation as a determinant of fear extinction in mice and characterize the cannabinoid CB1 receptor as an essential molecular correlate of this process.


Assuntos
Condicionamento Psicológico , Extinção Psicológica/fisiologia , Medo , Habituação Psicofisiológica/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Estimulação Acústica/métodos , Adaptação Fisiológica/genética , Animais , Comportamento Animal , Potenciais Evocados Auditivos/genética , Potenciais Evocados Auditivos/fisiologia , Habituação Psicofisiológica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor CB1 de Canabinoide/deficiência , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA