151
|
Borgkvist A, Marcellino D, Fuxe K, Greengard P, Fisone G. Regulation of DARPP-32 phosphorylation by Delta9-tetrahydrocannabinol. Neuropharmacology 2007; 54:31-5. [PMID: 17686497 DOI: 10.1016/j.neuropharm.2007.06.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Revised: 06/08/2007] [Accepted: 06/25/2007] [Indexed: 11/27/2022]
Abstract
CB1 receptor agonists increase the state of phosphorylation of the dopamine and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) at the cAMP-dependent protein kinase site, Thr 34. This effect, which occurs in the medium spiny neurons of the striatum, has been proposed to mediate the motor depressant action of cannabinoids. In this study, we have examined the effect produced by systemic administration of Delta(9)-tetrahydrocannabinol (THC), the major component of marihuana and hashish, on DARPP-32. We show that THC increases DARPP-32 phosphorylation at Thr 34 both in dorsal striatum and nucleus accumbens. Time-course and dose-response experiments indicate that DARPP-32 phosphorylation is maximal 30 min following administration of 10mg/kg of THC. The THC-mediated increase in DARPP-32 phosphorylation is reduced by administration of the CB1 receptor antagonist, SR141716A (3mg/kg). A similar attenuation of the effect of THC is also exerted by suppression of cAMP signaling achieved using the dopamine D1 receptor antagonist, SCH23390 (0.125 mg/kg), or the adenosine A2A receptor antagonist, KW6002 (3mg/kg). These results indicate that, in the striatum, THC promotes PKA-dependent phosphorylation of DARPP-32 in striatal medium spiny neurons expressing dopamine D1 and adenosine A2A receptors.
Collapse
Affiliation(s)
- Anders Borgkvist
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
152
|
Rubio M, McHugh D, Fernández-Ruiz J, Bradshaw H, Walker JM. Short-term exposure to alcohol in rats affects brain levels of anandamide, other N-acylethanolamines and 2-arachidonoyl-glycerol. Neurosci Lett 2007; 421:270-4. [PMID: 17574742 PMCID: PMC2966860 DOI: 10.1016/j.neulet.2007.05.052] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 05/19/2007] [Accepted: 05/23/2007] [Indexed: 01/08/2023]
Abstract
Chronic alcohol exposure leads to significant changes in the levels of endocannabinoids and their receptors in the brains of humans and laboratory animals, as well as in cultured neuronal cells. However, little is known about the effects of short-term periods of alcohol exposure. In the present study, we examined the changes in endocannabinoid levels (anandamide and 2-arachidonoylglycerol), as well as four additional N-acylethanolamines, in four brain regions of rats exposed to alcohol through the liquid diet for a period of 24h. The levels of N-acylethanolamines were diminished 24h after the onset of alcohol exposure. This was particularly evident for anandamide in the hypothalamus, amygdala and caudate-putamen, for N-palmitoylethanolamine in the caudate-putamen, for N-oleoylethanolamine in the hypothalamus, caudate-putamen and prefrontal cortex, and for N-stearoylethanolamine in the amygdala. The only exception was N-linoleoylethanolamine for which the levels increased in the amygdala after the exposure to alcohol. The levels of the other major endocannabinoid, 2-arachidonoylglycerol, were also reduced with marked effects in the prefrontal cortex. These results support the notion that short-term alcohol exposure reduces endocannabinoid levels in the brain accompanied by a reduction in several related N-acylethanolamines.
Collapse
Affiliation(s)
- Marina Rubio
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040-Madrid (Spain)
- Gill Center for Biomolecular Science and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN (USA)
| | - Douglas McHugh
- Gill Center for Biomolecular Science and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN (USA)
| | - Javier Fernández-Ruiz
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040-Madrid (Spain)
| | - Heather Bradshaw
- Gill Center for Biomolecular Science and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN (USA)
| | - J. Michael Walker
- Gill Center for Biomolecular Science and Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN (USA)
| |
Collapse
|
153
|
Sagredo O, García-Arencibia M, de Lago E, Finetti S, Decio A, Fernández-Ruiz J. Cannabinoids and neuroprotection in basal ganglia disorders. Mol Neurobiol 2007; 36:82-91. [PMID: 17952653 DOI: 10.1007/s12035-007-0004-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Accepted: 03/19/2007] [Indexed: 10/23/2022]
Abstract
Cannabinoids have been proposed as clinically promising neuroprotective molecules, as they are capable to reduce excitotoxicity, calcium influx, and oxidative injury. They are also able to decrease inflammation by acting on glial processes that regulate neuronal survival and to restore blood supply to injured area by reducing the vasoconstriction produced by several endothelium-derived factors. Through one or more of these processes, cannabinoids may provide neuroprotection in different neurodegenerative disorders including Parkinson's disease and Huntington's chorea, two chronic diseases that are originated as a consequence of the degeneration of specific nuclei of basal ganglia, resulting in a deterioration of the control of movement. Both diseases have been still scarcely explored at the clinical level for a possible application of cannabinoids to delay the progressive degeneration of the basal ganglia. However, the preclinical evidence seems to be solid and promising. There are two key mechanisms involved in the neuroprotection by cannabinoids in experimental models of these two disorders: first, a cannabinoid receptor-independent mechanism aimed at producing a decrease in the oxidative injury and second, an induction/upregulation of cannabinoid CB2 receptors, mainly in reactive microglia, that is capable to regulate the influence of these glial cells on neuronal homeostasis. Considering the relevance of these preclinical data and the lack of efficient neuroprotective strategies in both disorders, we urge the development of further studies that allow that the promising expectatives generated for these molecules progress from the present preclinical evidence till a real clinical application.
Collapse
Affiliation(s)
- Onintza Sagredo
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina III, Universidad Complutense, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
154
|
Ballesteros-Yáñez I, Valverde O, Ledent C, Maldonado R, DeFelipe J. Chronic cocaine treatment alters dendritic arborization in the adult motor cortex through a CB1 cannabinoid receptor–dependent mechanism. Neuroscience 2007; 146:1536-45. [PMID: 17467187 DOI: 10.1016/j.neuroscience.2007.03.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 02/18/2007] [Accepted: 03/04/2007] [Indexed: 11/30/2022]
Abstract
The CB1 cannabinoid receptors modulate the addictive processes associated with different drugs of abuse, including psychostimulants. Mice lacking CB1 receptors exhibit an important attenuation of the reinforcing responses produced by cocaine in an operant self-administration paradigm. We have investigated the effect of chronic cocaine treatment on dendrite structure and spine density of the principal cortical neuron, the pyramidal neuron, in CB1 knockout mice and wild type littermates. Layer III pyramidal cells of the motor cortex were injected intracellularly in fixed cortical slices and their morphometric parameters analyzed. Under basal conditions, the field area of the dendritic arbors was more extensive and dendritic spine density was higher in wild type mice than in CB1 knockout mice. Chronic treatment of cocaine diminished the size and length of the basal dendrites and spine density on pyramidal cells from wild type mice. However, the total number of spines in the pyramidal cells of CB1 knockout mice augmented slightly following chronic cocaine treatment, although no changes in the morphology of the dendritic arbor were observed. Our data demonstrate that microanatomy and synaptic connectivity are affected by cocaine, the magnitude and nature of these changes depend on the presence of CB1 receptors.
Collapse
|
155
|
Sarchielli P, Pini LA, Coppola F, Rossi C, Baldi A, Mancini ML, Calabresi P. Endocannabinoids in chronic migraine: CSF findings suggest a system failure. Neuropsychopharmacology 2007; 32:1384-90. [PMID: 17119542 DOI: 10.1038/sj.npp.1301246] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Based on experimental evidence of the antinociceptive action of endocannabinoids and their role in the modulation of trigeminovascular system activation, we hypothesized that the endocannabinoid system may be dysfunctional in chronic migraine (CM). We examined whether the concentrations of N-arachidonoylethanolamide (anandamide, AEA), palmitoylethanolamide (PEA), and 2-arachidonoylglycerol (2-AG) in the CSF of patients with CM and with probable CM and probable analgesic-overuse headache (PCM+PAOH) are altered compared with control subjects. The above endocannabinoids were measured by high-performance liquid chromatography (HPLC), and quantified by isotope dilution gas-chromatography/mass-spectrometry. Calcitonin gene-related peptide (CGRP) levels were also determined by RIA method and the end products of nitric oxide (NO), the nitrites, by HPLC. CSF concentrations of AEA were significantly lower and those of PEA slightly but significantly higher both in patients with CM and PCM+PAOH than in nonmigraineur controls (p<0.01 and p<0.02, respectively). A negative correlation was found between AEA and CGRP levels in CM and PCM+PAOH patients (r=0.59, p<0.01 and r=-0.65, p<0.007; respectively). A similar trend was observed between this endocannabinoid and nitrite levels. Reduced levels of AEA in the CSF of CM and PCM+PAOH patients may reflect an impairment of the endocannabinoid system in these patients, which may contribute to chronic head pain and seem to be related to increased CGRP and NO production. These findings support the potential role of the cannabinoid (CB)1 receptor as a possible therapeutic target in CM.
Collapse
Affiliation(s)
- Paola Sarchielli
- Neurologic Clinic, Department of Medical and Surgical Specialties and Public Health, University of Perugia, Perugia, Italy.
| | | | | | | | | | | | | |
Collapse
|
156
|
Nisoli E, Brunani A, Borgomainerio E, Tonello C, Dioni L, Briscini L, Redaelli G, Molinari E, Cavagnini F, Carruba MO. D2 dopamine receptor (DRD2) gene Taq1A polymorphism and the eating-related psychological traits in eating disorders (anorexia nervosa and bulimia) and obesity. Eat Weight Disord 2007; 12:91-6. [PMID: 17615493 DOI: 10.1007/bf03327583] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Food is considered a reinforcing agent, like a variety of substances such as alcohol and other drugs of abuse that produce pleasure. Psychopathological traits related to food intake are demonstrated in eating disorders as in obesity with different genetic aspects for these diseases. Recently, the prevalence of TaqA1 allele has been associated to alcohol, drug abuse and carbohydrate preference. For this reason, the aim of this study was to evaluate if the presence of A1 allele, in eating disorders and obesity, is associated with some particular psycho-pathological characteristics. METHODS We studied the presence of TaqA1 in Italian subjects affected by obesity (n=71), anorexia (n=28), bulimia (n=20) and in control group (n=54). The Eating Disorders Inventory (EDI test) was used to evaluate the psychological profiles. Patients without alcohol and drugs abuse were selected (>125 ml/day). RESULTS The A1+ allele, both in A1/A1 and A1/A2 genotypes, was not differently distributed among disease groups; on the contrary two EDI subscales (Drive for thinness and Ineffectiveness) resulted associated with A1+ allele without effect of the eating disease or obesity. CONCLUSION These results confirm that the presence of A1+ allele is not simply related to body weight but the A1+ allele might be a marker of a genetic psychological condition in people with high risk to develop pathological eating behaviour.
Collapse
Affiliation(s)
- E Nisoli
- Center for the Study and Research on Obesity, Department of Preclinical Sciences, Ospedale Sacco, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Bosier B, Tilleux S, Najimi M, Lambert DM, Hermans E. Agonist selective modulation of tyrosine hydroxylase expression by cannabinoid ligands in a murine neuroblastoma cell line. J Neurochem 2007; 102:1996-2007. [PMID: 17540007 DOI: 10.1111/j.1471-4159.2007.04679.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Functional interactions between catecholamines and cannabinoid transmission systems could explain the influence of Delta(9)-tetrahydrocannabinol on several central activities. Hence, the presence of cannabinoid CB(1) receptors in tyrosine hydroxylase (TH) containing cells has been suggested, providing clue for a direct control of catecholamines synthesis. In the present study, we evidenced the constitutive expression of functional cannabinoid CB(1) receptors in N1E-115 neuroblastoma and reported on the use of this model to examine the influence of diverse cannabinoid ligands on TH expression. Exposure of the cells to the high-affinity agonist HU 210 (5 h) resulted in a significant decrease in TH content (pEC(50): 6.40). In contrast, no change was observed after a similar treatment with the structurally unrelated agonist CP 55,940. Besides, the use of a luciferase reporter assay revealed that these two agonists showed opposite influences on TH gene promoter activity. Thus, in cells expressing pTH-luc constructs, inhibition and induction of luciferase activity were respectively observed with HU 210 (pEC(50): 8.95) and CP 55,940 (pEC(50): 9.09). Pharmacological characterisation revealed that these reciprocal responses were both related to the specific activation of cannabinoid CB(1) receptor, suggesting an agonist-dependent modulation of distinct signalling pathways. While these data points out the possible pharmacological manipulation of TH expression by cannabinoid ligands, such approach should take into account the existence of agonist selective trafficking of cannabinoid CB(1) receptor signalling.
Collapse
MESH Headings
- Animals
- CHO Cells
- Cannabinoid Receptor Modulators/metabolism
- Cannabinoids/pharmacology
- Catecholamines/biosynthesis
- Cell Line, Tumor
- Cricetinae
- Cricetulus
- Cyclohexanols/pharmacology
- Dronabinol/analogs & derivatives
- Dronabinol/pharmacology
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/genetics
- Ligands
- Mice
- Models, Biological
- Neuroblastoma
- Neurons/drug effects
- Neurons/enzymology
- Neuroprotective Agents/pharmacology
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- Protein Transport/drug effects
- Protein Transport/physiology
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Tyrosine 3-Monooxygenase/drug effects
- Tyrosine 3-Monooxygenase/genetics
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Barbara Bosier
- Unité de Chimie Pharmaceutique et de Radiopharmacie (UCL 7340), Université catholique de Louvain, Brussels, BelgiumLaboratoire de Pharmacologie expérimentale (UCL 5410), Université catholique de Louvain, Brussels, Belgium
| | - Sébastien Tilleux
- Unité de Chimie Pharmaceutique et de Radiopharmacie (UCL 7340), Université catholique de Louvain, Brussels, BelgiumLaboratoire de Pharmacologie expérimentale (UCL 5410), Université catholique de Louvain, Brussels, Belgium
| | - Mustapha Najimi
- Unité de Chimie Pharmaceutique et de Radiopharmacie (UCL 7340), Université catholique de Louvain, Brussels, BelgiumLaboratoire de Pharmacologie expérimentale (UCL 5410), Université catholique de Louvain, Brussels, Belgium
| | - Didier M Lambert
- Unité de Chimie Pharmaceutique et de Radiopharmacie (UCL 7340), Université catholique de Louvain, Brussels, BelgiumLaboratoire de Pharmacologie expérimentale (UCL 5410), Université catholique de Louvain, Brussels, Belgium
| | - Emmanuel Hermans
- Unité de Chimie Pharmaceutique et de Radiopharmacie (UCL 7340), Université catholique de Louvain, Brussels, BelgiumLaboratoire de Pharmacologie expérimentale (UCL 5410), Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
158
|
Abstract
PURPOSE OF REVIEW Endocannabinoids are defined as endogenous agonists of cannabinoid receptors, that is, of the two G-protein-coupled receptors for the Cannabis psychoactive principle Delta-tetra-hydrocannabinol. Two such endogenous mediators have been most thoroughly studied so far: anandamide and 2-arachidonoylglycerol. Here we review the mechanisms for the regulation of their levels under physiological and pathological conditions, and recent findings on their role in disease. RECENT FINDINGS It is becoming increasingly clear that, although both anandamide and 2-arachidonoyl-glycerol are produced and degraded 'on demand', the levels of these two compounds appear to be regulated in different, and sometimes even opposing, ways, often using redundant molecular mechanisms. Alterations of endocannabinoid levels have been found in both animal models of pain, neurological and neurodegenerative states, gastrointestinal disorders and inflammatory conditions, and in blood, cerebrospinal fluid and bioptic samples from patients with various diseases. SUMMARY Endocannabinoid levels appear to be transiently elevated as an adaptive reaction to re-establish normal homeostasis when this is acutely and pathologically perturbed. In some chronic conditions, however, this system also contributes to the progress or symptoms of the disorder. As a consequence, new therapeutic drugs are being designed from both stimulants and blockers of endocannabinoid action.
Collapse
Affiliation(s)
- Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.
| | | |
Collapse
|
159
|
Pandolfo P, Pamplona FA, Prediger RDS, Takahashi RN. Increased sensitivity of adolescent spontaneously hypertensive rats, an animal model of attention deficit hyperactivity disorder, to the locomotor stimulation induced by the cannabinoid receptor agonist WIN 55,212-2. Eur J Pharmacol 2007; 563:141-8. [PMID: 17374533 DOI: 10.1016/j.ejphar.2007.02.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Revised: 02/05/2007] [Accepted: 02/06/2007] [Indexed: 11/25/2022]
Abstract
Converging evidence points to adolescence as a critical period for the onset of a wide range of neuropsychiatric disorders, including attention deficit hyperactivity disorder (ADHD) and drug abuse. Spontaneously hypertensive rats (SHR) are generally considered to be a suitable genetic model for the study of ADHD, since they display hyperactivity, impulsivity, poorly sustained attention, cognitive deficits and increased novelty seeking. Despite the high prevalence of ADHD among adolescents, studies using SHR have mainly been performed on adult animals. The aim of the present study was to evaluate the effect of acute intraperitoneal (i.p.) administration of the cannabinoid receptor agonist WIN 55,212-2 (0.25-2.5 mg/kg) on locomotor activity and anxiety-like behavior in male adolescent and adult SHR and Wistar rats using the open field and elevated plus-maze tests. WIN 55,212-2 at doses of 0.25 and 1.25 mg/kg (i.p.) selectively promoted locomotor stimulation in adolescent SHR in the open field, but not in adult SHR or Wistar rats (regardless of age). The effect of WIN 55,212-2 (0.25 mg/kg, i.p.) on locomotion of adolescent SHR was reversed by pretreatment with the selective cannabinoid CB1 receptor antagonist AM 251 (0.25 mg/kg, i.p.). Moreover, although the present doses of WIN 55,212-2 had no effect on anxiety-related behaviors in any of the animal groups evaluated in the open field (central locomotion) or elevated plus-maze (time and entries in open arms), the highest dose of WIN 55,212-2 tested (2.5 mg/kg, i.p.) significantly decreased the number of closed-arm entries (an index of locomotor activity) of adolescent rats of both the Wistar and SHR strains in the elevated plus-maze. The present results indicate strain- and age-related effects of cannabinoids on locomotor activity in rats, extending the notion that adolescence and ADHD represent risk factors for the increased sensitivity to the effects of drugs.
Collapse
Affiliation(s)
- Pablo Pandolfo
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, UFSC, Florianópolis-SC 88049-900, Brazil
| | | | | | | |
Collapse
|
160
|
Marco EM, Granstrem O, Moreno E, Llorente R, Adriani W, Laviola G, Viveros MP. Subchronic nicotine exposure in adolescence induces long-term effects on hippocampal and striatal cannabinoid-CB1 and mu-opioid receptors in rats. Eur J Pharmacol 2007; 557:37-43. [PMID: 17174300 DOI: 10.1016/j.ejphar.2006.11.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 10/31/2006] [Accepted: 11/06/2006] [Indexed: 11/21/2022]
Abstract
There is evidence for the existence of functional interactions between nicotine and cannabinoids and opioid compounds in adult experimental animals. However, there is scarce information about these relationships in young animals. In the present study we evaluated short and long-term effects of a subchronic nicotine treatment [0.4 mg/kg daily i.p. injections from postnatal day (PND) 34 to PND 43], upon hippocampal and striatal cannabinoid-CB(1) and mu-opioid receptors in Wistar rats of both genders. Rats were sacrificed 2 h after the last nicotine injection (short-term effects, PND 43) or one month later (long-term effects, PND 75). Hippocampal and striatal cannabinoid CB(1) and mu-opioid receptors were quantified by Western blotting. The subchronic nicotine treatment induced a region-dependent long-lasting effect in cannabinoid CB(1) receptor: a significant increase in hippocampal cannabinoid CB(1) receptors and a significant decrease in striatal cannabinoid CB(1) receptors, with these effects being similar in males and females. With respect to mu-opioid receptors, subchronic nicotine induced a significant down-regulation in hippocampal and striatal mu-opioid receptors in the long-term, and within the striatum the effects were more marked in adult males than in females. The present results indicate that juvenile nicotine taking may have implications for the endocannabinoid and endogenous opioid function and for the behaviors served by those systems, this includes possible modification of the response of adults to different psychotropic drugs, i.e. cannabis and morphine/heroin when taken later in life.
Collapse
Affiliation(s)
- Eva M Marco
- Dept. Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
161
|
de Lago E, Ortega-Gutiérrez S, Ramos JA, López Rodríguez ML, Fernández-Ruiz J. Neurochemical effects of the endocannabinoid uptake inhibitor UCM707 in various rat brain regions. Life Sci 2006; 80:979-88. [PMID: 17173937 DOI: 10.1016/j.lfs.2006.11.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2006] [Revised: 10/17/2006] [Accepted: 11/19/2006] [Indexed: 11/26/2022]
Abstract
To date, UCM707, (5Z,8Z,11Z,14Z)-N-(3-furylmethyl)eicosa-5,8,11,14-tetraenamide, has the highest potency and selectivity in vitro and in vivo as inhibitor of the endocannabinoid uptake. Its biochemical, pharmacological and therapeutic properties have been intensely studied recently, but the information on its capability to modify neurotransmitter activity, which obviously underlies the above properties, is still limited. In the present study, we conducted a time-course experiment in rats aimed at examining the neurochemical effects of UCM707 in several brain regions following a subchronic administration (5 injections during 2.5 days) of this inhibitor in a dose of 5 mg/kg weight. In the hypothalamus, the administration of UCM707 did not modify GABA contents but reduced norepinephrine levels at 5 h after administration, followed by an increase at 12 h. Similar trends were observed for dopamine, whereas serotonin content remained elevated at 1 and, in particular, 5 and 12 h after administration. In the case of the basal ganglia, UCM707 reduced GABA content in the substantia nigra but only at longer (5 or 12 h) times after administration. There were no changes in serotonin content, but a marked reduction in its metabolite 5HIAA was recorded in the substantia nigra. The same pattern was found for dopamine, contents of which were not altered by UCM707 in the caudate-putamen, but its major metabolite DOPAC exhibited a marked decrease at 5 h. In the cerebellum, UCM707 reduced GABA, serotonin and norepinephrine content, but only the reduction found for norepinephrine at 5 h reached statistical significance. The administration of UCM707 did not modify the contents of these neurotransmitters in the hippocampus and the frontal cortex. Lastly, in the case of limbic structures, the administration of UCM707 markedly reduced dopamine content in the nucleus accumbens at 5 h, whereas GABA content remained unchanged in this structure and also in the ventral-tegmental area and the amygdala. By contrast, norepinephrine and serotonin content increased at 5 h in the nucleus accumbens, but not in the other two limbic structures. In summary, UCM707 administered subchronically modified the contents of serotonin, GABA, dopamine and/or norepinephrine with a pattern strongly different in each brain region. So, changes in GABA transmission (decrease) were restricted to the substantia nigra, but did not appear in other regions, whereas dopamine transmission was also altered in the caudate-putamen and the nucleus accumbens. By contrast, norepinephrine and serotonin were altered by UCM707 in the hypothalamus, cerebellum (only norepinephrine), and nucleus accumbens, exhibiting biphasic effects in some cases.
Collapse
Affiliation(s)
- Eva de Lago
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
| | | | | | | | | |
Collapse
|
162
|
Changes of dopamine transporter function in striatum during acute morphine addiction and its abstinence in rhesus monkey. Chin Med J (Engl) 2006. [DOI: 10.1097/00029330-200611010-00007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
163
|
Forget B, Barthélémy S, Saurini F, Hamon M, Thiébot MH. Differential involvement of the endocannabinoid system in short- and long-term expression of incentive learning supported by nicotine in rats. Psychopharmacology (Berl) 2006; 189:59-69. [PMID: 16969683 DOI: 10.1007/s00213-006-0525-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Accepted: 07/19/2006] [Indexed: 10/24/2022]
Abstract
RATIONALE We previously reported that the CB1 cannabinoid receptor antagonist, rimonabant, impaired the acquisition and the short-term (24 h), but not long-term (3 weeks), expression of conditioned place preference (CPP) induced by nicotine in rats. OBJECTIVE To assess the time interval of efficacy of a single pretest injection of rimonabant to abolish nicotine-CPP, and the effects of chronic CB1 receptor blockade on long-term expression of nicotine-CPP. MATERIALS AND METHODS Wistar rats were conditioned to nicotine (0.06 mg/kg, subcutaneous) using an unbiased one-compartment procedure. Two test sessions were conducted 24 h (without injection) and 1, 2, or 3 weeks later. Rimonabant (3 mg/kg, intraperitoneal) or vehicle was administered daily between the two test sessions. In addition, the CB1-stimulated [(35)S]GTP-gamma-S binding was assessed in rats from the 3-week experiment. RESULTS The capacity of a single injection of rimonabant (3 mg/kg, 30 min pretest) to block the expression of nicotine-CPP disappeared within 1 week after conditioning. Daily administrations of rimonabant for 6, 13, or 20 days post-acquisition did not impair nicotine-CPP but allowed an additional pretest injection of rimonabant to retain its capacity to abolish long-term expression of nicotine-CPP. The CB1 receptor-mediated G-protein signaling was not altered in various brain areas of rats given rimonabant for 3 weeks. CONCLUSIONS The endocannabinoid system is essential to the expression of nicotine-CPP during less than 1 week after conditioning but not later. However, endocannabinoid-dependent mechanisms are critically involved in the development of the neuroadaptive changes responsible for the shift from CB1-dependent to CB1-independent expression of nicotine incentive learning.
Collapse
|
164
|
Degroot A, Köfalvi A, Wade MR, Davis RJ, Rodrigues RJ, Rebola N, Cunha RA, Nomikos GG. CB1 receptor antagonism increases hippocampal acetylcholine release: site and mechanism of action. Mol Pharmacol 2006; 70:1236-45. [PMID: 16855179 DOI: 10.1124/mol.106.024661] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Evidence indicates that blockade of cannabinoid receptors increases acetylcholine (ACh) release in brain cortical regions. Although it is assumed that this type of effect is mediated through CB1 receptor (CB1R) antagonism, several in vitro functional studies recently have suggested non-CB1R involvement. In addition, neither the precise neuroanatomical site nor the exact mechanisms underlying this effect are known. We thoroughly examined these issues using a combination of systemic and local administration of CB1R antagonists, different methods of in vivo microdialysis, CB1R knockout (KO) mice, tissue measurements of ACh, and immunochemistry. First, we showed that systemic injections of the CB1R antagonists N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboximide hydrochloride (SR-141716A) and N-(piperidin-1-yl)-5-(4-iodophenyl)-1-(2, 4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (AM251) dose-dependently increased hippocampal ACh efflux. Likewise, local hippocampal, but not septal, infusions of SR141716A or AM251 increased hippocampal ACh release. It is noteworthy that the stimulatory effects of systemically administered CB1R antagonists on hippocampal ACh release were completely abolished in CB1R KO mice. CB1R KO mice had similar basal but higher stress-enhanced hippocampal ACh levels compared with wild-type controls. It is interesting that dopamine D1 receptor antagonism counteracted the stimulatory effect of CB1R blockade on hippocampal ACh levels. Finally, immunohistochemical methods revealed that a high proportion of CB1R-positive nerve terminals were found in hippocampus and confirmed the colocalization of CB1 receptors with cholinergic and dopaminergic nerve terminals. In conclusion, hippocampal ACh release may specifically be controlled through CB1Rs located on both cholinergic and dopaminergic neuronal projections, and CB1R antagonism increases hippocampal ACh release, probably through both a direct disinhibition of ACh release and an indirect increase in dopaminergic neurotransmission at the D1 receptors.
Collapse
MESH Headings
- Acetylcholine/metabolism
- Animals
- Dopamine Plasma Membrane Transport Proteins/metabolism
- Dose-Response Relationship, Drug
- Hippocampus/drug effects
- Hippocampus/metabolism
- Immunoassay/methods
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microdialysis/methods
- Models, Biological
- Neurons/physiology
- Piperidines/administration & dosage
- Pyrazoles/administration & dosage
- Rats
- Rats, Wistar
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/genetics
- Receptors, Dopamine D1/antagonists & inhibitors
- Rimonabant
- Vesicular Acetylcholine Transport Proteins/metabolism
Collapse
Affiliation(s)
- Aldemar Degroot
- Eli Lilly and Company, Lilly Corporate Center, Neuroscience Discovery Research, Indianapolis, IN, USA
| | | | | | | | | | | | | | | |
Collapse
|
165
|
Abstract
Changes in synaptic efficacy are thought to be crucial to experience-dependent modifications of neural function. The diversity of mechanisms underlying these changes is far greater than previously expected. In the last five years, a new class of use-dependent synaptic plasticity that requires retrograde signaling by endocannabinoids (eCB) and presynaptic CB1 receptor activation has been identified in several brain structures. eCB-mediated plasticity encompasses many forms of transient and long-lasting synaptic depression and is found at both excitatory and inhibitory synapses. In addition, eCBs can modify the inducibility of non-eCB-mediated forms of plasticity. Thus, the eCB system is emerging as a major player in synaptic plasticity. Given the wide distribution of CB1 receptors in the CNS, the list of brain structures and synapses expressing eCB-mediated plasticity is likely to expand.
Collapse
Affiliation(s)
- Vivien Chevaleyre
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | |
Collapse
|
166
|
Abstract
The recent identification of cannabinoid receptors and their endogenous lipid ligands has triggered an exponential growth of studies exploring the endocannabinoid system and its regulatory functions in health and disease. Such studies have been greatly facilitated by the introduction of selective cannabinoid receptor antagonists and inhibitors of endocannabinoid metabolism and transport, as well as mice deficient in cannabinoid receptors or the endocannabinoid-degrading enzyme fatty acid amidohydrolase. In the past decade, the endocannabinoid system has been implicated in a growing number of physiological functions, both in the central and peripheral nervous systems and in peripheral organs. More importantly, modulating the activity of the endocannabinoid system turned out to hold therapeutic promise in a wide range of disparate diseases and pathological conditions, ranging from mood and anxiety disorders, movement disorders such as Parkinson's and Huntington's disease, neuropathic pain, multiple sclerosis and spinal cord injury, to cancer, atherosclerosis, myocardial infarction, stroke, hypertension, glaucoma, obesity/metabolic syndrome, and osteoporosis, to name just a few. An impediment to the development of cannabinoid medications has been the socially unacceptable psychoactive properties of plant-derived or synthetic agonists, mediated by CB(1) receptors. However, this problem does not arise when the therapeutic aim is achieved by treatment with a CB(1) receptor antagonist, such as in obesity, and may also be absent when the action of endocannabinoids is enhanced indirectly through blocking their metabolism or transport. The use of selective CB(2) receptor agonists, which lack psychoactive properties, could represent another promising avenue for certain conditions. The abuse potential of plant-derived cannabinoids may also be limited through the use of preparations with controlled composition and the careful selection of dose and route of administration. The growing number of preclinical studies and clinical trials with compounds that modulate the endocannabinoid system will probably result in novel therapeutic approaches in a number of diseases for which current treatments do not fully address the patients' need. Here, we provide a comprehensive overview on the current state of knowledge of the endocannabinoid system as a target of pharmacotherapy.
Collapse
Affiliation(s)
- Pál Pacher
- Laboratory of Physiological Studies, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2S-24, Bethesda, MD 20892-9413, USA
| | | | | |
Collapse
|
167
|
Mendizábal V, Zimmer A, Maldonado R. Involvement of kappa/dynorphin system in WIN 55,212-2 self-administration in mice. Neuropsychopharmacology 2006; 31:1957-66. [PMID: 16292318 DOI: 10.1038/sj.npp.1300957] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Self-administration procedures have not yet provided evidence that freely moving mice can reliably acquire and maintain an operant behavior to self-administer cannabinoid agonists. The aim of the present work was to establish a model of cannabinoid operant intravenous self-administration in freely moving mice given the relevance of this species for the use of genetically modified animals. In addition, the possible involvement of the kappa/dynorphin system in cannabinoid self-administration was evaluated by using pro-dynorphin knockout mice. Outbred CD1 wild-type mice as well as pro-dynorphin knockout and wild-type mice were trained to self-administer the cannabinoid receptor agonist WIN 55,212-2 under an FR1 schedule of reinforcement. Two cannabinoid training doses (6.25 and 12.5 microg/kg/infusion) were used in the acquisition studies in outbred mice. Animals acquired a reliable operant responding to self-administer WIN 55,212-2 (12.5 microg/kg/infusion), but required as many as 15 sessions to attain this behavior. Interestingly, when a previous injection of WIN 55,212-2 (0.1 mg/kg, i.p.) was administered in the home-cage 24 h before the first session, mice acquired operant responding for cannabinoid self-administration by the fourth session. When the kappa-opioid agonist antagonist nor-binaltorphimine (5 mg/kg s.c.) was administered 4 h before the first session, the time required to acquire a reliable cannabinoid self-administration was also significantly reduced. Finally, a shift to the left in the dose-intake curve to self-administer WIN 55,212-2 was observed in pro-dynorphin knockout mice when compared to wild-type mice. These results indicate that the activation of the kappa/dynorphin opioid system after WIN 55,212-2 administration could counteract cannabinoid rewarding effects.
Collapse
Affiliation(s)
- Victoria Mendizábal
- Laboratori de Neurofarmacologia, Facultat de Ciències de la Salut i de la Vida, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | |
Collapse
|
168
|
Marco EM, Llorente R, Moreno E, Biscaia JM, Guaza C, Viveros MP. Adolescent exposure to nicotine modifies acute functional responses to cannabinoid agonists in rats. Behav Brain Res 2006; 172:46-53. [PMID: 16730079 DOI: 10.1016/j.bbr.2006.04.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Revised: 04/12/2006] [Accepted: 04/13/2006] [Indexed: 11/20/2022]
Abstract
We have studied functional interactions between nicotine and the cannabinoid receptor agonist CP 55,940 (CP) in the modulation of behavioural and corticosterone responses of male and female adolescent Wistar rats. The animals underwent a subchronic nicotine treatment (0.4 mg/kg i.p., once daily) during the periadolescent period (postnatal days 34-43). Twenty-four hours after the last injection of nicotine an acute dose of CP (1 or 100 microg/kg i.p.) was administered. Thirty minutes after the cannabinoid injection, the animals were tested individually in the holeboard immediately followed by the elevated plus-maze. We also measured corticosterone levels by radioimmunoassay. In males, neither CP (1 microg/kg) nor nicotine induced any modification in anxiety when administered alone. However, the combination of the two drugs resulted in a significant anxiogenic-like effect. In females, the lower dose of CP was anxiogenic and nicotine, which did not induce any effect per se, prevented this response. In the holeboard, subchronic nicotine and the acute cannabinoid treatment interacted in the modulation of horizontal activity and the nature of this interaction also showed a clear sexual dimorphism. Both, the cannabinoid agonist and nicotine increased corticosterone concentrations and the animals receiving the two drugs showed higher levels than the animals receiving the cannabinoid alone. The data provide evidence for the existence of functional interactions between nicotine and cannabinoids in the modulation of behavioural responses and adrenocortical activity in adolescent rats.
Collapse
Affiliation(s)
- E M Marco
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense, Ciudad Universitaria, C/Jose Antonio Novais 2, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
169
|
Abstract
Mammalian reproduction is a complicated process designed to diversify and strengthen the genetic complement of the offspring and to safeguard regulatory systems at various steps for propagating procreation. An emerging concept in mammalian reproduction is the role of endocannabinoids, a group of endogenously produced lipid mediators, that bind to and activate cannabinoid receptors. Although adverse effects of cannabinoids on fertility have been implicated for years, the mechanisms by which they exert these effects were not clearly understood. With the identification of cannabinoid receptors, endocannabinoid ligands, their key synthetic and hydrolytic pathways, and the generation of mouse models missing cannabinoid receptors, a wealth of information on the significance of cannabinoid/endocannabinoid signaling in spermatogenesis, fertilization, preimplantation embryo development, implantation, and postimplantation embryonic growth has been generated. This review focuses on various aspects of the endocannabinoid system in male and female fertility. It is hoped that a deeper insight would lead to potential clinical applications of the endocannabinoid signaling as a target for correcting infertility and improving reproductive health in humans.
Collapse
Affiliation(s)
- Haibin Wang
- Department of Pediatrics, Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
170
|
Murillo-Rodríguez E, Millán-Aldaco D, Palomero-Rivero M, Mechoulam R, Drucker-Colín R. Cannabidiol, a constituent ofCannabis sativa, modulates sleep in rats. FEBS Lett 2006; 580:4337-45. [PMID: 16844117 DOI: 10.1016/j.febslet.2006.04.102] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 04/10/2006] [Accepted: 04/17/2006] [Indexed: 11/23/2022]
Abstract
Delta(9)-tetrahydrocannabinol (Delta(9)-THC) and cannabidiol (CBD) are two major constituents of Cannabis sativa. Delta(9)-THC modulates sleep, but no clear evidence on the role of CBD is available. In order to determine the effects of CBD on sleep, it was administered intracerebroventricular (icv) in a dose of 10 microg/5 microl at the beginning of either the lights-on or the lights-off period. We found that CBD administered during the lights-on period increased wakefulness (W) and decreased rapid eye movement sleep (REMS). No changes on sleep were observed during the dark phase. Icv injections of CBD (10 microg/5microl) induced an enhancement of c-Fos expression in waking-related brain areas such as hypothalamus and dorsal raphe nucleus (DRD). Microdialysis in unanesthetized rats was carried out to characterize the effects of icv administration of CBD (10 microg/5 microl) on extracellular levels of dopamine (DA) within the nucleus accumbens. CBD induced an increase in DA release. Finally, in order to test if the waking properties of CBD could be blocked by the sleep-inducing endocannabinoid anandamide (ANA), animals received ANA (10 microg/2.5 microl, icv) followed 15 min later by CBD (10 microg/2.5 microl). Results showed that the waking properties of CBD were not blocked by ANA. In conclusion, we found that CBD modulates waking via activation of neurons in the hypothalamus and DRD. Both regions are apparently involved in the generation of alertness. Also, CBD increases DA levels as measured by microdialysis and HPLC procedures. Since CBD induces alertness, it might be of therapeutic value in sleep disorders such as excessive somnolence.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Depto de Neurociencias, Instituto de Fisiología Celular, Ciudad Universitaria, Circuito Interior, Universidad Nacional Autónoma de México, México DF, CP 04510, Mexico.
| | | | | | | | | |
Collapse
|
171
|
Antel J, Gregory PC, Nordheim U. CB1 Cannabinoid Receptor Antagonists for Treatment of Obesity and Prevention of Comorbid Metabolic Disorders. J Med Chem 2006; 49:4008-16. [PMID: 16821760 DOI: 10.1021/jm058238r] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jochen Antel
- Solvay Pharmaceuticals Research Laboratories, Hans-Boeckler-Allee 20, D-30173 Hannover, Germany.
| | | | | |
Collapse
|
172
|
Gerald TM, Ward GR, Howlett AC, Franklin SO. CB1 knockout mice display significant changes in striatal opioid peptide and D4 dopamine receptor gene expression. Brain Res 2006; 1093:20-4. [PMID: 16684513 DOI: 10.1016/j.brainres.2006.03.088] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Revised: 03/14/2006] [Accepted: 03/22/2006] [Indexed: 11/17/2022]
Abstract
Antagonism of the CB(1) cannabinoid receptor (CB(1) receptor) by rimonabant (SR141716) reduces self-administration of alcohol and other drugs of abuse in animal models. These findings suggest that the CB(1) receptor may be a target for genetic differences that modify the salient features of rewarding drugs. In the present study, wild-type (CB(1) (+/+)) are compared to transgenic mice deficient in CB(1) receptors (CB(1) (-/-)). The goal was to investigate the influences of the cannabinoid receptor system on opioid peptide gene expression and on dopamine receptor gene expression which is commonly influenced by substances of abuse. We demonstrate using reverse transcription and real-time polymerase chain reaction (PCR) that striatal mRNA for preproenkephalin (PPENK) and preprodynorphin (PPDYN) in the CB(1) (-/-) striatum increases when compared to CB(1) (+/+). Real-time PCR analyses to evaluate D(2) and D(4) dopamine receptor gene expression in striatum isolated from CB(1) (+/+) and CB(1) (-/-) revealed a nearly 2-fold increase in D(4) receptor mRNA in the striatum from CB(1) (-/-) mice and no significant change in D(2) expression. In contrast, treatment of C57BL/6 mice with the CB(1) receptor antagonist, rimonabant, produced a reduction of both D(2) and D(4) dopamine receptor expression in the striatum. These data suggest that genetic differences in CB(1) receptor may exert a modulatory effect on D(4) dopamine receptor and opioid peptide gene expression.
Collapse
MESH Headings
- Animals
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Dynorphins/metabolism
- Enkephalins/metabolism
- Gene Expression/drug effects
- Mice
- Mice, Knockout
- Opioid Peptides/metabolism
- Piperidines/pharmacology
- Protein Precursors/metabolism
- Pyrazoles/pharmacology
- RNA, Messenger/analysis
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, Dopamine D2/metabolism
- Receptors, Dopamine D4/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Rimonabant
Collapse
Affiliation(s)
- Tonya M Gerald
- Neuroscience of Drug Abuse Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | | | | | | |
Collapse
|
173
|
O’Connor J, Muly EC, Hemby SE. Molecular mapping of striatal subdivisions in juvenile Macaca Mulata. Exp Neurol 2006; 198:326-37. [PMID: 16455077 PMCID: PMC5076375 DOI: 10.1016/j.expneurol.2005.11.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 11/23/2005] [Accepted: 11/23/2005] [Indexed: 11/21/2022]
Abstract
The striatum of the primate brain can be subdivided into three distinct anatomical subregions: caudate (CAU), putamen (PUT), and ventral striatum (VS). Although these subregions share several anatomical connections, cell morphological, and histochemical features, they differ considerably in their vulnerability to different neurological and psychiatric diseases, and these brain regions have significantly different functions in health and disease. In order to better understand the molecular underpinnings of the different disease and functional vulnerabilities, transcriptional profiles were generated from the CAU, PUT, and VS of five juvenile rhesus macaques (Macaca mulatta) using human cDNA neuromicroarrays containing triplicate spots of 1227 cDNAs. Differences in microarray gene expression were assessed using z score analysis and 1.5-fold change between paired subregions. Clustering of genes based on dissimilarity of expression patterns between regions revealed subregion specific expression profiles encoding G-protein-coupled receptor signaling transcripts, transcription factors, kinases and phosphatases, and cell signaling and signal transduction transcripts. Twelve transcripts were examined using quantitative real-time PCR (qPCR), and 81% demonstrated alterations similar to those seen with microarray analysis, some of which were statistically significant. Subregion specific transcription profiles support the anatomical differentiation and potential disease vulnerabilities of the respective subregions.
Collapse
Affiliation(s)
- Joann O’Connor
- Molecular and Systems Pharmacology Program, Graduate Division of Biological and Behavioral Sciences, Emory University, Atlanta, GA 30322, USA
| | - Emil C. Muly
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, USA
- Yerkes National Primate Research Center, Division of Neuroscience, Emory University, Atlanta, GA 30329, USA
| | - Scott E. Hemby
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
174
|
Chakrabarti B, Kent L, Suckling J, Bullmore E, Baron-Cohen S. Variations in the human cannabinoid receptor (CNR1) gene modulate striatal responses to happy faces. Eur J Neurosci 2006; 23:1944-8. [PMID: 16623851 DOI: 10.1111/j.1460-9568.2006.04697.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Happy facial expressions are innate social rewards and evoke a response in the striatum, a region known for its role in reward processing in rats, primates and humans. The cannabinoid receptor 1 (CNR1) is the best-characterized molecule of the endocannabinoid system, involved in processing rewards. We hypothesized that genetic variation in human CNR1 gene would predict differences in the striatal response to happy faces. In a 3T functional magnetic resonance imaging (fMRI) scanning study on 19 Caucasian volunteers, we report that four single nucleotide polymorphisms (SNPs) in the CNR1 locus modulate differential striatal response to happy but not to disgust faces. This suggests a role for the variations of the CNR1 gene in underlying social reward responsivity. Future studies should aim to replicate this finding with a balanced design in a larger sample, but these preliminary results suggest neural responsivity to emotional and socially rewarding stimuli varies as a function of CNR1 genotype. This has implications for medical conditions involving hypo-responsivity to emotional and social stimuli, such as autism.
Collapse
Affiliation(s)
- Bhismadev Chakrabarti
- Autism Research Centre, Douglas House, 18 B, Trumpington Road, Cambridge CB2 2AH, UK.
| | | | | | | | | |
Collapse
|
175
|
Pickel VM, Chan J, Kearn CS, Mackie K. Targeting dopamine D2 and cannabinoid-1 (CB1) receptors in rat nucleus accumbens. J Comp Neurol 2006; 495:299-313. [PMID: 16440297 PMCID: PMC1698281 DOI: 10.1002/cne.20881] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The nucleus accumbens (Acb) shell and core are essential components of neural circuitry mediating the reward and motor effects produced by activation of dopamine D2 or cannabinoid-1 (CB1) receptors. D2 receptors can form heterodimeric complexes with cannabinoid-1 (CB1) receptors and are also involved in control of the availability of both dopamine and endocannabinoids. Thus, the subcellular locations of D2 and CB1 receptors with respect to each other are implicit to their physiological actions in the Acb. We used electron microscopic immunocytochemistry to determine these locations in the Acb shell and core of rat brain. In each region, many neuronal profiles showed endomembrane and plasmalemmal distributions of one or both receptors. Approximately one-third of the labeled profiles were somata and dendrites, some of which showed overlapping subcellular distributions of D2 and CB1 immunoreactivities. The remaining labeled profiles were small axons and axon terminals containing CB1 and/or D2 receptors. Of the labeled terminals forming recognizable synapses, approximately 20% of those containing CB1 receptors contacted D2-labeled dendrites, while conversely, almost 15% of those containing D2 receptors contacted CB1-labeled dendrites. These results provide the first ultrastructural evidence that D2 and CB1 receptors in the Acb shell and core have subcellular distributions supporting both intracellular associations and local involvement of D2 receptors in making available endocannabinoids that are active on CB1 receptors in synaptic neurons. These distributions have direct relevance to the rewarding and euphoric as well as motor effects produced by marijuana and by addictive drugs enhancing dopamine levels in the Acb.
Collapse
Affiliation(s)
- Virgina M Pickel
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | | | | | |
Collapse
|
176
|
Köfalvi A, Rodrigues RJ, Ledent C, Mackie K, Vizi ES, Cunha RA, Sperlágh B. Involvement of cannabinoid receptors in the regulation of neurotransmitter release in the rodent striatum: a combined immunochemical and pharmacological analysis. J Neurosci 2006; 25:2874-84. [PMID: 15772347 PMCID: PMC6725145 DOI: 10.1523/jneurosci.4232-04.2005] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Despite the profound effect of cannabinoids on motor function, and their therapeutic potential in Parkinson's and Huntington's diseases, the cellular and subcellular distributions of striatal CB1 receptors are not well defined. Here, we show that CB1 receptors are primarily located on GABAergic (vesicular GABA transporter-positive) and glutamatergic [vesicular glutamate transporter-1 (VGLUT-1)- and VGLUT-2-positive] striatal nerve terminals and are present in the presynaptic active zone, in the postsynaptic density, as well as in the extrasynaptic membrane. Both the nonselective agonist WIN552122 [(R)-(+)-[2,3-dihydro-5-methyl-3[(4-morpholinyl)methyl] pyrrolo[1,2,3-de]-1,4-benzoxazinyl]-(1-naphthalenyl)methanone mesylate salt] (EC50, 32 nM) and the CB1-selective agonist ACEA [N-(2-chloroethyl)-5Z,8Z,11Z,14Z-eicosatetraenamide] inhibited [3H]GABA release from rat striatal slices. The effect of these agonists was prevented by the CB1-selective antagonists SR141716A [N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide] (1 microM) and AM251 [1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide trifluoroacetate salt] (1 microM), indicating that cannabinoids inhibit the release of GABA via activation of presynaptic CB1 receptors. Cannabinoids modulated glutamate release via both CB1 and non-CB1 mechanisms. Cannabinoid agonists and antagonists inhibited 25 mM K+-evoked [3H]glutamate release and sodium-dependent [3H]glutamate uptake. Partial involvement of CB1 receptors is suggested because low concentrations of SR141716A partly and AM251 fully prevented the effect of WIN552122 and CP55940 [5-(1,1-dimethylheptyl)-2-[5-hydroxy-2-(3-hydroxypropyl)cyclohexyl]phenol]. However, the effect of CB1 agonists and antagonists persisted in CB1 knock-out mice, indicating the involvement of non-CB1,CB1-like receptors. In contrast, cannabinoids did not modulate [3H]dopamine release or [3H]dopamine and [3H]GABA uptake. Our results indicate distinct modulation of striatal GABAergic and glutamatergic transmission by cannabinoids and will facilitate the understanding of the role and importance of the cannabinoid system in normal and pathological motor function.
Collapse
MESH Headings
- 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology
- Animals
- Benzoxazines
- Blotting, Western/methods
- Calcium/metabolism
- Corpus Striatum/cytology
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Corpus Striatum/ultrastructure
- Dopamine/metabolism
- Dose-Response Relationship, Drug
- Drug Interactions
- Excitatory Amino Acid Antagonists/pharmacology
- Glutamic Acid/metabolism
- Immunohistochemistry/methods
- Male
- Mice
- Mice, Knockout
- Morpholines/pharmacology
- Naphthalenes/pharmacology
- Neurotransmitter Agents/metabolism
- Piperidines/pharmacology
- Potassium/pharmacology
- Pyrazoles/pharmacology
- Rats
- Rats, Wistar
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/deficiency
- Receptor, Cannabinoid, CB1/physiology
- Rimonabant
- Synapses/drug effects
- Synapses/metabolism
- Synaptosomes/drug effects
- Synaptosomes/metabolism
- Tetrodotoxin/pharmacology
- Tritium/metabolism
- Tyrosine 3-Monooxygenase/metabolism
- Vesicular Glutamate Transport Protein 1/metabolism
- Vesicular Glutamate Transport Protein 2/metabolism
- gamma-Aminobutyric Acid/metabolism
Collapse
Affiliation(s)
- Attila Köfalvi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1450, Hungary
| | | | | | | | | | | | | |
Collapse
|
177
|
Justinova Z, Solinas M, Tanda G, Redhi GH, Goldberg SR. The endogenous cannabinoid anandamide and its synthetic analog R(+)-methanandamide are intravenously self-administered by squirrel monkeys. J Neurosci 2006; 25:5645-50. [PMID: 15944392 PMCID: PMC2562767 DOI: 10.1523/jneurosci.0951-05.2005] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Anandamide, an endogenous ligand for brain cannabinoid CB(1) receptors, produces many behavioral effects similar to those of Delta(9)-tetrahydrocannabinol (THC), the main psychoactive ingredient in marijuana. Reinforcing effects of THC have been demonstrated in experimental animals, but there is only indirect evidence that endogenous cannabinoids such as anandamide participate in brain reward processes. We now show that anandamide serves as an effective reinforcer of drug-taking behavior when self-administered intravenously by squirrel monkeys. We also show that methanandamide, a synthetic long-lasting anandamide analog, similarly serves as a reinforcer of drug-taking behavior. Finally, we show that the reinforcing effects of both anandamide and methanandamide are blocked by pretreatment with the cannabinoid CB(1) receptor antagonist rimonabant (SR141716). These findings strongly suggest that release of endogenous cannabinoids is involved in brain reward processes and that activation of cannabinoid CB(1) receptors by anandamide could be part of the signaling of natural rewarding events.
Collapse
Affiliation(s)
- Zuzana Justinova
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland 21224, USA
| | | | | | | | | |
Collapse
|
178
|
Thanos PK, Dimitrakakis ES, Rice O, Gifford A, Volkow ND. Ethanol self-administration and ethanol conditioned place preference are reduced in mice lacking cannabinoid CB1 receptors. Behav Brain Res 2006; 164:206-13. [PMID: 16140402 DOI: 10.1016/j.bbr.2005.06.021] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Revised: 06/06/2005] [Accepted: 06/12/2005] [Indexed: 10/25/2022]
Abstract
Cannabinoids are postulated to play a role in modulating the reinforcing effects of abused drugs, including alcohol. Experiment 1 examined alcohol self-administration in cannabinoid CB1 receptor knockout (KO), heterozygous (HT) and wild type (WT) mice in a two-bottle choice paradigm. Mice were trained in a limited 8 h access/day to 10% (v/v) EtOH (EtOH) versus water. After baseline drinking levels (% EtOH preference and total EtOH intake (g/kg)), results indicated that the CB1 knockout mice displayed significantly lower baseline EtOH consumption compared to wild type mice. Subsequently, treatment with SR141716A (5mg/kg) significantly attenuated EtOH intake in the WT and HT mice but had little effect on the knockout mice. Experiment 2 examined the CB1 WT and CB1 KO strains in a conditioned place preference (CPP) procedure between saline and 2g/kg EtOH. The CB1 WT mice spent significantly more time in the EtOH-paired versus saline-paired chambers, whereas no significant preference was observed in the CB1 KO mice. Finally, we observed that CB1 KO mice were significantly lighter than WT and HT and that SR141716A did not significantly alter body weight. These results demonstrate that the cannabinoid CB1 receptor is an essential component of the molecular pathways underlying the reinforcing effects of alcohol. Thus, medications targeting the CB1 receptors may be beneficial for the treatment of alcoholism.
Collapse
MESH Headings
- Alcohol Drinking/genetics
- Analysis of Variance
- Animals
- Central Nervous System Depressants/pharmacology
- Conditioning, Classical/drug effects
- Conditioning, Classical/physiology
- Environment
- Ethanol/pharmacology
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/physiology
- Reward
- Self Administration
Collapse
Affiliation(s)
- Panayotis K Thanos
- Behavioral Pharmacology Lab, Department of Medicine, Brookhaven National Laboratory, Building 490, 30 Bell Avenue, Upton, NY 11973-5000, USA.
| | | | | | | | | |
Collapse
|
179
|
Pagotto U, Marsicano G, Cota D, Lutz B, Pasquali R. The emerging role of the endocannabinoid system in endocrine regulation and energy balance. Endocr Rev 2006; 27:73-100. [PMID: 16306385 DOI: 10.1210/er.2005-0009] [Citation(s) in RCA: 607] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
During the last few years, the endocannabinoid system has emerged as a highly relevant topic in the scientific community. Many different regulatory actions have been attributed to endocannabinoids, and their involvement in several pathophysiological conditions is under intense scrutiny. Cannabinoid receptors, named CB1 receptor and CB2 receptor, first discovered as the molecular targets of the psychotropic component of the plant Cannabis sativa, participate in the physiological modulation of many central and peripheral functions. CB2 receptor is mainly expressed in immune cells, whereas CB1 receptor is the most abundant G protein-coupled receptor expressed in the brain. CB1 receptor is expressed in the hypothalamus and the pituitary gland, and its activation is known to modulate all the endocrine hypothalamic-peripheral endocrine axes. An increasing amount of data highlights the role of the system in the stress response by influencing the hypothalamic-pituitary-adrenal axis and in the control of reproduction by modifying gonadotropin release, fertility, and sexual behavior. The ability of the endocannabinoid system to control appetite, food intake, and energy balance has recently received great attention, particularly in the light of the different modes of action underlying these functions. The endocannabinoid system modulates rewarding properties of food by acting at specific mesolimbic areas in the brain. In the hypothalamus, CB1 receptor and endocannabinoids are integrated components of the networks controlling appetite and food intake. Interestingly, the endocannabinoid system was recently shown to control metabolic functions by acting on peripheral tissues, such as adipocytes, hepatocytes, the gastrointestinal tract, and, possibly, skeletal muscle. The relevance of the system is further strenghtened by the notion that drugs interfering with the activity of the endocannabinoid system are considered as promising candidates for the treatment of various diseases, including obesity.
Collapse
Affiliation(s)
- Uberto Pagotto
- Endocrinology Unit, Department of Internal Medicine and Gastroenterology, Sant' Orsola-Malpighi Hospital, Bologna, Italy, and Department of Physiological Chemistry, Johannes Gutenberg-University Mainz, Germany.
| | | | | | | | | |
Collapse
|
180
|
Abstract
Although used for more than 4000 years for recreational and medicinal purposes, Cannabis and its best-known pharmacologically active constituents, the cannabinoids, became a protagonist in medical research only recently. This revival of interest is explained by the finding in the 1990s of the mechanism of action of the main psychotropic cannabinoid, Delta9-tetrahydrocannabinol (THC), which acts through specific membrane receptors, the cannabinoid receptors. The molecular characterization of these receptors allowed the development of synthetic molecules with cannabinoid and noncannabinoid structure and with higher selectivity, metabolic stability, and efficacy than THC, as well as the development of antagonists that have already found pharmaceutical application. The finding of endogenous agonists at these receptors, the endocannabinoids, opened new therapeutic possibilities through the modulation of the activity of cannabinoid receptors by targeting the biochemical mechanisms controlling endocannabinoid tissue levels.
Collapse
Affiliation(s)
- Vincenzo Di Marzo
- Endocannabinoid Research Group, Institutes of Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli, Naples, Italy.
| | | |
Collapse
|
181
|
Bir LS, Ercan S. Effects of intrathecal anandamide on somatosensory evoked responses in rats. Exp Neurol 2006; 197:386-90. [PMID: 16289169 DOI: 10.1016/j.expneurol.2005.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 09/12/2005] [Accepted: 10/08/2005] [Indexed: 10/25/2022]
Abstract
Anandamide, endogenous ligand of cannabinoid receptors produces similar effects of cannabinoids via CB1 receptors in the central nervous system. Its effect on ascending pathways of somatosensory conduction and somatosensory cortex is not known. The aim of this study was to determine the effects of anandamide on somatosensory evoked potentials (SEP). In this study, 24 Wistar male rats were used. The rats were divided into 4 groups. At the beginning, sciatic nerve stimulated scalp SEP traces were obtained from all of the rats. Later, 0.02 cm(3) anhydrous ethanol, 100 microg/kg, 200 microg/kg and 400 microg/kg anandamide dissolved in anhydrous ethanol were injected intrathecally to the first (control), second, third and fourth groups, respectively. Five minutes later, second SEP traces were started. In every SEP trace, two negative waves (N1, N2) following positive deflections were obtained. The latency and amplitudes of these waves assessed were compared in each group. In control and second groups, the parameters of these waves before and after the injections were not significantly different. However, in the third and fourth groups, latencies of N1 and N2 after injections were found significantly longer. This effect was dose dependent. In any of the groups, no significant changes were detected in the amplitudes after injections. In conclusion, anandamide, when injected intrathecally in pharmacological doses caused an induction of moderate conduction delay in SEP systems.
Collapse
Affiliation(s)
- Levent Sinan Bir
- Department of Neurology, Pamukkale Universitesi Tip Fakültesi Hastanesi Nöroloji AD. B-105, 20070 Kinikli-Denizli, Turkey.
| | | |
Collapse
|
182
|
Hill MN, Gorzalka BB. Is there a role for the endocannabinoid system in the etiology and treatment of melancholic depression? Behav Pharmacol 2006; 16:333-52. [PMID: 16148438 DOI: 10.1097/00008877-200509000-00006] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
With advances in basic and clinical neuroscience, many gaps have appeared in the traditional monoamine theory of depression that have led to reformulation of the hypotheses concerning the neurobiology of depression. The more recent hypotheses suggest that melancholic depression is characterized by central glucocorticoid resistance that results in hypercortisolemia, which in turn leads to down-regulation of neurotrophins and subsequent neurodegeneration. Examining the neurobiology of depression from this perspective suggests that the endocannabinoid system may play a role in the etiology of melancholic depression. Specifically, pharmacological and genetic blockade of the cannabinoid CB1 receptor induces a phenotypic state that is analogous to melancholic depression, including symptoms such as reduced food intake, heightened anxiety, increased arousal and wakefulness, deficits in extinction of aversive memories and supersensitivity to stress. These similarities between melancholic depression and an endocannabinoid deficiency become more interesting in light of recent findings that endocannabinoid activity is down-regulated by chronic stress and possibly increased by some antidepressant regimens. We propose that an endocannabinoid deficiency may underlie some of the symptoms of melancholic depression, and that enhancement of this system may ultimately be a novel form of pharmacotherapy for treatment-resistant depression.
Collapse
Affiliation(s)
- M N Hill
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
183
|
Haller J, Szirmai M, Varga B, Ledent C, Freund TF. Cannabinoid CB1 receptor dependent effects of the NMDA antagonist phencyclidine in the social withdrawal model of schizophrenia. Behav Pharmacol 2006; 16:415-22. [PMID: 16148446 DOI: 10.1097/00008877-200509000-00014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Clinical and laboratory findings suggest that cannabinoid signalling is implicated in schizophrenia. However, the interaction remains poorly understood, as data are often contradictory. Here we investigated wild-type (WT) and cannabinoid CB1 receptor-knockout (CB1-KO) mice in the phencyclidine-induced social withdrawal model of schizophrenia. N-methyl-D-aspartate (NMDA) antagonists (including phencyclidine) induce psychotic symptoms in humans, and are used to model schizophrenia in a variety of experimental conditions. In WTs, 5 mg/kg phencyclidine increased locomotion and stereotyped behaviours, and decreased social interactions. These changes are consistent with a schizophrenia-like effect. In CB1-KOs, phencyclidine decreased locomotion, enhanced ataxia and stereotypy more markedly than in WTs, but did not affect social interactions. Locomotion showed a significant negative correlation with both ataxia and stereotypy, suggesting that in CB1-KOs, the locomotor suppressive effect of phencyclidine was secondary to changes in these variables. Our findings demonstrate that CB1 gene disruption dramatically alters the behavioural effects of the NMDA antagonist phencyclidine, suggesting that the CB1 receptor is involved in schizophrenia. As social disruption and stereotypy respectively are believed to model negative and positive symptoms of schizophrenia, our findings tentatively suggest that cannabinoids are differentially involved in these two symptom categories. These findings require verification by experiments involving CB1 receptor blockers, as the genetic and pharmacological blockade of receptors may not always provide similar results.
Collapse
Affiliation(s)
- J Haller
- Department of Behavioural Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
184
|
Abstract
The discovery of cannabinoid receptors, together with the development of selective cannabinoid receptor antagonists, has encouraged a resurgence of cannabinoid pharmacology. With the identification of endogenous agonists, such as anandamide, scientists have sought to uncover the biological role of endocannabinoid systems; initially guided by the long-established actions of cannabis and exogenous cannabinoids such as delta9-tetrahydrocannabinol (THC). In particular, considerable research has examined endocannabinoid involvement in appetite, eating behaviour and body weight regulation. It is now confirmed that endocannabinoids, acting at brain CB1 cannabinoid receptors, stimulate appetite and ingestive behaviours, partly through interactions with more established orexigenic and anorexigenic signals. Key structures such as the nucleus accumbens and hypothalamic nuclei are sensitive sites for the hyperphagic actions of these substances, and endocannabinoid activity in these regions varies in relation to nutritional status and feeding expression. Behavioural studies indicate that endocannabinoids increase eating motivation by enhancing the incentive salience and hedonic evaluation of ingesta. Moreover, there is strong evidence of an endocannabinoid role in energy metabolism and fuel storage. Recent developments point to potential clinical benefits of cannabinoid receptor antagonists in the management of obesity, and of agonists in the treatment of other disorders of eating and body weight regulation.
Collapse
Affiliation(s)
- T C Kirkham
- School of Psychology, University of Liverpool, Liverpool, England.
| |
Collapse
|
185
|
Abstract
Drug abuse continues to take an enormous economic and social toll on the world. Among the costs are reduced productivity, increased need for medical services and stress on families. Treatments that allow affected individuals to reduce compulsive drug use are lacking and novel approaches to their development will likely come from increased understanding of the consequences of chronic exposure to reinforcing drugs. The purpose of this review is to explore the role of lipids in drug abuse and to present a rationale for an increased focus on the interactions between drugs of abuse and lipids in the brain. Small molecular weight lipids function as neuromodulators in the brain and, as such, play a role in the synaptic plasticity that occurs following exposure to drugs of abuse. In addition, the membrane lipid bilayer consists of lipid subdomains and emerging evidence suggests that protein function can be altered by transient associations with these subdomains. Finally, lipidomics is a very new field devoted to the exploration of changes in cellular lipid constituents during phenotypic alterations. Enhanced research in all of these areas will likely provide useful insights into and, perhaps, therapeutic targets for the treatment of drug abuse.
Collapse
Affiliation(s)
- Cecilia J Hillard
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States.
| |
Collapse
|
186
|
Lam CS, Rastegar S, Strähle U. Distribution of cannabinoid receptor 1 in the CNS of zebrafish. Neuroscience 2005; 138:83-95. [PMID: 16368195 DOI: 10.1016/j.neuroscience.2005.10.069] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 10/21/2005] [Accepted: 10/25/2005] [Indexed: 12/11/2022]
Abstract
The cannabinoid receptor 1 (Cb1) mediates the psychoactive effect of marijuana. In mammals, there is abundant evidence advocating the importance of cannabinoid signaling; activation of Cb1 exerts diverse functions, chiefly by its ability to modulate neurotransmission. Thus, much attention has been devoted to understand its role in health and disease and to evaluate its therapeutic potential. Here, we have cloned zebrafish cb1 and investigated its expression in developing and adult zebrafish brain. Sequence analysis showed that there is a high degree of conservation, especially in residues demonstrated to be critical for function in mammals. In situ hybridization revealed that zebrafish cb1 appears first in the preoptic area at 24 hours post-fertilization. Subsequently, transcripts are detected in the dorsal telencephalon, hypothalamus, pretectum and torus longitudinalis. A similar pattern of expression is recapitulated in the adult brain. While cb1 is intensively stained in the medial zone of the dorsal telencephalon, expression elsewhere is weak by comparison. In particular, localization of cb1 in the telencephalic periventricular matrix is suggestive of the involvement of Cb1 in neurogenesis, bearing strong resemblance in terms of expression and function to the proliferative mammalian hippocampal formation. In addition, a gradient-like expression of cb1 is detected in the torus longitudinalis, a teleost specific neural tissue. In relation to dopaminergic neurons in the diencephalic posterior tuberculum (considered to be the teleostean homologue of the mammalian midbrain dopaminergic system), both cb1 and tyrosine hydroxylase-expressing cells occupy non-overlapping domains. However there is evidence that they are co-localized in the caudal zone of the hypothalamus, implying a direct modulation of dopamine release in this particular region. Collectively, our data indicate the propensity of zebrafish cb1 to participate in multiple neurological processes.
Collapse
Affiliation(s)
- C S Lam
- Institute for Toxicology and Genetics, Forschungszentrum Karlsruhe, Postfach 3640, 76021 Karlsruhe, University of Heidelberg, Baden-Wurtemberg, Germany
| | | | | |
Collapse
|
187
|
Mátyás F, Yanovsky Y, Mackie K, Kelsch W, Misgeld U, Freund TF. Subcellular localization of type 1 cannabinoid receptors in the rat basal ganglia. Neuroscience 2005; 137:337-61. [PMID: 16289348 DOI: 10.1016/j.neuroscience.2005.09.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 09/08/2005] [Accepted: 09/09/2005] [Indexed: 11/29/2022]
Abstract
Endocannabinoids, acting via type 1 cannabinoid receptors (CB1), are known to be involved in short-term synaptic plasticity via retrograde signaling. Strong depolarization of the postsynaptic neurons is followed by the endocannabinoid-mediated activation of presynaptic CB1 receptors, which suppresses GABA and/or glutamate release. This phenomenon is termed depolarization-induced suppression of inhibition (DSI) or excitation (DSE), respectively. Although both phenomena have been reported to be present in the basal ganglia, the anatomical substrate for these actions has not been clearly identified. Here we investigate the high-resolution subcellular localization of CB1 receptors in the nucleus accumbens, striatum, globus pallidus and substantia nigra, as well as in the internal capsule, where the striato-nigral and pallido-nigral pathways are located. In all examined nuclei of the basal ganglia, we found that CB1 receptors were located on the membrane of axon terminals and preterminal axons. Electron microscopic examination revealed that the majority of these axon terminals were GABAergic, giving rise to mostly symmetrical synapses. Interestingly, preterminal axons showed far more intense staining for CB1, especially in the globus pallidus and substantia nigra, whereas their terminals were only faintly stained. Non-varicose, thin unmyelinated fibers in the internal capsule also showed strong CB1-labeling, and were embedded in bundles of myelinated CB1-negative axons. The majority of CB1 receptors labeled by immunogold particles were located in the axonal plasma membrane (92.3%), apparently capable of signaling cannabinoid actions. CB1 receptors in this location cannot directly modulate transmitter release, because the release sites are several hundred micrometers away. Interestingly, both the CB1 agonist, WIN55,212-2, as well as its antagonist, AM251, were able to block action potential generation, but via a CB1 independent mechanism, since the effects remained intact in CB1 knockout animals. Thus, our electrophysiological data suggest that these receptors are unable to influence action potential propagation, thus they may not be functional at these sites, but are likely being transported to the terminal fields. The present data are consistent with a role of endocannabinoids in the control of GABA, but not glutamate, release in the basal ganglia via presynaptic CB1 receptors, but also call the attention to possible non-CB1-mediated effects of widely used cannabinoid ligands on action potential generation.
Collapse
Affiliation(s)
- F Mátyás
- Institute of Experimental Medicine, Hungarian Academy of Sciences, PO Box 67, Budapest H-1450, Hungary
| | | | | | | | | | | |
Collapse
|
188
|
Moreno M, Escuredo L, Muñoz R, Rodriguez de Fonseca F, Navarro M. Long-term behavioural and neuroendocrine effects of perinatal activation or blockade of CB1 cannabinoid receptors. Behav Pharmacol 2005; 16:423-30. [PMID: 16148447 DOI: 10.1097/00008877-200509000-00015] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The present work studied the long-term effects of chronic perinatal manipulation of cannabinoid CB1 receptors in male and female rats. Perinatal activation of cannabinoid CB1 receptors by chronic administration of delta9-tetrahydrocannabinol at different doses (0.1, 0.5, 2 mg/kg, p.o.) induced sexually dimorphic behavioural changes in adulthood, altering habituation of locomotion, immobility and exploratory activity. These behavioural effects were also accompanied by alterations in corticosterone levels in the adult period. Prenatal blockade of CB1 receptors by chronic administration of 3 mg/kg (s.c.) of SR141716A decreased immobility behaviour in male and female animals, without any significant changes in corticosterone plasma levels. Cannabinoid CB1 receptors appear to play an important role in the ontogeny of psychomotor behaviours, and activation or blockade of these receptors during stages of plasticity, such as the prenatal or perinatal periods, can induce long-term effects, as shown by sexually dimorphic changes in behavioural patterns in adulthood.
Collapse
MESH Headings
- Adrenocorticotropic Hormone/blood
- Animals
- Animals, Newborn
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Corticosterone/blood
- Dose-Response Relationship, Drug
- Dronabinol/pharmacology
- Female
- Male
- Motor Activity/drug effects
- Piperidines/pharmacology
- Pregnancy
- Prenatal Exposure Delayed Effects
- Pyrazoles/pharmacology
- Rats
- Rats, Wistar
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/physiology
- Rimonabant
- Sex Factors
- Time Factors
Collapse
Affiliation(s)
- M Moreno
- Departamento de Psicobiología, Universidad Complutense de Madrid, 28223, Spain.
| | | | | | | | | |
Collapse
|
189
|
Moreno M, Lopez-Moreno JA, Rodríguez de Fonseca F, Navarro M. Behavioural effects of quinpirole following withdrawal of chronic treatment with the CB1 agonist, HU-210, in rats. Behav Pharmacol 2005; 16:441-6. [PMID: 16148449 DOI: 10.1097/00008877-200509000-00017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The present study investigated spontaneous and quinpirole-induced motor responses of in rats, following withdrawal from chronic treatment with the potent cannabinoid agonist HU-210. Withdrawal from chronic HU-210 (20 microg/kg daily, 14 days) produced a decrease in spontaneous activity at 1 and 2 days and enhanced the hyperactivity induced by acute administration of the dopamine D2 agonist quinpirole (0.5 mg/kg) at 4 days after the end of HU-210 treatment. Administration of quinpirole on day 4 of withdrawal from chronic HU-210 enhanced stereotyped responses and induced jumping behaviour. These results suggest that withdrawal from chronic exposure to cannabinoid agonists could induce a time-dependent alteration in dopamine D2 psychomotor function, leading to a behavioural disorganization, comparable to acute psychotic episodes after continuous cannabinoids.
Collapse
Affiliation(s)
- M Moreno
- Departamento de Psicobiologia, Facultad de Psicologia, Instituto Universitario de Drogodependencias, Universidad Complutense de Madrid, Spain.
| | | | | | | |
Collapse
|
190
|
Abstract
Mood and anxiety disorders, the most prevalent of the psychiatric disorders, cause immeasurable suffering worldwide. Despite impressive advances in pharmacological therapies, improvements in efficacy and side-effect profiles are needed. The present literature review examines the role that the endocannabinoid system may play in these disorders and the potential value of targeting this system in the search for novel and improved medications. Cannabis and its major psychoactive component (-)-trans-delta9-tetrahydrocannabinol, have profound effects on mood and can modulate anxiety and mood states. Cannabinoid receptors and other protein targets in the central nervous system (CNS) that modulate endocannabinoid function have been described. The discovery of selective modulators of some of these sites that increase or decrease endocannabinoid neurotransmission, primarily through the most prominent of the cannabinoid receptors in the CNS, the CB1 receptors, combined with transgenic mouse technology, has enabled detailed investigations into the role of these CNS sites in the regulation of mood and anxiety states. Although data point to the involvement of the endocannabinoid system in anxiety states, the pharmacological evidence seems contradictory: both anxiolytic- and anxiogenic-like effects have been reported with both endocannabinoid neurotransmission enhancers and blockers. Due to advances in the development of selective compounds directed at the CB1 receptors, significant progress has been made on this target. Recent biochemical and behavioural findings have demonstrated that blockade of CB1 receptors engenders antidepressant-like neurochemical changes (increases in extracellular levels of monoamines in cortical but not subcortical brain regions) and behavioural effects consistent with antidepressant/antistress activity in rodents.
Collapse
Affiliation(s)
- J M Witkin
- Psychiatric Drug Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285-0510, USA.
| | | | | |
Collapse
|
191
|
Soria G, Mendizábal V, Touriño C, Robledo P, Ledent C, Parmentier M, Maldonado R, Valverde O. Lack of CB1 cannabinoid receptor impairs cocaine self-administration. Neuropsychopharmacology 2005; 30:1670-80. [PMID: 15742004 DOI: 10.1038/sj.npp.1300707] [Citation(s) in RCA: 176] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute rewarding properties are essential for the establishment of cocaine addiction, and multiple neurochemical processes participate in this complex behavior. In the present study, we used the self-administration paradigm to evaluate the role of CB1 cannabinoid receptors in several aspects of cocaine reward, including acquisition, maintenance, and motivation to seek the drug. For this purpose, both CB1 receptor knockout mice and wild-type littermates were trained to intravenously self-administer cocaine under different schedules. Several cocaine training doses (0.32, 1, and 3.2 mg/kg/infusion) were used in the acquisition studies. Only 25% of CB1 knockout mice vs 75% of their wild-type littermates acquired a reliable operant responding to self-administer the most effective dose of cocaine (1 mg/kg/infusion), and the number of sessions required to attain this behavior was increased in knockout mice. Animals reaching the acquisition criteria were evaluated for the motivational strength of cocaine as a reinforcer under a progressive ratio schedule. The maximal effort to obtain a cocaine infusion was significantly reduced after the genetic ablation of CB1 receptors. A similar result was obtained after the pharmacological blockade of CB1 receptors with SR141716A in wild-type mice. Moreover, the cocaine dose-response curve was flattened in the knockout group, suggesting that the differences observed between genotypes were related to changes in the reinforcing efficacy of the training dose of cocaine. Self-administration for water and food was not altered in CB1 knockout mice in any of the reinforcement schedules used, which emphasizes the selective impairment of drug reinforcement in these knockout mice. Finally, cocaine effects on mesolimbic dopaminergic transmission were evaluated by in vivo microdialysis in these mice. Acute cocaine administration induced a similar enhancement in the extracellular levels of dopamine in the nucleus accumbens of both CB1 knockout and wild-type mice. This work clearly demonstrates that CB1 receptors play an important role in the consolidation of cocaine reinforcement, although are not required for its acute effects on mesolimbic dopaminergic transmission.
Collapse
Affiliation(s)
- Guadalupe Soria
- Laboratori de Neurofarmacologia, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
192
|
Avraham Y, Israeli E, Gabbay E, Okun A, Zolotarev O, Silberman I, Ganzburg V, Dagon Y, Magen I, Vorobia L, Pappo O, Mechoulam R, Ilan Y, Berry EM. Endocannabinoids affect neurological and cognitive function in thioacetamide-induced hepatic encephalopathy in mice. Neurobiol Dis 2005; 21:237-45. [PMID: 16102970 DOI: 10.1016/j.nbd.2005.07.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2005] [Revised: 07/11/2005] [Accepted: 07/13/2005] [Indexed: 10/25/2022] Open
Abstract
Endocannabinoids function as neurotransmitters and neuromodulators in the central nervous system via specific receptors and apparently have a neuroprotective role. We assumed that the endocannabinoid system could be involved in the pathogenesis of hepatic encephalopathy (HE), a neuropsychiatric syndrome due to liver disease. We used a mouse model of a thioacetamide induced fulminant hepatic failure. We found that the levels of the endocannabinoid 2-arachidonoyl-glycerol (2-AG) were elevated in the brain. Treatment with either 2-AG or with the CB1 receptor antagonist, SR141716A, improved a neurological score, activity and cognitive function. Activation of the CB2 receptor by a selective agonist, HU308, also improved the neurological score. 2-AG activity could be blocked with the specific CB2 receptor antagonist SR144528A. The CB1 receptor agonist noladin ether was inactive. We conclude that the endocannabinoid system may play an important role in the pathogenesis of HE. Modulation of this system either by exogenous agonists specific for the CB2 receptors or possibly also by antagonists to the CB1 receptors may have therapeutic potential.
Collapse
Affiliation(s)
- Yosefa Avraham
- Department of Metabolism and Human Nutrition, Braun School of Public Health, Hadassah-Hebrew University Medical School, Jerusalem, Israel 91120.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Pierce RC, Kumaresan V. The mesolimbic dopamine system: the final common pathway for the reinforcing effect of drugs of abuse? Neurosci Biobehav Rev 2005; 30:215-38. [PMID: 16099045 DOI: 10.1016/j.neubiorev.2005.04.016] [Citation(s) in RCA: 603] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Revised: 04/05/2005] [Accepted: 04/19/2005] [Indexed: 11/23/2022]
Abstract
In this review we will critically assess the hypothesis that the reinforcing effect of virtually all drugs of abuse is primarily dependent on activation of the mesolimbic dopamine system. The focus is on five classes of abused drugs: psychostimulants, opiates, ethanol, cannabinoids and nicotine. For each of these drug classes, the pharmacological and physiological mechanisms underlying the direct or indirect influence on mesolimbic dopamine transmission will be reviewed. Next, we evaluate behavioral pharmacological experiments that specifically assess the influence of activation of the mesolimbic dopamine system on drug reinforcement, with particular emphasis on animal experiments using drug self-administration paradigms. There is overwhelming evidence that all five classes of abused drugs increase dopamine transmission in limbic regions of the brain through interactions with a variety of transporters, ionotropic receptors and metabotropic receptors. Behavioral pharmacological experiments indicate that increased dopamine transmission is clearly both necessary and sufficient to promote psychostimulant reinforcement. For the other four classes of abused substances, self-administration experiments suggest that although increasing mesolimbic dopamine transmission plays an important role in the reinforcing effects of opiates, ethanol, cannabinoids and nicotine, there are also dopamine-independent processes that contribute significantly to the reinforcing effects of these compounds.
Collapse
Affiliation(s)
- R Christopher Pierce
- Department of Pharmacology, Boston University School of Medicine, 715 Albany Street, L603 Boston, MA 02118, USA.
| | | |
Collapse
|
194
|
Abstract
Anandamide (AEA) uptake has been described over the last decade to occur by facilitated diffusion, but a protein has yet to be isolated. In some cell types, it has recently been suggested that AEA, an uncharged hydrophobic molecule, passively diffuses through the plasma membrane in a process that is not protein-mediated. Since that observation, recent kinetics studies (using varying assay conditions) have both supported and denied the presence of an AEA transporter. In this review, we analyze the current literature exploring the mechanism of AEA uptake and endeavor to explain the reasons for the divergent views. One of the main variables among laboratories is the incubation time of the cells with AEA. Initial kinetics (at time points <1 min depending upon the cell type) isolate events that occur at the plasma membrane and are most useful to study saturability of uptake and effects of purported transport inhibitors upon uptake. Results with longer incubation times reflect events not only at the plasma membrane but also interactions at intracellular sites that may include enzyme(s), other proteins, or specialized lipid-binding domains. Furthermore, at long incubation times, antagonists to AEA receptors reduce AEA uptake. Another complicating factor in AEA transport studies is the nonspecific binding to plastic culture dishes. The magnitude of this effect may exceed AEA uptake into cells. Likewise, AEA may be released from plastic culture dishes (without cells) in such a manner as to mimic efflux from cells. AEA transport protocols using BSA, similar to the method used for fatty acid uptake studies, are gaining acceptance. This may improve AEA solution stability and minimize binding to plastic, although some groups report that BSA interferes with uptake. In response to criticisms that many transport inhibitors also inhibit the fatty acid amide hydrolase (FAAH), new compounds have recently been synthesized. Following their characterization in FAAH+/+ and FAAH-/- cells and transgenic mice, several inhibitors have been shown to have physiological activity in FAAH-/- mice. Their targets are now being characterized with the possibility that a protein transporter for AEA may be characterized.
Collapse
Affiliation(s)
- Sherrye T Glaser
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973, United States
| | | | | |
Collapse
|
195
|
Centonze D, Rossi S, Prosperetti C, Tscherter A, Bernardi G, Maccarrone M, Calabresi P. Abnormal sensitivity to cannabinoid receptor stimulation might contribute to altered gamma-aminobutyric acid transmission in the striatum of R6/2 Huntington's disease mice. Biol Psychiatry 2005; 57:1583-9. [PMID: 15953496 DOI: 10.1016/j.biopsych.2005.03.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Revised: 02/24/2005] [Accepted: 03/04/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND One of the earliest neurochemical alterations observed in both Huntington's disease (HD) patients and HD animal models is the dysregulation of the endocannabinoid system, an alteration that precedes the development of identifiable striatal neuropathology. How this alteration impacts striatal synaptic transmission is unknown. METHODS We measured the effects of cannabinoid receptor stimulation on gamma-aminobutyric acid (GABA)-ergic synaptic currents recorded from striatal neurons of R6/2 HD mice in the early phase of their disease. RESULTS The sensitivity of striatal GABA synapses to cannabinoid receptor stimulation is severely impaired in R6/2 HD mice. In particular, whereas in control animals activation of cannabinoid CB1 receptors results in a significant inhibition of both evoked and spontaneous GABA-mediated synaptic events by a presynaptic mechanism, in R6/2 mice this treatment fails to reduce GABA currents but causes, in contrast, a slight increase of spontaneous inhibitory postsynaptic currents (sIPSCs). CONCLUSIONS Experimental HD was also associated with enhanced frequency of sIPSCs, a result consistent with the conclusion that loss of cannabinoid-mediated control of GABA transmission might contribute to hyperactivity of GABA synapses in the striatum of HD mice. Accordingly, spontaneous excitatory postsynaptic currents, which were not upregulated in R6/2 mice, were still sensitive to cannabinoid receptor stimulation.
Collapse
Affiliation(s)
- Diego Centonze
- Clinica Neurologica, Dipartimento di Neuroscienze, Università di Tor Vergata, Italy.
| | | | | | | | | | | | | |
Collapse
|
196
|
Patel S, Roelke CT, Rademacher DJ, Hillard CJ. Inhibition of restraint stress-induced neural and behavioural activation by endogenous cannabinoid signalling. Eur J Neurosci 2005; 21:1057-69. [PMID: 15787710 DOI: 10.1111/j.1460-9568.2005.03916.x] [Citation(s) in RCA: 228] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The role of endocannabinoid (eCB) signalling in restraint stress-induced neuronal activation was studied. Male mice exposed to 30 min of restraint exhibit increased Fos protein within prefrontal cortex (PFC), lateral septum (LS), nucleus accumbens (Acb) and medial amygdala. SR141716 (2 mg/kg) itself had no effect on Fos but pretreatment with SR141716 significantly potentiated restraint-induced Fos expression in cingulate, LS and Acb. SR141716 also significantly increased the time spent in active escape behaviours during the restraint. In restraint-habituated mice (mice exposed to four previous restraint episodes), the fifth restraint exposure resulted in decreased expression of active escape behaviours compared to the first exposure and only induced Fos protein in the central and medial amygdala. Administration of SR141716 prior to the fifth restraint episode resulted in greater potentiation of restraint-induced Fos induction than the first; significant increases occurred within all regions of PFC examined, LS and Acb. Brain regional eCB content was measured immediately after restraint. N-arachidonylethanolamine content within the amygdala was significantly decreased after both restraint episodes. 2-Arachidonylglycerol content was significantly increased in both the limbic forebrain and amygdala after the fifth restraint but not the first. Restraint had no effect on cerebellar eCB content. These data suggest that eCB activation of CB(1) receptors opposes the behavioural and neuronal responses to aversive stimuli. Because repeated homotypic stress increased both limbic 2-AG and resulted in a greater effect of SR141716 on limbic Fos expression, we hypothesize that increased CB(1) receptor activity contributes to the expression of habituation to homotypic stress.
Collapse
Affiliation(s)
- Sachin Patel
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | | | | |
Collapse
|
197
|
van der Stelt M, Fox SH, Hill M, Crossman AR, Petrosino S, Di Marzo V, Brotchie JM. A role for endocannabinoids in the generation of parkinsonism and levodopa-induced dyskinesia in MPTP-lesioned non-human primate models of Parkinson's disease. FASEB J 2005; 19:1140-2. [PMID: 15894565 DOI: 10.1096/fj.04-3010fje] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endocannabinoids and cannabinoid CB1 receptors play a role in the control of movement by modulating GABA, glutamate, and other neurotransmitters throughout the basal ganglia. Roles for abnormalities in endocannabinoid signaling in Parkinson's disease (PD) and the major side effect of current treatments, levodopa-induced dyskinesia (LID), have been suggested by rodent studies. Here we show that signaling by endocannabinoids contributes to the pathophysiology of parkinsonism and LID in MPTP-lesioned, non-human primate models of Parkinson's disease. In MPTP-lesioned marmosets previously treated with levodopa to establish LID, attenuation of CB1 signaling by systemic administration of rimonabant (1 and 3 mg/kg) had anti-parkinsonian actions, equivalent to a 71% increase in motor activity at 3 mg/kg. Rimonabant did not elicit dyskinesia. Co-administration of levodopa (8 mg/kg) and rimonabant (1 and 3 mg/kg) resulted in significantly less dyskinesia than levodopa alone, without significantly affecting the anti-parkinsonian action of levodopa. These data suggest that enhanced endocannabinoid signaling may be involved in the pathophysiology of both parkinsonism and LID. To define potential mechanisms by which such a role might be mediated, we determined the levels of the endocannabinoids anandamide and 2-arachidonyl glycerol (2-AG) throughout the basal ganglia in normal and three groups of MPTP-lesioned cynomolgus monkeys (untreated; acutely treated with L-DOPA, non-dyskinetic; long-term treated, with levodopa-induced dyskinesia). In the untreated, MPTP-lesioned primate, parkinsonism was associated with increases in both 2-AG (+88%) and anandamide (+49%) in the striatum, and of 2-AG (+97%) in the substantia nigra, changes that are consistent with the previously suggested role for endocannabinoids in mechanisms attempting to compensate for loss of dopamine in untreated parkinsonism. Increased levels of anandamide (+34%) in the external globus pallidus of MPTP-lesioned animals were normalized by levodopa treatment and may contribute to the generation of parkinsonian symptoms. However, no clear alteration in endocannabinoid levels could be correlated with the expression of LID. These data highlight the potential roles played by endocannabinoids and CB1 in PD and LID and suggest the need for further research to pursue the multiple therapeutic opportunities for manipulating this system in movement disorders.
Collapse
Affiliation(s)
- Mario van der Stelt
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Pozzuoli, NA, Italy
| | | | | | | | | | | | | |
Collapse
|
198
|
Ashton CH, Moore PB, Gallagher P, Young AH. Cannabinoids in bipolar affective disorder: a review and discussion of their therapeutic potential. J Psychopharmacol 2005; 19:293-300. [PMID: 15888515 DOI: 10.1177/0269881105051541] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Bipolar affective disorder is often poorly controlled by prescribed drugs. Cannabis use is common in patients with this disorder and anecdotal reports suggest that some patients take it to alleviate symptoms of both mania and depression. We undertook a literature review of cannabis use by patients with bipolar disorder and of the neuropharmacological properties of cannabinoids suggesting possible therapeutic effects in this condition. No systematic studies of cannabinoids in bipolar disorder were found to exist, although some patients claim that cannabis relieves symptoms of mania and/or depression. The cannabinoids Delta(9)-tetrahydrocannabinol (THC) and cannabidiol (CBD) may exert sedative, hypnotic, anxiolytic, antidepressant, antipsychotic and anticonvulsant effects. Pure synthetic cannabinoids, such as dronabinol and nabilone and specific plant extracts containing THC, CBD, or a mixture of the two in known concentrations, are available and can be delivered sublingually. Controlled trials of these cannabinoids as adjunctive medication in bipolar disorder are now indicated.
Collapse
Affiliation(s)
- C H Ashton
- Department of Psychiatry, University of Newcastle upon Tyne, Royal Victoria Infirmary, Newcastle upon Tyne, UK.
| | | | | | | |
Collapse
|
199
|
Chhatwal JP, Davis M, Maguschak KA, Ressler KJ. Enhancing cannabinoid neurotransmission augments the extinction of conditioned fear. Neuropsychopharmacology 2005; 30:516-24. [PMID: 15637635 DOI: 10.1038/sj.npp.1300655] [Citation(s) in RCA: 277] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The endogenous cannabinoid (eCB) system represents a major therapeutic target for the treatment of a variety of anxiety-related disorders. A recent study has demonstrated that pharmacologic or genetic disruption of CB1-receptor-mediated neurotransmission decreases the extinction of conditioned fear in mice. Here, we examined whether CB1 blockade would similarly disrupt extinction in rats, using fear-potentiated startle as a measure of conditioned fear. We also examined whether pharmacologic enhancement of CB1 activation would lead to enhancements in extinction. Our results indicate that systemic administration of the CB1 antagonist rimonabant (SR141716A) prior to extinction training led to significant, dose-dependent decreases in extinction. While the administration of the CB1 agonist WIN 55,212-2 did not appear to affect extinction, administration of AM404, an inhibitor of eCB breakdown and reuptake, led to dose-dependent enhancements in extinction. In addition to showing decreased fear 1 and 24 h after extinction training, AM404-treated animals showed decreased shock-induced reinstatement of fear. Control experiments demonstrated that the effects of AM404 could not be attributed to alterations in the expression of conditioned fear, locomotion, shock reactivity, or baseline startle, as these parameters seemed unchanged by AM404. Furthermore, coadministration of rimonabant with AM404 blocked this enhancement of extinction, suggesting that AM404 was acting to increase CB1 receptor activation during extinction training. These results demonstrate that the eCB system can be modulated to enhance emotional learning, and suggest that eCB modulators may be therapeutically useful as adjuncts for exposure-based psychotherapies such as those used to treat Post-Traumatic Stress Disorder and other anxiety disorders.
Collapse
MESH Headings
- Amygdala/physiology
- Amygdala/physiopathology
- Animals
- Cannabinoid Receptor Modulators/metabolism
- Extinction, Psychological/physiology
- Fear/physiology
- In Situ Hybridization
- Male
- Rats
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/physiology
- Synaptic Transmission/physiology
Collapse
Affiliation(s)
- Jasmeer P Chhatwal
- Emory University School of Medicine, Department of Psychiatry and Behavioral Sciences, Center for Behavioral Neuroscience, Yerkes Research Center, Atlanta, GA 30329, USA
| | | | | | | |
Collapse
|
200
|
Houchi H, Babovic D, Pierrefiche O, Ledent C, Daoust M, Naassila M. CB1 receptor knockout mice display reduced ethanol-induced conditioned place preference and increased striatal dopamine D2 receptors. Neuropsychopharmacology 2005; 30:339-49. [PMID: 15383833 DOI: 10.1038/sj.npp.1300568] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cannabinoids and ethanol activate the same reward pathways, and recent advances in the understanding of the neurobiological basis of alcoholism suggest that the CB1 receptor system may play a key role in the reinforcing effects of ethanol and in modulating ethanol intake. In the present study, male CB1 receptors knockout mice generated on a CD1 background displayed decreased ethanol-induced conditioned place preference (CPP) compared to wild-type (CB1(+/+)) mice. Ethanol (0.5, 1.0, 1.5, and 2.0 g/kg) induced significant CPP in CB1(+/+) mice at all doses tested, whereas it induced significant CPP only at the highest dose of ethanol (2.0 g/kg) in CB1(-/-) mice. However, there was no genotypic difference in cocaine (20 mg/kg)-induced CPP. There was also no genotypic difference, neither in cocaine (10-50 mg/kg) nor in D-amphetamine (1.2-5 mg/kg)-induced locomotor effects. In addition, mutant and wild-type mice did not differ in sensitivity to the anxiolytic effects of ethanol (1.5 g/kg) when tested using the elevated plus maze. Interestingly, this decrease in ethanol efficacy to induce CPP in CB1(-/-) mice was correlated with an increase in D2/D3 receptors, as determined by [3H]raclopride binding, whereas there was no difference in D1-like receptors, as determined by [3H]SCH23390 binding, measured in the striatum from drug-naive mice. This increase in D2/D3 binding sites observed in CB1 knockout mice was associated with an altered locomotor response to the D2/D3 agonist quinpirole (low doses 0.02-0.1 mg/kg) but not to an alteration of quinpirole (0.1-1.0 mg/kg)-induced CPP compared to wild-type mice. Altogether, the present results indicate that lifelong deletion of CB1 receptors reduced ethanol-induced CPP and that these reduced rewarding effects of ethanol are correlated to an overexpression of striatal dopamine D2 receptors.
Collapse
MESH Headings
- Alcohol Drinking/genetics
- Alcohol Drinking/psychology
- Animals
- Anti-Anxiety Agents/pharmacology
- Benzazepines/pharmacology
- Central Nervous System Depressants/pharmacology
- Cocaine/pharmacology
- Conditioning, Operant/drug effects
- Dextroamphetamine/pharmacology
- Dopamine Agonists/pharmacology
- Dopamine Antagonists/pharmacology
- Dopamine Uptake Inhibitors/pharmacology
- Ethanol/pharmacology
- Mice
- Mice, Knockout
- Motor Activity/drug effects
- Neostriatum/drug effects
- Neostriatum/metabolism
- Quinpirole/pharmacology
- Raclopride/pharmacology
- Radioligand Assay
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/physiology
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/drug effects
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Reward
Collapse
Affiliation(s)
- Hakim Houchi
- Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP), Jeune Equipe, Université de Picardie Jules Verne, Faculté de Pharmacie, 1 rue des Louvels, Amiens, France
| | | | | | | | | | | |
Collapse
|