Canna~Fangled Abstracts

New insights on endocannabinoid transmission in psychomotor disorders

By August 3, 2013No Comments

pub med big

New insights on endocannabinoid transmission in psychomotor disorders

The publisher’s final edited version of this article is available at Prog Neuropsychopharmacol Biol Psychiatry

Abstract

The endocannabinoids are lipid signaling molecules that bind to cannabinoid CB1 and CB2 receptors and other metabotropic and ionotropic receptors. Anandamide and 2-arachidonoyl glycerol, the two best-characterized examples, are released on demand in a stimulus-dependent manner by cleavage of membrane phospholipid precursors. Together with their receptors and metabolic enzymes, the endocannabinoids play a key role in modulating neurotransmission and synaptic plasticity in the basal ganglia and other brain areas involved in the control of motor functions and motivational aspects of behavior.

This mini-review provides an update on the contribution of the endocannabinoid system to the regulation of psychomotor behaviors and its possible involvement in the pathophysiology of Parkinson’s disease and schizophrenia.

Keywords: cannabinoid, endocannabinoid, basal ganglia, Parkinson, schizophrenia, dyskinesia

1. The endocannabinoid system

The endocannabinoid (EC) system consists of a family of lipid signaling molecules (endocannabinoids) and their associated metabolic enzymes and receptors, which modulate various physiological processes, including vasodilation, immune responses, synaptic transmission, cognition, pain and motor activity to name a few.

In addition to the well-known cannabinoid CB1/CB2 receptors and their endogenous ligands, anandamide (AEA) and 2-arachidonoyl glycerol (2-AG), other molecular entities, such as noladin-ether, N-arachidonoyl-dopamine and virhodamine, as well as non-CB1/CB2 receptors are now considered part of the EC system (Hanus et al., 2001Bisogno et al., 2000Porter et al., 2002Kreitzer and Stella, 2009). The complexity of this system has clear implications for the design and translational applications of future cannabinoid-based therapies.

This mini-review addresses recent discoveries on EC transmission within the central nervous system (CNS), focusing in particular on the contribution of traditional cannabinoid and non-CB1/CB2 receptors to the pathophysiology of Parkinson’s disease (PD) and schizophrenia.

1.1 Endocannabinoids and their metabolizing enzymes

The endocannabinoids (ECs) are naturally occurring lipids that activate cannabinoid CB1/CB2 receptors and mimic the pharmacological effects of the psychoactive constituent of marijuana, Δ9-tretrahydrocannabinol (THC). To date, arachidonoylethanolamine (AEA) and 2-AG are the two most studied ECs. Details on the EC biosynthetic enzymes and their CNS distribution have been covered by other articles and reviews (Simon and Cravatt, 2006Liu et al., 2008Nyilas et al., 2008Ueda et al., 2011) and will not be discussed here.

AEA is a partial agonist at both cannabinoid receptor subtypes and can also bind to other non-CB1/CB2 receptors, such peroxisome proliferator-activated receptors (PPAR), TRPV1 channels and the orphan receptor GPR55 (see below).

The biological actions of AEA are terminated via a carrier-mediated uptake, whose molecular identity remains controversial (Fegley et al., 2004Glaser et al., 2003Hillard and Jarrahian, 2003), followed by enzymatic hydrolysis via a fatty acid amide hydrolase (FAAH) (Cravatt et al., 1996Wei et al., 2006). Administration of exogenous AEA to FAAH−/− mice may lead to the production of prostaglandin-like compounds (prostamides) through a COX-2-dependent pathway (Weber et al., 2004). Although AEA has low affinity for COX-2, this metabolic pathway may become physiologically relevant under conditions promoting COX-2 upregulation, such as neurotoxic insults and neurodegenerative disorders characterized by an inflammatory component, such as Parkinson’s disease (Vila et al., 2001Teismann et al., 2003).

Several lipoxygenases (such as, 12-LOX and 15-LOX) and P450 may also convert AEA into signaling lipids that activate classic cannabinoid receptors, as well as non-CB1/CB2 receptors (Kozak and Marnett, 2002; Snider et al., 2009).

2-AG, which is a full agonist at cannabinoid receptors, acts as a retrograde messenger on pre-synaptic CB1 receptors located on excitatory and inhibitory synapses, and as an autocrine mediator of post-synaptic slow self-inhibition (SSI) in neocortical interneurons (Kreitzer and Regehr, 2001Wilson and Nicoll, 2001Freund et al., 2003Marinelli et al., 2008).

Unlike AEA, 2-AG does not bind to TRPV1 or PPAR receptors, but can activate GPR55 receptors in vitro and a not-yet identified G-protein-coupled receptors that controls cell migration and viability (Ryberg et al., 2007;Pertwee et al., 2010).

2-AG is uptaken intracellularly through the AEA transporter, and can be metabolized by either FAAH, or the serine hydrolase MAGL, which represents the main 2-AG hydrolyzing enzyme in neurons (Beltramo and Piomelli, 2000;Dinh et al., 2002Muccioli et al., 2007Long et al., 2009Schlosburg et al., 2010). Pharmacological inhibition of MAGL increases 2-AG levels in the brain (Hohmann et al., 2005), potentiates its effects in vitro and in vivo (Long et al., 2009Makara et al., 2005), and significantly reduces brain arachidonic acid and associated eicosanoids under basal and neuroinflammatory conditions, suggesting that MAGL is a CNS metabolic node coupling EC to prostaglandin signaling (Nomura et al., 2011).

MAGL genetic ablation has been shown to alter EC-mediated synaptic plasticity in mouse hippocampus and cerebellum via 2-AG-induced persistent activation and consequential desensitization of CB1 receptors (Zhong et al., 2011Pan et al., 2011). Interestingly, although MAGL−/− mice have normal locomotor activity, they show enhanced learning behavior, suggesting the involvement of MAGL in the regulation of cognitive function (Chanda et al., 2010Pan et al., 2011).

Recently, Marrs and co-workers (2010) showed that the knockdown of the serine hydrolase alpha-beta-hydrolase domain 6 (ABHD6) reduced 2-AG hydrolysis in vitro and increased the efficacy of 2-AG-induced stimulation of cell migration. Also, inhibition of either ABHD6 or MAGL had similar effects on the CB1-dependent stimulation of long-term depression in mouse cortical excitatory synapses, suggesting that ABHD6 may control the amount of 2-AG reaching pre-synaptic CB1 receptors (Marrs et al., 2010).

1.2 Cannabinoid and GPR55 receptors

The two main metabotropic cannabinoid receptors, CB1 and CB2, are Gi/o-coupled receptors (GPCR) that initiate, upon activation, signaling events typically associated with this class of G proteins, i.e. inhibition of cAMP accumulation and protein kinase (PKA) activity (Pertwee et al., 2010). Stimulation of CB1 receptors has been shown to inhibit N and P/Q-type voltage-gated Ca2+ channels and M-type K+ channels (Twitchell et al., 1997;Schweitzer, 2000), and to activate A-type and inwardly rectifying K+currents, which have been implicated in the CB1-mediated depression of GABA and glutamate release (Mu et al., 1999Kreitzer and Regehr, 2001;Wilson et al., 2001Gerdeman and Lovinger, 2001). CB1 receptors can also indirectly modulate the activity of dopaminergic pathways via pre- and post-synaptic mechanisms (for review, see Laviolette and Grace, 2006).

Distinct cannabinoid ligands, and/or concomitant activation of other GPCR, may promote the coupling of CB1 receptors to different Gi isoforms (Glass and Felder, 1997Mukhopadhyay and Howlett, 2005Shoemaker et al., 2005), as well as the formation of heterodimers with dopamine D2 and mu-opioid receptors (Hojo et al., 2008Kearn et al., 2005). Different cannabinoid agonists may also stabilize unique cannabinoid receptor conformations, thus leading to functional selectivity in downstream signaling and diverging effects on receptor internalization and desensitization (Straikeret al., 2011Atwood et al., 2012).

CB1 receptors are mainly localized pre-synaptically, which is consistent with their proposed modulatory role of inhibitory and excitatory neurotransmission (Piomelli, 2003). Within the striatum, a brain area relevant to the pathophysiology of PD and schizophrenia, most studies agree that CB1 receptors are expressed on parvalbumin-positive GABAergic interneurons, on cholinergic subpopulations (Fusco et al., 2004;Uchigashima et al., 2007), on collaterals from GABAergic medium spiny neurons (MSN), and on glutamatergic, but not dopaminergic, afferents (Gerdeman and Lovinger, 2001Köfalvi et al., 2005Matyas et al., 2006;Pickel et al., 2006Uchigashima et al., 2007). CB1 are also expressed in MSN terminals projecting to the globus pallidus (medial and lateral segments) and to the substantia nigra, as well as on the projections of the subthalamic nucleus to the substantia nigra (Mailleux and Vanderhaeghen, 1992Julianet al., 2003Martin et al., 2008). By contrast, the presence of CB1 receptors in the somatodendritic area of MSN remains controversial (Köfalvi et al., 2005Matyas et al., 2006Rodriguez et al., 2001Uchigashima et al., 2007).

In cortical areas, CB1 receptors are localized in layers I and IV and, at lower density, in the intermediate layers (Herkenham et al., 1991Egerton et al., 2006). In primates and humans, CB1 receptors are highly expressed in the axon terminals of a subpopulation of GABAergic CCK-positive interneurons targeting the perisomatic-region of pyramidal neurons of the dorsolateral prefrontal cortex (Eggan et al., 2008Eggan et al., 2010). Optical density measurements of CB1 mRNA have shown the highest density in layer II, whereas weak or no expression have been observed in layers I, IV and V (Eggan et al., 2008). In contrast, immunohistochemistry studies indicate that CB1 expression increases progressively across layers II and III, forms a distinct band in layer IV, falls sharply in layer V and increases again in layer VI (Eggan et al., 2008). From a functional standpoint, this scenario is further complicated by the fact that the human CB1 receptor presents two splicing variants diverging in their amino-terminus sequences (Ryberg et al., 2005). When expressed in hippocampal neurons cultured from CB−/− mice, each variant exhibits distinct signaling properties and produces a less pronounced inhibition of synaptic transmission relative to rodent CB1 (Straiker et al., 2011).

Unlike CB1, CB2 receptors are primarily localized in immuno-competent cells, of which they modulate the mobility and function (Ramirez et al., 2005;Walter and Stella, 2004). Although small levels of CB2 have been detected in the basal ganglia and glial cells, their presence and distribution in neuronal populations of the brain is still debated (Sagredo et al., 2009Onaivi, 2011). Several studies have shown that CB2 receptors are generally upregulated in astrocytes and microglia in response to disease-related neuroinflammatory events, and after neurotoxic insults (Fernandez-Ruiz, 2009Palazuelos et al., 2009Price et al., 2009).

The persistence of cannabinoid-like effects after administration of cannabinoid agonists in CB1−/− and CB2−/− mice, as well as in mutant mice lacking CB1 receptors in neuronal subpopulations, clearly indicates that these drugs recognize other non-CB1/CB2 targets in the CNS (Marsicano et al., 2002Begg et al., 2005Brown, 2007). Among these targets, the orphan receptor GPR55 has received a lot of attention in the last decade (Godlewskiet al., 2009Sawzdargo et al., 1999). GPR55 mRNA is predominantly expressed in the striatum and, to a lesser extent, in the hippocampus, thalamus and cerebellum (Sawzdargo et al., 1999). These data, however, have not been validated by measuring the corresponding protein levels, and neither AEA nor 2-AG have shown consistent pharmacological effects upon stimulation of GPR55 receptors (Yin et al., 2009). Furthermore, GPR55 is activated, rather than inhibited, by the CB1 antagonists rimonabant and AM251, and blocked by the cannabinoid agonist CP55,940 (Johns et al., 2007Kapur et al., 2009Ryberg et al., 2007). Therefore, as GPR55 is phylogenetically distinct from CB1/CB2 receptors and is activated by the non-cannabinoid endogenous ligand, lysophosphatidylinositol, this receptor is currently viewed as a non-CB receptor with a binding side for cannabinoid ligands (McPartland et al., 2006Henstridge et al., 2011).

1.2 TRP channels and other non-CB1/CB2 receptors

Exogenous and endogenous cannabinoids interact with at least five distinct transient receptor potential (TRP) receptors, which are ligand-gated ion channels generating a cation inward flow upon activation (Starowicz et al., 2007Patapoutian et al., 2009). AEA binds to the VR1 subtype (TRPV1) with low-affinity, and inhibition of the AEA degrading enzyme FAAH has been shown to enhance the efficacy and potency of this EC at TRPV1 (Huang et al., 2002Szolcsanyi, 2000De Petrocellis et al., 2001). Interestingly, elevation of AEA concentrations by pharmacological or genetic inhibition of FAAH reduced 2-AG levels via TRPV1 receptors (Maccarrone et al., 2008). Similarly, direct stimulation of TRPV1 channels mimicked the effects of endogenous AEA on 2-AG levels through a glutathione-dependent pathway. Since AEA and 2-AG are considered the primary ECs for reducing glutamate and GABAergic inputs to striatal neurons (Gerdeman et al., 2002Gubelliniet al., 2002Jung et al., 2005), the ability of AEA to affect 2-AG levels via TRPV1 channels may represent a mechanism to integrate excitatory and inhibitory inputs in the basal ganglia.

Several cannabinoid compounds, including WIN55212-2, AEA, noladin ether and virodhamine, have been shown to activate PPAR receptors, which are characterized by a large ligand-binding domain and relative selectivity (Sunet al., 2007). PPAR are ligand-activated transcription factors that form heterodimers with the retinoid X receptor and enhance the expression of several target genes. The three isoforms, alfa, beta/delta and gamma, are expressed in neuronal and glial cells of the PNS and CNS (Cimini et al., 2005Moreno et al., 2004). In particular, PPARα have been found in TH+cells of the substantia nigra pars compacta and dorsal striatum (Galan-Rodriguez et al., 2009). Low levels of PPARγ expression have been detected in the ventral mesencephalon and striatum (Breidert et al., 2002), although it is unclear whether this immunoreactivity co-localizes with neuronal or glial markers.

2. Endocannabinoid transmission in the basal ganglia: Relevance for PD

In the basal ganglia, pharmacological, neurochemical and electrophysiological data indicate that ECs act as retrograde signaling molecules at GABAergic and glutamatergic synapses, and promote depolarization-induced suppression of excitation (DSE) and inhibition (DSI) at striatal synapses (Adermark et al., 2009Gerdeman et al., 2002Gubelliniet al., 2002Köfalvi et al., 2005Kreitzer and Malenka, 2007). Several lines of evidence also indicate the existence of a cross talk between the EC and dopaminergic systems. Indeed, ECs can affect dopamine release in vivo and modulate the firing activity of dopaminergic neurons by acting at TRPV1 or PPAR receptors (Cheer et al., 2004Price et al., 2007Solinas et al., 2006;Melis et al., 2008de Lago et al., 2004Marinelli et al., 2003). Also, stimulation of dopamine receptors has been shown to increase the levels of striatal AEA, which may serve as an inhibitory feedback signal countering dopamine-induced motor activity (Giuffrida et al., 1999Beltramo et al., 2000Ferrer et al., 2003).

2.1. Cannabinoid effects in PD

The EC ability to modulate neurotransmission and synaptic plasticity in the basal ganglia circuitries has spurred interest to develop cannabinoid–based therapies to treat PD, a neurodegerative disorder characterized by progressive loss of nigrostriatal neurons, maladaptive striatal plasticity and disabling motor disturbances (Dauer and Przedborski, 2003). Increased CB1 mRNA and receptor binding have been reported in primate and rodent models of PD (Lastres-Becker et al., 2001Romero et al., 2000). Although elevated EC levels have been found in the striatum of dopamine-depleted rats (Di Marzoet al., 2000Gubellini et al., 2002), other studies carried out in rats treated with the neurotoxin 6-hydroxidopamine (6-OHDA) have shown decreased AEA tone (Ferrer et al., 2003Kreitzer and Malenka, 2007Morgese et al., 2007). In these animals, administration of levodopa, the mainstay treatment for PD, failed to elevate AEA levels (Ferrer et al., 2003Morgese et al., 2007) and caused a further upregulation of striatal CB1 receptors (Zeng et al., 1999), suggesting that levodopa does not correct the EC abnormalities associated with nigro-striatal degeneration.

So far, studies on the effects of cannabinoid agonists and antagonists on PD motor symptoms have produced conflicting results, and there is no general consensus whether cannabinoid-based therapies might be beneficial in PD (Cao et al., 2007Meschler et al., 2001Mesnage et al., 2004Papa, 2008;van der Stelt et al., 2005). These discrepancies are possibly due to species-specific differences across PD models and/or to the specific physiological state of the animals at the time of the experiments, which may both affect EC transmission. Nevertheless, cannabinoid drugs may delay PD progression and the underlying neuroinflammatory process by modulating cell-mediated inflammatory and brain immune responses via cannabinoid receptor-dependent and -independent mechanisms (Molina-Holgado et al., 2003;Price et al., 2009Ramirez et al., 2005Sancho et al., 2003Walter and Stella, 2004).

Interestingly, chronic stimulation of CB2 receptors has been shown to protect against MPTP-induced nigrostriatal degeneration by inhibiting microglial activation infiltration, whereas CB2 genetic ablation exacerbated MPTP systemic toxicity (Price et al., 2009). These observations confirm previous reports in 6-OHDA-treated rats showing CB1-independent neuroprotective effects of cannabinoids (Garcia-Arencibia et al., 2007Lastres-Becker et al., 2005). They also suggest that, unlike other neurodegenerative conditions, such as cerebral ischemia and brain trauma, activation of CB2, rather than CB1 receptors may be a more effective pharmacological strategy to slow down or halt nigrostriatal degeneration (Marsicano et al., 2003Nagayama et al., 1999Panikashvili et al., 2001).

Cannabinoids can also exert neuroprotective actions via their anti-oxidant properties, or by encouraging the proliferation and differentiation of progenitor cells in neurogenic areas (Marsicano et al., 2002Lastres-Beckeret al., 2005Galve-Roperh et al., 2007).

Recent data point to MAGL as a metabolic node controlling brain prostaglandin production in neuroinflammatory states (Nomura et al., 2011). Specifically, MAGL Inhibition has been shown to suppress the inflammatory cascade associated with MPTP toxicity in a CB1/CB2-independent manner, and to protect against dopaminergic neuronal loss possibly by preventing 2-AG conversion into pro-inflammatory prostaglandins (Nomura et al., 2011). If confirmed in clinical settings, this approach appears particularly promising, as MAGL inhibitors do not show the gastrointestinal toxicity generally associated with other anti-inflammatory drugs, such as COX1 inhibitors. Long-term administration of MAGL inhibitors, however, may lead to CB1 desensitization and impair EC-dependent synaptic plasticity, which in turn may limit their application in the clinic (Stella, 2011).

2.2 Studies on levodopa-induced dyskinesias

In addition to their possible use as adjunctive therapy in PD, cannabinoid drugs are emerging as promising antidyskinetic agents, given their ability to reduce levodopa–induced abnormal involuntary movements (AIMs) in rodent and primate models (Morgese et al., 2007Walsh et al., 2010Cao et al., 2007). This beneficial effect may result from a combination of multiple downstream actions, including: 1) normalization of aberrant neurotransmission and synaptic plasticity (Chevaleyre et al., 2007Kreitzer and Malenka, 2007Mato et al., 2008Morgese et al., 2009Picconi et al., 2003); and 2) inhibition of levodopa-induced activation of cAMP/DARPP32 signaling, which is overactive in dyskinetic animals (Martinez et al., 2011;Picconi et al., 2003Santini et al., 2008). Interestingly, inhibition of presynaptic PKA activity is required for EC-mediated long-term depression in rat midbrain dopamine neurons (Haj-Dahmane and Shen, 2010). Thus, given the deficient AEA production observed in dyskinetic 6-OHDA-treated rats, activation of CB1 receptors could rebalance the striatal maladaptive plasticity by inhibiting levodopa-induced PKA hyperactivity and reducing glutamatergic input at striatal synapses, which is increased in dyskinetic animals (Gubellini et al., 2002).

Although some reports have shown that even CB1 antagonists can reduce levodopa-induced dyskinesias in reserpine-treated rats and MPTP-treated marmosets (Segovia et al., 2003van der Stelt et al., 2005), these results have not been confirmed in other animal models of PD (Cao et al., 2007Martinezet al., 2011Walsh et al., 2010). Similarly, the observation that DARPP-32 activation (which implies the phosphorylation of this protein at threonine 34) is required for the expression of cannabinoid-mediated motor effects (Andersson et al., 2005), has not been confirmed in dyskinetic 6-OHDA-treated rats. In these animals, indeed, the cannabinoid agonist WIN55212-2 significantly reduced levodopa-induced AIMs via activation of CB1 receptors, but reversed levodopa-induced DARPP-32 phosphorylation at Thr34 through a mechanism that was not fully reversed by CB1 antagonism (Martinez et al., 2011). Interestingly, Polissidis et al. (2010) observed that the dose of WIN55,212-2 used in the study of Martinez and co-workers (1 mg/kg, i.p.) induced opposite changes in striatal Thr-34 phosphorylation in different rat strains. The reasons for these discrepancies require further investigation, and reveal some limitations in generalizing cannabinoid function/effects across different animal species, which in turn may complicate the translation of these findings into therapeutic interventions.

In this context, a double-blind, placebo-controlled trial carried out in 19 PD patients failed to recognize any antidiskinetic effect of oral cannabis extracts (Carroll et al., 2004). This assessment, however, was based on data reported from patients, which are often inaccurate in identifying symptoms (Vitale et al., 2001). It is also important to point out that cannabis has a more complex pharmacological profile than synthetic cannabinoid agonists, as well as highly variable pharmacokinetics and pharmacodynamics, which may account for the lack of effect in the study of Carroll and co-workers.

Inhibition of the AEA degrading enzyme FAAH, a pharmacological approach that limits the activation of CB1 receptors to those brain areas where EC production and release occurs, failed to produce anti-dyskinetic effects in 6-OHDA-treated rats (Morgese et al., 2007). These findings suggest that AEA elevation is not sufficient to attenuate levodopa-induced dyskinesias, possibly because of its concomitant action at TRPV1 receptors (Ross, 2003). In support of this hypothesis, systemic administration of the FAAH inhibitor URB597 in combination with the TRPV1 antagonist capsazepine produced a significant anti-dyskinetic effect, suggesting that the beneficial actions of CB1stimulation may be counteracted by TRPV1 agonism. These data, however, differ from those reported by Lee et al. (2006), which showed that administration of URB597 alone, or stimulation of TRPV1 receptors by capsaicin, can attenuate levodopa-induced hyperactivity in reserpine-treated rats. As previously mentioned, these discrepancies may be attributed to differences in the biological and pathological aspects of PD reproduced by different animal models, and/or to the use of behavioral outcomes (i.e., vertical motor activity) that model stereotypies rather than dyskinesias (Cenciet al., 2002).

3. Endocannabinoid transmission in schizophrenia

Schizophrenia is a severe mental illness characterized by three main types of symptoms: positive (e.g., hallucinations, delusions), negative (e.g., social withdrawal, anhedonia), and cognitive deficits (e.g., impaired working memory and attention).

Since THC produced perceptual alterations similar to those observed in psychotic patients, Emrich and co-workers proposed that abnormalities in the EC system might contribute to the pathogenesis of schizophrenia, at least in a subgroup of patients (Emrich et al., 1997). This “cannabinoid hypothesis” of schizophrenia is consistent with the commonly accepted view that cannabis exposure has a negative impact on the course and expression of psychoses (D’Souza, 2007Sewell et al., 2009).

3.1 Studies on CB1 receptors

Recent investigations have established an association between some CB1receptor gene polymorphisms and the hebephrenic type of schizophrenia (Chavarria-Siles et al., 2008Ujike and Morita, 2004). By using radioactive cannabinoid agonists ([3H]CP55,940) or antagonists ([3H]SR141716A), post-mortem studies have shown increased CB1 binding in prefrontal cortex areas of schizophrenic patients (Dalton et al., 2011Dean et al., 2001Newell et al., 2006Zavitsanou et al., 2004). Although these reports support the “cannabinoid hypothesis” of schizophrenia, new experimental evidence has challenged these findings and suggested that CB1 abnormalities may be related, at best, to specific disease subtypes, or result from the chronic use of antipsychotic medications (Dalton et al., 2011Zuardi et al., 2011Uriguen et al., 2009). In addition, direct measurements of CB1 mRNA and protein have not confirmed the CB1 up-regulation in the anterior cingulate cortex (Koetheet al., 2007), but found decreased CB1 density in the dorsolateral prefrontal cortex (areas 9 and 46) (Eggan et al., 2008). Finally, a recent PET imaging study has suggested that CB1 binding increases with the severity of positive symptoms, but is inversely correlated to negative symptoms (Wong et al., 2010). These observations indicate that future clinical investigations should be ideally carried out in better-defined, and possibly drug-free, groups of subjects with similar symptom severity.

3.2 Endocannabinoid levels and schizophrenic symptoms

Several studies have reported increased AEA levels in the blood and cerebrospinal fluid (CSF) of schizophrenic patients (Leweke et al., 1999De Marchi et al., 2003Giuffrida et al., 2004), and clinical remission induced by the atypical antipsychotic olanzapine has been associated with a significant drop in circulating AEA (De Marchi et al., 2003). In other studies, patients treated with typical antipsychotics did not show elevated CSF AEA, whereas those receiving atypical antipsychotics (including olanzapine) had increased levels similar to those observed in drug-naïve schizophrenics (Giuffrida et al., 2004). Nevertheless, since these studies were carried out in acute paranoid schizophrenics, a patient population showing the highest CB1 density in the dorsolateral prefrontal cortex (Dalton et al., 2011), it is unclear whether these changes in AEA levels can be generalized to other subgroups of schizophrenic patients.

Surprisingly, the elevation of CSF AEA in drug-naïve schizophrenics has been negatively correlated to the severity of negative symptoms (Giuffrida et al., 2004Leweke et al., 2007). Thus, the idea that AEA might play a possible anti-psychotic role contrasts with the assumptions of the “cannabinoid hypothesis” of schizophrenia, and suggests that the association between cannabinoids and mental disorders is more complex than originally postulated.

3.3 Studies in animal models

So far, a limited number of animal models of schizophrenia have been used to clarify the contribution of EC dysfunction to schizophrenic symptoms. The psychostimulant phencyclidine (PCP) is known to produce behavior abnormalities that are almost indistinguishable from those observed in schizophrenia (Murray, 2002Steinpreis, 1996). Thus, PCP administration in rodents has been proposed as a valuable pharmacological approach to induce behavioral phenotypes modeling the three main categories of schizophrenic symptoms (Enomoto et al., 2007Jentsch and Roth, 1999). The psychotogenic effects of PCP, however, vary greatly according to the treatment regimen, and only chronic exposure to the drug produces schizophrenia-like behavioral deficits and neurochemical changes (Jentsch and Roth, 1999). Moreover, as PCP-induced symptoms persist for several weeks after drug discontinuation (Murray, 2002Qiao et al., 2001), it is preferable to add a washout period after chronic PCP to avoid possible interactions with the drugs to be tested. By using this approach, Giuffrida and Seillier (2009) showed that sub-chronic PCP administration (5 mg/kg, b.i.d., i.p., for 7 days, followed by a 7-day washout) produced not only a behavioral phenotype reminiscent of negative (social withdrawal) and positive (enhanced amphetamine-induced hyperactivity) symptoms, but also the characteristic cognitive deficit (impaired working memory) observed in the disease. In line with the clinical observations, several studies have also shown that atypical antipsychotics reverse social withdrawal and cognitive deficits in this model (Qiao et al., 2001Grayson et al., 2007Hashimoto et al., 2005McLean et al., 2008).

Globally, repeated PCP administration did not alter CB1 receptor expression in brain areas relevant to schizophrenia, such as the medial prefrontal cortex, anterior cingulate cortex, caudate-putamen, nucleus accumbens, hippocampus, and ventral tegmental area (VTA) (Seillier et al., 2010). In the latter region, Vigano et al. (2009) and Guidali et al. (2011) found increased CB1 expression using a different PCP regimen (2.58 mg/kg, once daily followed by a 72h-washout period), which does not produce social withdrawal (Egerton et al., 2008).

In PCP-treated rats, no changes in CSF or brain AEA levels were observed, with the exception of an increase of AEA in the nucleus accumbens (Seillier et al., 2010Guidali et al., 2011Vigano et al., 2009). The discrepancy between the EC levels measured in the PCP rat model versus drug-naive paranoid schizophrenics, in which CSF AEA is elevated, might reflect a specific feature of this patient subgroup, or depend on the different end-points used in these studies. Specifically, the human samples were obtained immediately after the first psychotic episode, which is accompanied by striatal hyperdopaminergia (Laruelle et al., 2003) and may lead in turn to increased AEA production (Giuffrida et al., 1999). On the other hand, the rat samples were collected from animals resting in their home cages, a condition that is unlikely to affect AEA levels, as EC release occurs in response to neuronal activity (Piomelli, 2003). In support of this observation, other studies have reported 2-AG elevation in the prefrontal cortex of PCP-treated rats sacrificed immediately after a memory test (Guidali et al., 2011Vigano et al., 2009), thus stressing the importance of the physiological state of the animal at the time of EC measurements.

As suggested by the inverse correlation between CSF AEA and the negative symptoms of schizophrenia (Giuffrida et al., 2004), elevation of AEA tone via administration of URB597 reversed social withdrawal in PCP-treated rats (Seillier et al., 2010). The same drug, however, decreased social interaction in control rats to an extent comparable to that observed after PCP treatment (Seillier et al., 2010). In agreement with these observations, Spano et al. (2010) showed that WIN55,212-2 self-administration attenuated PCP-induced deficits in sociability, but caused social withdrawal in saline-treated controls, strengthening the idea of a beneficial action of cannabinoids only in pathological conditions.

From this picture, it appears that the role played by the EC system in schizophrenia greatly varies depending on the type of symptoms (positive versus negative) and/or diagnosis (paranoid versus hebephrenic). In addition, the exposure to cannabinoid drugs produces clearly different effects in healthy versus pathological subjects. Therefore, further investigations in preclinical models, as well as in clinical settings, are still needed to understand the precise contributions of the EC system to psychoses.

 

Concluding remarks

Given their ability to modulate dopamine transmission and affect synaptic plasticity in cortico-striatal and limbic circuits, cannabinoid-base pharmacotherapies represent a promising treatment for psychomotor disorders, such as PD and schizophrenia. In addition, as cannabinoid drugs have clear anti-inflammatory properties and promote adult neuro- and glio-genesis (Solbrig et al., 2010), they may find possible applications in neurodegenerative syndromes, including NeuroAIDS and diseases of aging.

While it is still unclear what could be the net effect produced by stimulation of cannabinoid receptors on PD motor symptoms, direct cannabinoid agonists have shown anti-dyskinetic properties in preclinical studies, and manipulation of AEA levels has produced antipsychotic-like effects in animal models of schizophrenia.

Further work is required to explore the therapeutic potentials of targets other than traditional CB1/CB2 receptors, such as TRP channels, PPAR receptors and EC degrading enzymes, which may offer an opportunity for more specific pharmacological interventions than those provided by direct-acting cannabimimetics.

Highlights

  • Cannabinoid receptors are a promising pharmacological target for anti-dyskinetic and neuroprotective therapies in Parkinson’s disease
  • Animal models suggest the existence of disturbed endocannabinoid transmission in schizophrenia
  • In humans, altered endocannabinoid transmission may occur in specific schizophrenia subtypes.

Acknowledgments

This work was supported by the National Institute of Health, NS050401-07 and MH9113001-A1 to A.G.

Abbreviations

AEA
anandamide
2-AG
2-Arachidonoyl Glycerol
CSF
Cerebrospinal Fluid
DSE
Depolarization-induced Suppression of Excitation
DSI
Depolarization-induced Suppression of Inhibition
EC
Endocannabinoid
FAAH
Fatty Acid Amide Hydrolase
MSN
Medium Spiny Neurons
PPAR
Peroxisome Proliferator-activated Receptors
PCP
Phencyclidin
PD
Parkinson’s Disease
PET
Positron Emission Tomography
SSI
Slow Self Inhibition
THC
Tetrahydrocannabinol
TRP
Transient Receptor Potential receptors
VTA
Ventral Tegmental Area

Footnotes

Publisher’s Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

 

References

  • Adermark L, Talani G, Lovinger DM. Endocannabinoid-dependent plasticity at GABAergic and glutamatergic synapses in the striatum is regulated by synaptic activity. Eur J Neurosci. 2009;29:32–41.[PMC free article] [PubMed]
  • Andersson M, Usiello A, Borgkvist A, Pozzi L, Dominguez C, Fienberg AA, Svenningsson P, Fredholm BB, Borrelli E, Greengard P, Fisone G. Cannabinoid action depends on phosphorylation of dopamine- and cAMP-regulated phosphoprotein of 32 kDa at the protein kinase A site in striatal projection neurons. J Neurosci. 2005;25:8432–8438.[PubMed]
  • Atwood BK, Wager-Miller J, Haskins C, Straiker A, Mackie K. Functional selectivity in CB(2) cannabinoid receptor signaling and regulation: implications for the therapeutic potential of CB(2) ligands. Mol Pharmacol. 2012;81:250–263. [PMC free article][PubMed]
  • Begg M, Pacher P, Batkai S, Osei-Hyiaman D, Offertaler L, Mo FM, Liu J, Kunos G. Evidence for novel cannabinoid receptors.Pharmacol Ther. 2005;106:133–145. [PubMed]
  • Beltramo M, de Fonseca FR, Navarro M, Calignano A, Gorriti MA, Grammatikopoulos G, Sadile AG, Giuffrida A, Piomelli D. Reversal of dopamine D(2) receptor responses by an anandamide transport inhibitor. J Neurosci. 2000;20:3401–3407. [PubMed]
  • Beltramo M, Piomelli D. Carrier-mediated transport and enzymatic hydrolysis of the endogenous cannabinoid 2-arachidonylglycerol.NeuroReport. 2000;11:1231–1235. [PubMed]
  • Bisogno T, MacCarrone M, De Petrocellis L, Jarrahian A, Finazzi-Agro A, Hillard C, Di Marzo V. The uptake by cells of 2-arachidonoylglycerol, an endogenous agonist of cannabinoid receptors. Eur J Biochem. 2001;268:1982–1989. [PubMed]
  • Bisogno T, Melck D, Bobrov M, Gretskaya NM, Bezuglov VV, De Petrocellis L, Di Marzo V. N-acyl-dopamines: novel synthetic CB(1) cannabinoid-receptor ligands and inhibitors of anandamide inactivation with cannabimimetic activity in vitro and in vivo.Biochem J. 2000;351(Pt 3):817–824. [PMC free article] [PubMed]
  • Breidert T, Callebert J, Heneka MT, Landreth G, Launay JM, Hirsch EC. Protective action of the peroxisome proliferator-activated receptor-gamma agonist pioglitazone in a mouse model of Parkinson’s disease. J Neurochem. 2002;82:615–624. [PubMed]
  • Brown AJ. Novel cannabinoid receptors. Br J Pharmacol.2007;152:567–575. [PMC free article] [PubMed]
  • Cao X, Liang L, Hadcock JR, Iredale PA, Griffith DA, Menniti FS, Factor S, Greenamyre JT, Papa SM. Blockade of cannabinoid type 1 receptors augments the antiparkinsonian action of levodopa without affecting dyskinesias in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated rhesus monkeys. J Pharmacol Exp Ther.2007;323:318–326. [PubMed]
  • Carroll CB, Bain PG, Teare L, Liu X, Joint C, Wroath BA, Parkin SG, Fox P, Wright D, Hobart J, Zajicek JP. Cannabis for dyskinesia in Parkinson disease. Neurology. 2004;63:1245–1250. [PubMed]
  • Cenci MA, Whishaw IQ, Schallert T. Animal models of neurological deficits: how relevant is the rat? Nat Rev Neurosci. 2002;3:574–579.[PubMed]
  • Chanda PK, Gao Y, Mark L, Btesh J, Strassle BW, Lu P, et al. Monoacylglycerol lipase activity is a critical modulator of the tone and integrity of the endocannabinoid system. Mol Pharmacol.2010;78:996–1003. [PubMed]
  • Chavarria-Siles I, Contreras-Rojas J, Hare E, Walss-Bass C, Quezada P, Dassori A, Contreras S, Medina R, Ramirez M, Salazar R, Raventos H, Escamilla MA. Cannabinoid receptor 1 gene (CNR1) and susceptibility to a quantitative phenotype for hebephrenic schizophrenia. Am J Med Genet B Neuropsychiatr Genet.2008;147:279–284. [PubMed]
  • Cheer JF, Wassum KM, Heien ML, Phillips PE, Wightman RM. Cannabinoids enhance subsecond dopamine release in the nucleus accumbens of awake rats. J Neurosci. 2004;24:4393–4400.[PubMed]
  • Chevaleyre V, Heifets BD, Kaeser PS, Sudhof TC, Castillo PE. Endocannabinoid-mediated long-term plasticity requires cAMP/PKA signaling and RIM1alpha. Neuron. 2007;54:801–812.[PMC free article] [PubMed]
  • Cimini A, Benedetti E, Cristiano L, Sebastiani P, D’Amico MA, D’Angelo B, Di Loreto S. Expression of peroxisome proliferator-activated receptors (PPARs) and retinoic acid receptors (RXRs) in rat cortical neurons. Neuroscience. 2005;130:325–337. [PubMed]
  • Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature. 1996;384:83–87.[PubMed]
  • D’Souza DC. Cannabinoids and psychosis. Int Rev Neurobiol.2007;78:289–326. [PubMed]
  • Dalton VS, Long LE, Weickert CS, Zavitsanou K. Paranoid schizophrenia is characterized by increased CB1 receptor binding in the dorsolateral prefrontal cortex. Neuropsychopharmacology.2011;36:1620–1630. [PMC free article] [PubMed]
  • Dauer W, Przedborski S. Parkinson’s disease: mechanisms and models. Neuron. 2003;39:889–909. [PubMed]
  • de Lago E, de Miguel R, Lastres-Becker I, Ramos JA, Fernandez-Ruiz J. Involvement of vanilloid-like receptors in the effects of anandamide on motor behavior and nigrostriatal dopaminergic activity: in vivo and in vitro evidence. Brain Res. 2004;1007:152–159.[PubMed]
  • De Marchi N, De Petrocellis L, Orlando P, Daniele F, Fezza F, Di Marzo V. Endocannabinoid signalling in the blood of patients with schizophrenia. Lipids Health Dis. 2003;2:5. [PMC free article][PubMed]
  • De Petrocellis L, Bisogno T, Maccarrone M, Davis JB, Finazzi-Agro A, Di Marzo V. The activity of anandamide at vanilloid VR1 receptors requires facilitated transport across the cell membrane and is limited by intracellular metabolism. J Biol Chem. 2001;276:12856–12863.[PubMed]
  • Dean B, Sundram S, Bradbury R, Scarr E, Copolov D. Studies on [3H]CP-55940 binding in the human central nervous system: regional specific changes in density of cannabinoid-1 receptors associated with schizophrenia and cannabis use. Neuroscience.2001;103:9–15. [PubMed]
  • Di Marzo V, Hill MP, Bisogno T, Crossman AR, Brotchie JM. Enhanced levels of endogenous cannabinoids in the globus pallidus are associated with a reduction in movement in an animal model of Parkinson’s disease. FASEB J. 2000;14:1432–1438. [PubMed]
  • Dinh TP, Carpenter D, Leslie FM, Freund TF, Katona I, Sensi SL, Kathuria S, Piomelli D. Brain monoglyceride lipase participating in endocannabinoid inactivation. Proc Natl Acad Sci U S A.2002;99:10819–10824. [PMC free article] [PubMed]
  • Egerton A, Allison C, Brett RR, Pratt JA. Cannabinoids and prefrontal cortical function: insights from preclinical studies.Neurosci Biobehav Rev. 2006;30:680–695. [PubMed]
  • Egerton A, Reid L, McGregor S, Cochran SM, Morris BJ, Pratt JA. Subchronic and chronic PCP treatment produces temporally distinct deficits in attentional set shifting and prepulse inhibition in rats.Psychopharmacology (Berl) 2008;198:37–49. [PubMed]
  • Eggan SM, Hashimoto T, Lewis DA. Reduced cortical cannabinoid 1 receptor messenger RNA and protein expression in schizophrenia.Arch Gen Psychiatry. 2008;65:772–784. [PMC free article][PubMed]
  • Eggan SM, Melchitzky DS, Sesack SR, Fish KN, Lewis DA. Relationship of cannabinoid CB1 receptor and cholecystokinin immunoreactivity in monkey dorsolateral prefrontal cortex.Neuroscience. 2010;169:1651–1661. [PMC free article] [PubMed]
  • Emrich HM, Leweke FM, Schneider U. Towards a cannabinoid hypothesis of schizophrenia: cognitive impairments due to dysregulation of the endogenous cannabinoid system. Pharmacol Biochem Behav. 1997;56:803–807. [PubMed]
  • Enomoto T, Noda Y, Nabeshima T. Phencyclidine and genetic animal models of schizophrenia developed in relation to the glutamate hypothesis. Methods Find Exp Clin Pharmacol. 2007;29:291–301.[PubMed]
  • Fegley D, Kathuria S, Mercier R, Li C, Goutopoulos A, Makriyannis A, Piomelli D. Anandamide transport is independent of fatty-acid amide hydrolase activity and is blocked by the hydrolysis-resistant inhibitor AM1172. Proc Natl Acad Sci U S A. 2004;101:8756–8761.[PMC free article] [PubMed]
  • Fernandez-Ruiz J. The endocannabinoid system as a target for the treatment of motor dysfunction. Br J Pharmacol. 2009;156:1029–1040. [PMC free article] [PubMed]
  • Fernandez-Ruiz J, Gonzalez S. Cannabinoid control of motor function at the basal ganglia. In: Pertwee RG, editor. Handbook of Experimental Pharmacology. Springer-Verlag; Heidelberg: 2005. pp. 479–507. [PubMed]
  • Ferrer B, Asbrock N, Kathuria S, Piomelli D, Giuffrida A. Effects of levodopa on endocannabinoid levels in rat basal ganglia: implications for the treatment of levodopa-induced dyskinesias. Eur J Neurosci. 2003;18:1607–1614. [PubMed]
  • Freund TF, Katona I, Piomelli D. Role of endogenous cannabinoids in synaptic signaling. Physiol Rev. 2003;83:1017–1066. [PubMed]
  • Fusco FR, Martorana A, Giampa C, De March Z, Farini D, D’Angelo V, Sancesario G, Bernardi G. Immunolocalization of CB1 receptor in rat striatal neurons: a confocal microscopy study. Synapse.2004;53:159–167. [PubMed]
  • Galan-Rodriguez B, Suarez J, Gonzalez-Aparicio R, Bermudez-Silva FJ, Maldonado R, Robledo P, Rodriguez de Fonseca F, Fernandez-Espejo E. Oleoylethanolamide exerts partial and dose-dependent neuroprotection of substantia nigra dopamine neurons.Neuropharmacology. 2009;56:653–664. [PubMed]
  • Galve-Roperh I, Aguado T, Palazuelos J, Guzman M. The endocannabinoid system and neurogenesis in health and disease.Neuroscientist. 2007;13:109–114. [PubMed]
  • Garcia-Arencibia M, Gonzalez S, de Lago E, Ramos JA, Mechoulam R, Fernandez-Ruiz J. Evaluation of the neuroprotective effect of cannabinoids in a rat model of Parkinson’s disease: importance of antioxidant and cannabinoid receptor-independent properties. Brain Res. 2007;1134:162–170. [PubMed]
  • Gerdeman G, Lovinger DM. CB1 cannabinoid receptor inhibits synaptic release of glutamate in rat dorsolateral striatum. J Neurophysiol. 2001;85:468–471. [PubMed]
  • Gerdeman GL, Ronesi J, Lovinger DM. Postsynaptic endocannabinoid release is critical to long-term depression in the striatum. Nat Neurosci. 2002;5:446–451. [PubMed]
  • Giuffrida A, Leweke FM, Gerth CW, Schreiber D, Koethe D, Faulhaber J, Klosterkotter J, Piomelli D. Cerebrospinal anandamide levels are elevated in acute schizophrenia and are inversely correlated with psychotic symptoms. Neuropsychopharmacology.2004;29:2108–2114. [PubMed]
  • Giuffrida A, Parsons LH, Kerr TM, Rodriguez de Fonseca F, Navarro M, Piomelli D. Dopamine activation of endogenous cannabinoid signaling in dorsal striatum. Nat Neurosci. 1999;2:358–363.[PubMed]
  • Glaser ST, Abumrad NA, Fatade F, Kaczocha M, Studholme KM, Deutsch DG. Evidence against the presence of an anandamide transporter. Proc Natl Acad Sci U S A. 2003;100:4269–4274.[PMC free article] [PubMed]
  • Glass M, Felder CC. Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors augments cAMP accumulation in striatal neurons: evidence for a Gs linkage to the CB1 receptor. J Neurosci.1997;17:5327–5333. [PubMed]
  • Godlewski G, Offertaler L, Wagner JA, Kunos G. Receptors for acylethanolamides-GPR55 and GPR119. Prostaglandins Other Lipid Mediat. 2009;89:105–111. [PMC free article] [PubMed]
  • Grayson B, Idris NF, Neill JC. Atypical antipsychotics attenuate a sub-chronic PCP-induced cognitive deficit in the novel object recognition task in the rat. Behav Brain Res. 2007;184:31–38.[PubMed]
  • Gubellini P, Picconi B, Bari M, Battista N, Calabresi P, Centonze D, Bernardi G, Finazzi-Agro A, Maccarrone M. Experimental parkinsonism alters endocannabinoid degradation: implications for striatal glutamatergic transmission. J Neurosci. 2002;22:6900–6907.[PubMed]
  • Guidali C, Vigano D, Petrosino S, Zamberletti E, Realini N, Binelli G, Rubino T, Di Marzo V, Parolaro D. Cannabinoid CB1 receptor antagonism prevents neurochemical and behavioural deficits induced by chronic phencyclidine. Int J Neuropsychopharmacol.2011;14:17–28. [PubMed]
  • Haj-Dahmane S, Shen RY. Regulation of plasticity of glutamate synapses by endocannabinoids and the cyclic-AMP/protein kinase A pathway in midbrain dopamine neurons. J Physiol. 2010;588:2589–2604. [PMC free article] [PubMed]
  • Hanus L, Abu-Lafi S, Fride E, Breuer A, Vogel Z, Shalev DE, Kustanovich I, Mechoulam R. 2-arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci U S A. 2001;98:3662–3665. [PMC free article] [PubMed]
  • Hashimoto K, Fujita Y, Shimizu E, Iyo M. Phencyclidine-induced cognitive deficits in mice are improved by subsequent subchronic administration of clozapine, but not haloperidol. Eur J Pharmacol.2005;519:114–117. [PubMed]
  • Henstridge CM, Balenga NA, Kargl J, Andradas C, Brown AJ, Irving A, Sanchez C, Waldhoer M. Minireview: recent developments in the physiology and pathology of the lysophosphatidylinositol-sensitive receptor GPR55. Mol Endocrinol. 2011;25:1835–1848. [PubMed]
  • Herkenham M, Lynn AB, Johnson MR, Melvin LS, de Costa BR, Rice KC. Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci.1991;11:563–583. [PubMed]
  • Hillard CJ, Jarrahian A. Cellular accumulation of anandamide: consensus and controversy. Br J Pharmacol. 2003;140:802–808.[PMC free article] [PubMed]
  • Hohmann AG, Suplita RL, Bolton NM, Neely MH, Fegley D, Mangieri R, Krey JF, Walker JM, Holmes PV, Crystal JD, Duranti A, Tontini A, Mor M, Tarzia G, Piomelli D. An endocannabinoid mechanism for stress-induced analgesia. Nature. 2005;435:1108–1112. [PubMed]
  • Hojo M, Sudo Y, Ando Y, Minami K, Takada M, Matsubara T, Kanaide M, Taniyama K, Sumikawa K, Uezono Y. mu-Opioid receptor forms a functional heterodimer with cannabinoid CB1 receptor: electrophysiological and FRET assay analysis. J Pharmacol Sci. 2008;108:308–319. [PubMed]
  • Huang SM, Bisogno T, Trevisani M, Al-Hayani A, De Petrocellis L, Fezza F, Tognetto M, Petros TJ, Krey JF, Chu CJ, Miller JD, Davies SN, Geppetti P, Walker JM, Di Marzo V. An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc Natl Acad Sci U S A. 2002;99:8400–8405.[PMC free article] [PubMed]
  • Jentsch JD, Roth RH. The neuropsychopharmacology of phencyclidine: from NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia. Neuropsychopharmacology.1999;20:201–225. [PubMed]
  • Johns DG, Behm DJ, Walker DJ, Ao Z, Shapland EM, Daniels DA, Riddick M, Dowell S, Staton PC, Green P, Shabon U, Bao W, Aiyar N, Yue TL, Brown AJ, Morrison AD, Douglas SA. The novel endocannabinoid receptor GPR55 is activated by atypical cannabinoids but does not mediate their vasodilator effects. Br J Pharmacol. 2007;152:825–831. [PMC free article] [PubMed]
  • Julian MD, Martin AB, Cuellar B, Rodriguez De Fonseca F, Navarro M, Moratalla R, Garcia-Segura LM. Neuroanatomical relationship between type 1 cannabinoid receptors and dopaminergic systems in the rat basal ganglia. Neuroscience. 2003;119:309–318. [PubMed]
  • Jung KM, Mangieri R, Stapleton C, Kim J, Fegley D, Wallace M, Mackie K, Piomelli D. Stimulation of endocannabinoid formation in brain slice cultures through activation of group I metabotropic glutamate receptors. Mol Pharmacol. 2005;68:1196–1202. [PubMed]
  • Kapur A, Zhao P, Sharir H, Bai Y, Caron MG, Barak LS, Abood ME. Atypical responsiveness of the orphan receptor GPR55 to cannabinoid ligands. J Biol Chem. 2009;284:29817–29827.[PMC free article] [PubMed]
  • Kearn CS, Blake-Palmer K, Daniel E, Mackie K, Glass M. Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors enhances heterodimer formation: a mechanism for receptor cross-talk? Mol Pharmacol. 2005;67:1697–1704. [PubMed]
  • Koethe D, Llenos IC, Dulay JR, Hoyer C, Torrey EF, Leweke FM, Weis S. Expression of CB1 cannabinoid receptor in the anterior cingulate cortex in schizophrenia, bipolar disorder, and major depression. J Neural Transm. 2007;114:1055–1063. [PubMed]
  • Köfalvi A, Rodrigues RJ, Ledent C, Mackie K, Vizi ES, Cuhna RA, Sperlagh B. Involvement of cannabinoid receptors in the regulation of neurotransmitter release in the rodent striatum: a combined immunochemical and pharmacological analysis. The Journal of Neuroscience. 2005;25:2874–2884. [PubMed]
  • Kreitzer AC, Malenka RC. Endocannabinoid-mediated rescue of striatal LTD and motor deficits in Parkinson’s disease models.Nature. 2007;445:643–647. [PubMed]
  • Kreitzer AC, Regehr WG. Retrograde inhibition of presynaptic calcium influx by endogenous cannabinoids at excitatory synapses onto Purkinje cells. Neuron. 2001;29:717–727. [PubMed]
  • Kreitzer FR, Stella N. The therapeutic potential of novel cannabinoid receptors. Pharmacol Ther 2009 [PMC free article] [PubMed]
  • Laruelle M, Kegeles LS, Abi-Dargham A. Glutamate, dopamine, and schizophrenia: from pathophysiology to treatment. Ann N Y Acad Sci. 2003;1003:138–158. [PubMed]
  • Lastres-Becker I, Cebeira M, de Ceballos ML, Zeng BY, Jenner P, Ramos JA, Fernandez-Ruiz JJ. Increased cannabinoid CB1 receptor binding and activation of GTP-binding proteins in the basal ganglia of patients with Parkinson’s syndrome and of MPTP-treated marmosets. Eur J Neurosci. 2001;14:1827–1832. [PubMed]
  • Lastres-Becker I, Molina-Holgado E, Ramos JA, Mechoulam R, Fernandez-Ruiz J. Cannabinoids provide neuroprotection against 6-hydroxydopamine toxicity in vivo and in vitro: relevance to Parkinson’s disease. Neurobiol Dis. 2005;19:96–107. [PubMed]
  • Laviolette SR, Grace AA. The roles of cannabinoid and dopamine receptor systems in neural emotional learning circuits: implications for schizophrenia and addiction. Cell Mol Life Sci. 2006;63:1597–1613. [PubMed]
  • Lee J, Di Marzo V, Brotchie JM. A role for vanilloid receptor 1 (TRPV1) and endocannabinnoid signalling in the regulation of spontaneous and L-DOPA induced locomotion in normal and reserpine-treated rats. Neuropharmacology. 2006;51:557–565.[PubMed]
  • Leweke FM, Giuffrida A, Koethe D, Schreiber D, Nolden BM, Kranaster L, Neatby MA, Schneider M, Gerth CW, Hellmich M, Klosterkotter J, Piomelli D. Anandamide levels in cerebrospinal fluid of first-episode schizophrenic patients: impact of cannabis use.Schizophr Res. 2007;94:29–36. [PubMed]
  • Leweke FM, Giuffrida A, Wurster U, Emrich HM, Piomelli D. Elevated endogenous cannabinoids in schizophrenia. Neuroreport.1999;10:1665–1669. [PubMed]
  • Liu J, Wang L, Harvey-White J, Huang BX, Kim HY, Luquet S, Palmiter RD, Krystal G, Rai R, Mahadevan A, Razdan RK, Kunos G. Multiple pathways involved in the biosynthesis of anandamide.Neuropharmacology. 2008;54:1–7. [PMC free article] [PubMed]
  • Long JZ, Li W, Booker L, Burston JJ, Kinsey SG, Schlosburg JE, Pavon FJ, Serrano AM, Selley DE, Parsons LH, Lichtman AH, Cravatt BF. Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects. Nat Chem Biol. 2009;5:37–44.[PMC free article] [PubMed]
  • Maccarrone M, Rossi S, Bari M, De Chiara V, Fezza F, Musella A, Gasperi V, Prosperetti C, Bernardi G, Finazzi-Agro A, Cravatt BF, Centonze D. Anandamide inhibits metabolism and physiological actions of 2-arachidonoylglycerol in the striatum. Nat Neurosci.2008;11:152–159. [PubMed]
  • Mailleux P, Vanderhaeghen JJ. Distribution of neuronal cannabinoid receptor in the adult rat brain: a comparative receptor binding radioautography and in situ hybridization histochemistry.Neuroscience. 1992;48:655–668. [PubMed]
  • Makara JK, Mor M, Fegley D, Szabo SI, Kathuria S, Astarita G, Duranti A, Tontini A, Tarzia G, Rivara S, Freund TF, Piomelli D. Selective inhibition of 2-AG hydrolysis enhances endocannabinoid signaling in hippocampus. Nat Neurosci. 2005;8:1139–1141.[PubMed]
  • Marinelli S, Di Marzo V, Berretta N, Matias I, Maccarrone M, Bernardi G, Mercuri NB. Presynaptic facilitation of glutamatergic synapses to dopaminergic neurons of the rat substantia nigra by endogenous stimulation of vanilloid receptors. J Neurosci.2003;23:3136–3144. [PubMed]
  • Marinelli S, Pacioni S, Bisogno T, Di Marzo V, Prince DA, Huguenard JR, Bacci A. The endocannabinoid 2-arachidonoylglycerol is responsible for the slow self-inhibition in neocortical interneurons. J Neurosci. 2008;28:13532–13541. [PMC free article] [PubMed]
  • Marrs WR, Blankman JL, Horne EA, Thomazeau A, Lin YH, Coy J, Bodor AL, Muccioli GG, Hu SS, Woodruff G, Fung S, Lafourcade M, Alexander JP, Long JZ, Li W, Xu C, Moller T, Mackie K, Manzoni OJ, Cravatt BF, Stella N. The serine hydrolase ABHD6 controls the accumulation and efficacy of 2-AG at cannabinoid receptors. Nat Neurosci. 2010;13:951–957. [PMC free article] [PubMed]
  • Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, Cannich A, Azad SC, Cascio MG, Gutierrez SO, van der Stelt M, Lopez-Rodriguez ML, Casanova E, Schutz G, Zieglgansberger W, Di Marzo V, Behl C, Lutz B. CB1 cannabinoid receptors and on-demand defense against excitotoxicity. Science. 2003;302:84–88. [PubMed]
  • Marsicano G, Wotjak CT, Azad SC, Bisogno T, Rammes G, Cascio MG, Hermann H, Tang J, Hofmann C, Zieglgansberger W, Di Marzo V, Lutz B. The endogenous cannabinoid system controls extinction of aversive memories. Nature. 2002;418:530–534. [PubMed]
  • Martin AB, Fernandez-Espejo E, Ferrer B, Gorriti MA, Bilbao A, Navarro M, Rodriguez de Fonseca F, Moratalla R. Expression and function of CB1 receptor in the rat striatum: localization and effects on D1 and D2 dopamine receptor-mediated motor behaviors.Neuropsychopharmacology. 2008;33:1667–1679. [PubMed]
  • Martinez A, Macheda T, Morgese MG, Trabace L, Giuffrida A. The cannabinoid agonist WIN55212–2 decreases l-DOPA-induced PKA activation and dyskinetic behavior in 6-OHDA-treated rats. Neurosci Res 2011 [PMC free article] [PubMed]
  • Mato S, Lafourcade M, Robbe D, Bakiri Y, Manzoni OJ. Role of the cyclic-AMP/PKA cascade and of P/Q-type Ca++ channels in endocannabinoid-mediated long-term depression in the nucleus accumbens. Neuropharmacology. 2008;54:87–94. [PubMed]
  • Matyas F, Yanovsky Y, Mackie K, Kelsch W, Misgeld U, Freund TF. Subcellular localization of type 1 cannabinoid receptors in the rat basal ganglia. Neuroscience. 2006;137:337–361. [PubMed]
  • McLean SL, Beck JP, Woolley ML, Neill JC. A preliminary investigation into the effects of antipsychotics on sub-chronic phencyclidine-induced deficits in attentional set-shifting in female rats. Behav Brain Res. 2008;189:152–158. [PubMed]
  • McPartland JM, Matias I, Di Marzo V, Glass M. Evolutionary origins of the endocannabinoid system. Gene. 2006;370:64–74. [PubMed]
  • Melis M, Pillolla G, Luchicchi A, Muntoni AL, Yasar S, Goldberg SR, Pistis M. Endogenous fatty acid ethanolamides suppress nicotine-induced activation of mesolimbic dopamine neurons through nuclear receptors. J Neurosci. 2008;28:13985–13994. [PMC free article][PubMed]
  • Meschler JP, Howlett AC, Madras BK. Cannabinoid receptor agonist and antagonist effects on motor function in normal and 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP)-treated non-human primates. Psychopharmacology (Berl) 2001;156:79–85. [PubMed]
  • Mesnage V, Houeto JL, Bonnet AM, Clavier I, Arnulf I, Cattelin F, Le Fur G, Damier P, Welter ML, Agid Y. Neurokinin B, neurotensin, and cannabinoid receptor antagonists and Parkinson disease. Clin Neuropharmacol. 2004;27:108– 110. [PubMed]
  • Molina-Holgado F, Pinteaux E, Moore JD, Molina-Holgado E, Guaza C, Gibson RM, Rothwell NJ. Endogenous interleukin-1 receptor antagonist mediates anti-inflammatory and neuroprotective actions of cannabinoids in neurons and glia. J Neurosci.2003;23:6470–6474. [PubMed]
  • Moreno S, Farioli-Vecchioli S, Ceru MP. Immunolocalization of peroxisome proliferator-activated receptors and retinoid X receptors in the adult rat CNS. Neuroscience. 2004;123:131–145. [PubMed]
  • Morgese MG, Cassano T, Cuomo V, Giuffrida A. Anti-dyskinetic effects of cannabinoids in a rat model of Parkinson’s disease: role of CB(1) and TRPV1 receptors. Exp Neurol. 2007;208:110–119.[PMC free article] [PubMed]
  • Morgese MG, Cassano T, Gaetani S, Macheda T, Laconca L, Dipasquale P, Ferraro L, Antonelli T, Cuomo V, Giuffrida A. Neurochemical changes in the striatum of dyskinetic rats after administration of the cannabinoid agonist WIN55,212- 2.Neurochem Int. 2009;54:56–64. [PMC free article] [PubMed]
  • Mu J, Zhuang SY, Kirby MT, Hampson RE, Deadwyler SA. Cannabinoid receptors differentially modulate potassium A and D currents in hippocampal neurons in culture. J Pharmacol Exp Ther.1999;291:893–902. [PubMed]
  • Muccioli GG, Xu C, Odah E, Cudaback E, Cisneros JA, Lambert DM, Lopez Rodriguez ML, Bajjalieh S, Stella N. Identification of a novel endocannabinoid-hydrolyzing enzyme expressed by microglial cells. J Neurosci. 2007;27:2883–2889. [PubMed]
  • Mukhopadhyay S, Howlett AC. Chemically distinct ligands promote differential CB1 cannabinoid receptor-Gi protein interactions. Mol Pharmacol. 2005;67:2016–2024. [PubMed]
  • Murray JB. Phencyclidine (PCP): a dangerous drug, but useful in schizophrenia research. J Psychol. 2002;136:319–327. [PubMed]
  • Nagayama T, Sinor AD, Simon RP, Chen J, Graham SH, Jin K, Greenberg DA. Cannabinoids and neuroprotection in global and focal cerebral ischemia and in neuronal cultures. J Neurosci.1999;19:2987–2995. [PubMed]
  • Newell KA, Deng C, Huang XF. Increased cannabinoid receptor density in the posterior cingulate cortex in schizophrenia. Exp Brain Res. 2006;172:556–560. [PubMed]
  • Nomura DK, Morrison BE, Blankman JL, Long JZ, Kinsey SG, Marcondes MC, Ward AM, Hahn YK, Lichtman AH, Conti B, Cravatt BF. Endocannabinoid hydrolysis generates brain prostaglandins that promote neuroinflammation. Science. 2011;334:809–813.[PMC free article] [PubMed]
  • Nyilas R, Dudok B, Urban GM, Mackie K, Watanabe M, Cravatt BF, Freund TF, Katona I. Enzymatic machinery for endocannabinoid biosynthesis associated with calcium stores in glutamatergic axon terminals. J Neurosci. 2008;28:1058–1063. [PubMed]
  • Onaivi ES. Commentary: Functional Neuronal CB2 Cannabinoid Receptors in the CNS. Curr Neuropharmacol. 2011;9:205–208.[PMC free article] [PubMed]
  • Palazuelos J, Aguado T, Pazos MR, Julien B, Carrasco C, Resel E, Sagredo O, Benito C, Romero J, Azcoitia I, Fernandez-Ruiz J, Guzman M, Galve-Roperh I. Microglial CB2 cannabinoid receptors are neuroprotective in Huntington’s disease excitotoxicity. Brain.2009;132:3152–3164. [PubMed]
  • Pan B, Wang W, Zhong P, Blankman JL, Cravatt BF, Liu QS. Alteration of endocannabinoid signaling, synaptic plasticity, learning and memoryin monoacylglycerol lipase knock-out mice. J Neurosci.2011;31:13420–13430. [PMC free article] [PubMed]
  • Panikashvili D, Simeonidou C, Ben-Shabat S, Hanus L, Breuer A, Mechoulam R, Shohami E. An endogenous cannabinoid (2-AG) is neuroprotective after brain injury. Nature. 2001;413:527–531.[PubMed]
  • Papa SM. The cannabinoid system in Parkinson’s disease: multiple targets to motor effects. Exp Neurol. 2008;211:334–338. [PubMed]
  • Patapoutian A, Tate S, Woolf CJ. Transient receptor potential channels: targeting pain at the source. Nat Rev Drug Discov.2009;8:55–68. [PMC free article] [PubMed]
  • Pertwee RG, Howlett AC, Abood ME, Alexander SP, Di Marzo V, Elphick MR, Greasley PJ, Hansen HS, Kunos G, Mackie K, Mechoulam R, Ross RA. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB and CB. Pharmacol Rev. 2010;62:588–631.[PMC free article] [PubMed]
  • Picconi B, Centonze D, Hakansson K, Bernardi G, Greengard P, Fisone G, Cenci MA, Calabresi P. Loss of bidirectional striatal synaptic plasticity in L-DOPA-induced dyskinesia. Nat Neurosci.2003;6:501–506. [PubMed]
  • 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. [PMC free article] [PubMed]
  • Piomelli D. The molecular logic of endocannabinoid signalling.Nature Reviews Neuroscience. 2003;4:873–884. [PubMed]
  • Polissidis A, Chouliara O, Galanopoulos A, Rentesi G, Dosi M, Hyphantis T, Marselos M, Papadopoulou-Daifoti Z, Nomikos GG, Spyraki C, Tzavara ET, Antoniou K. Individual differences in the effects of cannabinoids on motor activity, dopaminergic activity and DARPP-32 phosphorylation in distinct regions of the brain. Int J Neuropsychopharmacol. 2010;13:1175–1191. [PubMed]
  • Porter AC, Sauer JM, Knierman MD, Becker GW, Berna MJ, Bao J, Nomikos GG, Carter P, Bymaster FP, Leese AB, Felder CC. Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J Pharmacol Exp Ther.2002;301:1020–1024. [PubMed]
  • Price DA, Martinez AA, Seillier A, Koek W, Acosta Y, Fernandez E, Strong R, Lutz B, Marsicano G, Roberts JL, Giuffrida A. WIN55,212–2, a cannabinoid receptor agonist, protects against nigrostriatal cell loss in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Eur J Neurosci. 2009;29:2177–2186.[PMC free article] [PubMed]
  • Price DA, Owens WA, Gould GG, Frazer A, Roberts JL, Daws LC, Giuffrida A. CB1-independent inhibition of dopamine transporter activity by cannabinoids in mouse dorsal striatum. J Neurochem.2007;101:389–396. [PubMed]
  • Qiao H, Noda Y, Kamei H, Nagai T, Furukawa H, Miura H, Kayukawa Y, Ohta T, Nabeshima T. Clozapine, but not haloperidol, reverses social behavior deficit in mice during withdrawal from chronic phencyclidine treatment. NeuroReport. 2001;12:11–15.[PubMed]
  • Ramirez BG, Blazquez C, Gomez del Pulgar T, Guzman M, de Ceballos ML. Prevention of Alzheimer’s disease pathology by cannabinoids: neuroprotection mediated by blockade of microglial activation. J Neurosci. 2005;25:1904–1913. [PubMed]
  • Rodriguez JJ, Mackie K, Pickel VM. Ultrastructural localization of the CB1 cannabinoid receptor in mu-opioid receptor patches of the rat Caudate putamen nucleus. J Neurosci. 2001;21:823–833.[PubMed]
  • Romero J, Berrendero F, Perez-Rosado A, Manzanares J, Rojo A, Fernandez-Ruiz JJ, de Yebenes JG, Ramos JA. Unilateral 6-hydroxydopamine lesions of nigrostriatal dopaminergic neurons increased CB1 receptor mRNA levels in the caudate-putamen. Life Sci. 2000;66:485–494. [PubMed]
  • Ross RA. Anandamide and vanilloid TRPV1 receptors. Br J Pharmacol. 2003;140:790–801. [PMC free article] [PubMed]
  • Ryberg E, Larsson N, Sjogren S, Hjorth S, Hermansson NO, Leonova J, Elebring T, Nilsson K, Drmota T, Greasley PJ. The orphan receptor GPR55 is a novel cannabinoid receptor. Br J Pharmacol.2007;152:1092–1101. [PMC free article] [PubMed]
  • Ryberg E, Vu HK, Larsson N, Groblewski T, Hjorth S, Elebring T, et al. Identification and characterization of a novel splice variant of the human CB1 receptor. FEBS Lett. 2005;579:259–264. [PubMed]
  • Sagredo O, Gonzalez S, Aroyo I, Pazos MR, Benito C, Lastres-Becker I, Romero JP, Tolon RM, Mechoulam R, Brouillet E, Romero J, Fernandez-Ruiz J. Cannabinoid CB2 receptor agonists protect the striatum against malonate toxicity: relevance for Huntington’s disease. Glia. 2009;57:1154–1167. [PMC free article] [PubMed]
  • Sancho R, Calzado MA, Di Marzo V, Appendino G, Munoz E. Anandamide inhibits nuclear factor-kappaB activation through a cannabinoid receptor-independent pathway. Mol Pharmacol.2003;63:429–438. [PubMed]
  • Santini E, Valjent E, Fisone G. Parkinson’s disease: levodopa-induced dyskinesia and signal transduction. FEBS J. 2008;275:1392–1399.[PubMed]
  • Sawzdargo M, Nguyen T, Lee DK, Lynch KR, Cheng R, Heng HH, George SR, O’Dowd BF. Identification and cloning of three novel human G protein-coupled receptor genes GPR52, PsiGPR53 and GPR55: GPR55 is extensively expressed in human brain. Brain Res Mol Brain Res. 1999;64:193–198. [PubMed]
  • Schlosburg JE, Blankman JL, Long JZ, Nomura DK, Pan B, Kinsey SG, Nguyen PT, Ramesh D, Booker L, Burston JJ, Thomas EA, Selley DE, Sim-Selley LJ, Liu QS, Lichtman AH, Cravatt BF. Chronic monoacylglycerol lipase blockade causes functional antagonism of the endocannabinoid system. Nat Neurosci. 2010;13:1113–1119.[PMC free article] [PubMed]
  • Schweitzer P. Cannabinoids decrease the K+ M-current in hippocampal CA1 neurons. The Journal of Neuroscience.2000;20:51–58. [PubMed]
  • Segovia G, Mora F, Crossman AR, Dsc, Brotchie JM. Effects of CB1 cannabinoid receptor modulating compounds on the hyperkinesia induced by high-dose levodopa in the reserpine-treated rat model of Parkinson’s disease. Movement Disorders. 2003;18:138–149.[PubMed]
  • Seillier A, Advani T, Cassano T, Hensler JG, Giuffrida A. Inhibition of fatty-acid amide hydrolase and CB1 receptor antagonism differentially affect behavioural responses in normal and PCP-treated rats. Int J Neuropsychopharmacol. 2010;13:373–386. [PubMed]
  • Seillier A, Giuffrida A. Evaluation of NMDA receptor models of schizophrenia: divergences in the behavioral effects of sub-chronic PCP and MK-801. Behav Brain Res. 2009;204:410–415. [PubMed]
  • Sewell RA, Ranganathan M, D’Souza DC. Cannabinoids and psychosis. Int Rev Psychiatry. 2009;21:152–162. [PubMed]
  • Shoemaker JL, Ruckle MB, Mayeux PR, Prather PL. Agonist-directed trafficking of response by endocannabinoids acting at CB2 receptors.J Pharmacol Exp Ther. 2005;315:828–838. [PubMed]
  • Simon GM, Cravatt BF. Endocannabinoid biosynthesis proceeding through glycerophospho-N-acyl ethanolamine and a role for alpha/beta-hydrolase 4 in this pathway. J Biol Chem.2006;281:26465–26472. [PubMed]
  • Snider NT, Nast JA, Tesmer LA, Hollenberg PF. A Cytochrome P450-derived Epoxygenated Metabolite of Anandamide is a Potent Cannabinoid Receptor 2 – Selective Agonist. Mol Pharmacol 2009[PMC free article] [PubMed]
  • Solbrig MV, Fan Y, Hermanowicz N, Morgese MG, Giuffrida A. A synthetic cannabinoid agonist promotes oligodendrogliogenesis during viral encephalitis in rats. Exp Neurol. 2010;226:231–241.[PMC free article] [PubMed]
  • Solinas M, Justinova Z, Goldberg SR, Tanda G. Anandamide administration alone and after inhibition of fatty acid amide hydrolase (FAAH) increases dopamine levels in the nucleus accumbens shell in rats. J Neurochem. 2006;98:408–419. [PubMed]
  • Spano MS, Fadda P, Frau R, Fattore L, Fratta W. Cannabinoid self-administration attenuates PCP-induced schizophrenia-like symptoms in adult rats. Eur Neuropsychopharmacol. 2010;20:25–36.[PubMed]
  • Starowicz K, Nigam S, Di Marzo V. Biochemistry and pharmacology of endovanilloids. Pharmacol Ther. 2007;114:13–33. [PubMed]
  • Steinpreis RE. The behavioral and neurochemical effects of phencyclidine in humans and animals: some implications for modeling psychosis. Behav Brain Res. 1996;74:45–55. [PubMed]
  • Stella N. Cell biology. Anatomy of prostaglandin signals. Science.2011;334:768–769. [PMC free article] [PubMed]
  • Straiker A, Wager-Miller J, Hutchens J, Mackie K. Differential signaling in human cannabinoid CB(1) receptors and their splice variants in autaptic hippocampal neurons. Br J Pharmacol. 2011 doi: 10.1111/j.1476-5381.2011.01744.x. [Epub ahead of print][PMC free article] [PubMed] [Cross Ref]
  • Sun Y, Alexander SP, Garle MJ, Gibson CL, Hewitt K, Murphy SP, Kendall DA, Bennett AJ. Cannabinoid activation of PPARalpha; a novel neuroprotective mechanism. Br J Pharmacol 2007[PMC free article] [PubMed]
  • Szolcsanyi J. Are cannabinoids endogenous ligands for the VR1 capsaicin receptor? Trends Pharmacol Sci. 2000;21:41–42. [PubMed]
  • Teismann P, Tieu K, Choi DK, Wu DC, Naini A, Hunot S, Vila M, Jackson-Lewis V, Przedborski S. Cyclooxygenase-2 is instrumental in Parkinson’s disease neurodegeneration. Proc Natl Acad Sci U S A.2003;100:5473–5478. [PMC free article] [PubMed]
  • Twitchell W, Brown S, Mackie K. Cannabinoids inhibit N- and P/Q-type calcium channels in cultured rat hippocampal neurons. J Neurophysiol. 1997;78:43–50. [PubMed]
  • Tzavara ET, Li DL, Moutsimilli L, Bisogno T, Di Marzo V, Phebus LA, Nomikos GG, Giros B. Endocannabinoids activate transient receptor potential vanilloid 1 receptors to reduce hyperdopaminergia-related hyperactivity: therapeutic implications. Biol Psychiatry.2006;59:508–515. [PubMed]
  • Uchigashima M, Narushima M, Fukaya M, Katona I, Kano M, Watanabe M. Subcellular arrangement of molecules for 2-arachidonoyl-glycerol-mediated retrograde signaling and its physiological contribution to synaptic modulation in the striatum. J Neurosci. 2007;27:3663–3676. [PubMed]
  • Ueda N, Tsuboi K, Uyama T, Ohnishi T. Biosynthesis and degradation of the endocannabinoid 2-arachidonoylglycerol.Biofactors. 2011;37:1–7. [PubMed]
  • Ujike H, Morita Y. New perspectives in the studies on endocannabinoid and cannabis: cannabinoid receptors and schizophrenia. J Pharmacol Sci. 2004;96:376–381. [PubMed]
  • Uriguen L, Garcia-Fuster MJ, Callado LF, Morentin B, La Harpe R, Casado V, Lluis C, Franco R, Garcia-Sevilla JA, Meana JJ. Immunodensity and mRNA expression of A2A adenosine, D2 dopamine, and CB1 cannabinoid receptors in postmortem frontal cortex of subjects with schizophrenia: effect of antipsychotic treatment. Psychopharmacology (Berl) 2009;206:313–324.[PubMed]
  • 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;19:1140–1142. [PubMed]
  • Vigano D, Guidali C, Petrosino S, Realini N, Rubino T, Di Marzo V, Parolaro D. Involvement of the endocannabinoid system in phencyclidine-induced cognitive deficits modelling schizophrenia.Int J Neuropsychopharmacol. 2009;12:599–614. [PubMed]
  • Vila M, Jackson-Lewis V, Guegan C, Wu DC, Teismann P, Choi DK, Tieu K, Przedborski S. The role of glial cells in Parkinson’s disease.Curr Opin Neurol. 2001;14:483–489. [PubMed]
  • Vitale C, Pellecchia MT, Grossi D, Fragassi N, Cuomo T, Di Maio L, Barone P. Unawareness of dyskinesias in Parkinson’s and Huntington’s diseases. Neurol Sci. 2001;22:105–106. [PubMed]
  • Walsh S, Gorman AM, Finn DP, Dowd E. The effects of cannabinoid drugs on abnormal involuntary movements in dyskinetic and non-dyskinetic 6-hydroxydopamine lesioned rats. Brain Res.2010;1363:40–48. [PubMed]
  • Walter L, Stella N. Cannabinoids and neuroinflammation. Br J Pharmacol. 2004;141:775–785. [PMC free article] [PubMed]
  • Weber A, Ni J, Ling KH, Acheampong A, Tang-Liu DD, Burk R, Cravatt BF, Woodward D. Formation of prostamides from anandamide in FAAH knockout mice analyzed by HPLC with tandem mass spectrometry. J Lipid Res. 2004;45:757–763.[PubMed]
  • Wei BQ, Mikkelsen TS, McKinney MK, Lander ES, Cravatt BF. A second fatty acid amide hydrolase with variable distribution among placental mammals. J Biol Chem. 2006;281:36569–36578.[PubMed]
  • Wilson RI, Kunos G, Nicoll RA. Presynaptic specificity of endocannabinoid signaling in the hippocampus. Neuron.2001;31:453–462. [PubMed]
  • Wilson RI, Nicoll RA. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature. 2001;410:588–592.[PubMed]
  • Wong DF, Kuwabara H, Horti AG, Raymont V, Brasic J, Guevara M, Ye W, Dannals RF, Ravert HT, Nandi A, Rahmim A, Ming JE, Grachev I, Roy C, Cascella N. Quantification of cerebral cannabinoid receptors subtype 1 (CB1) in healthy subjects and schizophrenia by the novel PET radioligand [11C]OMAR. Neuroimage. 2010;52:1505–1513. [PubMed]
  • Yin H, Chu A, Li W, Wang B, Shelton F, Otero F, Nguyen DG, Caldwell JS, Chen YA. Lipid G protein-coupled receptor ligand identification using beta-arrestin PathHunter assay. J Biol Chem.2009;284:12328–12338. [PMC free article] [PubMed]
  • Zavitsanou K, Garrick T, Huang XF. Selective antagonist [3H]SR141716A binding to cannabinoid CB1 receptors is increased in the anterior cingulate cortex in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:355–360.[PubMed]
  • Zeng BY, Dass B, Owen A, Rose S, Cannizzaro C, Tel BC, Jenner P. Chronic L-DOPA treatment increases striatal cannabinoid CB1 receptor mRNA expression in 6-hydroxydopamine-lesioned rats.Neurosci Lett. 1999;276:71–74. [PubMed]
  • Zhong P, Pan B, Gao X, Blankman JL, Cravatt BF, Liu WS. Genetic deletion of monoacylglycerol lipase alters endocannabinoid-mediated retrograde synaptic depression in the cerebellum. J Physiol.2011;589:4847–4855. [PMC free article] [PubMed]
  • Zuardi AW, Guimaraes FS, Hallak JE, Crippa JA. Is the highest density of CB1 receptors in paranoid schizophrenia a correlate of endocannabinoid system functioning? Expert Rev Neurother.2011;11:1111–1114. [PubMed]

potp font 1