Elsevier

Brain Research

Volume 1188, 10 January 2008, Pages 157-164
Brain Research

Research Report
Cannabidiol potentiates pharmacological effects of Δ9-tetrahydrocannabinol via CB1 receptor-dependent mechanism

https://doi.org/10.1016/j.brainres.2007.09.090Get rights and content

Abstract

Cannabidiol, a non-psychoactive component of cannabis, has been reported to have interactions with Δ9-tetrahydrocannabinol (Δ9-THC). However, such interactions have not sufficiently been clear and may have important implications for understanding the pharmacological effects of marijuana. In the present study, we investigated whether cannabidiol modulates the pharmacological effects of Δ9-THC on locomotor activity, catalepsy-like immobilisation, rectal temperature and spatial memory in the eight-arm radial maze task in mice. In addition, we measured expression level of cannabinoid CB1 receptor at striatum, cortex, hippocampus and hypothalamus. Δ9-THC (1, 3, 6 and 10 mg/kg) induced hypoactivity, catalepsy-like immobilisation and hypothermia in a dose-dependent manner. In addition, Δ9-THC (1, 3 and 6 mg/kg) dose-dependently impaired spatial memory in eight-arm radial maze. On the other hand, cannabidiol (1, 3, 10, 25 and 50 mg/kg) did not affect locomotor activity, catalepsy-like immobilisation, rectal temperature and spatial memory on its own. However, higher dose of cannabidiol (10 or 50 mg/kg) exacerbated pharmacological effects of lower dose of Δ9-THC, such as hypoactivity, hypothermia and impairment of spatial memory. Moreover, cannabidiol (50 mg/kg) with Δ9-THC (1 mg/kg) enhanced the expression level of CB1 receptor expression in hippocampus and hypothalamus. Cannabidiol potentiated pharmacological effects of Δ9-THC via CB1 receptor-dependent mechanism. These findings may contribute in setting the basis for interaction of cannabinoids and to find a cannabinoid mechanism in central nervous system.

Introduction

Cannabis contains about 60 different cannabinoids, including the psychoactive component, Δ9-tetrahydrocannabinol (Δ9-THC), and other major non-psychoactive components, such as cannabidiol, cannabinol and cannabigerol. Δ9-THC has been demonstrated to produce hypothermia, learning and memory impairment, impairment of the prepulse inhibition of the startle reflex, catalepsy-like immobilisation, aggressive behaviour, analgesia and hypoactivity (Wiley and Martin, 2002, Martin et al., 1991, Fujiwara, 2001, Mishima et al., 2001, Nagai et al., 2006, Egashira et al., 2006). These effects are at least partly caused by binding to cannabinoid receptor 1 (CB1 receptor) within the brain. On the other hand, cannabidiol, a non-psychoactive component of cannabis, has been found to be an anticonvulsant in animal models of epilepsy and in humans with epilepsy. Moreover, cannabidiol has been shown to exert cannabinoid-like effects against such conditions as spasm, anxiety, nausea and rheumatoid arthritis (Mecholam et al., 2002, Ashton et al., 2005). However, cannabidiol is generally known to have a very low affinity (in the micromolar range) for the cannabinoid CB1 and CB2 receptors, but also to have many pharmacological actions. These actions are thought to be dependent on a new cannabinoid receptor, such as an abnormal cannabidiol receptor, a non-CB1 and non-CB2 receptor, and G-protein-coupled receptor GPR55 (Begg et al., 2005, Baker et al., 2006). There are multiple cannabinoid receptors, most of them identified pharmacologically but yet to be cloned.

It has been suggested that coadministration of cannabidiol may alter the pharmacological effects of Δ9-THC, potentiating some therapeutic benefits of Δ9-THC, while attenuating some of its negative effects (Karniol et al., 1974, Russo and Guy, 2006, Zuardi et al., 1982). The potentially interesting modulatory effects of cannabidiol have been described in several reports. For example, the cataleptic effects of Δ9-THC were similarly reversed by equal doses of cannabidiol (Karniol and Carlini, 1973). Δ9-THC-induced hypothermia is potentiated by equal doses of cannabidiol (Fernandes et al., 1974), while higher doses of cannabidiol either antagonise (Borgen and Davis, 1974) or have no effect (Ham et al., 1975). In addition, the antinociceptive effects of Δ9-THC are adversely potentiated by a high dose of cannabidiol (i.e. 30 mg/kg) in mice (Varvel et al., 2006), while similar doses of cannabidiol have been found to antagonise the effects of Δ9-THC in a formic-acid-induced writhing test (Welburn et al., 1976). Moreover, administration of very high doses of cannabidiol have been shown to raise the Δ9-THC concentration in the blood and brain (Bornheim et al., 1995, Jones and Pertwee, 1972), an effect likely due to inhibition of hepatic microsomal Δ9-THC metabolism (Jaeger et al., 1996, Watanabe et al., 1987). This pharmacokinetic explanation may account for several potentiating effects, for which high doses of cannabidiol have been assessed. Therefore, it is important to assess the interaction between Δ9-THC and cannabidiol.

The present experiments were conducted to evaluate putative interactions between Δ9-THC and cannabidiol in behavioural tests, such as spontaneous locomotor activity, catalepsy-like immobilisation and rectal temperature. In addition, we assessed the potentiating effects of combining Δ9-THC with cannabidiol, compared to Δ9-THC alone, on spatial memory in the eight-arm radial maze task. Finally, we measured the expression levels of cannabinoid CB1 receptor in the striatum, cortex, hippocampus and hypothalamus.

Section snippets

Spontaneous locomotor activity, catalepsy-like immobilisation and rectal temperature

Δ9-THC (1, 3, 6 and 10 mg/kg) decreased both locomotor activity and rectal temperature in a dose-dependent manner. In addition, Δ9-THC dose-dependently produced catalepsy-like immobilisation. On the other hand, cannabidiol (1, 3, 10, 25 and 50 mg/kg) did not affect locomotor activity, catalepsy-like immobilisation and rectal temperature [Locomotor activity: F(4,36) = 5.559, P < 0.01; Δ9-THC 6 mg/kg, P < 0.05; Δ9-THC 10 mg/kg, P < 0.01 compared with vehicle; rectal temperature 1 h: F(4,22) = 13.023, P < 

Discussion

In the present study, Δ9-THC induced hypoactivity, catalepsy-like immobilisation and hypothermia in a dose-dependent manner (Table 1). In addition, Δ9-THC dose-dependently impaired spatial memory in the eight-arm radial maze task (Fig. 1). On the other hand, cannabidiol did not affect the above at any dose tested on its own (Table 1, Fig. 1). However, higher doses of cannabidiol exacerbated the pharmacological effects of lower doses of Δ9-THC on locomotor activity, rectal temperature and

Animals

Male ddY mice (25–35 g; Kiwa Experimental Animal Laboratory, Wakayama, Japan) were kept under a 12 h light/dark cycle (lights on from 07:00 to 19:00 h) in an air-conditioned room (23 ± 2 °C; humidity, 60 ± 5%), with food (CE-2; Clea Japan, Tokyo, Japan) and water available ad libitum. All procedures regarding animal care and use were performed in compliance with the regulations established by the Experimental Animal Care and Use Committee of Fukuoka University, Japan. All animals were housed in the

Acknowledgments

Part of this study was supported by a Grant-in-Aid for Scientific Research (No. 17590479) from the Ministry of Education, Science and Culture of Japan, the Advanced Materials Institute of Fukuoka University and The Naito Foundation.

References (47)

  • K. Mishima et al.

    Characteristics of learning and memory impairment induce by Δ9-tetrahydrocannabinol in rats

    Jpn. J. Pharmacol.

    (2001)
  • E. Murillo-Rodriguez et al.

    Cannabidiol, a constituent of Cannabis sativa, modulates sleep in rats

    FEBS. Lett.

    (2006)
  • H. Nagai et al.

    Antipsychotics improve Delta9-tetrahydrocannabinol0induced impairment of the prepulse inhibition of the startle reflex in mice

    Pharmacol. Biochem. Behav.

    (2006)
  • E. Russo et al.

    A tale of two cannabinoids: the therapeutic rationale for combining tetrahydrocannabinol and cannabidiol

    Med. Hypotheses

    (2006)
  • V. Vialou et al.

    Neurochemical characterization of pathways expressing plasma membrane monoamine transporter in the rat brain

    Neuroscience

    (2007)
  • I. Walaas et al.

    Biochemical evidence for glutamate as a transmitter in hippocampal efferents to the basal forebrain and hypothalamus in rat brain

    Neuroscience

    (1980)
  • K. Watanabe et al.

    Self-catalyzed inactivation of cytochrome P-450 during microsomal metabolism of cannabidiol

    Biochem. Pharmacol.

    (1987)
  • J.L. Wiley et al.

    Cannabinoid pharmacology: implications for additional cannabinoid receptor subtypes

    Chem. Phys. Lipids

    (2002)
  • C.H. Ashton et al.

    Cannabinoids in bipolar affective disorder: a review and discussion of their therapeutic potential

    J. Psychopharmacol.

    (2005)
  • T. Bisogno et al.

    Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide

    Br. J. Pharmacol.

    (2001)
  • L.A. Borgen et al.

    Cannabidiol interaction with Δ9-tetrahydrocannabinol

    Res. Commun. Chem. Pathol. Pharmacol.

    (1974)
  • L.M. Bornheim et al.

    Effect of cannabidiol pretreatment on the kinetics of tetrahydrocannabinol metabolites in mouse brain

    Drug Metab. Dispos.

    (1995)
  • E.J. Carrier et al.

    Inhibition of an equilibrative nucleoside transporter by cannabidiol: a mechanism of cannabinoid immunosuppression

    Proc. Natl. Acad. Sci. USA.

    (2006)
  • Cited by (0)

    View full text