Review
The enigmatic pharmacology of GPR55

https://doi.org/10.1016/j.tips.2008.12.004Get rights and content

Preliminary data presented at conferences and in the patent literature introduced the possibility the orphan receptor GPR55 might account for some of the well-documented non-CB1, non-CB2 effects reported for certain cannabinoid ligands. Several peer-reviewed publications have recently emerged in which the pharmacology of the cannabinoids at GPR55 has been probed in more depth. Despite this, the classification of GPR55 as a cannabinoid receptor remains a contentious issue. The weight of evidence points to GPR55 as a receptor that is activated by certain cannabinoid ligands and by the bioactive lipid l-α-lysophosphatidylinsoitol. It couples to G12 proteins, activates RhoA and mobilizes intracellular Ca2+, possibly in an agonist- and tissue-dependant manner, thus displaying ‘agonist functional selectivity’. Here, I review the recent literature in an effort to glean the key controversies and outstanding questions surrounding the interaction between cannabinoids and this orphan receptor.

Introduction

In 2005, both meeting presentations and the patent literature revealed that GPR55 might be activated by several cannabinoid ligands (for review, see Ref. [1]). GPR55 has the highest amino acid sequence homology with GPR23, GPR35, P2Y5 and CCR2 [2]. mRNA for this receptor has been detected in regions of the central nervous system (CNS) including the caudate nucleus, putamen, hippocampus, thalamus, pons, cerebellum, frontal cortex and hypothalamus 2, 3 and in the adrenal glands and gastrointestinal tract [3]. The endocannabinoid system comprises two established cannabinoid receptors (CB1 and CB2); GPR55 shares only 13.5% sequence identity with the CB1 receptor and 14.4% with the CB2 receptor. The levels of GPR55 mRNA expression are notably between three- and tenfold lower than those reported for the cannabinoid CB1 receptor [3]. To put this finding in perspective, it is important to note that CB1 receptor expression in the CNS is considerably higher than other G-protein-coupled receptors (GPCRs) [4]. Within the brain, the distribution of CB1 receptors is heterogeneous; they are found predominantly on nerve terminals where they attenuate neurotransmitter release [4]. The CB2 receptor is predominantly expressed on immune cells, although there is evidence for neuronal localization in the brainstem [5].

There is a family of endogenous cannabinoid ligands (endocannabinoids) and specific molecular machinery for the synthesis and inactivation of these ligands 6, 7. The most studied endocannabinoids are arachidonoylethanolamide (anandamide) and 2-arachidonoyl glycerol (2-AG), both of which are synthesized on demand [8]. 2-AG acts as a ‘retrograde’ signalling molecule in the CNS; it is synthesized postsynaptically and acts presynaptically to inhibit neurotransmitter release. The endocannabinoid system is involved in the pathophysiology of obesity, osteoporosis, nicotine addiction, mental illness, neuropathic and inflammatory pain, cardiovascular disorders and liver disease 6, 8. The most abundant constituents of cannabis (phytocannabinoids) include Δ9-tetrahydrocannabinol (Δ9THC), which is psychoactive, and (−)-cannabidiol (CBD), which is non-psychoactive. These compounds interact with various elements of the endocannabinoid system 6, 8.

The pharmacology of both the endocannabinoids and the phytocannabinoids is complex; their actions are mediated by cannabinoid CB1 and CB2 receptors and by putative non-CB1, non-CB2 receptors. One such novel target has been implicated in endothelium-dependent vasodilatation in isolated resistance vessels, in haemodynamic responses and in modulation of endothelial, microglial and immune-cell migration (for review, see Refs 9, 10, 11, 12). Such non-CB1, non-CB2 effects have been observed with a range of cannabinoid ligands including anandamide and virodhamine (which are eicosanoids) and the phytocannabinoid-like compounds O-1602 and the CBD analogue known as ‘abnormal-CBD’; these effects are antagonized by CBD and its analogue O-1918 and by micromolar concentrations of the CB1 receptor antagonist SR141716A (Rimonabant®). This novel target has been referred to as: the abnormal-CBD receptor, the CBx receptor or the endothelial anandamide receptor (for reviews, see Refs 10, 11). It is increasingly likely that more than one non-CB1, non-CB2 target might exist [9]. Clearly, the pharmacology surrounding the abnormal-CBD receptor might, in fact, encompass several novel hitherto unrecognized ‘cannabinoid’ receptors, one of which might be GPR55 (Box 1).

In the past two years, several peer-reviewed studies have been published in which the pharmacology of cannabinoids at GPR55 was probed in more depth 3, 13, 14, 15, 16, 17, 18. Despite this, GPR55 pharmacology remains a contentious issue 19, 20. The concept of ‘agonist functional selectivity’ is particularly pertinent to the debate surrounding GPR55 (for reviews, see Refs 21, 22). Canonical pharmacology implied that ligands interact with GPCRs in such way that, irrespective of the ligand structure or the measured functional response, this union culminates in the same endpoint. However, the scenario seems to be less simplistic and recent data demonstrate that the downstream signalling of a given GPCR might vary in both a ligand- and a system-dependant manner. Clearly, this might underlie the contradictory nature of the pharmacological findings surrounding GPR55 (Table 1).

Here, the recent literature is reviewed in an effort to glean the key controversies and questions surrounding the interaction of the cannabinoids with this orphan receptor. I will consider the GPR55 pharmacology of various classes of compound including the endocannabinoids, the bioactive lipid l-α-lysophosphatidylinsoitol (LPI), the phytocannabinoids and synthetic cannabinoid ligands. The characteristics of GPR55 downstream signalling and the agonist-specific nature of this signalling is reviewed. Furthermore, the debate surrounding the classification of the GPR55 as a ‘cannabinoid receptor’ will also be explored.

Section snippets

GPR55 pharmacology

The cannabinoids fall into three categories: the endocannabinoids, the phytocannabinoids and the synthetic small molecules that target CB1 and CB2 receptors. The GPR55 pharmacology of the endocannabinoids has potential physiological implications; these compounds have a key role in both physiology and in pathophysiology. Furthermore, hitherto unrecognized GPR55-mediated effects of the phytocannabinoids and of certain ligands that were previously classified as cannabinoid-receptor-selective might

Is GPR55 a cannabinoid receptor?

To summarize, the status of the endogenous cannabinoids as GPR55 ligands is debatable [19]; the standing of LPI as a GPR55 ligand is less controversial but it has not been comprehensively investigated. Against this background, debate surrounds the accuracy of the categorization of this receptor as a ‘cannabinoid’ receptor. It would perhaps be premature to define GPR55 as a cannabinoid receptor on the basis of the discordant findings with anandamide and 2-AG. Clearly the pharmacology of the

Downstream signalling

Several publications have investigated various aspects of the downstream signalling linked to GPR55. These are summarized in Figure 1. The G12 subfamily of G protein comprises Gα12 and Gα13. Using a peptide blocking approach, it has been demonstrated that obstruction of Gαi1/2, Gαi3 and Gαs had no effect on signalling initiated by GPR55 agonists in a [35S]GTPγS binding assay; however, the peptide directed against Gα13 abolish the agonist effect [3]. The authors ruled out Gαq owing to the

GPR55−/− mice

Two studies have been published employing mice generated by GlaxoSmithKline that are deficient in GPR55. This group made use of these mice to investigate the role of this receptor in the vasodilator effects of the CBD analogues O-1602 and abnormal-CBD [14]. The study sought to address the issue of the role of GPR55 as a mediator of the widely reported effects of certain cannabinoid ligands in blood vessels [28]; that is, effects that are not mediated by the CB1 or CB2 receptor 1, 9. Notably,

Conclusions and future directions

The pharmacology of GPR55 is enigmatic; reports conflict as to the nature of the interaction between endocannabinoids and this orphan receptor. The situation is confounded by the clear evidence that the GPR55 agonists O-1602 and abnormal-CBD activate additional targets in the cardiovascular system of GPR55−/− mice. Clearly, future research should be directed towards the identification and pharmacological characterisation of additional orphan receptors for cannabinoid ligands. Although

References (31)

  • P.C. Staton

    The putative cannabinoid receptor GPR55 plays a role in mechanical hyperalgesia associated with inflammatory and neuropathic pain

    Pain

    (2008)
  • C.W. Vaughan

    Stressed-out endogenous cannabinoids relieve pain

    Trends Pharmacol. Sci.

    (2006)
  • D. Baker

    In silico patent searching reveals a new cannabinoid receptor

    Trends Pharmacol. Sci.

    (2005)
  • E. Ryberg

    The orphan receptor GPR55 is a novel cannabinoid receptor

    Br. J. Pharmacol.

    (2007)
  • A.C. Howlett

    International Union of Pharmacology. XXVII. Classification of cannabinoid receptors

    Pharmacol. Rev

    (2002)
  • Cited by (248)

    • Targeting the endocannabinoid system in HAND

      2024, HIV-Associated Neurocognitive Disorders
    • The oncogenic lysophosphatidylinositol (LPI)/GPR55 signaling

      2022, Life Sciences
      Citation Excerpt :

      The higher expression of GPR55 mRNA in CNS includes the frontal cortex, cerebellum, hypothalamus, microglia, and nucleus accumbens [2,3,53,54]. Out of the CNS, its expression includes the spleen neutrophils, adrenal glands, tonsils, testes, thymus, and gastrointestinal tract, mesometrial decidua, bone, and adipose tissue [36,52,55,56]. In the CNS, the physiology of GPR55 includes areas and functions such as: (i) dorsal basal nuclei mediating motor skills [18] and procedural memory [53]; (ii) ventral basal nuclei mediating reinforcing actions of substances of abuse such as nicotine [54]; (iii) hippocampus involved in long term potentiation, neurogenesis [57], spatial memory formation [20], regulation of immune responses [58] and neuroprotection [59,60]; and (iv) limbic structures (e.g., periaqueductal gray) linked to anxiety-like processes [23], alcohol intake and defensive aggressive responses in alcohol pre-exposed rats [24], and nociception [61].

    View all citing articles on Scopus
    View full text