Inhibition of fatty acid amide hydrolase, a key endocannabinoid metabolizing enzyme, by analogues of ibuprofen and indomethacin

https://doi.org/10.1016/j.ejphar.2007.02.051Get rights and content

Abstract

There is evidence in the literature that the nonsteroidal anti-inflammatory drugs indomethacin and ibuprofen can interact with the cannabinoid system both in vitro and in vivo. In the present study, a series of analogues of ibuprofen and indomethacin have been investigated with respect to their ability to inhibit fatty acid amide hydrolase, the enzyme responsible for the hydrolysis of the endogenous cannabinoid anandamide. Of the fourteen compounds tested, the 6-methyl-pyridin-2-yl analogue of ibuprofen (“ibu-am5”) was selected for further study. This compound inhibited rat brain anandamide hydrolysis in a non-competitive manner, with IC50 values of 4.7 and 2.5 μM being found at pH 6 and 8, respectively. By comparison, the IC50 values for ibuprofen were 130 and 750 μM at pH 6 and 8, respectively. There was no measurable N-acylethanolamine hydrolyzing acid amidase activity in rat brain membrane preparations. In intact C6 glioma cells, ibu-am5 inhibited the hydrolysis of anandamide with an IC50 value of 1.2 μM. There was little difference in the potencies of ibu-am5 and ibuprofen towards cyclooxygenase-1 and -2 enzymes, and neither compound inhibited the activity of monoacylglycerol lipase. Ibu-am5 inhibited the binding of [3H]-CP55,940 to rat brain CB1 and human CB2 cannabinoid receptors more potently than ibuprofen, but the increase in potency was less than the corresponding increase in potency seen for inhibition of FAAH activity. It is concluded that ibu-am5 is an analogue of ibuprofen with a greater potency towards fatty acid amide hydrolase but with a similar cyclooxygenase inhibitory profile, and may be useful for the study of the therapeutic potential of combined fatty acid amide hydrolase–cyclooxygenase inhibitors.

Introduction

There is evidence indicating that the cannabinoid system can contribute to the in vivo pharmacological effects of nonsteroidal anti-inflammatory drugs (NSAIDs). Thus, the effects of spinally administered indomethacin in the formalin test of inflammatory pain are blocked by the CB1 cannabinoid receptor antagonist AM251, and are not seen in CB1-receptor knockout mice (Gühring et al, 2002). A similar result was seen for intrathecally administered flurbiprofen (Ates et al., 2003). Locally administered ibuprofen produces synergistic effects with the endogenous cannabinoid anandamide (arachidonoylethanolamide, AEA) in the formalin test, the synergy being prevented by AM251 (Guindon et al., 2006a).

The mechanisms involved in these effects are as yet unclear. Guindon et al. (2006b) reported that the combination of locally administered ibuprofen and anandamide produced increases in the paw levels of AEA and the related N-acylethanolamines palmitoylethanolamide and oleoylethanolamide over and above those seen with either compound per se. Similar results (and a similar synergistic effect) were seen with the cyclooxygenase-2 (COX-2) inhibitor rofecoxib (Guindon et al., 2006b). Gühring et al. (2002) and Ates et al. (2003) suggested that indomethacin and flurbiprofen may allow for an increased synthesis of endocannabinoids from arachidonic acid by blocking the cyclooxygenase (COX) catalyzed removal of arachidonic acid.

A key pathway for removal of anandamide and other N-acylethanolamines such as palmitoylethanolamide (PEA) is the enzyme fatty acid amide hydrolase (FAAH). Loss of activity of this enzyme, either by genetic deletion or by the use of selective inhibitors such as URB597 leads to increased levels of N-acylethanolamines and potentially beneficial effects in models of inflammation and inflammatory pain (see e.g. Lichtman et al., 2004, Holt et al., 2005, Jayamanne et al., 2006). In 1996, Paria et al. reported that indomethacin reduced the rate of anandamide hydrolysis in the mouse uterus (Paria et al., 1996). More systematic studies have since found that many NSAIDs, including indomethacin and ibuprofen, inhibit the activity of FAAH (Fowler et al., 1997, Fowler et al., 1999, Fowler et al., 2003), particularly at low pH (Holt et al., 2001, Holt and Fowler, 2003, Fowler et al., 2003), such as is seen in inflamed tissue (Häbler, 1929, Andersson et al., 1999). AEA is also a substrate for COX-2 (see Kozak and Marnett, 2002), and so it is possible that the effects of NSAIDs upon inflammation and inflammatory pain may not only be due to a decreased production of prostaglandin, but also a reduced removal of AEA (and other anti-inflammatory N-acylethanolamines) by COX-2 and/or FAAH (Holt et al., 2005, Guindon et al., 2006b). Certainly, when it is considered that AEA and PEA can also interact with other targets that are involved in the inflammation and pain, such as TRPV1 receptors (Zygmunt et al., 1999) and the peroxisome proliferator-activated receptor γ and α isoforms (Bouaboula et al., 2005, Lo Verme et al., 2005), and that the relative roles played by CB- and TRPV1-meditated responses to AEA are different in normal and inflamed tissue (Singh Tahim et al., 2005), the situation becomes very complicated indeed.

The pharmacological properties of FAAH inhibitors, together with the potential role of FAAH in the action of NSAIDs raise the possibility that compounds with dual actions upon FAAH and COX-2 may be useful novel compounds for the treatment of inflammation and inflammatory pain. An obvious template for the identification of such compounds is the NSAIDs themselves. However, structure–activity relationships of analogues of clinically used NSAIDs with respect to FAAH inhibition have not been examined in the literature, in contrast to the situation for the relative COX-1 and -2 potencies of such compounds (see e.g. Kalgutar et al., 2000a, Kalgutar et al., 2000b). In consequence, in the present study, a series of ibuprofen and indomethacin analogues have been investigated with respect to their ability to inhibit FAAH. The ibuprofen analogues have not previously been characterized biochemically, but have been shown to produce analgesic effects comparable to ibuprofen in the acetic acid writhing model, and in some cases to possess considerably less ulcerogenic properties (Cocco et al., 2003). Five of the indomethacin analogues tested show selectivity for COX-2 vs. COX-1 (Kalgutar et al., 2000a, Kalgutar et al., 2000b) but have not been tested vis-à-vis the endocannabinoid system. Finally, indomethacin morpholinamide has been reported to be a CB2 receptor-selective inverse agonist (Gallant et al., 1996, New and Wong, 2003).

Section snippets

Materials

Anandamide [ethanolamine-1-3H] (specific activity 60 Ci/mmol), anandamide [arachidonoyl 5,6,8,9,11,12,14,15-3H] (specific activity 200 Ci/mmol), palmitoylethanolamide [ethanolamine-1-3H] ([3H]-PEA; specific activity 20 Ci/mmol) and 2-oleoylglycerol [glycerol-1,2,3-3H] (specific activity 20 Ci/mmol) were purchased from American Radiolabeled Chemicals, Inc. St. Louis, MO, U.S.A. [3H]-CP55,940 ((−)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-trans-4-(3-hydroxypropyl) cyclohexanol) [side chain

Inhibition of [3H]-AEA hydrolysis in brain membranes by ibuprofen and related compounds

Initial assays of [3H]-AEA were undertaken with the ibuprofen analogues using mouse brain homogenates. The mouse is rather appropriate in this respect, since it has been shown that genetic deletion of FAAH results in a > 99% loss in the ability of brain homogenates to hydrolyze AEA at an assay pH of 7.2 (Cravatt et al., 2001). Assuming no dramatic mouse strain differences, an inhibition of the hydrolytic activity of [3H]-AEA by test compounds in the mouse brain homogenates represents an

Discussion

In the present study a series of indomethacin and ibuprofen analogues were tested with the aim of finding a compound with a dual action upon FAAH and COX-2, but with a more pronounced FAAH action than the original parent compounds. With respect to the indomethacin series, only one compound showed an increased potency towards FAAH, in contrast to the situation for COX-2 (Kalgutar et al., 2000a, Kalgutar et al., 2000b). Furthermore, the FAAH inhibition produced by this compound was not complete,

Acknowledgements

The authors are indebted to Britt Jacobsson and Ingrid Persson for expert technical assistance with some of the experiments reported here. C.F. would like to thank the Swedish Research Council (Grant no. 12158, medicine), Gun and Bertil Stohne's Foundation, Konung Gustaf V's and Drottning Victorias Foundation, Stiftelsen för Gamla Tjänarinnor and the Research Funds of the Medical Faculty, Umeå University for financial support. The postdoctoral research of S.V. was supported by a grant from the

References (43)

  • K.R. Kozak et al.

    Oxidative metabolism of endocannabinoids. Prostaglandins Leukot

    Essent. Fatty Acids

    (2002)
  • A.H. Lichtman et al.

    Mice lacking fatty acid amide hydrolase exhibit a cannabinoid receptor-mediated phenotypic hypoalgesia

    Pain

    (2004)
  • D.C. New et al.

    BML-190 and AM251 act as inverse agonists at the human cannabinoid CB2 receptor: signalling via cAMP and inositol phosphates

    FEBS Lett.

    (2003)
  • B. Paylor et al.

    The potency of the fatty acid amide hydrolase inhibitor URB597 is dependent upon the assay pH

    Pharmacol. Res.

    (2006)
  • A. Sandberg et al.

    Measurement of saturable and non-saturable components of anandamide uptake into P19 embryonic carcinoma cells in the presence of fatty acid-free bovine serum albumin

    Chem. Phys. Lipids

    (2005)
  • P.C. Schmid et al.

    Properties of rat liver N-acylethanolamine amidohydrolase

    J. Biol. Chem.

    (1985)
  • A. Singh Tahim et al.

    Inflammatory mediators convert anandamide into a potent activator of the vanilloid type 1 transient receptor potential receptor in nociceptive primary sensory neurons

    Neuroscience

    (2005)
  • G. Tiger et al.

    Pharmacological properties of rat brain fatty acid amidohydrolase in different subcellular fractions using palmitoylethanolamide as substrate

    Biochem. Pharmacol.

    (2000)
  • N. Ueda et al.

    Purification and characterization of an acid amidase selective for N-palmitoylethanolamine, a putative endogenous anti-inflammatory substance

    J. Biol. Chem.

    (2001)
  • S.E. Andersson et al.

    Tissue and intracellular pH in normal periarticular soft tissue and during different phases of antigen induced arthritis in the rat

    J. Rheumatol.

    (1999)
  • M. Ates et al.

    Intrathecally applied flurbiprofen produces an endocannabinoid-dependent antinociception in the rat formalin test

    Eur. J. Neurosci.

    (2003)
  • Cited by (61)

    • Novel propanamides as fatty acid amide hydrolase inhibitors

      2017, European Journal of Medicinal Chemistry
    • Potent multitarget FAAH-COX inhibitors: Design and structure-activity relationship studies

      2016, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Agents designed to achieve this objective include nitric oxide (NO) donors-NSAIDs [16,17], COX-2 inhibitors–NO–donors [18,19], hydrogen sulfide (H2S) donors-NSAIDs [20–22], as well as compounds that block distinct enzymes of the AA pathway, such as COX/lipoxygenase [23,24] and COX-2/soluble epoxy hydrolase (sEH) [25]. Another potential multitarget strategy to treat inflammation is the concomitant inhibition of COX and fatty acid amide hydrolase (FAAH) [26] [27–33], a serine hydrolase that deactivates a family of analgesic and anti-inflammatory lipid amides that are produced by host-defense cells and other cells in the body [34,35]. These lipid mediators include the endocannabinoid anandamide (arachidonoylethanolamide) – which engages cannabinoid-1 (CB1) and CB2 receptors to suppress neutrophil migration [36] and prevent immune-cell recruitment [37,38] – as well as the endogenous peroxisome proliferator-activate receptor-α (PPAR-α) agonists, palmitoylethanolamide (PEA) and oleoylethanolamide (OEA) [39–41].

    • Inhibition of FAAH, TRPV1, and COX2 by NSAID-serotonin conjugates

      2014, Bioorganic and Medicinal Chemistry Letters
    View all citing articles on Scopus
    1

    Present address: Medical Products Agency, SE-751 03 Uppsala, Sweden.

    2

    Present address: Department of Medical Biosciences, Umeå University, SE-901 87 Umeå, Sweden.

    3

    Present address: Unité de Chimie pharmaceutique et de Radiopharmacie, Université catholique de Louvain, Avenue Mounier, 73, UCL-CMFA 73.40, B-1200 Brussels, Belgium.

    View full text