Dynamic regulation of the endocannabinoid system: implications for analgesia

Devi Rani Sagar, A Gemma Gaw, Bright N Okine, Stephen G Woodhams, Amy Wong, David A Kendall, Victoria Chapman, Devi Rani Sagar, A Gemma Gaw, Bright N Okine, Stephen G Woodhams, Amy Wong, David A Kendall, Victoria Chapman

Abstract

The analgesic effects of cannabinoids are well documented, but these are often limited by psychoactive side-effects. Recent studies indicate that the endocannabinoid system is dynamic and altered under different pathological conditions, including pain states. Changes in this receptor system include altered expression of receptors, differential synthetic pathways for endocannabinoids are expressed by various cell types, multiple pathways of catabolism and the generation of biologically active metabolites, which may be engaged under different conditions. This review discusses the evidence that pain states alter the endocannabinoid receptor system at key sites involved in pain processing and how these changes may inform the development of cannabinoid-based analgesics.

Figures

Figure 1
Figure 1
Proposed biosynthetic pathways for the generation of AEA from its arachidonoyl containing NAPE (NArPE) precursor. NAPEs are formed from phosphatidyl choline and phosphatidyl ethanolamine membrane precursors by an as yet uncharacterised N-acyl transferase enzyme. The most widely accepted route of AEA biosynthesis is via NAPE-PLD [142,143]. This enzyme is also responsible for the generation of other NAEs including OEA and PEA from their corresponding NAPE precursor. The serine hydrolase αβh4 can generate lysoNAPE and glycerophospho-N-acyl ethanolamine (GpNAE), including glycerophospho-N-arachidonoyl ethanolamine (GpAEA), glycerophospho-N-oleoyl ethanolamine (GpOEA) and glycerophospho-N-palmitoyl ethanolamine (GpPEA) intermediates that are subsequently hydrolysed by a metal dependant phosphodiesterase to produce AEA, OEA and PEA, respectively. In mouse brain, this enzyme has been identified as GDE1 [144]. LPS induced synthesis of AEA involves the generation of phosphorylated AEA (pAEA) via PLC which is then converted to AEA by phosphatases. In mouse brain, this phosphatase has been identified as PTPN22 [83]. Whether this third pathway contributes to the synthesis of other NAEs such as OEA and PEA remains to be determined.
Figure 2
Figure 2
Proposed metabolic pathways for the breakdown of AEA and 2-AG via hydrolase and oxygenase pathways.
Figure 3
Figure 3
A: The endocannabinoid AEA and related NAEs PEA and OEA are broken down by FAAH, 2-AG is primarily metabolized by MAGL. AEA is a ligand at CB1, CB2 and TRPV1 receptors and the nuclear receptor PPAR-α. OEA and PEA are ligands for PPAR-α. 2-AG is a ligand at CB1and CB2. Both AEA and 2-AG can be metabolized by COX2, LOX and CYP450 to form biologically active metabolites, some of which are ligands for CB1, CB2 and PPAR-α. B: Under pathological conditions, such as inflammatory or neuropathic pain, the presence of infiltrating immune cells or the activation of microglia provides another source of endocannabinoid synthesis and catabolism, as well as providing additional/or alternative receptor sites of action of the endocannabinoids, NAEs and their metabolites.

References

    1. Mechoulam R, Gaoni Y. The absolute configuration of delta-1-tetrahydrocannabinol, the major active constituent of hashish. Tetrahedron Lett. 1967;12:1109–1111. doi: 10.1016/S0040-4039(00)90646-4.
    1. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a Cannabinoid Receptor and Functional Expression of the Cloned Cdna. Nature. 1990;346:561–564. doi: 10.1038/346561a0.
    1. Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA, Felder CC, Herkenham M, Mackie K, Martin BR, et al. International Union of Pharmacology. XXVII. Classification of Cannabinoid Receptors. Pharmacol Rev. 2002;54:161–202. doi: 10.1124/pr.54.2.161.
    1. Egertova M, Elphick MR. Localisation of cannabinoid receptors in the rat brain using antibodies to the intracellular C-terminal tail of CB1. Journal of Comparative Neurology. 2000;422:159–171. doi: 10.1002/(SICI)1096-9861(20000626)422:2<159::AID-CNE1>;2-1.
    1. Brown AJ. Novel cannabinoid receptors. Br J Pharmacol. 2007;152:567–575. doi: 10.1038/sj.bjp.0707481.
    1. Johns DG, Behm DJ, Walker DJ, Ao Z, Shapland EM, Daniels DA, Riddick M, Dowell S, Staton PC, Green P, et al. The novel endocannabinoid receptor GPR55 is activated by atypical cannabinoids but does not mediate their vasodilator effects. Br J Pharmacol. 2007;152:825–831. doi: 10.1038/sj.bjp.0707419.
    1. Lauckner JE, Jensen JB, Chen HY, Lu HC, Hille B, Mackie K. GPR55 is a cannabinoid receptor that increases intracellular calcium and inhibits M current. Proc Natl Acad Sci USA. 2008;105:2699–2704. doi: 10.1073/pnas.0711278105.
    1. 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. doi: 10.1038/sj.bjp.0707460.
    1. Ross RA. The enigmatic pharmacology of GPR55. Trends Pharmacol Sci. 2009;30:156–163. doi: 10.1016/j.tips.2008.12.004.
    1. Caterina MJ, Julius D. The vanilloid receptor: a molecular gateway to the pain pathway. Annu Rev Neurosci. 2001;24:487–517. doi: 10.1146/annurev.neuro.24.1.487.
    1. Potenzieri C, Brink TS, Simone DA. Excitation of cutaneous C nociceptors by intraplantar administration of anandamide. Brain Res. 2009;1268:38–47. doi: 10.1016/j.brainres.2009.02.061.
    1. Singh Tahim A, Santha P, Nagy I. Inflammatory mediators convert anandamide into a potent activator of the vanilloid type 1 transient receptor potential receptor in nociceptive primary sensory neurons. Neuroscience. 2005;136:539–548. doi: 10.1016/j.neuroscience.2005.08.005.
    1. Alexander SP, Kendall DA. The complications of promiscuity: endocannabinoid action and metabolism. Br J Pharmacol. 2007;152:602–623. doi: 10.1038/sj.bjp.0707456.
    1. Lozovaya N, Min R, Tsintsadze V, Burnashev N. Dual modulation of CNS voltage-gated calcium channels by cannabinoids: Focus on CB1 receptor-independent effects. Cell Calcium. 2009;46:154–162. doi: 10.1016/j.ceca.2009.07.007.
    1. Maingret F, Patel AJ, Lazdunski M, Honore E. The endocannabinoid anandamide is a direct and selective blocker of the background K(+) channel TASK-1. EMBO J. 2001;20:47–54. doi: 10.1093/emboj/20.1.47.
    1. Kim HI, Kim TH, Shin YK, Lee CS, Park M, Song JH. Anandamide suppression of Na+ currents in rat dorsal root ganglion neurons. Brain Res. 2005;1062:39–47. doi: 10.1016/j.brainres.2005.09.004.
    1. Spivak CE, Lupica CR, Oz M. The endocannabinoid anandamide inhibits the function of alpha4beta2 nicotinic acetylcholine receptors. Mol Pharmacol. 2007;72:1024–1032. doi: 10.1124/mol.107.036939.
    1. Racz I, Bilkei-Gorzo A, Markert A, Stamer F, Gothert M, Zimmer A. Anandamide effects on 5-HT(3) receptors in vivo. Eur J Pharmacol. 2008;596:98–101. doi: 10.1016/j.ejphar.2008.08.012.
    1. Costa B, Comelli F, Bettoni I, Colleoni M, Giagnoni G. The endogenous fatty acid amide, palmitoylethanolamide, has anti-allodynic and anti-hyperalgesic effects in a murine model of neuropathic pain: involvement of CB(1), TRPV1 and PPARgamma receptors and neurotrophic factors. Pain. 2008.
    1. Rockwell CE, Snider NT, Thompson JT, Heuvel JP Vanden, Kaminski NE. Interleukin-2 suppression by 2-arachidonyl glycerol is mediated through peroxisome proliferator-activated receptor gamma independently of cannabinoid receptors 1 and 2. Mol Pharmacol. 2006;70:101–111.
    1. Russo R, LoVerme J, La Rana G, D'Agostino G, Sasso O, Calignano A, Piomelli D. Synergistic antinociception by the cannabinoid receptor agonist anandamide and the PPAR-alpha receptor agonist GW7647. Eur J Pharmacol. 2007;566:117–119. doi: 10.1016/j.ejphar.2007.03.007.
    1. Sun Y, Alexander SP, Garle MJ, Gibson CL, Hewitt K, Murphy SP, Kendall DA, Bennett AJ. Cannabinoid activation of PPAR alpha; a novel neuroprotective mechanism. Br J Pharmacol. 2007;152:734–743. doi: 10.1038/sj.bjp.0707478.
    1. O'Sullivan SE. Cannabinoids go nuclear: evidence for activation of peroxisome proliferator-activated receptors. Br J Pharmacol. 2007;152:576–582. doi: 10.1038/sj.bjp.0707423.
    1. Cuzzocrea S, Mazzon E, Di Paola R, Peli A, Bonato A, Britti D, Genovese T, Muia C, Crisafulli C, Caputi AP. The role of the peroxisome proliferator-activated receptor-alpha (PPAR-alpha) in the regulation of acute inflammation. J Leukoc Biol. 2006;79:999–1010. doi: 10.1189/jlb.0605341.
    1. Genovese T, Mazzon E, Di Paola R, Cannavo G, Muia C, Bramanti P, Cuzzocrea S. Role of endogenous ligands for the peroxisome proliferators activated receptors alpha in the secondary damage in experimental spinal cord trauma. Exp Neurol. 2005;194:267–278. doi: 10.1016/j.expneurol.2005.03.003.
    1. Jhaveri MD, Richardson D, Robinson I, Garle MJ, Patel A, Sun Y, Sagar DR, Bennett AJ, Alexander SP, Kendall DA, et al. Inhibition of fatty acid amide hydrolase and cyclooxygenase-2 increases levels of endocannabinoid related molecules and produces analgesia via peroxisome proliferator-activated receptor-alpha in a model of inflammatory pain. Neuropharmacology. 2008;55:85–93. doi: 10.1016/j.neuropharm.2008.04.018.
    1. Sagar DR, Kendall DA, Chapman V. Inhibition of fatty acid amide hydrolase produces PPAR-alpha-mediated analgesia in a rat model of inflammatory pain. Br J Pharmacol. 2008;155:1297–1306. doi: 10.1038/bjp.2008.335.
    1. Staumont-Salle D, Abboud G, Brenuchon C, Kanda A, Roumier T, Lavogiez C, Fleury S, Remy P, Papin JP, Bertrand-Michel J, et al. Peroxisome proliferator-activated receptor alpha regulates skin inflammation and humoral response in atopic dermatitis. J Allergy Clin Immunol. 2008;121:962–968. doi: 10.1016/j.jaci.2007.12.1165.
    1. Galiegue S, Mary S, Marchand J, Dussossoy D, Carriere D, Carayon P, Bouaboula M, Shire D, Le Fur G, Casellas P. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem. 1995;232:54–61. doi: 10.1111/j.1432-1033.1995.tb20780.x.
    1. Herkenham M, Lynn AB, Little MD, Johnson MR, Melvin LS, Decosta BR, Rice KC. Cannabinoid receptor localization in brain. Proc Natl Acad Sci USA. 1990;87:1932–1936. doi: 10.1073/pnas.87.5.1932.
    1. Herkenham M, Lynn AB, Johnson MR, Melvin LS, Decosta BR, Rice KC. Characterization and Localization of Cannabinoid Receptors in Rat-Brain - a Quantitative Invitro Autoradiographic Study. Journal of Neuroscience. 1991;11:563–583.
    1. Tsou K, Brown S, Sanudo-Pena MC, Mackie K, Walker JM. Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system. Neuroscience. 1998;83:393–411. doi: 10.1016/S0306-4522(97)00436-3.
    1. Chin CL, Tovcimak AE, Hradil VP, Seifert TR, Hollingsworth PR, Chandran P, Zhu CZ, Gauvin D, Pai M, Wetter J, et al. Differential effects of cannabinoid receptor agonists on regional brain activity using pharmacological MRI. Br J Pharmacol. 2008;153:367–379. doi: 10.1038/sj.bjp.0707506.
    1. Beltramo M, Bernardini N, Bertorelli R, Campanella M, Nicolussi E, Fredduzzi S, Reggiani A. CB2 receptor-mediated antihyperalgesia: possible direct involvement of neural mechanisms. Eur J Neurosci. 2006;23:1530–1538. doi: 10.1111/j.1460-9568.2006.04684.x.
    1. Gong JP, Onaivi ES, Ishiguro H, Liu QR, Tagliaferro PA, Brusco A, Uhl GR. Cannabinoid CB2 receptors: immunohistochemical localization in rat brain. Brain Res. 2006;1071:10–23. doi: 10.1016/j.brainres.2005.11.035.
    1. Van Sickle MD, Duncan M, Kingsley PJ, Mouihate A, Urbani P, Mackie K, Stella N, Makriyannis A, Piomelli D, Davison JS, et al. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science. 2005;310:329–332. doi: 10.1126/science.1115740.
    1. Pertwee RG. Cannabinoid receptors and pain. Prog Neurobiol. 2001;63:569–611. doi: 10.1016/S0301-0082(00)00031-9.
    1. Iversen L, Chapman V. Cannabinoids: a real prospect for pain relief? Curr Opin Pharmacol. 2002;2:50–55. doi: 10.1016/S1471-4892(01)00120-5.
    1. Walker JM, Huang SM. Cannabinoid analgesia. Pharmacol Ther. 2002;95:127–135. doi: 10.1016/S0163-7258(02)00252-8.
    1. Hohmann AG, Tsou K, Walker JM. Cannabinoid modulation of wide dynamic range neurons in the lumbar dorsal horn of the rat by spinally administered WIN55,212-2. Neurosci Lett. 1998;257:119–122. doi: 10.1016/S0304-3940(98)00802-7.
    1. Kelly S, Chapman V. Selective cannabinoid CB1 receptor activation inhibits spinal nociceptive transmission in vivo. J Neurophysiol. 2001;86:3061–3064.
    1. Kelly S, Chapman V. Cannabinoid CB(1) receptor inhibition of mechanically evoked responses of spinal neurones in control rats, but not in rats with hindpaw inflammation. Eur J Pharmacol. 2003;474:209–216. doi: 10.1016/S0014-2999(03)02085-5.
    1. Kelly S, Jhaveri MD, Sagar DR, Kendall DA, Chapman V. Activation of peripheral cannabinoid CB1 receptors inhibits mechanically evoked responses of spinal neurons in noninflamed rats and rats with hindpaw inflammation. Eur J Neurosci. 2003;18:2239–2243. doi: 10.1046/j.1460-9568.2003.02957.x.
    1. Welch SP, Stevens DL. Antinociceptive activity of intrathecally administered cannabinoids alone, and in combination with morphine, in mice. J Pharmacol Exp Ther. 1992;262:10–18.
    1. Welch SP, Huffman JW, Lowe J. Differential blockade of the antinociceptive effects of centrally administered cannabinoids by SR141716A. J Pharmacol Exp Ther. 1998;286:1301–1308.
    1. Lichtman AH, Cook SA, Martin BR. Investigation of brain sites mediating cannabinoid-induced antinociception in rats: evidence supporting periaqueductal gray involvement. J Pharmacol Exp Ther. 1996;276:585–593.
    1. Finn DP, Jhaveri MD, Beckett SR, Roe CH, Kendall DA, Marsden CA, Chapman V. Effects of direct periaqueductal grey administration of a cannabinoid receptor agonist on nociceptive and aversive responses in rats. Neuropharmacology. 2003;45:594–604. doi: 10.1016/S0028-3908(03)00235-1.
    1. Martin WJ, Coffin PO, Attias E, Balinsky M, Tsou K, Walker JM. Anatomical basis for cannabinoid-induced antinociception as revealed by intracerebral microinjections. Brain Res. 1999;822:237–242. doi: 10.1016/S0006-8993(98)01368-7.
    1. Meng ID, Manning BH, Martin WJ, Fields HL. An analgesia circuit activated by cannabinoids. Nature. 1998;395:381–383. doi: 10.1038/26481.
    1. Agarwal N, Pacher P, Tegeder I, Amaya F, Constantin CE, Brenner GJ, Rubino T, Michalski CW, Marsicano G, Monory K, et al. Cannabinoids mediate analgesia largely via peripheral type 1 cannabinoid receptors in nociceptors. Nat Neurosci. 2007;10:870–879. doi: 10.1038/nn1916.
    1. Kelly S, Chapman V. Spinal administration of capsazepine inhibits noxious evoked responses of dorsal horn neurons in non-inflamed and carrageenan inflamed rats. Brain Res. 2002;935:103–108. doi: 10.1016/S0006-8993(02)02552-0.
    1. Scott DA, Wright CE, Angus JA. Evidence that CB-1 and CB-2 cannabinoid receptors mediate antinociception in neuropathic pain in the rat. Pain. 2004;109:124–131. doi: 10.1016/j.pain.2004.01.020.
    1. Clayton N, Marshall FH, Bountra C, O'Shaughnessy CT. CB1 and CB2 cannabinoid receptors are implicated in inflammatory pain. Pain. 2002;96:253–260. doi: 10.1016/S0304-3959(01)00454-7.
    1. Elmes SJ, Winyard LA, Medhurst SJ, Clayton NM, Wilson AW, Kendall DA, Chapman V. Activation of CB1 and CB2 receptors attenuates the induction and maintenance of inflammatory pain in the rat. Pain. 2005;118:327–335. doi: 10.1016/j.pain.2005.09.005.
    1. Richardson JD, Kilo S, Hargreaves KM. Cannabinoids reduce hyperalgesia and inflammation via interaction with peripheral CB1 receptors. Pain. 1998;75:111–119. doi: 10.1016/S0304-3959(97)00213-3.
    1. Akopian AN, Ruparel NB, Patwardhan A, Hargreaves KM. Cannabinoids desensitize capsaicin and mustard oil responses in sensory neurons via TRPA1 activation. J Neurosci. 2008;28:1064–1075. doi: 10.1523/JNEUROSCI.1565-06.2008.
    1. Guindon J, Hohmann AG. Cannabinoid CB2 receptors: a therapeutic target for the treatment of inflammatory and neuropathic pain. Br J Pharmacol. 2008;153:319–334. doi: 10.1038/sj.bjp.0707531.
    1. Jhaveri MD, Sagar DR, Elmes SJ, Kendall DA, Chapman V. Cannabinoid CB(2) Receptor-Mediated Anti-nociception in Models of Acute and Chronic Pain. Mol Neurobiol. 2007;36:26–35. doi: 10.1007/s12035-007-8007-7.
    1. Ibrahim MM, Rude ML, Stagg NJ, Mata HP, Lai J, Vanderah TW, Porreca F, Buckley NE, Makriyannis A, Malan TP., Jr CB2 cannabinoid receptor mediation of antinociception. Pain. 2006;122:36–42. doi: 10.1016/j.pain.2005.12.018.
    1. Malan TP, Jr, Ibrahim MM, Deng H, Liu Q, Mata HP, Vanderah T, Porreca F, Makriyannis A. CB2 cannabinoid receptor-mediated peripheral antinociception. Pain. 2001;93:239–245. doi: 10.1016/S0304-3959(01)00321-9.
    1. Valenzano KJ, Tafesse L, Lee G, Harrison JE, Boulet JM, Gottshall SL, Mark L, Pearson MS, Miller W, Shan S, et al. Pharmacological and pharmacokinetic characterization of the cannabinoid receptor 2 agonist, GW40 utilizing rodent models of acute and chronic pain, anxiety, ataxia and catalepsy. Neuropharmacology. 5833;48:658–672. doi: 10.1016/j.neuropharm.2004.12.008.
    1. Elmes SJ, Jhaveri MD, Smart D, Kendall DA, Chapman V. Cannabinoid CB2 receptor activation inhibits mechanically evoked responses of wide dynamic range dorsal horn neurons in naive rats and in rat models of inflammatory and neuropathic pain. Eur J Neurosci. 2004;20:2311–2320. doi: 10.1111/j.1460-9568.2004.03690.x.
    1. Ibrahim MM, Porreca F, Lai J, Albrecht PJ, Rice FL, Khodorova A, Davar G, Makriyannis A, Vanderah TW, Mata HP, Malan TP., Jr CB2 cannabinoid receptor activation produces antinociception by stimulating peripheral release of endogenous opioids. Proc Natl Acad Sci USA. 2005;102:3093–3098. doi: 10.1073/pnas.0409888102.
    1. Sagar DR, Kelly S, Millns PJ, O'Shaughnessey CT, Kendall DA, Chapman V. Inhibitory effects of CB1 and CB2 receptor agonists on responses of DRG neurons and dorsal horn neurons in neuropathic rats. Eur J Neurosci. 2005;22:371–379. doi: 10.1111/j.1460-9568.2005.04206.x.
    1. Jhaveri MD, Elmes SJ, Richardson D, Barrett DA, Kendall DA, Mason R, Chapman V. Evidence for a novel functional role of cannabinoid CB receptors in the thalamus of neuropathic rats. Eur J Neurosci. 2008;27:1722–1730. doi: 10.1111/j.1460-9568.2008.06162.x.
    1. Romero-Sandoval A, Nutile-McMenemy N, DeLeo JA. Spinal microglial and perivascular cell cannabinoid receptor type 2 activation reduces behavioral hypersensitivity without tolerance after peripheral nerve injury. Anesthesiology. 2008;108:722–734. doi: 10.1097/ALN.0b013e318167af74.
    1. Yamamoto W, Mikami T, Iwamura H. Involvement of central cannabinoid CB2 receptor in reducing mechanical allodynia in a mouse model of neuropathic pain. European Journal of Pharmacology. 2008;583:56–61. doi: 10.1016/j.ejphar.2008.01.010.
    1. Racz I, Nadal X, Alferink J, Banos JE, Rehnelt J, Martin M, Pintado B, Gutierrez-Adan A, Sanguino E, Manzanares J, et al. Crucial role of CB(2) cannabinoid receptor in the regulation of central immune responses during neuropathic pain. J Neurosci. 2008;28:12125–12135. doi: 10.1523/JNEUROSCI.3400-08.2008.
    1. Leichsenring A, Andriske M, Backer I, Stichel CC, Lubbert H. Analgesic and antiinflammatory effects of cannabinoid receptor agonists in a rat model of neuropathic pain. Naunyn Schmiedebergs Arch Pharmacol. 2009;379:627–636. doi: 10.1007/s00210-008-0386-4.
    1. Racz I, Nadal X, Alferink J, Banos JE, Rehnelt J, Martin M, Pintado B, Gutierrez-Adan A, Sanguino E, Bellora N, et al. Interferon-gamma is a critical modulator of CB(2) cannabinoid receptor signaling during neuropathic pain. J Neurosci. 2008;28:12136–12145. doi: 10.1523/JNEUROSCI.3402-08.2008.
    1. Malan TP, Jr, Ibrahim MM, Lai J, Vanderah TW, Makriyannis A, Porreca F. CB2 cannabinoid receptor agonists: pain relief without psychoactive effects? Curr Opin Pharmacol. 2003;3:62–67. doi: 10.1016/S1471-4892(02)00004-8.
    1. Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, Gibson D, Mandelbaum A, Etinger A, Mechoulam R. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258:1946–1949. doi: 10.1126/science.1470919.
    1. Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski NE, Schatz AR, Gopher A, Almog S, Martin BR, Compton DR, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol. 1995;50:83–90. doi: 10.1016/0006-2952(95)00109-D.
    1. 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 USA. 2001;98:3662–3665. doi: 10.1073/pnas.061029898.
    1. 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. doi: 10.1124/jpet.301.3.1020.
    1. Huang SM, Bisogno T, Trevisani M, Al-Hayani A, De Petrocellis L, Fezza F, Tognetto M, Petros TJ, Krey JF, Chu CJ, et al. An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc Natl Acad Sci USA. 2002;99:8400–8405. doi: 10.1073/pnas.122196999.
    1. LoVerme J, Russo R, La Rana G, Fu J, Farthing J, Mattace-Raso G, Meli R, Hohmann A, Calignano A, Piomelli D. Rapid broad-spectrum analgesia through activation of peroxisome proliferator-activated receptor-alpha. J Pharmacol Exp Ther. 2006;319:1051–1061. doi: 10.1124/jpet.106.111385.
    1. Guzman M, Lo Verme J, Fu J, Oveisi F, Blazquez C, Piomelli D. Oleoylethanolamide stimulates lipolysis by activating the nuclear receptor peroxisome proliferator-activated receptor alpha (PPAR-alpha) J Biol Chem. 2004;279:27849–27854. doi: 10.1074/jbc.M404087200.
    1. Guindon J, Desroches J, Beaulieu P. The antinociceptive effects of intraplantar injections of 2-arachidonoyl glycerol are mediated by cannabinoid CB2 receptors. Br J Pharmacol. 2007;150:693–701. doi: 10.1038/sj.bjp.0706990.
    1. Ahn K, McKinney MK, Cravatt BF. Enzymatic pathways that regulate endocannabinoid signaling in the nervous system. Chem Rev. 2008;108:1687–1707. doi: 10.1021/cr0782067.
    1. Egertova M, Simon GM, Cravatt BF, Elphick MR. Localization of N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD) expression in mouse brain: A new perspective on N-acylethanolamines as neural signaling molecules. J Comp Neurol. 2008;506:604–615. doi: 10.1002/cne.21568.
    1. Leung D, Saghatelian A, Simon GM, Cravatt BF. Inactivation of N-acyl phosphatidylethanolamine phospholipase D reveals multiple mechanisms for the biosynthesis of endocannabinoids. Biochemistry. 2006;45:4720–4726. doi: 10.1021/bi060163l.
    1. Liu J, Wang L, Harvey-White J, Huang BX, Kim HY, Luquet S, Palmiter RD, Krystal G, Rai R, Mahadevan A, et al. Multiple pathways involved in the biosynthesis of anandamide. Neuropharmacology. 2008;54:1–7. doi: 10.1016/j.neuropharm.2007.05.020.
    1. Bisogno T, Ligresti A, Di Marzo V. The endocannabinoid signalling system: biochemical aspects. Pharmacol Biochem Behav. 2005;81:224–238. doi: 10.1016/j.pbb.2005.01.027.
    1. Hashimotodani Y, Ohno-Shosaku T, Maejima T, Fukami K, Kano M. Pharmacological evidence for the involvement of diacylglycerol lipase in depolarization-induced endocanabinoid release. Neuropharmacology. 2008;54:58–67. doi: 10.1016/j.neuropharm.2007.06.002.
    1. Bisogno T, Howell F, Williams G, Minassi A, Cascio MG, Ligresti A, Matias I, Schiano-Moriello A, Paul P, Williams EJ, et al. Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J Cell Biol. 2003;163:463–468. doi: 10.1083/jcb.200305129.
    1. Sugiura T, Kishimoto S, Oka S, Gokoh M. Biochemistry, pharmacology and physiology of 2-arachidonoylglycerol, an endogenous cannabinoid receptor ligand. Prog Lipid Res. 2006;45:405–446. doi: 10.1016/j.plipres.2006.03.003.
    1. Yoshida T, Fukaya M, Uchigashima M, Miura E, Kamiya H, Kano M, Watanabe M. Localization of diacylglycerol lipase-alpha around postsynaptic spine suggests close proximity between production site of an endocannabinoid, 2-arachidonoyl-glycerol, and presynaptic cannabinoid CB1 receptor. J Neurosci. 2006;26:4740–4751. doi: 10.1523/JNEUROSCI.0054-06.2006.
    1. Nyilas R, Gregg LC, Mackie K, Watanabe M, Zimmer A, Hohmann AG, Katona I. Molecular architecture of endocannabinoid signaling at nociceptive synapses mediating analgesia. Eur J Neurosci. 2009;29:1964–1978. doi: 10.1111/j.1460-9568.2009.06751.x.
    1. Chapman V. The cannabinoid CB1 receptor antagonist, SR141716A, selectively facilitates nociceptive responses of dorsal horn neurones in the rat. Br J Pharmacol. 1999;127:1765–1767. doi: 10.1038/sj.bjp.0702758.
    1. Maione S, De Petrocellis L, de Novellis V, Moriello AS, Petrosino S, Palazzo E, Rossi FS, Woodward DF, Di Marzo V. Analgesic actions of N-arachidonoyl-serotonin, a fatty acid amide hydrolase inhibitor with antagonistic activity at vanilloid TRPV1 receptors. Br J Pharmacol. 2007;150:766–781. doi: 10.1038/sj.bjp.0707145.
    1. Beaulieu P, Bisogno T, Punwar S, Farquhar-Smith WP, Ambrosino G, Di Marzo V, Rice AS. Role of the endogenous cannabinoid system in the formalin test of persistent pain in the rat. Eur J Pharmacol. 2000;396:85–92. doi: 10.1016/S0014-2999(00)00226-0.
    1. Walker JM, Huang SM, Strangman NM, Tsou K, Sanudo-Pena MC. Pain modulation by release of the endogenous cannabinoid anandamide. Proc Natl Acad Sci USA. 1999;96:12198–12203. doi: 10.1073/pnas.96.21.12198.
    1. Drew GM, Lau BK, Vaughan CW. Substance P drives endocannabinoid-mediated disinhibition in a midbrain descending analgesic pathway. J Neurosci. 2009;29:7220–7229. doi: 10.1523/JNEUROSCI.4362-08.2009.
    1. Mitrirattanakul S, Ramakul N, Guerrero AV, Matsuka Y, Ono T, Iwase H, Mackie K, Faull KF, Spigelman I. Site-specific increases in peripheral cannabinoid receptors and their endogenous ligands in a model of neuropathic pain. Pain. 2006;126:102–114. doi: 10.1016/j.pain.2006.06.016.
    1. Guasti L, Richardson D, Jhaveri M, Eldeeb K, Barrett D, Elphick MR, Alexander SP, Kendall D, Michael GJ, Chapman V. Minocycline treatment inhibits microglial activation and alters spinal levels of endocannabinoids in a rat model of neuropathic pain. Mol Pain. 2009;5:35. doi: 10.1186/1744-8069-5-35.
    1. Inoue K. The function of microglia through purinergic receptors: neuropathic pain and cytokine release. Pharmacol Ther. 2006;109:210–226. doi: 10.1016/j.pharmthera.2005.07.001.
    1. Carrier EJ, Kearn CS, Barkmeier AJ, Breese NM, Yang W, Nithipatikom K, Pfister SL, Campbell WB, Hillard CJ. Cultured rat microglial cells synthesize the endocannabinoid 2-arachidonylglycerol, which increases proliferation via a CB2 receptor-dependent mechanism. Mol Pharmacol. 2004;65:999–1007. doi: 10.1124/mol.65.4.999.
    1. Muccioli GG, Stella N. Microglia produce and hydrolyze palmitoylethanolamide. Neuropharmacology. 2008;54:16–22. doi: 10.1016/j.neuropharm.2007.05.015.
    1. 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. doi: 10.1523/JNEUROSCI.4830-06.2007.
    1. 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. doi: 10.1038/384083a0.
    1. Vandevoorde S, Lambert DM. The multiple pathways of endocannabinoid metabolism: a zoom out. Chem Biodivers. 2007;4:1858–1881. doi: 10.1002/cbdv.200790156.
    1. Deutsch DG, Chin SA. Enzymatic synthesis and degradation of anandamide, a cannabinoid receptor agonist. Biochem Pharmacol. 1993;46:791–796. doi: 10.1016/0006-2952(93)90486-G.
    1. 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. doi: 10.1074/jbc.M606646200.
    1. 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 USA. 2002;99:10819–10824. doi: 10.1073/pnas.152334899.
    1. Vandevoorde S, Jonsson KO, Labar G, Persson E, Lambert DM, Fowler CJ. Lack of selectivity of URB602 for 2-oleoylglycerol compared to anandamide hydrolysis in vitro. Br J Pharmacol. 2007;150:186–191. doi: 10.1038/sj.bjp.0706971.
    1. Tsuboi K, Takezaki N, Ueda N. The N-acylethanolamine-hydrolyzing acid amidase (NAAA) Chem Biodivers. 2007;4:1914–1925. doi: 10.1002/cbdv.200790159.
    1. Fowler CJ. The contribution of cyclooxygenase-2 to endocannabinoid metabolism and action. Br J Pharmacol. 2007;152:594–601. doi: 10.1038/sj.bjp.0707379.
    1. Samad TA, Moore KA, Sapirstein A, Billet S, Allchorne A, Poole S, Bonventre JV, Woolf CJ. Interleukin-1beta-mediated induction of Cox-2 in the CNS contributes to inflammatory pain hypersensitivity. Nature. 2001;410:471–475. doi: 10.1038/35068566.
    1. Kozak KR, Crews BC, Ray JL, Tai HH, Morrow JD, Marnett LJ. Metabolism of prostaglandin glycerol esters and prostaglandin ethanolamides in vitro and in vivo. J Biol Chem. 2001;276:36993–36998. doi: 10.1074/jbc.M105854200.
    1. Kozak KR, Marnett LJ. Oxidative metabolism of endocannabinoids. Prostaglandins Leukot Essent Fatty Acids. 2002;66:211–220. doi: 10.1054/plef.2001.0359.
    1. Burstein SH, Rossetti RG, Yagen B, Zurier RB. Oxidative metabolism of anandamide. Prostaglandins Other Lipid Mediat. 2000;61:29–41. doi: 10.1016/S0090-6980(00)00053-8.
    1. Matias I, Chen J, De Petrocellis L, Bisogno T, Ligresti A, Fezza F, Krauss AH, Shi L, Protzman CE, Li C, et al. Prostaglandin ethanolamides (prostamides): in vitro pharmacology and metabolism. J Pharmacol Exp Ther. 2004;309:745–757. doi: 10.1124/jpet.103.061705.
    1. 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. doi: 10.1194/jlr.M300475-JLR200.
    1. Sang N, Zhang J, Chen C. PGE2 glycerol ester, a COX-2 oxidative metabolite of 2-arachidonoyl glycerol, modulates inhibitory synaptic transmission in mouse hippocampal neurons. J Physiol. 2006;572:735–745.
    1. Sang N, Zhang J, Chen C. COX-2 oxidative metabolite of endocannabinoid 2-AG enhances excitatory glutamatergic synaptic transmission and induces neurotoxicity. J Neurochem. 2007;102:1966–1977. doi: 10.1111/j.1471-4159.2007.04668.x.
    1. Hu SS, Bradshaw HB, Chen JS, Tan B, Walker JM. Prostaglandin E2 glycerol ester, an endogenous COX-2 metabolite of 2-arachidonoylglycerol, induces hyperalgesia and modulates NFkappaB activity. Br J Pharmacol. 2008;153:1538–1549. doi: 10.1038/bjp.2008.33.
    1. 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;75:965–972. doi: 10.1124/mol.108.053439.
    1. Chen JK, Chen J, Imig JD, Wei S, Hachey DL, Guthi JS, Falck JR, Capdevila JH, Harris RC. Identification of novel endogenous cytochrome p450 arachidonate metabolites with high affinity for cannabinoid receptors. J Biol Chem. 2008;283:24514–24524. doi: 10.1074/jbc.M709873200.
    1. Cowart LA, Wei S, Hsu MH, Johnson EF, Krishna MU, Falck JR, Capdevila JH. The CYP4A isoforms hydroxylate epoxyeicosatrienoic acids to form high affinity peroxisome proliferator-activated receptor ligands. J Biol Chem. 2002;277:35105–35112. doi: 10.1074/jbc.M201575200.
    1. Fang X, Hu S, Xu B, Snyder GD, Harmon S, Yao J, Liu Y, Sangras B, Falck JR, Weintraub NL, Spector AA. 14,15-Dihydroxyeicosatrienoic acid activates peroxisome proliferator-activated receptor-alpha. Am J Physiol Heart Circ Physiol. 2006;290:H55–63. doi: 10.1152/ajpheart.00427.2005.
    1. Cravatt BF, Demarest K, Patricelli MP, Bracey MH, Giang DK, Martin BR, Lichtman AH. Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase. Proc Natl Acad Sci USA. 2001;98:9371–9376. doi: 10.1073/pnas.161191698.
    1. Lichtman AH, Shelton CC, Advani T, Cravatt BF. Mice lacking fatty acid amide hydrolase exhibit a cannabinoid receptor-mediated phenotypic hypoalgesia. Pain. 2004;109:319–327. doi: 10.1016/j.pain.2004.01.022.
    1. Jayamanne A, Greenwood R, Mitchell VA, Aslan S, Piomelli D, Vaughan CW. Actions of the FAAH inhibitor URB597 in neuropathic and inflammatory chronic pain models. Br J Pharmacol. 2006;147:281–288. doi: 10.1038/sj.bjp.0706510.
    1. Chang L, Luo L, Palmer JA, Sutton S, Wilson SJ, Barbier AJ, Breitenbucher JG, Chaplan SR, Webb M. Inhibition of fatty acid amide hydrolase produces analgesia by multiple mechanisms. Br J Pharmacol. 2006;148:102–113. doi: 10.1038/sj.bjp.0706699.
    1. Lichtman A, Leung D, Shelton C, Saghatelian A, Hardouin C, Boger D, Cravatt B. Reversible Inhibitors of Fatty Acid Amide Hydrolase That Promote Analgesia: Evidence for an Unprecedented Combination of Potency and Selectivity. Journal of Pharmacology and Experimental Therapeutics. 2004;311:441–448. doi: 10.1124/jpet.104.069401.
    1. Kathuria S, Gaetani S, Fegley D, Valino F, Duranti A, Tontini A, Mor M, Tarzia G, La Rana G, Calignano A, et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nature Medicine. 2003;9:76–81. doi: 10.1038/nm803.
    1. Fegley D, Gaetani S, Duranti A, Tontini A, Mor M, Tarzia G, Piomelli D. Characterization of the fatty acid amide hydrolase inhibitor cyclohexyl carbamic acid 3'-carbamoyl-biphenyl-3-yl ester (URB597): effects on anandamide and oleoylethanolamide deactivation. J Pharmacol Exp Ther. 2005;313:352–358. doi: 10.1124/jpet.104.078980.
    1. Russo R, Loverme J, La Rana G, Compton T, Parrot J, Duranti A, Tontini A, Mor M, Tarzia G, Calignano A, Piomelli D. The fatty-acid amide hydrolase inhibitor URB597 (cyclohexyl carbamic acid 3'-carbamoyl-biphenyl-3-yl ester) reduces neuropathic pain after oral administration in mice. J Pharmacol Exp Ther. 2007;322:236–242. doi: 10.1124/jpet.107.119941.
    1. Zhang D, Saraf A, Kolasa T, Bhatia P, Zheng GZ, Patel M, Lannoye GS, Richardson P, Stewart A, Rogers JC, et al. Fatty acid amide hydrolase inhibitors display broad selectivity and inhibit multiple carboxylesterases as off-targets. Neuropharmacology. 2007;52:1095–1105. doi: 10.1016/j.neuropharm.2006.11.009.
    1. Karbarz MJ, Luo L, Chang L, Tham CS, Palmer JA, Wilson SJ, Wennerholm ML, Brown SM, Scott BP, Apodaca RL, et al. Biochemical and Biological Properties of 4-(3-phenyl-[1,2,4]thiadiazol-5-yl)-piperazine-1-carboxylic acid phenylamide, a Mechanism-Based Inhibitor of Fatty Acid Amide Hydrolase. Anesthesia and Analgesia. 2009;108:316–329. doi: 10.1213/ane.0b013e31818c7cbd.
    1. Ahn K, Johnson DS, Mileni M, Beidler D, Long JZ, McKinney MK, Weerapana E, Sadagopan N, Liimatta M, Smith SE, et al. Discovery and Characterization of a Highly Selective FAAH Inhibitor that Reduces Inflammatory Pain. Chemistry & Biology. 2009;16:411–420. doi: 10.1016/j.chembiol.2009.02.013.
    1. Kinsey SG, Long JZ, O'Neal ST, Abdullah RA, Poklis JL, Boger DL, Cravatt BF, Lichtman AH. Blockade of endocannabinoid-degrading enzymes attenuates neuropathic pain. J Pharmacol Exp Ther. 2009;330:902–910. doi: 10.1124/jpet.109.155465.
    1. Jhaveri MD, Richardson D, Kendall DA, Barrett DA, Chapman V. Analgesic effects of fatty acid amide hydrolase inhibition in a rat model of neuropathic pain. J Neurosci. 2006;26:13318–13327. doi: 10.1523/JNEUROSCI.3326-06.2006.
    1. Comelli F, Giagnoni G, Bettoni I, Colleoni M, Costa B. The inhibition of monoacylglycerol lipase by URB602 showed an anti-inflammatory and anti-nociceptive effect in a murine model of acute inflammation. Br J Pharmacol. 2007;152:787–794. doi: 10.1038/sj.bjp.0707425.
    1. Long JZ, Li WW, Booker L, Burston JJ, Kinsey SG, Schlosburg JE, Pavon FJ, Serrano AM, Selley DE, Parsons LH, et al. Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects. Nature Chemical Biology. 2009;5:37–44. doi: 10.1038/nchembio.129.
    1. Bisogno T, Ortar G, Petrosino S, Morera E, Palazzo E, Nalli M, Maione S, Di Marzo V. Development of a potent inhibitor of 2-arachidonoylglycerol hydrolysis with antinociceptive activity in vivo. Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 2009;1791:53–60. doi: 10.1016/j.bbalip.2008.10.007.
    1. D'Agostino G, La Rana G, Russo R, Sasso O, Iacono A, Esposito E, Mattace Raso G, Cuzzocrea S, Loverme J, Piomelli D, et al. Central administration of palmitoylethanolamide reduces hyperalgesia in mice via inhibition of NF-kappaB nuclear signalling in dorsal root ganglia. Eur J Pharmacol. 2009;613:54–59. doi: 10.1016/j.ejphar.2009.04.022.
    1. Petrosino S, Palazzo E, de Novellis V, Bisogno T, Rossi F, Maione S, Di Marzo V. Changes in spinal and supraspinal endocannabinoid levels in neuropathic rats. Neuropharmacology. 2007;52:415–422. doi: 10.1016/j.neuropharm.2006.08.011.
    1. Minami T, Nakano H, Kobayashi T, Sugimoto Y, Ushikubi F, Ichikawa A, Narumiya S, Ito S. Characterization of EP receptor subtypes responsible for prostaglandin E2-induced pain responses by use of EP1 and EP3 receptor knockout mice. Br J Pharmacol. 2001;133:438–444. doi: 10.1038/sj.bjp.0704092.
    1. Minami T, Uda R, Horiguchi S, Ito S, Hyodo M, Hayaishi O. Allodynia evoked by intrathecal administration of prostaglandin F2 alpha to conscious mice. Pain. 1992;50:223–229. doi: 10.1016/0304-3959(92)90166-9.
    1. Piomelli D. The molecular logic of endocannabinoid signalling. Nat Rev Neurosci. 2003;4:873–884. doi: 10.1038/nrn1247.
    1. Schmid HH. Pathways and mechanisms of N-acylethanolamine biosynthesis: can anandamide be generated selectively? Chem Phys Lipids. 2000;108:71–87. doi: 10.1016/S0009-3084(00)00188-2.
    1. Simon GM, Cravatt BF. Anandamide biosynthesis catalyzed by the phosphodiesterase GDE1 and detection of glycerophospho-N-acyl ethanolamine precursors in mouse brain. J Biol Chem. 2008;283:9341–9349. doi: 10.1074/jbc.M707807200.

Source: PubMed

3
Abonnere