AJA Asian Journal of Anesthesiology

Advancing, Capability, Improving lives

Review Article
Volume 51, Issue 4, Pages 161-170
Lih-Chu Chiou 1.2.3.4 , Sherry Shu-Jung Hu 5 , Yu-Cheng Ho 1
4113 Views

Outline


Abstract

Marijuana has been used to relieve pain for centuries, but its analgesic mechanism has only been understood during the past two decades. It is mainly mediated by its constituents, cannabinoids, through activating central cannabinoid 1 (CB1) receptors, as well as peripheral CB1 and CB2 receptors. CB2-selective agonists have the benefit of lacking CB1 receptor-mediated CNS side effects. Anandamide and 2-arachidonoylglycerol (2-AG) are two intensively studied endogenous lipid ligands of cannabinoid receptors, termed endocannabinoids, which are synthesized on demand and rapidly degraded. Thus, inhibitors of their degradation enzymes, fatty acid amide hydrolase and monoacylglycerol lipase (MAGL), respectively, may be superior to direct cannabinoid receptor ligands as a promising strategy for pain relief. In addition to the antinociceptive properties of exogenous cannabinoids and endocannabinoids, involving their biosyn.

The putative role of 2-AG generated after activating the above neurotransmitter receptors in stress-induced analgesia is also discussed.

Keywords

Cannabinoids; Endocannabinoids: anandamide; Neuropeptides: orexin; Pain; Receptors: metabotropic glutamate;


1. Cannabinoids

Marijuana has been used for relieving pain for more than four centuries, but its analgesic mechanism was only understood during the past two decades, after cannabinoid receptors were discovered. The main active component of marijuana is Δ9-tetrahydrocannabinol (Δ9-THC),1 which exerts several pharmacological actions, including analgesia, immobilization, heightened sensory awareness, euphoria, hypothermia, impairment of short-term memory, and suppression of immune responses.2 Compounds mimicking the effects of Δ9-THC through activating cannabinoid receptors are termed cannabinoids with three categories. Phytocannabinoids are the constituents isolated from marijuana.3 Endocannabinoids are endogenous ligands of cannabinoid receptors.4 Synthetic cannabinoids are compounds developed for potential medical uses, based on the concept that they would mimic the therapeutic effects of phytocannabinoids while having no psychoactivity.

2. CB1 and CB2 receptors

The first cannabinoid receptor, CB1, was cloned by Matsuda et al5 from a rat brain cDNA library. Thereafter, Munro's group6 cloned another cannabinoid receptor, CB2, from an HL-60 cell cDNA library. Both CB1 and CB2 receptors are seven-transmembrane G-protein-coupled receptors, mainly coupled to Gi/o proteins,7 and share 44% overall identity (68% identity for the transmembrane domains).

CB1 receptors are enriched in the nervous systems and moderately expressed in peripheral tissues.589 In both rat and human brains, CB1 receptors are densely distributed in the frontal cerebral cortex, basal ganglia, cerebellum, hippocampus, hypothalamus, and anterior cingulate cortex, but rarely in the brainstem nuclei.10 The latter finding may account for the low toxicity of cannabinoids when given in accidental overdose.

At the cellular level, CB1 receptors are mainly localized to axons and nerve terminals and are largely absent from neuronal somas or dendrites.11 This ultra-structural finding, suggesting a predominantly presynaptic localization of CB1 receptors, is consistent with the functional finding that activation of CB1 receptors inhibits calcium channels and activates potassium channels, leading to inhibition of neurotransmitter release.1213

The first cannabinoid receptor, CB1, was cloned by Matsuda et al5 from a rat brain cDNA library. Thereafter, Munro's group6 cloned another cannabinoid receptor, CB2, from an HL-60 cell cDNA library. Both CB1 and CB2 receptors are seven-transmembrane G-protein-coupled receptors, mainly coupled to Gi/o proteins,7 and share 44% overall identity (68% identity for the transmembrane domains).

CB1 receptors are enriched in the nervous systems and moderately expressed in peripheral tissues.589 In both rat and human brains, CB1 receptors are densely distributed in the frontal cerebral cortex, basal ganglia, cerebellum, hippocampus, hypothalamus, and anterior cingulate cortex, but rarely in the brainstem nuclei.10 The latter finding may account for the low toxicity of cannabinoids when given in accidental overdose.

At the cellular level, CB1 receptors are mainly localized to axons and nerve terminals and are largely absent from neuronal somas or dendrites.11 This ultra-structural finding, suggesting a predominantly presynaptic localization of CB1 receptors, is consistent with the functional finding that activation of CB1 receptors inhibits calcium channels and activates potassium channels, leading to inhibition of neurotransmitter release.1213

CB2 receptors are mainly distributed in immune cells.614 Although originally thought absent from the CNS, CB2 receptors were later found to be expressed in microglia, dorsal root ganglion, spinal cord, and sparsely in several brain regions, such as the cerebellum, cortex, and brainstem.1516Nevertheless, CB2 receptors in the CNS can be strongly induced in sensory neurons and spinal cord in neuropathic and inflammatory pain models, as well as in spinal microglia and macrophages in human postmortem spinal cord specimens of multiple sclerosis and amyotrophic lateral sclerosis.17

3. Endocannabinoids

The finding of cannabinoid receptors in the brain motivated the search for their endogenous ligands to elucidate their functional roles. Indeed, several endogenous arachidonic acid derivatives have been identified to have cannabimimetic actions and are termed endocannabinoids; they include anandamide (N-arachidonoylethanolamine),18 2-arachidonoylglycerol (2-AG),1920 noladin ether (2-arachidonoylglyceryl ether),21 virodhamine (O-arachidonoylethanolamine),22 and N-arachidonoyldopamine.23 These endocannabinoids, like other lipid mediators, such as prostaglandins, are synthesized and then released locally on demand in activity-dependent and receptor-regulated manners.24 Released endocannabinoids are rapidly inactivated by uptake and enzymatic degradation. Agents that interfere with the inactivation of endocannabinoids may provide a pharmacological means to modify cannabinoid-mediated functions.2526 Anandamide and 2-AG are the two best studied endocannabinoids. Despite similar chemical structures, they have distinct pathways for biosynthesis and degradation.

3.1. Anandamide

3.1.1. Anandamide synthesis

Anandamide is an N-acylethanolamine and can be formed by a two-step biosynthesis scheme catalyzed by N-acetyltransferase and N-arachidonoyl phosphatidylethanolamine-phospholipase D (NAPE-PLD).2728 Anandamide can also be generated from NAPE through an α/β-hydrolase-4 (ABHD4) and glycerophosphodiesterase 1 (GDE1) enzymatic pathway.293031 However, basal levels of brain anandamide in mice lacking the putative synthetic enzymes, either NAPE-PLD32 or GDE1,33 or both GDE1 and NAPE-PLD,33were not different from those in wild type mice. Thus, additional biosynthetic pathways are suggested. Because basal levels of anandamide were measured in these studies, it is not known if activity-dependent anandamide levels differ between the genotypes.

3.1.2. Anandamide degradation

In contrast to its obscure synthetic pathway, anandamide is known to be rapidly inactivated by the degradation enzyme, fatty acid amide hydrolase (FAAH). It is a 579 amino acid serine amidohydrolase, widely distributed in the brain.3435 FAAH is mainly located postsynaptically, complimentary to the presynaptically located CB1 receptors, in many, but not all, brain regions.353637

3.1.3. Anandamide uptake

In addition to the degradation enzyme, a membrane transporter has been proposed to play a role in the uptake of anandamide. However, its existence remains controversial3839 because several putative anandamide transporter inhibitors, such as AM404, VDM11, and LY218240, which enhance the effects of anandamide,4041 also inhibit FAAH.424344

3.1.4. Anandamide and type 1 transient receptor potential vanilloid channels

Unlike 2-AG, which primarily acts at cannabinoid receptors, anandamide activates both cannabinoid receptors and type 1 transient receptor potential vanilloid (TRPV1) channels.45 The TRPV1 channel is a voltage-gated cation channel which is distributed in both the peripheral and the central nervous system. Capsaicin, the constituent of chili pepper, is a potent agonist. Anandamide and capsaicin have similar binding affinities in displacing [3H]-resiniferatoxin from TRPV1.46 Hence, activation of TRPV1 by anandamide may complicate the interpretation of the effects of experiments using FAAH or anandamide transporter inhibitors.

3.2. 2-AG

3.2.1. 2-AG synthesis

2-AG is a monoacylglycerol (MAG) and is mainly formed by the hydrolysis of diacylglycerol (DAG) through DAG lipases (DAGLs). DAG can be generated through postsynaptic depolarization-induced Ca2+ influx and/or activation of Gq-protein-coupled receptors (GqPCRs) through phospholipase C (PLC).4748DAGLs are postsynaptic integral membrane proteins and exist as two isoforms, α and β.4950 DAGLα is the major synthetic enzyme for 2-AG in the brain.51

3.2.2. 2-AG degradation

2-AG is mainly degraded by a cytosolic serine hydrolase, MAG lipase (MAGL),5253 which is localized in presynaptic terminals, where CB1receptors are also expressed.52 FAAH, the main degradation enzyme of anandamide, metabolizes 2-AG in vitro5455 but not efficiently in vivo.56

4. Cannabinoids are antinociceptive

After the identification of CB1 and CB2 receptors, subtype-selective agonists and antagonists, as well as knockout mice, were actively developed. With these tools, the analgesic mechanisms of cannabinoids were explored. Systemic administration of cannabinoids can produce an analgesic effect with an efficacy comparable to opioids in acute pain animal models,57 and was even more effective than opioids in some chronic pain models.5859 These analgesic effects are mainly mediated by CB1 receptors in the CNS,596061and by both CB162 and CB26364 receptors in the periphery.

4.1. CB1 receptor-mediated central site of action

In the CNS, the analgesic effect of cannabinoids is mainly mediated through CB1 receptors located in structures that mediate nociceptive neurotransmission, including the spinal dorsal horn, periaqueductal gray (PAG),65 dorsal raphe nuclei,66 and thalamic ventroposterolateral nucleus.67 At the spinal level, electrophysiological and c-fos expression studies support that cannabinoids inhibit spinal dorsal neuronal activity to produce analgesia.59616268 Among the supraspinal areas involved in the modulation of nociception, the midbrain PAG has been extensively studied.

4.2. The midbrain PAG is a supraspinal site of action

Activation of the PAG produces analgesia,69 through activating the downstream rostroventral medulla (RVM), which sends inhibitory projections to the dorsal horn of the spinal cord.70 This PAG-RVM-spinal dorsal horn circuit constitutes a key endogenous descending pain inhibitory pathway. Studies using microinjection techniques in pain models suggest that the PAG is the site for cannabinoids producing CB1-mediated analgesic effects.6671Immunohistochemical72 and electrophysiological73 studies support that cannabinoids produce analgesic effects in the PAG via a disinhibition (inhibition of GABAergic transmission) mechanism mediated by CB1receptors.

4.3. CB2 receptor-mediated peripheral site of action

In the periphery, the antinociceptive effect of cannabinoids can be mediated both by CB162 and CB2 receptors. Peripherally mediated CB2 antinociception has been an especially attractive therapeutic target, as compounds that selectively activate CB2 receptors have the merit of avoiding CB1-mediated CNS side effects, such as hypomotility, catalepsy, hypothermia, and cognitive impairment. Intraplantar (i.pl.) administration of the CB2-preferring agonists, AM1241 or GW405833, effectively reduced nociceptive responses in several inflammatory pain models.747576 These antinociceptive effects were blocked by i.pl. injection of a CB2, but not a CB1 antagonist74 and were absent in CB2 receptor-knockout mice.7677 However, activation of CB2 receptors may also have a proinflammatory effect.78 In addition, CB2 receptors were strongly induced in sensory neurons and the spinal cord in neuropathic and inflamed rats.7980 This high inducibility of CB2 receptors renders them an attractive potential therapeutic target for pain treatment. A small scale, proof-of-concept clinical trial (32 patients, single site, double-blind, two-way crossover) using a CB2 receptor agonist, LY2828360 (80 mg/kg), for the treatment of osteoarthritis was conducted by Eli Lilly (Indianapolis, IN, USA). However, it failed to meet the primary endpoint.81

5. The role of endocannabinoids in pain regulation

Our understanding of the role of endocannabinoids in pain regulation comes from studies using both pharmacological and genetic approaches by blocking or silencing CB1 and CB2 receptors, or by inhibiting or silencing degradation or synthetic enzymes of anandamide and 2-AG. Table 1 summarizes the effects of inactivating the endocannabinoid system on nociceptive responses in various pain models.

5.1. CB1 receptor-mediated

5.1.1. CB1 antagonists

Systemic administration of SR141716A, the first CB1 antagonist,82 was initially found to have no effect in several pain models (Table 1). Later, it was reported to increase nociceptive responses when given by intrathecal (i.t.) or intraperitoneal (i.p.), but not i.pl., routes of administration, although replication of these findings has been variable (Table 1). Because SR141716A also acts as an inverse agonist of CB1 receptors,83 this might lead to pronociceptive effects in the above studies. The finding that systemic administration of AM4113,84 a neutral CB1 antagonist, produced no changes in nociception suggests there is little tonic endocannabinoid modulation of pain thresholds. However, i.t. injection of CB1 receptor antisense nucleotide caused hyperalgesia in the hot-plate test in mice,8586 suggesting an analgesic tone produced by endocannabinoids via CB1 receptors, at least at the spinal level.

5.1.2. CB1 knockout mice

Pain sensitivity has been examined in three lines of CB1 knockout mice of distinct genetic backgrounds, C57BL/6J,87 CD1,88 and 129/SvJ.89 Knockout of CB1 in C57BL/6J mice resulted in lower pain sensitivity in the hot-plate and formalin tests, but not the tail-flick test. However, the antinociceptive responses in these mutants may be confounded by their impaired locomotor activity.87 The other two lines of CB1 knockout mice displayed normal pain sensitivity in several nociceptive tests (Table 1). These results also suggest that tonic levels of endocannabinoids play a minor role in setting pain thresholds.

5.2. CB2 receptor-mediated

5.2.1. CB2 antagonists

SR144528, the first CB2 receptor antagonist, was used to investigate the role of CB2 receptor activation in the effect of endocannabinoids on pain regulation, mainly in inflammatory pain models (Table 1). In the formalin test, it was hyperalgesic when given by Intravenous (i.v.) injection63 but was ineffective by i.p. administration.9091 This CB2 antagonist induced hyperalgesia and enhanced edema when given by i.p. injection in the carrageenan model,92 but displayed anti-inflammatory effects by oral administration in the same model.78 Again, the inverse agonist property of SR144528 might also explain its hyperalgesic effects in these studies.

5.2.2. CB2 knockout mice

CB2 knockout mice, as compared with wild type mice, had increased thermal nociceptive responses in the plantar test, while having normal pain sensitivity in the hot-plate and tail-immersion assays.77 In a neuropathic pain model, CB2 knockout mice displayed enhanced nociceptive responses in the contralateral paw compared to control mice, which was also associated with enhanced interferon-γ and microglial expression in the contralateral spinal dorsal horn.9394 These results suggest that endocannabinoids play a CB2receptor-mediated tonic analgesic role, but also exert a proinflammatory tone during neuropathic pain state.

5.3. Role of anandamide in pain regulation

5.3.1. FAAH inhibitors

Several lines of evidence suggest that inhibition of FAAH, the degradation enzyme of anandamide, effectively reduced nociceptive responses in various pain models (Table 1). A series of irreversible carbamate FAAH inhibitors, such as URB532 and URB597, has been developed and patented9596 for pain treatment. Later, an α-ketooxazole reversible FAAH inhibitor, OL-135, was demonstrated to have high in vivo efficacy for reducing nociceptive responses in tail-immersion, hot-plate, formalin tests97 and neuropathic pain models.98Thereafter, a highly selective urea-based irreversible FAAH inhibitor, PF-3845, was developed by Pfizer (NYC, NY, USA) and shown to be an efficacious analgesic in inflammatory99100 and neuropathic101 pain models. These FAAH inhibitors significantly increased anandamide levels in the brain and mainly induced a CB1 receptor-mediated antinociception, suggesting that endogenous anandamide, when protected from degradation, can produce antinociception through CB1 receptors. Chronic systemic treatment with PF-3845 produced a persistent analgesic effect without tolerance in mice.101Interestingly, this compound was also effective when given by i.pl. administration.100 Recently, a peripherally acting FAAH inhibitor, URB937, was shown to be effective in inflammatory pain, neuropathic pain, and arthritis pain models.102 URB597 and URB937 were also found to be effective in the cisplatin (a chemotherapeutic agent) neuropathic pain model.103 An orally bioavailable urea-based irreversible carbamate FAAH inhibitor, PF-04457845, was developed by Pfizer and displayed significant CB1 receptor-dependent reductions in inflammatory and arthritis pain models.104105

The promising findings with FAAH inhibitors in reducing nociceptive responses in various animal pain models (Table 1) motivated advancing an FAAH inhibitor to a clinical pain trial. However, in a randomized, placebo-controlled clinical trial, chronic administration of PF-04457845 did not have significant analgesic effects in patients with osteoarthritis, although their plasma levels of endocannabinoids were significantly elevated and no significant side effects were reported.106 Although this Phase II clinical trial was terminated early due to ineffectiveness in an interim analysis, it remains to be further elucidated if FAAH inhibitors would be clinically effective for other pain indications,107 such as migraine, cancer pain or neuropathic pain induced by chemotherapy,103 diabetes, or herpes virus. The possible species difference in the FAAH between human and rodents should also be investigated.106 Interestingly, a noncovalent, reversible and noncompetitive FAAH inhibitor, AZ513, was published by AstraZenica,108 with a higher potency in inhibiting human FAAH than rat FAAH. It is not known if this compound will be more promising if there is any clinical trial in the future.

5.3.2. FAAH knockout mice

FAAH knockout mice exhibited reduced CB1-mediated pain sensitivity, but locomotor activity and body temperature were similar to those of wild type mice.34 This suggests that FAAH plays an important role in regulating endogenous anandamide-mediated analgesic tone but not in its motor- or thermo-regulation. CB1-mediated nociceptive responses in acute and inflammatory pain models were significantly reduced in FAAH knockout mice, as seen in animals treated with FAAH inhibitors. However, FAAH knockout mice had unchanged sensitivity in neuropathic pain models, as compared with wild type mice (Table 1).

5.4. Role of 2-AG in pain regulation

5.4.1. MAGL inhibitors

URB602, the first MAGL inhibitor, given by i.pl. injection, significantly reduced mechanical and thermal allodynia in a neuropathic pain model.109JZL184, a much more potent and selective MAGL inhibitor,110 was also effective in reducing nociceptive responses in several inflammatory and neuropathic pain models by either i.p., i.t., or i.pl. injection (Table 1). This suggests that acute administration of MAGL inhibitors has an analgesic effect.

5.4.2. MAGL knockout mice

Brain 2-AG levels were elevated more than 10-fold in MAGL knockout mice101111 close to the 8-fold elevation induced by acute administration of JZL184.110 However, because of this elevation of 2-AG desensitized CB1receptors in MAGL knockout mice, fewer and less active CB1 receptors existed in the brains of these mutants.101111 Similarly, treatment with a high dose of JZL184 for 6 consecutive days also led to desensitization of CB1 receptors, loss of analgesic activity, and cross-tolerance to the antiallodynic effects of CB1agonists and FAAH inhibitors.101111 This is significantly different from the finding that CB1 receptor function remained normal in mice lacking FAAH or which were chronically treated with FAAH inhibitors.34101112 The difference between FAAH and MAGL inhibition may be because 2-AG has a higher efficacy than anandamide at CB1 receptors.113114115 Thus, after long-term inhibition of its degradation enzyme, the enhanced and prolonged action of 2-AG, but not anandamide, leads to CB1 receptor desensitization. Interestingly, lower doses of MAGL inhibitors produce significant therapeutic benefits in preclinical models, while avoiding desensitization and tolerance of CB1 signaling.116117118

6. GqPCR activation-initiated 2-AG retrograde disinhibition in the PAG: A novel analgesic mechanism

A major pathway to generate 2-AG is by GqPCR activation. Activation of postsynaptic GqPCRs results in phospholipid hydrolysis by PLCβ to yield DAG. DAG is then hydrolyzed by DAGLα located at postsynaptic membranes to generate 2-AG, which then diffuses retrogradely across the synapse to activate presynaptic CB1 receptors and inhibit transmitter release.119 This neurotransmission inhibitory mechanism mediated by a GqPCR-PLC-DAGL-2-AG retrograde signaling cascade has been reported in several brain regions,119 including the PAG. Stimulation of the type 5 metabotropic glutamate receptor (mGluR5),120121122123124125 M1/M3 muscarinic acetylcholine receptor (M1/M3 mAChR),126 and orexin 1 receptor (OX1R),127has been reported to initiate the GqPCR-PLC-DAGL-2-AG signaling-mediated retrograde inhibition of GABAergic transmission (disinhibition) in PAG slices. This 2-AG-mediated disinhibition mechanism may contribute to the analgesic effects induced by activation of these GqPCRs.

6.1. mGluR5 activation

There are eight subtypes of mGluRs classified into three groups according to their coupling signaling pathways: Group I (mGluR1/5), Group II (mGluR2/3), and Group III (mGluR4/6-8).128129 Only Group I mGluRs belong to the GqPCR family.130 Drew and Vaughan131 found that activation of all three groups of mGluRs inhibits PAG GABAergic transmission, but only the effect of activating Group I mGluRs, specifically mGluR5, is mediated through endocannabinoids.123

6.2. Inhibiting glutamate transport

As postsynaptic mGluR5s are mainly located perisynaptically,132 they are primarily activated by spillover glutamate, such as when glutamate transporters are inhibited or overwhelmed, subsequently leading to disinhibition of the PAG through the mGluR5-PLC-DAGL-2-AG signaling.123

6.3. mGluR5 agonists

Recently, Gregg and colleagues,120 found that microinjection of an mGluR5 agonist into the dorsolateral PAG (dlPAG) triggered the release of 2-AG, but not anandamide. This effect was reversed by intra-dorsolateral PAG (intra-dlPAG) injection of a CB1 antagonist and DAGL inhibitor, or by expressing siRNA to silence DAGLα in the dlPAG. Immunohistochemical staining showed that mGluR5s were colocalized with DAGLα in the postsynaptic dendritic site, which is juxtaposed to the presynaptic localization of CB1receptors. These data support the fact that postsynaptic mGluR5-DAGLα cascade triggers retrograde 2-AG signaling in the PAG in vivo.

6.3.1. Activating NK1, NTS1/2, and CCK1 receptors

In addition to inhibiting glutamate transporter or directly applying mGluR agonists, there are several ways to release glutamate in sufficient amounts to activate perisynaptic mGluR5s, leading to disinhibition of the PAG through the mGluR5-PLC-DAGL-2-AG signaling. Indeed, Vaughan and colleagues proved that this 2-AG-mediated disinhibition signaling triggered by mGluR5 activation in ventrolateral PAG (vlPAG) slices can be induced by several analgesic neuropeptides.122124125 Substance P,122 neurotensin,125 and cholecystokinin (CCK),124 activated neurokinin 1, neurotensin 1/2, cholecystokinin 1 receptors, respectively, on glutamatergic somas to release glutamate, which then activated perisynaptic mGluR5s, and produced disinhibition in the PAG through 2-AG. It remains to be elucidated by in vivostudies if this PAG disinhibition mechanism mediated by the mGluR5-PLC-DAGL-2-AG signaling contributes to the antinociceptive effects of intra-PAG injection of substance P,133 neurotensin,134 and CCK.135

6.3.2. TRPV1 activation

Capsaicin, when injected into the PAG, induced antinociception, in contrast to its pronociceptive effect in the periphery.136137 In a study examining the analgesic mechanism of capsaicin at the supraspinal level, we recently found that capsaicin can also induce substantial glutamate release in the vlPAG to activate the mGluR5-PLC-DAGL-2-AG disinhibition mechanism, leading to analgesia.121 We found that capsaicin markedly increased the frequency of miniature excitatory postsynaptic currents (mEPSCs), but decreased evoked inhibitory postsynaptic currents (eIPSCs) in PAG slices. This IPSC inhibitory effect of capsaicin was antagonized by TRPV1, mGluR5, and CB1 antagonists and by a DAGL inhibitor. These results suggest that capsaicin activates the TRPV1 channels on glutamatergic terminals to release massive amounts of glutamate to activate postsynaptic mGluR5s, leading to disinhibition in the PAG through the mGluR5-PLC-DAGL-2-AG signaling. Finally, we proved that this mechanism contributes to the analgesic effect induced by intra-ventrolateral PAG (intra-vlPAG) injection of capsaicin,121 because this analgesic effect was reversed by intra-vlPAG injection of AM251 and 2-methyl-6-(phenylethynyl)pyridine, an mGluR5 antagonist. In agreement with our findings, Starowicz et al137 also found that intra-vlPAG capsaicin induced analgesia by releasing glutamate from the PAG to activate the OFF neurons in the RVM.

6.4. M1/M3 mAChR activation

The PAG receives dense cholinergic projections from the pontine tegmentum.138 Microinjection of cholinergic agonists into the PAG produces analgesia and related behaviors.139140 In PAG slices, Lau and Vaughan126found that carbachol suppressed IPSCs in PAG slices. This effect was mimicked by inhibiting acetylcholinesterase, occluded by a CB1 agonist, and reduced by blocking M1/M3 mAChRs and CB1 receptors, and inhibiting DAGL. These results suggest that activation of postsynaptic M1/M3 mAChR, like other GqPCRs, initiates the GqPCR-PLC-DAGL-2-AG disinhibition mechanism in the PAG.

6.5. OX1 receptor activation

Orexin A and orexin B, also named hypocretin 1 and 2, respectively, are a pair of hypothalamic neuropeptides and exert their biological functions through two GqPCRs, OX1 and OX2 receptors.141142 Orexin-expressing neurons are localized in the perifornical area and the lateral hypothalamus (LH),141142but project widely throughout the brain, including the PAG.143144145Orexins have been implicated in a myriad of physiological functions, such as sleep, reward, energy homeostasis, autonomic central control, and pain.146147 However, the mechanism(s) of how orexins regulate pain, especially at the supraspinal level, remained unclear until our findings in 2011.127 We found that orexin A depressed IPSCs in PAG slices and that this effect was inhibited by OX1 and CB1, but not OX2 antagonists, as well as by PLC and DAGL inhibitors. Moreover, the effect of orexin A was mimicked by a cannabinoid agonist and enhanced by an MAGL inhibitor. These results suggest that activation of postsynaptic OX1 receptors, like mGluR5s and M1/M3 mAChRs, initiates the GqPCR-PLC-DAGL-2-AG disinhibition mechanism in the PAG. We also proved that this mechanism contributes to the antinociceptive effect induced by intra-vlPAG injection of orexin A in the rat hot-plate test.127

Taken together, in the PAG, mGluR5 can be activated directly by an mGluR5 agonist,120 or by endogenous spillover glutamate: (1) when glutamate transport is inhibited;123 (2) when glutamatergic terminals are depolarized by TRPV1 activation by capsaicin121 or anandamide;148149 and (3) when glutamatergic cell bodies were excited by substance P via NK1 receptors,122neurotensin via NTS1/2 receptors,125 or CCK via CCK1 receptors.124 After mGluR5 activation, 2-AG was generated through the PLC-DAGL enzymatic cascade to produce retrograde inhibition of GABAergic transmission in the PAG, leading to analgesia.

7. Functional role of endocannabinoid analgesia in the PAG

7.1. Stress-induced analgesia

In 2005, Hohmann et al150 demonstrated that stress-induced analgesia (SIA), a phenomenon believed to represent the evolutionary impetus for the development of central pain inhibition mechanisms in humans and animals,151 is associated with the rapid formation of 2-AG and anandamide in the dlPAG. SIA induced by foot shock stress was blocked by CB1 antagonists and enhanced by the MAGL or FAAH inhibitor given by intra-dlPAG injection, but not affected by the CB2, opioid, or TRPV1 antagonist. Valverde et al152 also reported that forced swim stress-induced analgesia was absent in CB1 knockout mice.

Olango et al153 and Butler et al154 also demonstrated that endocannabinoids are involved in fear-conditioned analgesia (FCA) in rats re-exposed to the context previously associated with foot shock. They found that intra-dlPAG injection of the CB1 antagonist attenuated, and the FAAH inhibitor enhanced, FCA in rats injected with i.pl. formalin. Elevated anandamide in the dlPAG was specifically associated with FCA.153

Together, these results suggest that endocannabinoids, either anandamide or 2-AG, can be induced to release by various stressors, either electrical foot shock, swimming or fear, or during a pain state to activate the CB1 receptors in the PAG, serving as an endogenous analgesic protector. However, how a stress triggers endocannabinoid release to induce SIA remains unclear. The disinhibition mechanism mediated by 2-AG in the PAG through the GqPCR-PLC-DAGL signaling might be such a mechanism.

7.2. mGluR5 involvement

Recently, Gregg and colleagues120 found that intra-dlPAG injection of an mGluR5 agonist increased PAG 2-AG and enhanced SIA. This effect was reversed by intra-dlPAG injection of the CB1 receptor antagonist and DAGL inhibitors, and by silencing DAGLα in the dlPAG.120 These data suggest that the mGluR5-PLC-DAGLα-2-AG disinhibition mechanism in the PAG contributes to SIA.

7.3. Orexin involvement

Two studies have suggested that endogenous orexins play a role in SIA. Watanabe et al155 reported that SIA induced by electrical foot shock was absent in prepro-orexin knockout mice. Xie et al156 found that in mice with hypothalamic orexin neurons degenerated by ataxin-3 expression, restraint stress-induced antinociception was significantly reduced, as compared to wild type controls. The reduction of SIA in orexin/ataxin-3 mice can be reversed by intracerebroventricular (i.c.v.) injection of orexin A.

We recently found that a 30-min restraint stress in mice induced analgesia in the hot-plate test accompanied with increased c-fos expression in LH orexin neurons. This SIA was blocked by intra-vlPAG injection of OX1 and CB1antagonists, respectively. These results suggest that restraint stress activates hypothalamic orexin neurons, releasing orexins to activate OX1 receptors in the vlPAG, initiating the GqPCR-PLC-DAGL-2-AG disinhibition mechanism in the PAG, leading to analgesia.157

7.4. Substance P/neurotensin involvements

Substance P is antinociceptive at the supraspinal level, in contrast to a pronociceptive action in the periphery. The PAG is one of the sites of action because intra-PAG injection of substance P induced antinociception.133Chemical (carbachol) stimulation of LH triggered substance P release in the PAG, leading to analgesia.158 Interestingly, activation of LH by restraint stress also released orexins, which in turn acted in the PAG to induce analgesia through 2-AG mediated disinhibition.157 It is interesting to know if substance P also plays a role in endocannabinoid-mediated SIA. Moreover, both orexin and substance P were released after LH activation, so it will be interesting to examine if there are interactions between these two neuropeptide systems during SIA.

Similarly, neurotensin was also suggested to be involved in SIA.159 It will also be interesting to see if neurotensin plays a role in endocannabinoid-mediated SIA.

7.5. Analgesic mechanism of acetaminophen

Acetaminophen, one of the most popular analgesic drugs, has been used for more than a century. Its mode of analgesic action is still a matter of debate. In 2005–2006, a new look for the analgesic effect of acetaminophen, involving the CB1 receptor, was proposed by two groups simultaneously.160161162Hogestatt et al162 demonstrated that N-arachidonoylphenolamine, known as AM404, can be formed in the brain after systemic administration of acetaminophen in rats. They suggested that after systemic administration, acetaminophen is deacetylated in the liver into p-aminophenol, which is then conjugated with arachidonic acid to form AM404 in the brain by FAAH, the degradation enzyme of anandamide. Further studies showed that the analgesic effects of acetaminophen were reduced by CB1 antagonists,160161 and absent in CB1 knockout mice.161

AM404 has complicated pharmacological properties. Like anandamide, it is a C20 unsaturated fatty acid amide, originally developed as a potent inhibitor for anandamide uptake,40 but it is also an FAAH inhibitor. Furthermore, AM404 is also a TRPV1 agonist, as potent as capsaicin.163 Recently, Mallet et al164 found that the TRPV1 channel in the brain is involved in acetaminophen-induced antinociception. They found that the oral antinociceptive effects in several pain models induced by acetaminophen at a clinically effective dose range observed in wild type mice, were absent in TRPV1 or FAAH knockout mice. The analgesic effect of AM404 (i.c.v.) was also absent in TRPV1 knockout mice.

Together, these studies suggest that the analgesic effects of oral acetaminophen, or its central active metabolite, AM404, are mediated by TRPV1 channels and CB1 receptors. However, TRPV1 activation usually causes neuroexcitation, whereas CB1R activation decreases neurotransmitter release. We recently proved that the mechanism we reported for the analgesic mechanism of capsaicin,121 can explain how acetaminophen can produce a CB1R-mediated analgesic effect via TRPV1 activation.165 We found that oral acetaminophen or intra-PAG microinjection of AM404 produced comparable antinociceptive effects in the rat hot-plate test. Both antinociceptive effects were antagonized by intra-PAG microinjection of antagonists of TRPV1, mGluR5, or CB1R. It is suggested that after oral administration, acetaminophen is deacetylated in the liver into p-aminophenol, which is then converted into AM404 in the brain. AM404 then activates TRPV1, like capsaicin, in the vlPAG, to release a great amount of glutamate, which activates the mGluR5-PLC-DAGLα-2-AG disinhibition mechanism in the PAG, leading to analgesia.

8. Conclusions and perspectives

After the identification of cannabinoid receptors and endocannabinoids, accumulating studies have emerged during the past two decades that have aimed to elucidate the regulatory mechanisms of the cannabinoid system in pain control, with a goal of developing new pain therapies. CB2 selective agonists have the merit of avoiding CB1 receptor-mediated CNS side effects. Inhibitors of anandamide and 2-AG degradation enzymes, FAAH and MAGL, respectively, have the merit of focusing action at generating sites. Despite promising preclinical results, the first clinical trial with an FAAH inhibitor failed. The efficacy of MAGL inhibitors remains to be determined. Because of its synthesis on demand, endogenous 2-AG levels can be elevated in the PAG upon GqPCR activation and lead to analgesia via PAG disinhibition. Several endogenous analgesic neuropeptides may serve as endogenous analgesics during SIA via this GqPCR-PLC-DAGL-2-AG disinhibition mechanism in the PAG. As an effective endogenous pain relief mechanism during evolution, it offers a promising strategy for analgesic drug development.

Acknowledgments

This study was supported by the grants to LCC from the National Science Council, Taipei, Taiwan (NSC 101-2325-B002-048 and NSC 102-2321-B002-066) and National Health Research Institutes, Miaoli, Taiwan (NHRI-EX102-10251NI).


References

1
Y. Gaoni, R. Mechoulam
Isolation, structure and partial synthesis of an active constituent of hashish
J Am Chem Soc, 86 (1964), pp. 1646-1647
2
R. Mechoulam
Cannabinoids as therapeutic agents
Chapman and Hall/CRC, Florida (1986)
Article  
3
E.B. Russo, J.M. McPartland
Cannabis is more than simply delta(9)-tetrahydrocannabinol
Psychopharmacology (Berl), 165 (2003), pp. 431-432 author reply 433–434
4
R.G. Pertwee
Inverse agonism and neutral antagonism at cannabinoid CB1 receptors
Life Sci, 76 (2005), pp. 1307-1324
5
L.A. Matsuda, S.J. Lolait, M.J. Brownstein, A.C. Young, T.I. Bonner
Structure of a cannabinoid receptor and functional expression of the cloned cDNA
Nature, 346 (1990), pp. 561-564
6
S. Munro, K.L. Thomas, M. Abu-Shaar
Molecular characterization of a peripheral receptor for cannabinoids
Nature, 365 (1993), pp. 61-65
7
M.H. Rhee, M. Bayewitch, T. Avidor-Reiss, R. Levy, Z. Vogel
Cannabinoid receptor activation differentially regulates the various adenylyl cyclase isozymes
J Neurochem, 71 (1998), pp. 1525-1534
8
C.M. Gerard, C. Mollereau, G. Vassart, M. Parmentier
Molecular cloning of a human cannabinoid receptor which is also expressed in testis
Biochem J, 279 (Pt 1) (1991), pp. 129-134
9
A.C. Howlett, C.S. Breivogel, S.R. Childers, S.A. Deadwyler, R.E. Hampson, L.J. Porrino
Cannabinoid physiology and pharmacology: 30 years of progress
Neuropharmacology, 47 (Suppl. 1) (2004), pp. 345-358
10
M. Herkenham, A.B. Lynn, M.D. Little, M.R. Johnson, L.S. Melvin, B.R. de Costa, et al.
Cannabinoid receptor localization in brain
Proc Natl Acad Sci U S A, 87 (1990), pp. 1932-1936
11
I. Katona, B. Sperlagh, A. Sik, A. Käfalvi, E.S. Vizi, K. Mackie, et al.
Presynaptically located CB1 cannabinoid receptors regulate GABA release from axon terminals of specific hippocampal interneurons
J Neurosci, 19 (1999), pp. 4544-4558
12
K. Mackie, Y. Lai, R. Westenbroek, R. Mitchell
Cannabinoids activate an inwardly rectifying potassium conductance and inhibit Q-type calcium currents in AtT20 cells transfected with rat brain cannabinoid receptor
J Neurosci, 15 (1995), pp. 6552-6561
Article  
13
M. Shen, T.M. Piser, V.S. Seybold, S.A. Thayer
Cannabinoid receptor agonists inhibit glutamatergic synaptic transmission in rat hippocampal cultures
J Neurosci, 16 (1996), pp. 4322-4334
14
S. Galiegue, S. Mary, J. Marchand, D. Dussossoy, D. Carrière, P. Carayon, et al.
Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations
Eur J Biochem, 232 (1995), pp. 54-61
15
M.D. Van Sickle, M. Duncan, P.J. Kingsley, A. Mouihate, P. Urbani, K. Mackie, et al.
Identification and functional characterization of brainstem cannabinoid CB2 receptors
Science, 310 (2005), pp. 329-332
16
J.S. Walczak, V. Pichette, F. Leblond, K. Desbiens, P. Beaulieu
Characterization of chronic constriction of the saphenous nerve, a model of neuropathic pain in mice showing rapid molecular and electrophysiological changes
J Neurosci Res, 83 (2006), pp. 1310-1322
17
B.K. Atwood, K. Mackie
CB2: a cannabinoid receptor with an identity crisis
Br J Pharmacol, 160 (2010), pp. 467-479
18
W.A. Devane, L. Hanus, A. Breuer, R.G. Pertwee, L.A. Stevenson, G. Griffin, et al.
Isolation and structure of a brain constituent that binds to the cannabinoid receptor
Science, 258 (1992), pp. 1946-1949
19
R. Mechoulam, S. Ben-Shabat, L. Hanus, M. Ligumsky, N.E. Kaminski, A.R. Schatz, et al.
Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors
Biochem Pharmacol, 50 (1995), pp. 83-90
20
T. Sugiura, S. Kondo, A. Sukagawa, S. Nakane, A. Shinoda, K. Itoh, et al.
2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain
Biochem Biophys Res Commun, 215 (1995), pp. 89-97
21
L. Hanus, S. Abu-Lafi, E. Fride, A. Breuer, Z. Vogel, D.E. Shalev, et al.
2-Arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor
Proc Natl Acad Sci U S A, 98 (2001), pp. 3662-3665
22
A.C. Porter, J.M. Sauer, M.D. Knierman, G.W. Becker, M.J. Berna, J. Bao, et al.
Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor
J Pharmacol Exp Ther, 301 (2002), pp. 1020-1024
23
S.M. Huang, T. Bisogno, M. Trevisani, A. Al-Hayani, L. De Petrocellis, F. Fezza, et al.
An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors
Proc Natl Acad Sci U S A, 99 (2002), pp. 8400-8405
24
V. Di Marzo, A. Fontana, H. Cadas, S. Schinelli, G. Cimino, J.C. Schwartz, et al.
Formation and inactivation of endogenous cannabinoid anandamide in central neurons
Nature, 372 (1994), pp. 686-691
25
D. Piomelli, A. Giuffrida, A. Calignano, F. Rodriguez de Fonseca
The endocannabinoid system as a target for therapeutic drugs
Trends Pharmacol Sci, 21 (2000), pp. 218-224
26
V. Di Marzo
Endocannabinoids: synthesis and degradation
Rev Physiol Biochem Pharmacol, 160 (2008), pp. 1-24
27
P.C. Schmid, P.V. Reddy, V. Natarajan, H.H. Schmid
Metabolism of N-acylethanolamine phospholipids by a mammalian phosphodiesterase of the phospholipase D type
J Biol Chem, 258 (1983), pp. 9302-9306
28
Y. Okamoto, J. Wang, J. Morishita, N. Ueda
Biosynthetic pathways of the endocannabinoid anandamide
Chem Biodivers, 4 (2007), pp. 1842-1857
29
Y.X. Sun, K. Tsuboi, Y. Okamoto, T. Tonai, M. Murakami, I. Kudo, et al.
Biosynthesis of anandamide and N-palmitoylethanolamine by sequential actions of phospholipase A2 and lysophospholipase D
Biochem J, 380 (Pt 3) (2004), pp. 749-756
30
G.M. Simon, B.F. Cravatt
Endocannabinoid biosynthesis proceeding through glycerophospho-N-acyl ethanolamine and a role for alpha/beta hydrolase 4 in this pathway
J Biol Chem, 281 (2006), pp. 26465-26472
31
J. Liu, L. Wang, J. Harvey-White, B.X. Huang, H.Y. Kim, S. Luquet, et al.
Multiple pathways involved in the biosynthesis of anandamide
Neuropharmacology, 54 (2008), pp. 1-7
32
D. Leung, A. Saghatelian, G.M. Simon, B.F. Cravatt
Inactivation of N-acyl phosphatidylethanolamine phospholipase D reveals multiple mechanisms for the biosynthesis of endocannabinoids
Biochemistry, 45 (2006), pp. 4720-4726
33
G.M. Simon, B.F. Cravatt
Characterization of mice lacking candidate N-acyl ethanolamine biosynthetic enzymes provides evidence for multiple pathways that contribute to endocannabinoid production in vivo
Mol Biosyst, 6 (2010), pp. 1411-1418
34
B.F. Cravatt, K. Demarest, M.P. Patricelli, M.H. Bracey, D.K. Giang, B.R. Martin, et al.
Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase
Proc Natl Acad Sci U S A, 98 (2001), pp. 9371-9376
35
M. Egertova, B.F. Cravatt, M.R. Elphick
Comparative analysis of fatty acid amide hydrolase and cb(1) cannabinoid receptor expression in the mouse brain: evidence of a widespread role for fatty acid amide hydrolase in regulation of endocannabinoid signaling
Neuroscience, 119 (2003), pp. 481-496
36
M. Egertova, D.K. Giang, B.F. Cravatt, M.R. Elphick
A new perspective on cannabinoid signalling: complementary localization of fatty acid amide hydrolase and the CB1 receptor in rat brain
Proc R Soc Lond B Biol Sci, 265 (1998), pp. 2081-2085
37
K. Tsou, M.I. Nogueron, S. Muthian, M.C. Sañudo-Pena, C.J. Hillard, D.G. Deutsch, et al.
Fatty acid amide hydrolase is located preferentially in large neurons in the rat central nervous system as revealed by immunohistochemistry
Neurosci Lett, 254 (1998), pp. 137-140
38
S.T. Glaser, M. Kaczocha, D.G. Deutsch
Anandamide transport: a critical review
Life Sci, 77 (2005), pp. 1584-1604
39
C.J. Fowler
Transport of endocannabinoids across the plasma membrane and within the cell
Febs J, 280 (2013), pp. 1895-1904
40
M. Beltramo, N. Stella, A. Calignano, S.Y. Lin, A. Makriyannis, D. Piomelli
Functional role of high-affinity anandamide transport, as revealed by selective inhibition
Science, 277 (1997), pp. 1094-1097
41
V. Di Marzo, M. Bifulco, L. De Petrocellis
The endocannabinoid system and its therapeutic exploitation
Nat Rev Drug Discov, 3 (2004), pp. 771-784
42
A. Jarrahian, S. Manna, W.S. Edgemond, W.B. Campbell, C.J. Hillard
Structure-activity relationships among N-arachidonylethanolamine (Anandamide) head group analogues for the anandamide transporter
J Neurochem, 74 (2000), pp. 2597-2606
43
S. Vandevoorde, C.J. Fowler
Inhibition of fatty acid amide hydrolase and monoacylglycerol lipase by the anandamide uptake inhibitor VDM11: evidence that VDM11 acts as an FAAH substrate
Br J Pharmacol, 145 (2005), pp. 885-893
44
J.P. Alexander, B.F. Cravatt
The putative endocannabinoid transport blocker LY2183240 is a potent inhibitor of FAAH and several other brain serine hydrolases
J Am Chem Soc, 128 (2006), pp. 9699-9704
45
P.M. Zygmunt, J. Petersson, D.A. Andersson, H. Chuang, M. Sørgård, V. Di Marzo, et al.
Vanilloid receptors on sensory nerves mediate the vasodilator action of anandamide
Nature, 400 (1999), pp. 452-457
46
L. De Petrocellis, T. Bisogno, M. Maccarrone, J.B. Davis, A. Finazzi-Agro, V. Di Marzo
The activity of anandamide at vanilloid VR1 receptors requires facilitated transport across the cell membrane and is limited by intracellular metabolism
J Biol Chem, 276 (2001), pp. 12856-12863
47
Y. Hashimotodani, T. Ohno-Shosaku, H. Tsubokawa, H. Ogata, K. Emoto, T. Maejima, et al.
Phospholipase Cbeta serves as a coincidence detector through its Ca(2+) dependency for triggering retrograde endocannabinoid signal
Neuron, 45 (2005), pp. 257-268
48
Y. Hashimotodani, T. Ohno-Shosaku, M. Kano
Ca(2+)-assisted receptor-driven endocannabinoid release: mechanisms that associate presynaptic and postsynaptic activities
Curr Opin Neurobiol, 17 (2007), pp. 360-365
49
T. Bisogno, F. Howell, G. Williams, A. Minassi, M.G. Cascio, A. Ligresti, 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, 163 (2003), pp. 463-468
50
F. Matyas, G.M. Urban, M. Watanabe, K. Mackie, A. Zimmer, T.F. Freund, et al.
Identification of the sites of 2-arachidonoylglycerol synthesis and action imply retrograde endocannabinoid signaling at both GABAergic and glutamatergic synapses in the ventral tegmental area
Neuropharmacology, 54 (2008), pp. 95-107
51
Y. Gao, D.V. Vasilyev, M.B. Goncalves, F.V. Howell, C. Hobbs, M. Reisenberg, et al.
Loss of retrograde endocannabinoid signaling and reduced adult neurogenesis in diacylglycerol lipase knock-out mice
J Neurosci, 30 (2010), pp. 2017-2024
52
T.P. Dinh, T.F. Freund, D. Piomelli
A role for monoglyceride lipase in 2-arachidonoylglycerol inactivation
Chem Phys Lipids, 121 (2002), pp. 149-158
53
T.P. Dinh, S. Kathuria, D. Piomelli
RNA interference suggests a primary role for monoacylglycerol lipase in the degradation of the endocannabinoid 2-arachidonoylglycerol
Mol Pharmacol, 66 (2004), pp. 1260-1264
54
V. Di Marzo, T. Bisogno, T. Sugiura, D. Melck, L. De Petrocellis
The novel endogenous cannabinoid 2-arachidonoylglycerol is inactivated by neuronal- and basophil-like cells: connections with anandamide
Biochem J, 331 (Pt 1) (1998), pp. 15-19
55
S.K. Goparaju, N. Ueda, K. Taniguchi, S. Yamamoto
Enzymes of porcine brain hydrolyzing 2-arachidonoylglycerol, an endogenous ligand of cannabinoid receptors
Biochem Pharmacol, 57 (1999), pp. 417-423
56
J.L. Blankman, G.M. Simon, B.F. Cravatt
A comprehensive profile of brain enzymes that hydrolyze the endocannabinoid 2-arachidonoylglycerol
Chem Biol, 14 (2007), pp. 1347-1356
57
J.M. Walker, N.M. Strangman, S.M. Huang
Cannabinoids and pain
Pain Res Manag Summer, 6 (2001), pp. 74-79
58
A.S. Bloom, W.L. Dewey, L.S. Harris, K.K. Brosius
9-nor-9beta-hydroxyhexahydrocannabinol, a cannabinoid with potent antinociceptive activity: comparisons with morphine
J Pharmacol Exp Ther, 200 (1977), pp. 263-270
59
K. Tsou, K.A. Lowitz, A.G. Hohmann, W.J. Martin, C.B. Hathaway, D.A. Bereiter, et al.
Suppression of noxious stimulus-evoked expression of Fos protein-like immunoreactivity in rat spinal cord by a selective cannabinoid agonist
Neuroscience, 70 (1996), pp. 791-798
60
A.G. Hohmann, W.J. Martin, K. Tsou, J.M. Walker
Inhibition of noxious stimulus-evoked activity of spinal cord dorsal horn neurons by the cannabinoid WIN 55,212-2
Life Sci, 56 (1995), pp. 2111-2118
61
A.G. Hohmann, K. Tsou, J.M. Walker
Intrathecal cannabinoid administration suppresses noxious stimulus-evoked Fos protein-like immunoreactivity in rat spinal cord: comparison with morphine
Zhongguo Yao Li Xue Bao, 20 (1999), pp. 1132-1136
62
N. Agarwal, P. Pacher, I. Tegeder, F. Amaya, C.E. Constantin, G.J. Brenner, et al.
Cannabinoids mediate analgesia largely via peripheral type 1 cannabinoid receptors in nociceptors
Nat Neurosci, 10 (2007), pp. 870-879
63
A. Calignano, G. La Rana, A. Giuffrida, D. Piomelli
Control of pain initiation by endogenous cannabinoids
Nature, 394 (1998), pp. 277-281
64
J.D. Richardson, S. Kilo, K.M. Hargreaves
Cannabinoids reduce hyperalgesia and inflammation via interaction with peripheral CB1 receptors
Pain, 75 (1998), pp. 111-119
65
M. Herkenham, A.B. Lynn, M.R. Johnson, L.S. Melvin, B.R. de Costa, K.C. Rice
Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study
J Neurosci, 11 (1991), pp. 563-583
66
W.J. Martin, S.L. Patrick, P.O. Coffin, K. Tsou, J.M. Walker
An examination of the central sites of action of cannabinoid-induced antinociception in the rat
Life Sci, 56 (1995), pp. 2103-2109
67
W.J. Martin, A.G. Hohmann, J.M. Walker
Suppression of noxious stimulus-evoked activity in the ventral posterolateral nucleus of the thalamus by a cannabinoid agonist: correlation between electrophysiological and antinociceptive effects
J Neurosci, 16 (1996), pp. 6601-6611
68
A.G. Nackley, R.L. Suplita 2nd, A.G. Hohmann
A peripheral cannabinoid mechanism suppresses spinal fos protein expression and pain behavior in a rat model of inflammation
Neuroscience, 117 (2003), pp. 659-670
69
D.V. Reynolds
Surgery in the rat during electrical analgesia induced by focal brain stimulation
Science, 164 (1969), pp. 444-445
70
M.J. Millan
Descending control of pain
Prog Neurobiol, 66 (2002), pp. 355-474
71
A.H. Lichtman, S.A. Cook, B.R. Martin
Investigation of brain sites mediating cannabinoid-induced antinociception in rats: evidence supporting periaqueductal gray involvement
J Pharmacol Exp Ther, 276 (1996), pp. 585-593
72
K. Tsou, S. Brown, M.C. Sanudo-Pena, K. Mackie, J.M. Walker
Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system
Neuroscience, 83 (1998), pp. 393-411
73
C.W. Vaughan, M. Connor, E.E. Bagley, M.J. Christie
Actions of cannabinoids on membrane properties and synaptic transmission in rat periaqueductal gray neurons in vitro
Mol Pharmacol, 57 (2000), pp. 288-295
74
T.P. Malan Jr., M.M. Ibrahim, H. Deng, Q. Liu, H.P. Mata, T. Vanderah, et al.
CB2 cannabinoid receptor-mediated peripheral antinociception
Pain, 93 (2001), pp. 239-245
75
A.G. Nackley, A. Makriyannis, A.G. Hohmann
Selective activation of cannabinoid CB(2) receptors suppresses spinal fos protein expression and pain behavior in a rat model of inflammation
Neuroscience, 119 (2003), pp. 747-757
76
G.T. Whiteside, S.L. Gottshall, J.M. Boulet, S.M. Chaffer, J.E. Harrison, M.S. Pearson, et al.
A role for cannabinoid receptors, but not endogenous opioids, in the antinociceptive activity of the CB2-selective agonist, GW405833
Eur J Pharmacol, 528 (2005), pp. 65-72
77
M.M. Ibrahim, M.L. Rude, N.J. Stagg, H.P. Mata, J. Lai, T.W. Vanderah, et al.
CB(2) cannabinoid receptor mediation of antinociception
Pain, 122 (2006), pp. 36-42
78
H. Iwamura, H. Suzuki, Y. Ueda, T. Kaya, T. Inaba
In vitro and in vivo pharmacological characterization of JTE-907, a novel selective ligand for cannabinoid CB2 receptor
J Pharmacol Exp Ther, 296 (2001), pp. 420-425
79
J. Zhang, C. Hoffert, H.K. Vu, T. Groblewski, S. Ahmad, D. O'Donnell
Induction of CB2 receptor expression in the rat spinal cord of neuropathic but not inflammatory chronic pain models
Eur J Neurosci, 17 (2003), pp. 2750-2754
80
G. Wotherspoon, A. Fox, P. McIntyre, S. Colley, S. Bevan, J. Winter
Peripheral nerve injury induces cannabinoid receptor 2 protein expression in rat sensory neurons
Neuroscience, 135 (2005), pp. 235-245
81
A. Pereira, A. Chappell, J. Dethy, H. Hoeck, L. Arendt-Nielsen, S. Verfaille, et al.
A proof-of-concept (POC) study including experimental pain models (EPMs) to assess the effects of a CB2 agonist (LY2828360) in the treatment of patients with osteoarthritic (OA) knee pain
Clin Pharmacol Thera, 93 (S1) (2013), pp. S56-S57 No. PII-11
82
M. Rinaldi-Carmona, F. Barth, M. Heaulme, D. Shire, B. Calandra, C. Congy, et al.
SR141716A, a potent and selective antagonist of the brain cannabinoid receptor
FEBS Lett, 350 (1994), pp. 240-244
83
R.S. Landsman, T.H. Burkey, P. Consroe, W.R. Roeske, H.I. Yamamura
SR141716A is an inverse agonist at the human cannabinoid CB1 receptor
Eur J Pharmacol, 334 (1997), pp. R1-R2
84
K.S. Sink, P.J. McLaughlin, J.A. Wood, C. Brown, P. Fan, V.K. Vemuri, et al.
The novel cannabinoid CB1 receptor neutral antagonist AM4113 suppresses food intake and food-reinforced behavior but does not induce signs of nausea in rats
Neuropsychopharmacology, 33 (2008), pp. 946-955
85
J.D. Richardson, L. Aanonsen, K.M. Hargreaves
SR 141716A, a cannabinoid receptor antagonist, produces hyperalgesia in untreated mice
Eur J Pharmacol, 319 (1997), pp. R3-R4
86
A. Dogrul, L.R. Gardell, S. Ma, M.H. Ossipov, F. Porreca, J. Lai
‘Knock-down’ of spinal CB1 receptors produces abnormal pain and elevates spinal dynorphin content in mice
Pain, 100 (2002), pp. 203-209
87
A. Zimmer, A.M. Zimmer, A.G. Hohmann, M. Herkenham, T.I. Bonner
Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1 receptor knockout mice
Proc Natl Acad Sci U S A, 96 (1999), pp. 5780-5785
88
C. Ledent, O. Valverde, G. Cossu, F. Petitet, J.F. Aubert, F. Beslot, et al.
Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice
Science, 283 (1999), pp. 401-404
89
M.M. Ibrahim, H. Deng, A. Zvonok, D.A. Cockayne, J. Kwan, H.P. Mata, et al.
Activation of CB2 cannabinoid receptors by AM1241 inhibits experimental neuropathic pain: pain inhibition by receptors not present in the CNS
Proc Natl Acad Sci U S A, 100 (2003), pp. 10529-10533
90
P. Beaulieu, T. Bisogno, S. Punwar, W.P. Farquhar-Smith, G. Ambrosino, V. Di Marzo, et al.
Role of the endogenous cannabinoid system in the formalin test of persistent pain in the rat
Eur J Pharmacol, 396 (2000), pp. 85-92
91
A.H. Lichtman, C.C. Shelton, T. Advani, B.F. Cravatt
Mice lacking fatty acid amide hydrolase exhibit a cannabinoid receptor-mediated phenotypic hypoalgesia
Pain, 109 (2004), pp. 319-327
92
N. Clayton, F.H. Marshall, C. Bountra, C.T. O'Shaughnessy
CB1 and CB2 cannabinoid receptors are implicated in inflammatory pain
Pain, 96 (2002), pp. 253-260
93
I. Racz, X. Nadal, J. Alferink, J.E. Baños, J. Rehnelt, M. Martín, et al.
Interferon-gamma is a critical modulator of CB(2) cannabinoid receptor signaling during neuropathic pain
J Neurosci, 28 (2008), pp. 12136-12145
94
I. Racz, X. Nadal, J. Alferink, J.E. Baños, J. Rehnelt, M. Martín, et al.
Crucial role of CB(2) cannabinoid receptor in the regulation of central immune responses during neuropathic pain
J Neurosci, 28 (2008), pp. 12125-12135
95
S. Sit, K. Xie
Inventors. Bisarylimidazolyl fatty acid amide hydrolase inhibitors
Bristol-Myers Squibb Company, US20020188009-A1, Princeton, NJ (2002)
96
S. Sit, K. Xie
Inventors. (Oxime) carbamoyl fatty acid amide hydrolase inhibitors
Bristol-Myers Squibb Company, US2003003222, Princeton, NJ (2003)
97
A.H. Lichtman, D. Leung, C.C. Shelton, A. Saghatelian, C. Hardouin, D.L. Boger, et al.
Reversible inhibitors of fatty acid amide hydrolase that promote analgesia: evidence for an unprecedented combination of potency and selectivity
J Pharmacol Exp Ther, 311 (2004), pp. 441-448
98
S.G. Kinsey, J.Z. Long, S.T. O'Neal, R.A. Abdullah, J.L. Poklis, D.L. Boger, et al.
Blockade of endocannabinoid-degrading enzymes attenuates neuropathic pain
J Pharmacol Exp Ther, 330 (2009), pp. 902-910
99
K. Ahn, D.S. Johnson, M. Mileni, D. Beidler, J.Z. Long, M.K. McKinney, et al.
Discovery and characterization of a highly selective FAAH inhibitor that reduces inflammatory pain
Chem Biol, 16 (2009), pp. 411-420
100
L. Booker, S.G. Kinsey, R.A. Abdullah, J.L. Blankman, J.Z. Long, C. Ezzili, et al.
The fatty acid amide hydrolase (FAAH) inhibitor PF-3845 acts in the nervous system to reverse LPS-induced tactile allodynia in mice
Br J Pharmacol, 165 (2012), pp. 2485-2496
101
J.E. Schlosburg, J.L. Blankman, J.Z. Long, D.K. Nomura, B. Pan, S.G. Kinsey, et al.
Chronic monoacylglycerol lipase blockade causes functional antagonism of the endocannabinoid system
Nat Neurosci, 13 (2010), pp. 1113-1119
102
O. Sasso, R. Bertorelli, T. Bandiera, R. Scarpelli, G. Colombano, A. Armirotti, et al.
Peripheral FAAH inhibition causes profound antinociception and protects against indomethacin-induced gastric lesions
Pharmacol Res, 65 (2012), pp. 553-563
103
J. Guindon, Y. Lai, S.M. Takacs, H.B. Bradshaw, A.G. Hohmann
Alterations in endocannabinoid tone following chemotherapy-induced peripheral neuropathy: effects of endocannabinoid deactivation inhibitors targeting fatty-acid amide hydrolase and monoacylglycerol lipase in comparison to reference analgesics following cisplatin treatment
Pharmacol Res, 67 (2013), pp. 94-109
104
K. Ahn, S.E. Smith, M.B. Liimatta, D. Beidler, N. Sadagopan, D.T. Dudley, et al.
Mechanistic and pharmacological characterization of PF-04457845: a highly potent and selective fatty acid amide hydrolase inhibitor that reduces inflammatory and noninflammatory pain
J Pharmacol Exp Ther, 338 (2011), pp. 114-124
105
D.S. Johnson, C. Stiff, S.E. Lazerwith, S.R. Kesten, L.K. Fay, M. Morris, et al.
Discovery of pf-04457845: a highly potent, orally bioavailable, and selective urea FAAH inhibitor
ACS Med Chem Lett, 2 (2011), pp. 91-96
106
J.P. Huggins, T.S. Smart, S. Langman, L. Taylor, T. Young
An efficient randomised, placebo-controlled clinical trial with the irreversible fatty acid amide hydrolase-1 inhibitor PF-04457845, which modulates endocannabinoids but fails to induce effective analgesia in patients with pain due to osteoarthritis of the knee
Pain, 153 (2012), pp. 1837-1846
107
V. Di Marzo
Inhibitors of endocannabinoid breakdown for pain: not so FA(AH)cile, after all
Pain, 153 (2012), pp. 1785-1786
108
C.W. Scott, G. Tian, X.H. Yu, K.A. Paschetto, D.E. Wilkins, L. Meury, et al.
Biochemical characterization and in vitro activity of AZ513, a noncovalent, reversible, and noncompetitive inhibitor of fatty acid amide hydrolase
Eur J Pharmacol, 667 (2011), pp. 74-79
109
J. Desroches, J. Guindon, C. Lambert, P. Beaulieu
Modulation of the anti-nociceptive effects of 2-arachidonoyl glycerol by peripherally administered FAAH and MGL inhibitors in a neuropathic pain model
Br J Pharmacol, 155 (2008), pp. 913-924
110
J.Z. Long, W. Li, L. Booker, J.J. Burston, S.G. Kinsey, J.E. Schlosburg, et al.
Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects
Nat Chem Biol, 5 (2009), pp. 37-44
111
P.K. Chanda, Y. Gao, L. Mark, J. Btesh, B.W. Strassle, P. Lu, et al.
Monoacylglycerol lipase activity is a critical modulator of the tone and integrity of the endocannabinoid system
Mol Pharmacol, 78 (2010), pp. 996-1003
112
A.H. Lichtman, E.G. Hawkins, G. Griffin, B.F. Cravatt
Pharmacological activity of fatty acid amides is regulated, but not mediated, by fatty acid amide hydrolase in vivo
J Pharmacol Exp Ther, 302 (2002), pp. 73-79
113
C.J. Hillard
Biochemistry and pharmacology of the endocannabinoids arachidonylethanolamide and 2-arachidonylglycerol
Prostaglandins Other Lipid Mediat, 61 (2000), pp. 3-18
114
T. Sugiura, S. Kondo, S. Kishimoto, T. Miyashita, S. Nakane, T. Kodaka, et al.
Evidence that 2-arachidonoylglycerol but not N-palmitoylethanolamine or anandamide is the physiological ligand for the cannabinoid CB2 receptor. Comparison of the agonistic activities of various cannabinoid receptor ligands in HL-60 cells
J Biol Chem, 275 (2000), pp. 605-612
115
K. Mackie, W.A. Devane, B. Hille
Anandamide, an endogenous cannabinoid, inhibits calcium currents as a partial agonist in N18 neuroblastoma cells
Mol Pharmacol., 44 (1993), pp. 498-503
116
A. Busquets-Garcia, E. Puighermanal, A. Pastor, R. de la Torre, R. Maldonado, A. Ozaita
Differential role of anandamide and 2-arachidonoylglycerol in memory and anxiety-like responses
Biol Psychiatry, 70 (2011), pp. 479-486
117
S. Ghosh, L.E. Wise, Y. Chen, R. Gujjar, A. Mahadevan, B.F. Cravatt, et al.
The monoacylglycerol lipase inhibitor JZL184 suppresses inflammatory pain in the mouse carrageenan model
Life Sci, 92 (2013), pp. 498-505
118
S.G. Kinsey, L.E. Wise, D. Ramesh, R. Abdullah, D.E. Selley, B.F. Cravatt, et al.
Repeated low-dose administration of the monoacylglycerol lipase inhibitor JZL184 retains cannabinoid receptor type 1-mediated antinociceptive and gastroprotective effects
J Pharmacol Exp Ther, 345 (2013), pp. 492-501
119
M. Kano, T. Ohno-Shosaku, Y. Hashimotodani, M. Uchigashima, M. Watanabe
Endocannabinoid-mediated control of synaptic transmission
Physiol Rev, 89 (2009), pp. 309-380
120
L.C. Gregg, K.M. Jung, J.M. Spradley, R. Nyilas, R.L. Suplita 2nd, A. Zimmer, et al.
Activation of type 5 metabotropic glutamate receptors and diacylglycerol lipase-alpha initiates 2-arachidonoylglycerol formation and endocannabinoid-mediated analgesia
J Neurosci, 32 (2012), pp. 9457-9468
121
H.T. Liao, H.J. Lee, Y.C. Ho, L.C. Chiou
Capsaicin in the periaqueductal gray induces analgesia via metabotropic glutamate receptor-mediated endocannabinoid retrograde disinhibition
Br J Pharmacol, 163 (2011), pp. 330-345
122
G.M. Drew, B.K. Lau, C.W. Vaughan
Substance P drives endocannabinoid-mediated disinhibition in a midbrain descending analgesic pathway
J Neurosci, 29 (2009), pp. 7220-7229
123
G.M. Drew, V.A. Mitchell, C.W. Vaughan
Glutamate spillover modulates GABAergic synaptic transmission in the rat midbrain periaqueductal grey via metabotropic glutamate receptors and endocannabinoid signaling
J Neurosci, 28 (2008), pp. 808-815
124
V.A. Mitchell, H.J. Jeong, G.M. Drew, C.W. Vaughan
Cholecystokinin exerts an effect via the endocannabinoid system to inhibit GABAergic transmission in midbrain periaqueductal gray
Neuropsychopharmacology, 36 (2011), pp. 1801-1810
125
V.A. Mitchell, H. Kawahara, C.W. Vaughan
Neurotensin inhibition of GABAergic transmission via mGluR-induced endocannabinoid signalling in rat periaqueductal grey
J Physiol, 587 (Pt 11) (2009), pp. 2511-2520
126
B.K. Lau, C.W. Vaughan
Muscarinic modulation of synaptic transmission via endocannabinoid signalling in the rat midbrain periaqueductal gray
Mol Pharmacol, 74 (2008), pp. 1392-1398
127
Y.C. Ho, H.J. Lee, L.W. Tung, Y.Y. Liao, S.Y. Fu, S.F. Teng, et al.
Activation of orexin 1 receptors in the periaqueductal gray of male rats leads to antinociception via retrograde endocannabinoid (2-arachidonoylglycerol)-induced disinhibition
J Neurosci, 31 (2011), pp. 14600-14610
128
G. Riedel, W. Wetzel, K.G. Reymann
Comparing the role of metabotropic glutamate receptors in long-term potentiation and in learning and memory
Prog Neuropsychopharmacol Biol Psychiatry, 20 (1996), pp. 761-789
129
P.J. Conn, J.P. Pin
Pharmacology and functions of metabotropic glutamate receptors
Annu Rev Pharmacol Toxicol, 37 (1997), pp. 205-237
130
J.P. Pin, R. Duvoisin
The metabotropic glutamate receptors: structure and functions
Neuropharmacology, 34 (1995), pp. 1-26
131
G.M. Drew, C.W. Vaughan
Multiple metabotropic glutamate receptor subtypes modulate GABAergic neurotransmission in rat periaqueductal grey neurons in vitro
Neuropharmacology, 46 (2004), pp. 927-934
132
Z. Nusser, E. Mulvihill, P. Streit, P. Somogyi
Subsynaptic segregation of metabotropic and ionotropic glutamate receptors as revealed by immunogold localization
Neuroscience, 61 (1994), pp. 421-427
133
A. Rosen, Y.X. Zhang, I. Lund, T. Lundeberg, L.C. Yu
Substance P microinjected into the periaqueductal gray matter induces antinociception and is released following morphine administration
Brain Res, 1001 (2004), pp. 87-94
134
P.W. Kalivas, L. Jennes, C.B. Nemeroff, A.J. Prange Jr.
Neurotensin: topographical distribution of brain sites involved in hypothermia and antinociception
J Comp Neurol, 210 (1982), pp. 225-238
135
X.H. Chen, E.B. Geller, M.W. Adler
CCK(B) receptors in the periaqueductal grey are involved in electroacupuncture antinociception in the rat cold water tail-flick test
Neuropharmacology, 37 (1998), pp. 751-757
136
E. Palazzo, V. de Novellis, I. Marabese, D. Cuomo, F. Rossi, L. Berrino, et al.
Interaction between vanilloid and glutamate receptors in the central modulation of nociception
Eur J Pharmacol, 439 (2002), pp. 69-75
137
K. Starowicz, S. Maione, L. Cristino, E. Palazzo, I. Marabese, F. Rossi, et al.
Tonic endovanilloid facilitation of glutamate release in brainstem descending antinociceptive pathways
J Neurosci, 27 (2007), pp. 13739-13749
138
N.J. Woolf, J.B. Harrison, J.S. Buchwald
Cholinergic neurons of the feline pontomesencephalon. II. Ascending anatomical projections
Brain Res, 520 (1990), pp. 55-72
139
C.R. Monassi, A. Hoffmann, L. Menescal-de-Oliveira
Involvement of the cholinergic system and periaqueductal gray matter in the modulation of tonic immobility in the guinea pig
Physiol Behav, 62 (1997), pp. 53-59
140
A.P. Guimaraes, F.S. Guimaraes, W.A. Prado
Modulation of carbachol-induced antinociception from the rat periaqueductal gray
Brain Res Bull, 51 (2000), pp. 471-478
141
T. Sakurai, A. Amemiya, M. Ishii, I. Matsuzaki, R.M. Chemelli, H. Tanaka, et al.
Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior
Cell, 92 (1998), pp. 573-585
142
L. de Lecea, T.S. Kilduff, C. Peyron, X. Gao, P.E. Foye, P.E. Danielson, et al.
The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity
Proc Natl Acad Sci U S A, 95 (1998), pp. 322-327
143
C. Peyron, D.K. Tighe, A.N. van den Pol, L. de Lecea, H.C. Heller, J.G. Sutcliffe, et al.
Neurons containing hypocretin (orexin) project to multiple neuronal systems
J Neurosci, 18 (1998), pp. 9996-10015
144
T.L. Horvath, C. Peyron, S. Diano, A. Ivanov, G. Aston-Jones, T.S. Kilduff, et al.
Hypocretin (orexin) activation and synaptic innervation of the locus coeruleus noradrenergic system
J Comp Neurol, 415 (1999), pp. 145-159
145
A.N. van den Pol
Hypothalamic hypocretin (orexin): robust innervation of the spinal cord
J Neurosci, 19 (1999), pp. 3171-3182
146
N. Tsujino, T. Sakurai
Orexin/hypocretin: a neuropeptide at the interface of sleep, energy homeostasis, and reward system
Pharmacol Rev, 61 (2009), pp. 162-176
147
L.C. Chiou, H.J. Lee, Y.C. Ho, S.P. Chen, Y.Y. Liao, C.H. Ma, et al.
Orexins/hypocretins: pain regulation and cellular actions
Curr Pharm Des, 16 (2010), pp. 3089-3100
148
S. Maione, T. Bisogno, V. de Novellis, E. Palazzo, L. Cristino, M. Valenti, et al.
Elevation of endocannabinoid levels in the ventrolateral periaqueductal grey through inhibition of fatty acid amide hydrolase affects descending nociceptive pathways via both cannabinoid receptor type 1 and transient receptor potential vanilloid type-1 receptors
J Pharmacol Exp Ther, 316 (2006), pp. 969-982
149
H. Kawahara, G.M. Drew, M.J. Christie, C.W. Vaughan
Inhibition of fatty acid amide hydrolase unmasks CB1 receptor and TRPV1 channel-mediated modulation of glutamatergic synaptic transmission in midbrain periaqueductal grey
Br J Pharmacol, 163 (2011), pp. 1214-1222
150
A.G. Hohmann, R.L. Suplita, N.M. Bolton, M.H. Neely, D. Fegley, R. Mangieri, et al.
An endocannabinoid mechanism for stress-induced analgesia
Nature, 435 (2005), pp. 1108-1112
151
H. Flor, N. Birbaumer, R. Schulz, S.M. Grusser, R.F. Mucha
Pavlovian conditioning of opioid and nonopioid pain inhibitory mechanisms in humans
Eur J Pain, 6 (2002), pp. 395-402
152
O. Valverde, C. Ledent, F. Beslot, M. Parmentier, B.P. Roques
Reduction of stress-induced analgesia but not of exogenous opioid effects in mice lacking CB1 receptors
Eur J Neurosci, 12 (2000), pp. 533-539
153
W.M. Olango, M. Roche, G.K. Ford, B. Harhen, D.P. Finn
The endocannabinoid system in the rat dorsolateral periaqueductal grey mediates fear-conditioned analgesia and controls fear expression in the presence of nociceptive tone
Br J Pharmacol, 165 (2012), pp. 2549-2560
154
R.K. Butler, G.K. Ford, M. Hogan, M. Roche, K.M. Doyle, J.P. Kelly, et al.
Fear-induced suppression of nociceptive behaviour and activation of Akt signalling in the rat periaqueductal grey: role of fatty acid amide hydrolase
J Psychopharmacol, 26 (2012), pp. 83-91
155
S. Watanabe, T. Kuwaki, M. Yanagisawa, Y. Fukuda, M. Shimoyama
Persistent pain and stress activate pain-inhibitory orexin pathways
Neuroreport, 16 (2005), pp. 5-8
156
X. Xie, J.P. Wisor, J. Hara, T.L. Crowder, R. LeWinter, T.V. Khroyan, et al.
Hypocretin/orexin and nociceptin/orphanin FQ coordinately regulate analgesia in a mouse model of stress-induced analgesia
J Clin Invest, 118 (2008), pp. 2471-2481
157
L.C. Chiou, S.F. Teng, L.Y. Chang
Restraint stress induces analgesia through endogenous orexins via OX1 receptor-mediated endocannabinoid retrograde disinhibition in the ventrolateral periaqueductal gray
Annual meeting of neuroscience, San Diego, CA (November, 2010) Prog. No. 844.1
158
J.E. Holden, J.A. Pizzi, Y. Jeong
An NK1 receptor antagonist microinjected into the periaqueductal gray blocks lateral hypothalamic-induced antinociception in rats
Neurosci Lett, 453 (2009), pp. 115-119
159
X. Gui, R.E. Carraway, P.R. Dobner
Endogenous neurotensin facilitates visceral nociception and is required for stress-induced antinociception in mice and rats
Neuroscience, 126 (2004), pp. 1023-1032
160
A. Ottani, S. Leone, M. Sandrini, A. Ferrari, A. Bertolini
The analgesic activity of paracetamol is prevented by the blockade of cannabinoid CB1 receptors
Eur J Pharmacol, 531 (2006), pp. 280-281
161
C. Mallet, L. Daulhac, J. Bonnefont, C. Ledent, M. Etienne, E. Chapuy, et al.
Endocannabinoid and serotonergic systems are needed for acetaminophen-induced analgesia
Pain, 139 (2008), pp. 190-200
162
E.D. Hogestatt, B.A. Jonsson, A. Ermund, D.A. Andersson, H. Björk, J.P. Alexander, et al.
Conversion of acetaminophen to the bioactive N-acylphenolamine AM404 via fatty acid amide hydrolase-dependent arachidonic acid conjugation in the nervous system
J Biol Chem, 280 (2005), pp. 31405-31412
163
P.M. Zygmunt, H. Chuang, P. Movahed, D. Julius, E.D. Hogestatt
The anandamide transport inhibitor AM404 activates vanilloid receptors
Eur J Pharmacol, 396 (2000), pp. 39-42
164
C. Mallet, D.A. Barriere, A. Ermund, B.A.G. Jönsson, A. Eschalier, P.M. Zygmunt, et al.
TRPV1 in brain is involved in acetaminophen-induced antinociception
PLoS One, 5 (2010), p. e12748
CrossRef  
165
L.C. Chiou, H.J. Lee, L.W. Tung
Acetaminophen induces analgesia via the mGluR5-mediated endocannabinoid retrograde disinhibition mechanism initiated by TRPV1 activation in the periaqueductal gray 639.29/C30
42nd Annual Meeting of Neuroscience (2012) New Orleans
166
B. Costa, M. Colleoni
SR141716A induces in rats a behavioral pattern opposite to that of CB1 receptor agonists
Zhongguo Yao Li Xue Bao., 20 (1999), pp. 1103-1108
167
V. Chapman
The cannabinoid CB1 receptor antagonist, SR141716A, selectively facilitates nociceptive responses of dorsal horn neurones in the rat
Br J Pharmacol, 127 (1999), pp. 1765-1767
168
N.M. Strangman, S.L. Patrick, A.G. Hohmann, K. Tsou, J.M. Walker
Evidence for a role of endogenous cannabinoids in the modulation of acute and tonic pain sensitivity
Brain Res, 813 (1998), pp. 323-328
169
J.D. Richardson, L. Aanonsen, K.M. Hargreaves
Hypoactivity of the spinal cannabinoid system results in NMDA-dependent hyperalgesia
J Neurosci, 18 (1998), pp. 451-457
170
S. Kathuria, S. Gaetani, D. Fegley, F. Valiño, A. Duranti, A. Tontini, et al.
Modulation of anxiety through blockade of anandamide hydrolysis
Nat Med, 9 (2003), pp. 76-81
171
V.L. Haller, D.L. Stevens, S.P. Welch
Modulation of opioids via protection of anandamide degradation by fatty acid amide hydrolase
Eur J Pharmacol, 600 (2008), pp. 50-58
172
P. Hasanein, S. Shahidi, A. Komaki, N. Mirazi
Effects of URB597 as an inhibitor of fatty acid amide hydrolase on modulation of nociception in a rat model of cholestasis
Eur J Pharmacol, 591 (2008), pp. 132-135
173
R. Russo, J. Loverme, G. La Rana, T.R. Compton, J. Parrott, A. Duranti, et al.
The fatty acid amide hydrolase inhibitor URB597 (cyclohexylcarbamic acid 3'-carbamoylbiphenyl-3-yl ester) reduces neuropathic pain after oral administration in mice
J Pharmacol Exp Ther, 322 (2007), pp. 236-242
174
S.G. Kinsey, J.Z. Long, B.F. Cravatt, A.H. Lichtman
Fatty acid amide hydrolase and monoacylglycerol lipase inhibitors produce anti-allodynic effects in mice through distinct cannabinoid receptor mechanisms
J Pain, 11 (2010), pp. 1420-1428
175
J.Z. Long, D.K. Nomura, R.E. Vann, D.M. Walentiny, L. Booker, X. Jin, et al.
Dual blockade of FAAH and MAGL identifies behavioral processes regulated by endocannabinoid crosstalk in vivo
Proc Natl Acad Sci U S A, 106 (2009), pp. 20270-20275
176
S.G. Woodhams, A. Wong, D.A. Barrett, A.J. Bennett, V. Chapman, S.P. Alexander
Spinal administration of the monoacylglycerol lipase inhibitor JZL184 produces robust inhibitory effects on nociceptive processing and the development of central sensitization in the rat
Br J Pharmacol, 167 (2012), pp. 1609-1619
177
I.A. Khasabova, A. Chandiramani, C. Harding-Rose, D.A. Simone, V.S. Seybold
Increasing 2-arachidonoyl glycerol signaling in the periphery attenuates mechanical hyperalgesia in a model of bone cancer pain
Pharmacol Res, 64 (2011), pp. 60-67
178
J. Guindon, A. Guijarro, D. Piomelli, A.G. Hohmann
Peripheral antinociceptive effects of inhibitors of monoacylglycerol lipase in a rat model of inflammatory pain
Br J Pharmacol, 163 (2011), pp. 1464-1478

References

Close