You are on page 1of 16

Neuropharmacology 124 (2017) 105e120

Contents lists available at ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

Invited review

The cannabinoid system and pain


Stephen G. Woodhams a, *, Victoria Chapman a, David P. Finn b, Andrea G. Hohmann c, d, e, f,
Volker Neugebauer g
a
Arthritis UK Pain Centre, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
b
Pharmacology & Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland
Galway, University Road, Galway, Ireland
c
Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
d
Program in Neuroscience, Indiana University, Bloomington, IN, USA
e
Interdisciplinary Biochemistry Graduate Program, Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
f
Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
g
Department of Pharmacology and Neuroscience, Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health
Sciences Center, Lubbock, TX 79430, USA

a r t i c l e i n f o a b s t r a c t

Article history: Chronic pain states are highly prevalent and yet poorly controlled by currently available analgesics,
Received 6 April 2017 representing an enormous clinical, societal, and economic burden. Existing pain medications have sig-
Received in revised form nificant limitations and adverse effects including tolerance, dependence, gastrointestinal dysfunction,
31 May 2017
cognitive impairment, and a narrow therapeutic window, making the search for novel analgesics ever
Accepted 14 June 2017
more important. In this article, we review the role of an important endogenous pain control system, the
Available online 15 June 2017
endocannabinoid (EC) system, in the sensory, emotional, and cognitive aspects of pain. Herein, we briefly
cover the discovery of the EC system and its role in pain processing pathways, before concentrating on
Chemical compounds studied in this article:
AA-5-HT (PubChem CID: 10027372)
three areas of current major interest in EC pain research; 1. Pharmacological enhancement of endo-
BIA-102474 (PubChem CID: 46831476) cannabinoid activity (via blockade of EC metabolism or allosteric modulation of CB1 receptors); 2.
JZL184 (PubChem CID: 25021165) The EC System and stress-induced modulation of pain; and 3. The EC system & medial prefrontal
JZL195 (PubChem CID: 46232606) cortex (mPFC) dysfunction in pain states. Whilst we focus predominantly on the preclinical data, we
MJN110 (PubChem CID: 71722059) also include extensive discussion of recent clinical failures of endocannabinoid-related therapies, the
PF-04457845 (PubChem CID: 24771824) future potential of these approaches, and important directions for future research on the EC system and
URB597 (PubChem CID: 1383884) pain.
URB937 (PubChem CID: 53394762)
This article is part of the Special Issue entitled “A New Dawn in Cannabinoid Neurobiology”.
Keywords: © 2017 Elsevier Ltd. All rights reserved.
Cannabinoid
Endocannabinoid
Pain
FAAH
MAGL
Stress
Stress-induced analgesia
Stress-induced hyperalgesia
mGluR5
mPFC
Amygdala
Cortical control of pain

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
1.1. Pain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106

* Corresponding author.
E-mail address: stephen.woodhams@nottingham.ac.uk (S.G. Woodhams).

http://dx.doi.org/10.1016/j.neuropharm.2017.06.015
0028-3908/© 2017 Elsevier Ltd. All rights reserved.
106 S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120

1.2. Pain & the EC system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106


2. Blockade of EC metabolism as an analgesic approach: past issues and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
2.1. FAAH inhibition for analgesia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
2.2. MAGL inhibition for analgesia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
2.3. The future of EC metabolic enzyme inhibitors: broadening the appeal . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
3. Positive allosteric modulators of cannabinoid CB1 receptor signalling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
4. The EC system and stress-induced modulation of pain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
4.1. Stress-induced analgesia (SIA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
4.2. Stress-induced hyperalgesia (SIH) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
5. The EC system & medial prefrontal cortex (mPFC) dysfunction in pain states: CB1 and mGluR5 in a model of arthritic pain . . . . . . . . . . . . . . . . . . . . . 113
5.1. mPFC deactivation in pain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
5.2. Synaptic mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
5.3. Role of CB1 and mGluR5 interaction in control of cortical feedforward inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114
5.4. Role of CB1 and mGluR5 interaction in control of amygdala function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114
5.5. Role of CB1 and mGluR5 interaction in cortical control of pain behaviour . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114
6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
7. Future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115

1. Introduction the existence of a second ubiquitous endogenous pain control


pathway; the endocannabinoid (EC) system. In the past two de-
1.1. Pain cades, numerous tools to perturb the EC system have been devel-
oped, and a wealth of research has demonstrated the potential
Pain is complex phenomena comprising an unpleasant sensory efficacy of this approach for pain relief (reviewed in Guindon and
and emotional experience associated with actual or potential tissue Hohmann, 2009; Sagar et al., 2009, 2012; Woodhams et al., 2015).
damage (Loeser and Treede, 2008), serving a vital protective However, global targeting of the EC system is also associated with
evolutionary function. Whilst acute pain can be considered adap- undesirable results, including deleterious effects on memory (Hall
tive, chronic pain in the absence of injury, following its resolution, and Solowij, 1998), cognition (Pattij et al., 2008), and mood
or following damage to the nervous system (neuropathic pain) is a (Rubino et al., 2015), and the development of tolerance and
pathological condition of enormous clinical, societal, and economic dependence (Lichtman and Martin, 2005; Tappe-Theodor et al.,
significance (Apkarian et al., 2009). Chronic pain is the most 2007). This subject has been the topic of many excellent review
commonly presented clinical complaint in the USA, afflicting ~10% articles in the past, and therefore in this Special Issue of Neuro-
of the adult population (Nahin, 2015), and representing the greatest pharmacology, we briefly cover the history of EC research before
economic burden of any pathological condition with an estimated focussing on three areas of current interest in the field of canna-
annual cost of $565e635 billion in this region alone (Gaskin and binoid pain research e pharmacological enhancement of EC system
Richard, 2012). Despite its prevalence, current treatments for pain activity via enzyme inhibitors (Section 2) or by allosteric modula-
provide inadequate duration and/or extent of relief (Vardeh et al., tion of CB1 (Section 3), the role of the EC system in stress-induced
2016), highlighting the urgent need for effective novel analgesic modulation of pain (Section 4), and actions on the cognitive aspects
agents. Opioids, nonsteroidal anti-inflammatory drugs, selective of pain via coupling of endocannabinoids and metabotropic gluta-
COX2 inhibitors (Coxibs), anti-depressants, anticonvulsants and mate receptors in the medial pre-frontal cortex (mPFC; Section 5).
local anaesthetics are all used clinically in the treatment of pain
(Guindon et al., 2007b). Opioids, such as morphine derived from
the opium poppy, have been utilised in pain relief for millennia 1.2. Pain & the EC system
(Holden et al., 2005) whereas synthetic opioids (tramadol, fentanyl,
remifentanil) are mainstays for neuropathic and post-operative The subjective experience of pain involves integration of sen-
pain (Guindon et al., 2007b). These agents, nonetheless have sig- sory, emotional, and cognitive aspects. This cannot be reported by
nificant limitations including constipation, tolerance and depen- the non-human animals on which basic pain research is conducted,
dence which have contributed to an epidemic of addiction and and thus it is important to make the distinction between subjective
drug-related deaths in the US in recent years (Kolodny et al., pain and the measurable neuronal events and behavioural outputs
2015). In the past few decades, a new target for pain relief, also underlying it, termed nociception (Loeser and Treede, 2008).
taking advantage of an ancient pain relieving medication and an Nociceptive signalling begins with the transduction of a noxious
endogenous pain control pathway, has emerged. Preparations of stimulus (thermal, mechanical, or chemical) in the periphery into
the Cannabis sativa plant have been used as analgesics for centuries, neuronal activity in specialised classes of sensory afferent neurons.
but it was only in the 1960s that the major active constituent (D9- The resultant action potentials travel to cell bodies located in dorsal
tetrahydrocannabinol) was identified (Mechoulam and Gaoni, root ganglia (DRG), then to a synapse in the superficial dorsal horn
1967). It was not until the 1990s that the molecular targets medi- of the spinal cord. Here, local processing integrates peripheral input
ating its effects were discovered (the cannabinoid receptors CB1 & with descending supraspinal modulation, before transmitting the
CB2 - Devane et al., 1988; Munro et al., 1993), and the mechanisms output via several ascending pathways to the brainstem, thalamus,
and sites of action elucidated (Gregg et al., 2012; Herkenham et al., and other higher brain regions involved in the sensory and affective
1990; Hohmann et al., 2005; Martin et al., 1999; Walker and components of pain (Millan, 1999). The components of the EC
Hohmann, 2005; Walker and Huang, 2002). This work revealed system, described in detail elsewhere in this Special Issue, comprise
the G protein-coupled cannabinoid receptors CB1 & CB2, their
S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120 107

endogenous ligands anandamide (AEA) and 2-arachidonyl glycerol demand in response to high levels of activity, and produce short-
(2-AG), and their respective major synthetic (N-acylphosphatidy- term antinociceptive effects via their actions as retrograde trans-
lethanolamine phospholipase D [NAPE-PLD] & diacyglyerol lipase a mitters at presynaptic inhibitory CB1 G protein-coupled receptors
[DAGLa]) and degradative (fatty acid amide hydrolase [FAAH] & (GPCRs), with duration of effect limited by their rapid enzymatic
monoacyglycerol lipase [MAGL]) enzymes. These components are degradation. Endocannabinoids play a key role in the resolution of
expressed almost ubiquitously throughout nociceptive pathways, acute pain states (Alkaitis et al., 2010), and are elevated at various
and thus targeting the system via exogenous cannabinoid ligands sites in nociceptive pathways in chronic pain (Guindon et al., 2013;
or enhancement of endogenous signalling can regulate nociceptive Sagar et al., 2009, 2012), highlighting their role as endogenous
signalling at multiple sites; in the periphery (reviewed in Guindon analgesics.
and Beaulieu, 2009), the dorsal horn of the spinal cord (Hohmann, However, this rather simplistic picture is complicated by the
2002; Nyilas et al., 2009; Richardson et al., 1998; Sagar et al., 2010; ubiquity of EC system expression e components are localized not
Woodhams et al., 2012) and in supraspinal pain-associated regions just on excitatory neurons, but also inhibitory neurons, peripheral
of the brain, as summarized in Fig. 1. Sections 2 & 3 detail effects of immune cells, and glial cells in the central nervous system
systemic or local modulation of EC activity at peripheral and central (Egertova et al., 2003; Egertova and Elphick, 2000; Gong et al.,
sites, whilst Sections 4 & 5 focus on the role of the EC system in 2006; Gregg et al., 2012; Hegyi et al., 2009; Horva th et al., 2014).
supraspinal pain-associated regions. In neural circuits, endo- Furthermore, cannabinoid ligands and their metabolites are
cannabinoids act as short-term circuit breakers (Katona and promiscuous (Alexander and Kendall, 2007), with some excitatory
Freund, 2008). Endocannabinoids (i.e. 2-AG) are generated on- actions such as AEA agonism at TRPV1 (Ross, 2003), alongside

Fig. 1. Schematic of Nociceptive Pathways & Sites of EC System Expression.


Nociceptive stimuli are conducted from the periphery to the dorsal horn of the spinal cord, and then transmitted to the supraspinal regions via the spinothalamic tract (STT, blue)
and spinoparabrachial tract (SPBT, red). The major descending modulatory control pathway (DMCP, purple) is displayed on the right. This pathway crosses the midline at the level
of the medulla. Coloured areas indicate the position of synapses, and therefore sites of additional neuronal processing and endocannabinoid signalling, in each pathway. The
positions of laminae IeVI in the dorsal horn are indicated by dotted lines, while the black region in the brain represents the lateral ventricles.
Thal., thalamus; VMH, ventromedial hypothalamus; PbN, parabrachial nucleus; PAF, primary afferent fibre; PAG, periaqueductal grey matter; RVM, rostroventral medulla; Pyr.,
pyramidal tract; DRG, dorsal root ganglion.
Adapted from (Hunt and Mantyh, 2001).
108 S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120

inhibitory actions at CB1 and CB2 and the nuclear family of perox- produces CB1-mediated antinociception in the CFA model in rats
isome proliferator-activated receptors (PPARs, reviewed in (Wilson et al., 2005). Both CB1 and CB2 have been implicated in the
O'Sullivan, 2007; O'Sullivan, 2016). EC activity can therefore induce antinociceptive effects of FAAH inhibition in this model (Ahn et al.,
a complex interplay of actions, the result of which can be anti- 2011; Jayamanne et al., 2006), and a CB2 component is likely of
nociceptive (Alkaitis et al., 2010) or pronociceptive (Pernia-Andrade increased importance in models of inflammatory pain due to po-
et al., 2009) depending on the site of expression and the underlying tential involvement of CB2-expressing peripheral immune cells and
physiological state (Zeilhofer, 2010). glial cells of the CNS. Indeed, several distinct classes of FAAH in-
hibitors reduce nociceptive behaviour following hindpaw admin-
2. Blockade of EC metabolism as an analgesic approach: past istration of LPS, and a detailed investigation of the mechanism
issues and future directions implicates both CB1 and CB2, but not TRPV1, PPARa, or m-opioid
receptors (Booker et al., 2012).
As EC levels are known to be elevated specifically at sites of FAAH Inhibition in Models of Neuropathic Pain: The efficacy
injury or excessive nociceptive signalling (Sagar et al., 2012), the of FAAH inhibition is not limited to inflammatory pain states, and
use of specific enzyme inhibitors to augment their effects has numerous studies have demonstrated antinociceptive effects of
received particular interest in the field of pain research. This FAAH inhibition in models of neuropathic pain. In the chronic
approach targets areas of high EC turnover whilst sidestepping the constriction injury (CCI) model in mice, several studies have
undesirable effects of global cannabinoid receptor activation asso- demonstrated antinociceptive effects of FAAH inhibitors which
ciated with application of exogenous cannabinoid ligands appear to require both CB1 and CB2 (Kinsey et al., 2009, 2010; Russo
(reviewed in Ameri, 1999). In the following section we review the et al., 2007), as antagonism or genetic deletion of either receptor
historical preclinical successes of this approach, the recent setbacks blocks the anti-allodynic effect (but also see discussion of Carey
in translating into the clinic, and the promising future directions. et al., 2016 below). In contrast, the antinociceptive effects of
FAAH inhibition in the partial sciatic nerve ligation (PSNL) model in
2.1. FAAH inhibition for analgesia mice were dependent only on CB1 (Desroches et al., 2014), whilst
that seen in the spinal nerve ligation (SNL) model in rats required
Anadamide (AEA) was the first endocannabinoid to be identified both CB1 and CB2 (Chang et al., 2006; Jhaveri et al., 2006; Karbarz
(Devane et al., 1992), and thus formed the central focus of the early et al., 2009). Interestingly, another study failed to find any effect
years of endocannabinoid pain research, in which its efficacy as an of FAAH inhibition in the PSNL model in rats (Jayamanne et al.,
analgesic was established in multiple preclinical models of acute 2006), suggesting that there may be species- and model-specific
pain (Calignano et al., 1998; Walker et al., 1999). Fatty-acid amide mechanisms which complicate the interpretation of these data.
hydrolase (FAAH) was identified as the major enzyme degrading Central and Peripheral Mechanisms of FAAH Inhibition: To
fatty-acid amides which bind to cannabinoid receptors, such as investigate the major site of action of FAAH inhibition, several
AEA, but also the related compounds oleoylethanolamine (OEA) studies have used local administration of compounds into the
and palmitoylethanolamine (PEA) which do not (Cravatt et al., hindpaw, spinal cord, or brain. Local administration of a FAAH in-
1996). This discovery led to the development of several classes of hibitor into the hindpaw in the rat carrageenan model is anti-
compounds capable of inhibiting FAAH and thus promoting sig- nociceptive, but not anti-inflammatory, blocking both pain
nalling of AEA (PF3845 - Ahn et al., 2009; PF-04457845 - Ahn et al., behaviour and the receptive field expansion of dorsal horn neurons
2011; OL135 - Chang et al., 2006; JNJ-1661010 - Karbarz et al., 2009; (Jhaveri et al., 2008; Sagar et al., 2008), a marker of central sensi-
URB597 - Kathuria et al., 2003) and other fatty-acid amides. These tisation, and thus development of an inflammatory pain state
compounds show high specificity for FAAH, significantly elevating (Latremoliere and Woolf, 2009). Development of the peripherally-
levels of AEA, OEA, and PEA in the central nervous system and restricted FAAH inhibitor URB937 revealed a significant CB1-
peripheral tissues. Whilst OEA and PEA are not endocannabinoids mediated peripheral component to the antinociceptive effects of
as they have no affinity for cannabinoid receptors, PEA has well- FAAH inhibition in models of inflammatory pain, since the effects
established anti-inflammatory properties via peroxisome were completely blocked by administration of a CB1 antagonist
proliferator-activated receptors (PPARs) (Alhouayek and Muccioli, (Clapper et al., 2010; Sasso et al., 2015b), in line with the suggestion
2014; Lambert et al., 2002; LoVerme et al., 2006), and PEA and that exogenous cannabinoids exert much of their pain relieving
OEA are capable of elevating levels of AEA through substrate effects via peripheral CB1 receptors (Agarwal et al., 2007). It should
competition at FAAH (Di Marzo et al., 1994). Thus, these N-acyle- be noted, however, that spinal and supraspinal mechanisms have
thanolamines are of interest to the field of cannabinoid pain also been described for FAAH inhibition in models of acute pain. In
research. the surgical incision model of acute resolving pain in rats, spinal
FAAH Inhibition in Models of Inflammatory Pain: Due to the levels of AEA are reduced at early time points coinciding with
dual analgesic and anti-inflammatory properties of FAAH sub- maximal mechanical hypersensitivity, returning to baseline as
strates, inhibitors of FAAH have been preferentially evaluated as a nociceptive behaviour subsides (Alkaitis et al., 2010), whilst
therapeutic approach in preclinical models of inflammatory pain. administration of a FAAH inhibitor to the periaqueductal grey
These models involve application of noxious substances to the matter altered thresholds to thermal stimuli in a biphasic manner
hindpaw, resulting in inflammation (oedema) and measureable (Maione et al., 2006). Global deletion of FAAH in mice results in 15-
nociceptive behaviour, including allodynia (heightened responses fold elevations in brain levels of AEA, reduced sensitivity to thermal
to non-noxious levels of cutaneous stimulation) and hyperalgesia and inflammatory pain, and augmented responses to exogenous
(heightened responses to noxious levels of cutaneous stimulation), AEA which are mediated by CB1 (Cravatt et al., 2001). However,
although the underlying signalling mechanisms generating recent evidence has also demonstrated a pronociceptive phenotype
inflammation differ. Both brain impermeant (Clapper et al., 2010) when FAAH KO mice are challenged with the TRPV1 agonist
and brain permeant inhibitors of FAAH have been shown to sup- capsaicin (Carey et al., 2016), and show elevations in numerous
press inflammatory pain induced by formalin, carrageenan, and endovanilloids and other potentially bioactive lipids suggesting
Complete Freund's Adjuvant (CFA) (for review, see Guindon and that persistent elevation of fatty-acid amides could potentially lead
Hohmann, 2009). Systemic FAAH inhibition is anti-inflammatory to sensitisation (e.g. of TRPV1 channels) in certain pathological pain
via CB2 in the carrageenan model in mice (Holt et al., 2005), and states.
S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120 109

FAAH Inhibitors in the Clinic e a bridge too FAAH?: The development of novel analgesics.
wealth of preclinical data demonstrating efficacy of FAAH inhibi-
tion as an analgesic strategy has led to completion of Phase I trials 2.2. MAGL inhibition for analgesia
in humans (PF-04,457,845 - Li et al., 2012; V158866 - Pawsey et al.,
2016). However, despite being well-tolerated and producing sig- Although discovered a few years after AEA, 2-AG is now
nificant elevations in peripheral blood levels of AEA, PF-04,457,845 considered to be the major endocannabinoid ligand in the CNS
failed to produce significant analgesia in a Phase II trial in late-stage (Stella et al., 1997), responsible for most of the well-characterized
osteoarthritis (OA) patients (Huggins et al., 2012). Tolerance to synaptic properties of CB1 activation (reviewed in Katona and
persistently elevated AEA levels may occur with chronic FAAH in- Freund, 2012). Monoacyglycerol lipase (MAGL) was identified as
hibition, as indicated by a lack of effect of chronic FAAH inhibition the major enzyme responsible for terminating 2-AG signalling
in the rat carrageenan model of inflammatory pain (Okine et al., (Dinh et al., 2002, 2004), and though other enzymes do contribute
2012), or it could be that AEA levels at key central signalling sites to 2-AG metabolism in vivo (Blankman et al., 2007; Marrs et al.,
are already maximally elevated in these patients and/or result in 2010), ~85% of hydrolytic activity in brain can be attributed to
sensitisation of TRPV1 receptors. Better stratification of patients MAGL. Initially, attempts to develop selective MAGL inhibitors were
and targeting of pain conditions may result in more positive results. unsuccessful due to off-target interactions with other serine hy-
In the case of OA, for example, it may be beneficial to assess efficacy drolases, including FAAH (King et al., 2007; Vandevoorde et al.,
in earlier stages of disease when tolerance to chronically-elevated 2007). Local injections of the O-biphenylcarbamate URB602, an
endocannabinoids has not occurred. Furthermore, recent clinical MAGL preferring inhibitor, reduced MGL activity, selectively
assessments of the efficacy of cannabinoids in pain states (reviewed elevated levels of 2-AG without altering levels of AEA, and pro-
in Lichtman and Chapman, 2011), have suggested that beneficial duced antinociception, but was not potent enough to be used sys-
effects on pain comorbidities including mood disturbances, loss of temically (Hohmann et al., 2005). However, in 2009 a compound
appetite, and disturbed sleep, may outweigh those on sensory as- with 300-fold selectivity for MAGL over FAAH was developed from
pects of pain. Conditions such as fibromyagia, complex regional activity-based protein profiling of a diverse library of carbamates
pain syndrome, MS, or cancer pain in which such factors play a and lead optimization by the Cravatt group (Long et al., 2009a).
more prominent role may therefore be better clinical targets for JZL184, administered systemically, produces 8-fold elevations in
these compounds. brain 2-AG without markedly elevating AEA levels, and increases
Another FAAH inhibitor, BIA-102474, led to severe neurological acute thermal and mechanical pain thresholds in mice. Similar to
toxicity in a more recent Phase I trial, resulting in the death of one FAAH inhibition, substantial antinociception has been demon-
of the participants and permanent neurological damage in 5 strated in rodent models of peripheral inflammatory pain (Ghosh
others (Kaur et al., 2016; Moore, 2016). This tragic outcome would et al., 2013; Guindon et al., 2011; Woodhams et al., 2012), visceral
have the potential to kill any further clinical interest in FAAH in- and gastrointestinal pain (Busquets-Garcia et al., 2011; Kinsey et al.,
hibition, were it not for several important considerations. After 2011), neuropathic pain (Kinsey et al., 2009, 2010), chemotherapy-
extensive review of the data arising from this trial, it now seems induced neuropathy (Guindon et al., 2013), and bone cancer pain
that these shocking events reflect problems with the design of the (Khasabova et al., 2011). In models of acute pain, effects of MAGL
trial and the specific molecule utilised, rather than with targeting inhibition appear to be largely mediated by CB1 (Long et al., 2009a)
FAAH in general (Kaur et al., 2016). These issues relate to the though a more prominent CB2-mediated component has been
supramaximal dose administered, the decision to continue dosing identified in inflammatory and neuropathic pain models (Guindon
other participants after adverse events first arose, the relatively et al., 2007a, 2011; Guindon and Hohmann, 2008), perhaps un-
low selectivity of the compound, potential irreversible in- surprisingly since 2-AG is a full agonist at CB2 whilst AEA is only a
teractions with non-FAAH serine hydrolases, its long half-life, and weak partial agonist (Gonsiorek et al., 2000). However, no sooner
the lack of toxicity studies specifically relating to its known me- had a specific MAGL inhibitor been developed, than findings
tabolites. Firstly, it should be noted that the dose administered questioning the long-term efficacy of this approach appeared. Full
was 20e50 times that required for full FAAH inhibition (Be gaud inhibition of MAGL via JZL184 produces many cannabinoid-like
et al., 2016), and furthermore that this dose was administered behaviours (Long et al., 2009a), suggesting that this approach
repeatedly, allowing for accumulated tissue concentrations at may share some of the unwanted side-effects of cannabinoids.
which off-target effects became inevitable. BIA-102474 reportedly Furthermore, several studies have now revealed that sustained
showed 50e100 fold selectivity for FAAH over other serine hy- global elevation of 2-AG via genetic deletion of MAGL or persistent
drolases, and adverse events arose only on the 5th and 6th days of blockade of MAGL activity with enzyme inhibitors produces func-
50 mg administration when concentrations likely exceeded those tional antagonism of the brain EC system, resulting in profound
at which the drug was selective. Single administrations of up to downregulation and desensitization of CB1 receptors in
100 mg showed no ill effects, strongly suggesting that the toxicity nociception-associated regions, and a loss of analgesic phenotype
arose from accumulation of the drug. Although the cause of (Chanda et al., 2010; Imperatore et al., 2015; Navia-Paldanius et al.,
toxicity has not been fully established, and it remains possible that 2015; Schlosburg et al., 2010). Chronic MAGL blockade may also
they may have arisen due to impurities in the batch of compound result in physical dependence, since a CB1 antagonist precipitated
utilised in the trial, it seems likely that irreversible, off-target ef- behavioural symptoms of withdrawal following repeated high dose
fects of BIA-102474 or one of its metabolites at alternative serine JZL184 treatment (Schlosburg et al., 2009).
hydrolases was responsible. The lack of similar effects in multiple However, these problems may not present an insurmountable
trials of chemically disparate classes of FAAH inhibitors argue obstacle. Chronic partial inhibition of MAGL produces sustained
strongly against a FAAH-mediated mechanism (Huggins et al., analgesia in the absence of cannabinoid side effects in mice
2012; Li et al., 2012; Pawsey et al., 2016). In light of these trial (Busquets-Garcia et al., 2011; Kinsey et al., 2011). Furthermore, a
and compound-specific issues and the wealth of preclinical data new generation of MAGL inhibitors have been developed with more
supporting efficacy of compounds directed at FAAH, we believe attractive therapeutic profiles (Ignatowska-Jankowska et al., 2014;
that FAAH inhibition still remains a promising target for the MJN110 - Niphakis et al., 2013a). These compounds have greater
110 S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120

selectivity for MAGL over FAAH than JZL184, producing anti- pronociceptive effects via the activation, but not desensitization, of
nociceptive effects in mouse models of acute and chronic pain with TRPV1 when AEA levels are highly elevated (Maione et al., 2006;
reduced cannabimimetic properties (Ignatowska-Jankowska et al., Ross, 2003). Dual FAAH inhibition in combination with TRPV1
2014, 2015b). antagonism is therefore an attractive therapeutic target (Fowler
et al., 2009). Indeed, TRPV1 antagonism in its own right was once
2.3. The future of EC metabolic enzyme inhibitors: broadening the a promising analgesic strategy (Ghilardi et al., 2005; Swanson et al.,
appeal 2004), but clinical development of TRPV1 inhibitors was stalled due
to adverse hyperthermic effects (Gavva, 2009; Gavva et al., 2008).
FAAH and MAGL degrade a variety of lipid signalling molecules In contrast, the dual FAAH inhibitor/TRPV1 antagonist compounds
which do not bind to cannabinoid receptors, and therefore, FAAH AA-5-HT (Hohmann et al., 2005; Maione et al., 2007) and OMDM-
and MAGL inhibitors are not selective for the endocannabinoid 198 (Maione et al., 2013) produce antinociception in the absence of
system. Moreover, endocannabinoids also undergo oxidative hyperthermia. AA-5-HT has antinociceptive efficacy in the formalin
metabolism by a variety of different enzymes such as cyclo- and carrageenan models of inflammation, and the CCI and SNI
oxygenases (for reviews, see Guindon and Hohmann, 2009; models of neuropathic pain (Costa et al., 2010; de Novellis et al.,
Starowicz and Di Marzo, 2013). These observations raise the pos- 2008; de Novellis et al., 2011; Maione et al., 2007; Malek et al.,
sibility that compounds targeting multiple enzymes may produce 2016), whilst OMDM-198 is effective in the formalin and carra-
better analgesic efficacy than targeting a single enzyme in isolation. geenan models of inflammatory pain (Maione et al., 2013), and a rat
Complete ablation of EC catabolic enzyme activity may, therefore, model of osteoarthritic pain (Malek et al., 2015). A significant
not be the most effective therapeutic strategy for pain relief, as supraspinal component to these effects has been described via
evidenced by the recent clinical failure of a FAAH inhibitor in microinjection of AA-5-HT into the PAG, resulting in CB1-, and
osteoarthritic pain, and the uncovering of functional antagonism TRPV1-dependent analgesia, and a restoration of the excitation-
following chronic absence of MAGL activity. However, clinical trials inhibition balance in prefrontal cortex (de Novellis et al., 2008; de
fail for many reasons, including choice of target indication and lack Novellis et al., 2011). Thus, this approach may even be effective at
of demonstration of target engagement, and there remains signif- neutralising the affective component of pain (see Section 3) and the
icant hope in this area of analgesic drug development due to the cortical dysfunctions associated with chronic pain states (see Sec-
high safety profile observed with well-characterized FAAH in- tion 4), however further research is still ongoing to determine the
hibitors in humans. Many recent preclinical studies have utilised clinical potential of these preliminary data.
combined inhibition of FAAH and MAGL, or inhibitors of other pain- FAAH/COX2 Inhibition: Cyclooxygenase enzymes in CNS neu-
related enzymes, and/or pain-relieving drugs to produce analgesia rons play a critical role in inflammatory pains state through their
in the absence of side-effects. Multifunctional compounds often actions in generating prostanoids (Vardeh et al., 2009), and are the
have improved safety profiles compared to highly specific single central target for the non-steroidal anti-inflammatory drug
target molecules, and this work suggests that a more broad- (NSAID) class of analgesics. However, COX2 can also metabolise
spectrum approach may finally allow translation of the pre- AEA when other catabolic pathways are blocked (Fowler, 2007),
clinical promise of EC-directed therapies to the clinic. Finally, the reducing AEA tone, and potentially contributing to the clinical
allosteric modulatory site (Price et al., 2005) on the cannabinoid failure of FAAH (Starowicz and Di Marzo, 2013). Strong evidence
CB1 receptor is also a promising target for small molecule analgesic demonstrates that NSAIDs can act as FAAH inhibitors (Fowler et al.,
drug development (Ignatowska-Jankowska et al., 2015a). 1999; Seidel et al., 2003), and co-administration of FAAH inhibitors
Combined FAAH/MAGL Inhibition: Alongside JZL184, Long and and NSAIDs produces synergistic antinociceptive effects in pre-
colleagues developed an analogue compound with dual inhibitory clinical models of pain (Grim et al., 2014; Guindon and Beaulieu,
properties at MAGL and FAAH (Long et al., 2009b). Systemic 2006; Guindon et al., 2006), providing a powerful rationale for
administration of JZL195 produces greater cannabimimetic effects the design of compounds with dual activity at these two enzymes.
than full inhibition of MAGL or FAAH alone (Long et al., 2009b), but One such compound, ARN2508, has recently been developed and
unlike synthetic cannabinoid agonists, the effective dose for anti- showed efficacy in a model of intestinal inflammation (Sasso et al.,
nociception is significantly lower than that producing unwanted 2015a). Notably, this drug produced antinociception in the absence
side-effects, indicating a potential therapeutic window (Adamson of gastric damage, and may thus avoid a major side-effect of chronic
Barnes et al., 2016). Similarly, a recent study employed full FAAH NSAID treatment. In addition, substrate-specific inhibitors of COX2
inhibition with partial MAGL inhibition via co-administration of PF- have been developed which selectively block the hydrolysis of AEA,
3845 and JZL184 (Ghosh et al., 2015), producing a synergistic but spare other molecules (Hermanson et al., 2013, 2014). These
augmentation of antinociceptive potency in models of neuropathic compounds have shown efficacy at treating preclinical models of
and inflammatory pain, in the absence of cannabimimetic side ef- stress-induced neuropsychiatric disorders (Gamble-George et al.,
fects, and with an apparent lack of tolerance and dependence 2016), highlighting the link between the endocannabinoid system
following chronic dosing. In accordance with these data, a novel and stress, which forms the basis of a subsequent section of this
dual FAAH/MAGL inhibitor with markedly greater potency at FAAH review.
than MAGL (SA-57 - Niphakis et al., 2013b), produces anti-
nociception in mouse models of neuropathic and inflammatory 3. Positive allosteric modulators of cannabinoid CB1 receptor
pain (Wilkerson et al., 2016a). Interestingly, this compound can be signalling
used in combination with morphine to produce synergistic anal-
gesia, and even appears to alleviate tolerance to opioid analgesia. A The identification of allosteric binding site(s) (Price et al., 2005)
similar effect has also been noted using low dose MAGL inhibition on the CB1 GPCR has facilitated drug discovery efforts aimed at
via MJN110 in combination with morphine in a rodent model of harnessing the therapeutic potential of endocannabinoid signal-
neuropathic pain (Wilkerson et al., 2016b), suggesting this is an ling. Positive allosteric modulators enhance the affinity and/or ef-
exciting avenue for future research. ficacy of the endogenous ligand at the classical (orthosteric)
Combined FAAH Inhibition/TRPV1 Antagonism: FAAH inhibi- binding site. Because allosteric modulators bind to sites distinct
tion can produce biphasic effects in pain models, with anti- from the orthosteric binding site, they might be expected to show a
nociception via AEA actions at CB1 at low concentrations, and more limited spectrum of unwanted cannabimimetic effects
S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120 111

compared to direct agonists like THC. Positive allosteric modulators et al., 1998; see Fig. 1). Stress and fear have been shown to alter
that have been evaluated for antinociceptive efficacy in the pub- levels of endocannabinoids in these brain regions (Carrier et al.,
lished literature include lipoxin A4 (Pamplona et al., 2012) and 2005; Gregg et al., 2012; Hill et al., 2005, 2013; Hohmann et al.,
ZCZ011 (Ignatowska-Jankowska et al., 2015a). Of these, the best 2005; Morena et al., 2015; Olango et al., 2012; Patel et al., 2005;
characterized is ZCZ011, as Lipoxin A4 is an endogenous lipid Rademacher et al., 2008). In this section, we will review the role
mediator that could not be administered systemically. Nonetheless, of the endocannabinoid system in SIA and SIH, with an emphasis on
following intraventricular administration Lipoxin A4 produces the sites and mechanisms involved.
typical CB1-mediated cannabimimetic effects following intraven-
tricular administration (i.e. reduces locomotor activity in the open 4.1. Stress-induced analgesia (SIA)
field, produces catalepsy in the ring test, hypothermia) and pro-
duces antinociception in the hotplate test (Pamplona et al., 2012). SIA is a form of adaptive pain suppression, an evolutionarily
ZCZ011 suppressed neuropathic and inflammatory pain behaviour conserved response to stress that has survival value (Amit and
in mice through a CB1 mechanism (Ignatowska-Jankowska et al., Galina, 1986; Butler and Finn, 2009; Ford and Finn, 2008). Early
2015a). ZCZ011 also increased the potency of orthosteric cannabi- studies indicated that SIA was mediated via both opioid and non-
noid agonists in the tetrad (antinociception in tail-flick, hypother- opioid mechanisms, and in 2000 it was shown that CB1 receptor
mia, catalepsy and locomotor activity) did not exhibit the knock-out mice do not exhibit opioid-mediated antinociception
discriminative stimulus effects of CP55,940 or ananadamide following a forced swim in water at 34  C (Valverde et al., 2000),
(Ignatowska-Jankowska et al., 2015b). Antinociceptive efficacy was suggesting that the endocannabinoid and opioid systems interact
also preserved following 6 days of repeated dosing (Ignatowska- to mediate this form of SIA. In 2004, Finn and colleagues demon-
Jankowska et al., 2015b). Moreover, ZCZ011 did not produce strated that systemic administration of the CB1 receptor antagonist/
conditioned place preference or aversion (Ignatowska-Jankowska inverse agonist rimonabant completely prevented the suppression
et al., 2015b). of formalin-evoked nociceptive behaviour expressed in rats upon
GAT211 and GAT229 are other recently described CB1 positive re-exposure to an aversively conditioned context previously paired
allosteric modulators (Laprairie et al., 2017). The in vivo profile of with footshock (i.e. fear-conditioned analgesia; FCA) (Finn et al.,
GAT211 has only recently been reported (Slivicki et al., in press). 2004). A series of studies from Hohmann and colleagues demon-
GAT211 suppresses both inflammatory and neuropathic pain strated a key role for the EC system in an opioid-independent form
without producing typical CB1-mediated cannabimimetic effects of unconditioned SIA in rats (footshock followed immediately by
(Slivicki et al., in press). Moreover, GAT211 shows synergistic anti- tail-flick testing) and identified some of the brain regions involved.
nociceptive effects with inhibitors of both FAAH and MAGL, sug- Systemic administration of CB1 receptor antagonists (Hohmann
gesting it is not acting in a probe-specific manner to enhance 2-AG et al., 2005), or their direct microinjection into the dorsolateral
over anandamide signalling or vice versa (Laprairie et al., 2017; PAG (Hohmann et al., 2005; Suplita et al., 2005), brainstem RVM
Slivicki et al., in press). Unlike orthosteric CB1 agonists and MAGL (Suplita et al., 2005), basolateral amygdala (BLA) (Connell et al.,
inhibitors, GAT211 did not produce tolerance over a 20 day interval 2006), but not spinal cord (Suplita et al., 2006), attenuated SIA.
of once daily dosing (Slivicki et al., in press). Moreover, GAT211 also Footshock stress was shown to increase the formation of AEA and
failed to produce conditioned place preference or reward (Slivicki 2-AG in the dorsolateral PAG (Gregg et al., 2012; Hohmann et al.,
et al., in press). The similar features shared by GAT211 and 2005) and either systemic or intra-PAG administration of drugs
ZCZ011 support the importance of allosteric modulatory site(s) on which inhibit the enzymatic degradation or transport of endo-
the CB1 receptor as a target for analgesic drug development. cannabinoids potentiated SIA (Hohmann et al., 2005; Suplita et al.,
Although preclinical studies are still at an early stage CB1 positive 2005). Potentiation of SIA was also observed following direct in-
allosteric modulators show potential for suppressing chronic pain jection of a FAAH inhibitor into the RVM (Suplita et al., 2005) or
in a manner that is safe, effective and lacks unwanted side effects of intrathecal injection of FAAH and MAGL inhibitors (Suplita et al.,
orthosteric CB1 agonists. It should also be noted that several 2006). Thus, although endocannabinoids at the spinal level are
exogenous and endogenous substances have also been identified as capable of modulating this form of unconditioned SIA, mediation of
negative allosteric modulators of CB1, suggesting complex regula- this behavioural response was critically dependent on endo-
tion of CB1 signalling. This topic is covered in detail elsewhere in cannabinoid-CB1 signalling in supra-spinal sites including the PAG
this Special Issue (Khurana et al., 2017). and RVM, key components of the descending pain pathway.
The ability of footshock stress to trigger the mobilization of
4. The EC system and stress-induced modulation of pain endocannabinoids such as 2-AG led to the use of this model of
stress antinociception to probe the biochemical mechanisms un-
The intensity of perceived pain does not necessarily correlate derlying on demand formation of 2-AG in vivo (Gregg et al., 2012).
with the degree of tissue damage, injury or inflammation, and the The results of behavioural, pharmacological studies, and immuno-
importance of modulation of pain by context and emotion is now histochemical studies, along with RNA interference, quantitative
widely recognized. Stress, fear, and anxiety can modulate pain PCR and lipidomic analyses of endocannabinoid content also show
(Asmundson and Katz, 2009; Burke et al., 2015; Butler and Finn, that activation of type 5 metabotropic glutamate receptors
2009; Fitzgibbon et al., 2016; Ford and Finn, 2008; Jennings et al., (mGluR5) leads to a Ca2þ-dependent increase in the activity of the
2014; Olango and Finn, 2014; Rhudy and Meagher, 2000, 2001; enzyme diacylglycerol lipase-a (DAGL-a), which in turn initiates
Wiech and Tracey, 2009). Negative emotions with low to moder- both 2-arachidonoylglycerol formation in the dorsolateral PAG and
ate arousal tend to exacerbate pain through the phenomenon of endocannabinoid mediated analgesia (Gregg et al., 2012). Stimu-
stress-induced hyperalgesia (SIH), while negative emotions with lation of mGluR5 receptors in the dlPAG with the group 1 mGluR
high arousal tend to inhibit pain through the phenomenon of agonist (S)-3,5-dihydroxyphenylglycine (DHPG) initiated 2-AG
stress-induced analgesia (SIA) (de Wied and Verbaten, 2001; formation and enhanced endocannabinoid-mediated stress anti-
Dougher, 1979; Meagher et al., 2001; Rhudy and Meagher, 2000, nociception through a mechanism that required both presynaptic
2001, 2003). Cannabinoid receptors are localized in brain regions CB1 receptors and DAGL-a. Pharmacological inhibition of DAGL
involved in the modulation of pain, stress, and emotion including activity in the dlPAG with tetrathydrolipstin decreased levels of 2-
the RVM, PAG, amygdala and PFC (Herkenham et al., 1991; Tsou AG in the PAG with no change in levels of anandamide. Moreover,
112 S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120

virally mediated RNA silencing of DAGL-a but not the DAGL-b iso- visceral hypersensitivity (Shen et al., 2010). In the same study, a
form suppressed both 2-AG formation and stress antinociception. stress-induced up-regulation of CB1 receptors was demonstrated in
These effects of RNA silencing of DAGL-a mRNA were mimicked by the colon. In another study, visceral motor response increased
multiple pharmacological inhibitors of DAGL injected into the same significantly in water avoidance stressed rats, indicating hyper-
site. Finally, 2-AG is likely to act as a retrograde messenger in the algesia (Hong et al., 2009). Treatment of water avoidance stressed
dlPAG because CB1 was confined to presynaptic terminal whereas rats with the cannabinoid receptor agonist, WIN 55,212-2, also
DAGL-a, which resided in dendritic spines, colocalized with prevented the development of visceral hyperalgesia. Levels of
mGluR5 (Gregg et al., 2012). A similar molecular architecture of 2- anandamide in the dorsal root ganglia of stressed rats were
AG signalling and CB1-mediated potentiation of stress anti- increased, while CB1 receptor expression was decreased (Hong
nociception was found in the lumbar spinal cord (Nyilas et al., et al., 2009). These results suggest that endocannabinoid signal-
2009). Thus, stress-induced synthesis of 2-AG within the dorso- ling through CB1 may play an important role in stress-induced
lateral PAG and lumbar spinal cord is dependent on mGluR5- visceral hyperalgesia.
induced stimulation of postsynaptic DAGLa (Gregg et al., 2012; Chronic unpredictable stress (CUS), a widely used model for
Nyilas et al., 2009). Although intra-BLA administration of rimona- inducing anxiety and depressive-like behaviour in mice, enhances
bant suppressed unconditioned SIA, inhibitors of endocannabinoid thermal (hot plate test) and mechanical (von Frey) hyperalgesia
hydrolysis had no effect on SIA when injected into this brain region (Shi et al., 2010). CUS has also been shown to induce long-lasting
(Connell et al., 2006). Unlike SIA, FCA was not prevented by intra- widespread hyperalgesia in mice following repeated intramus-
BLA rimonabant (Roche et al., 2007, 2010), but was attenuated by cular injection of nerve growth factor (NGF) which induces spinal
intra-BLA administration of another CB1 receptor antagonist/in- sensitisation accompanied by hyperalgesia (Lomazzo et al., 2015).
verse agonist, AM251 (Rea et al., 2013). The model of FCA used by The FAAH and MAGL inhibitors URB597 and JZL184 attenuated the
Finn and co-workers has an opioid-mediated component. Thus, CUS-induced anxiety-related behaviour in the light-dark box and
enhancement of FCA in this model, by systemic administration of thermal hyperalgesia in the hot plate test. URB597 significantly
the FAAH inhibitor URB597, is prevented by co-administration of reduced the widespread hyperalgesia induced via combined CUS
the opioid receptor antagonist naloxone, as well as by rimonabant and NGF in this study, while JZL184 had no significant effect
and the selective CB2 receptor antagonist SR144528 (Butler et al., (Lomazzo et al., 2015). These data suggest a role for a FAAH sub-
2008). Like SIA, it has been shown that FCA in rats is fully attenu- strate in this form of SIH.
ated by intra-dlPAG injection of the CB1 receptor antagonist/inverse Genetic background plays a key role in determining the effect of
agonist rimonabant and is associated with increased tissue levels of stress on pain. The Wistar-Kyoto (WKY) rat displays increased
AEA in this PAG column (Olango et al., 2012). The ventral hippo- sensitivity to noxious stimuli and exhibits an anxio-depressive
campus is another locus of endocannabinoid-mediated FCA in rats phenotype and hyper-sensitivity to stress, compared with other
(Ford et al., 2011). Additional work in mice has suggested that in- rat strains, including Sprague-Dawley (SD) rats (Burke et al., 2010;
teractions between the endocannabinoid system and the chol- O'Mahony et al., 2011). Recently, impairment in pain-related
ecystokininergic (CCK) system (particularly CCK 2 receptors) are mobilization of AEA and 2-AG, along with their synthesizing en-
important for expression of an opioid-dependent form of uncon- zymes, NAPE-PLD and DAGL, respectively, has been demonstrated
ditioned SIA (Kurrikoff et al., 2008). in the RVM of WKY rats compared with SD rats, following intra-
By contrast, endocannabinoid-mediated SIA was attenuated in plantar injection of formalin (Rea et al., 2014). Systemic adminis-
rats tolerant to the cannabinoid receptor agonists WIN55,212-2 or tration of AM251 potentiated, while systemic administration of the
D9-THC (Hohmann et al., 2005; Suplita et al., 2008) but not in rats FAAH inhibitor URB597 attenuated, hyperalgesia to formalin in-
rendered tolerant to morphine (Hohmann et al., 2005), suggesting jection in WKY rats, but not SD rats, an effect mediated by CB1
that endocannabinoid mediation of this form of SIA occurs inde- receptors in the RVM. These data suggest that impaired endo-
pendently of m-opioid receptors. Furthermore, rats acutely exposed cannabinoid-CB1-dependent signalling in the RVM underlies the
to footshock were hypersensitive to the antinociceptive effects of hyper-sensitivity to noxious stimuli in WKY rat model of negative
WIN55,212-2 and D9-THC and, in turn, acute D9-THC and affective state (Rea et al., 2014). More recently, further work from
WIN55,212-2 administration potentiated SIA, suggesting a bidi- Finn and colleagues has identified a role for non-cannabinoid re-
rectional sensitisation between endocannabinoid-mediated SIA ceptor targets of the endocannabinoids in hyperalgesia in WKY rats,
and exogenous cannabinoid-induced antinociception. In summary, specifically PPARg (Okine et al., 2017), and TRPV1 (Madasu et al.,
it is clear that the endocannabinoid system plays a key role in 2016) in the PAG. The role of the EC system in the effects of
mediating non-opioid and opioid-dependent forms of endogenous repeated exposure to forced swim stress on formalin-evoked
pain suppression in response to either unconditioned or condi- nociceptive behaviour in stress normo-responsive (SD) and stress
tioned stressors. hyper-responsive (WKY) rat strains has also been investigated.
Formalin-evoked nociceptive behaviour was increased in SD rats
4.2. Stress-induced hyperalgesia (SIH) following ten days of forced swim stress (Jennings et al., 2016).
Anandamide levels were reduced in the contralateral amygdala
Stress and anxiety do not invariably suppress pain; they can also (relative to formalin injection) of SD rats but not WKY rats. Strain
enhance nociception and exacerbate pain in a phenomenon differences in components of the endocannabinoid system within
referred to as stress-induced hyperalgesia (SIH). Despite the well- the amygdala were also observed. For example, decreased levels of
established role of the endocannabinoid system in stress, anxiety, anandamide and 2-AG were reported in the ipsilateral amygdala of
and pain (Finn, 2010), few studies have investigated the role of SD, but not WKY, rats. Lower levels of CB1 receptor mRNA were seen
endocannabinoids in SIH. Systemic administration of the CB1 re- in the ipsilateral, but not contralateral, amygdala of WKY rats. These
ceptor agonist arachidonyl-2-chloro ethylamine (ACEA) signifi- data indicate a role for the endocannabinoid system in the amyg-
cantly reduced the enhanced visceromotor reflex to colorectal dala in SIH, as well as implicating it in the strain differences seen
distention (i.e. number of abdominal contractions) and also atten- between WKY and SD rats (Jennings et al., 2016). Additional studies
uated changes in electromyogram response in rats stressed by are warranted to fully understand the role of the endocannabinoid
partial restraint, whereas, the CB1 receptor antagonist/inverse system in SIH, particularly in neuropathic pain models and studies
agonist (rimonabant) had opposing effects on this stress-induced in human subjects. Preclinical studies could, for example,
S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120 113

investigate the effects of pharmacological or genetic manipulation amygdala output region (central nucleus, CeA) as well as to extra-
of the endocannabinoid system on stress-induced enhancement of amygdalar brain regions such as the mPFC. In contrast to pain-
hyperalgesia in rodent models of neuropathic pain. Clinical studies related deactivation of the mPFC, the amygdala (CeA and BLA) de-
could assess the potential utility of endocannabinoids (and related velops synaptic plasticity and hyperactivity in pain states, which
lipids) as biomarkers for stress-induced exacerbation of chronic critically contribute to the emotional-affective aspects of pain and
pain, and co-morbidity of chronic pain with affective disorders, in the facilitation of pain sensitivity (Neugebauer, 2015). Increased
addition to investigating whether mechanisms elucidated in ro- activity in the amygdala has also been described in experimental
dents translate to human patients and might be modulated phar- and clinical human pain states (reviewed by Neugebauer, 2015).
macologically for therapeutic benefit. Taken together, the data to Increased BLA output has been causally linked to mPFC deacti-
date, reviewed above, suggest that endocannabinoid-CB1 receptor vation. Pharmacological studies in an arthritis pain model (knee
signalling in key components of the descending pain pathway joint monoarthritis induced by intraarticular kaolin/carrageenan,
mediates SIA, while a deficit in endocannabinoid-CB1 receptor 5e6 h post-induction; Neugebauer et al., 2007) showed that
signalling may underlie SIH. normalizing activity of BLA neurons with stereotaxic intra-
amygdala injection of a corticotropin-releasing factor 1 (CRF1) re-
5. The EC system & medial prefrontal cortex (mPFC) ceptor antagonist partially restored the decreased activity of layer V
dysfunction in pain states: CB1 and mGluR5 in a model of mPFC pyramidal neurons (Ji et al., 2010). The mechanism by which
arthritic pain BLA projection neurons decrease mPFC pyramidal cell activity is
glutamate-driven activation of GABAergic mPFC interneurons,
Alongside key involvement in the sensory and stress-induced resulting in feedforward inhibition of mPFC pyramidal cells (Fig. 2).
aspects of pain processing, the EC system is also involved in the Whole-cell patch-clamp recordings of layer V pyramidal cells in the
accompanying affective-emotional and cognitive dysfunctions infralimbic and prelimbic mPFC showed increased inhibitory syn-
which underlie the negative mood aspects of chronic pain in hu- aptic transmission (inhibitory postsynaptic currents, IPSCs) in brain
man patients. In the final section of this review, we introduce slices from rats with a knee joint monoarthritis (5e6 h post-
mPFC-amygdala interactions as a recently identified major player in induction by intraarticular kaolin/carrageenan) compared to con-
chronic pain states, and the potential for alleviating dysfunctions by trols (Ji et al., 2010; Kiritoshi et al., 2016). IPSCs were evoked by
targeting the functional coupling of cannabinoid and metabotropic focal electrical stimulation of fiber tracts containing anterogradely
glutamate receptors. labeled BLA axons, and by more selective optogenetic activation of
BLA terminals in the mPFC expressing a light sensitive channel
5.1. mPFC deactivation in pain (channel rhodopsin 2, ChR2) 4 weeks after injection of viral vector
(rAAV5/CaMKIIa-ChR2(H134R)-eYFP) into the BLA (Ji et al., 2010;
The mPFC is responsible for executive functions and is closely
connected to other limbic areas playing key roles in emotion, such
as the amygdala. The infralimbic mPFC (area 25) exerts top-down
control of amygdala function to inhibit aversive behaviours in a
process referred to as “fear extinction” (Herry et al., 2010; Likhtik
et al., 2005; Marek et al., 2013; Orsini and Maren, 2012; Pape and
Pare, 2010; Sotres-Bayon and Quirk, 2010). Decreased infralimbic
activity has been implicated in extinction deficits (Chang and
Maren, 2010; Hefner et al., 2008; Kim et al., 2010; Sierra-Mercado
et al., 2011). Accumulating evidence suggests that dysfunction in
mPFC-amygdala interactions plays an important role in pain
(Neugebauer, 2015). Functional and structural abnormalities in the
mPFC have been detected in human pain patients (Apkarian et al.,
2004b; Mayer et al., 2005) and in acute and chronic models of
pain states (Cardoso-Cruz et al., 2013; Ji et al., 2010; Metz et al.,
2009). Decreased activity of output neurons (pyramidal cells) in
the infralimbic and prelimbic (area 32) regions of the mPFC has
been demonstrated in rodent models of arthritis (Ji and
Neugebauer, 2011, 2014; Ji et al., 2010) and neuropathic pain
(Wang et al., 2015; Zhang et al., 2015). Brain slice physiology studies
show that decreased infralimbic and prelimbic mPFC output is the
consequence of abnormally enhanced glutamatergic activation of
parvalbumin-expressing GABAergic interneurons (Ji and
Neugebauer, 2011; Ji et al., 2010; Kiritoshi et al., 2016; Zhang
et al., 2015). Mechanisms of this abnormal feedforward inhibition,
however, remained to be determined, and form the subject of the
following two sections.
Fig. 2. Schematic of mGLuR5-CB1 coupling in mPFC-amygdala interaction in pain
states.
5.2. Synaptic mechanisms The amygdala (green dashed box) sends glutamatergic (Glu) projections from the
basolateral nucleus, (BLA) to mPFC (blue dashed box) pyramidal cells activating
A major source of input to the infralimbic and prelimbic mPFC mGluR5, and to mPFC GABAergic interneurons inhibiting mPFC pyramidal cells
are long-range projections from the basolateral amygdala (BLA) (“feedforward inhibition”). mGluR5 couples to 2-AG synthesis via DAGLa. 2-AG release
acts on presynaptic CB1 to inhibit synaptic inhibition hence increasing mPFC output.
(Cheriyan et al., 2016; Little and Carter, 2013; Senn et al., 2014). The mPFC pyramidal cells send glutamatergic projections to GABAergic interneurons in the
BLA is a major site for the integration of sensory and other inputs in amygdala (intercalated cells, ITC) to control amygdala output from the central nucleus
the amygdala, forwarding this highly processed information to the (CeA).
114 S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120

Kiritoshi et al., 2016). Increased feedforward inhibition of prelimbic explain the decreased mPFC output seen in this model. The
layer V pyramidal cells was also detected 10 days after induction of mechanism underlying the loss of CB1-mediated control of synaptic
SNI neuropathic pain in the mouse (Zhang et al., 2015). inhibition appears to be a lack of available 2-AG, rather than of
Feedforward inhibition of mPFC pyramidal cells involves acti- functional CB1 receptors or impaired release of endocannabinoids.
vation of GABAergic interneurons mediated by metabotropic This has been demonstrated by the restoration of mGluR5-CB1
glutamate receptor subtype mGluR1, but not mGluR5 (Ji and facilitation of mPFC output via use of specific enzyme inhibitors.
Neugebauer, 2011; Sun and Neugebauer, 2011). Since blockade of Increasing availability of 2-AG in the postsynaptic cell via inhibition
mGluR1 restored mPFC activity only partially (Ji and Neugebauer, of the postsynaptic 2-AG hydrolyzing enzyme ABHD6 with intra-
2011), other mechanisms and targets to increase and restore cellular WWL70, MAGL via JZL184, or by blocking GABAergic inhi-
mPFC activity were explored. The EC system has subsequently bition with intracellular picrotoxin were all effective at restoring
emerged as a likely candidate, based on the evidence for an mGLuR5-CB1 disinhibition of mPFC output (Kiritoshi et al., 2016).
important role of CB1 receptors in the infralimbic mPFC in fear A rescue strategy has been devised to increase 2-AG availability,
extinction (Lin et al., 2009; Marsicano et al., 2002). thus removing abnormal inhibition and increasing mPFC output. In
the arthritic pain state (kaolin/carrageenan model), the combined
5.3. Role of CB1 and mGluR5 interaction in control of cortical application of VU0360172 and ACEA into the mPFC restored DSI,
feedforward inhibition decreased IPSCs and increased output of mPFC pyramidal cells
measured as synaptically-evoked spiking (E-S coupling) in rat brain
ECs act as retrograde signalling molecules on presynaptic CB1 slices (Kiritoshi et al., 2013, 2016). This combination strategy also
receptors to inhibit excitatory or inhibitory synaptic transmission increased background and evoked activity (action potentials) of
(Di Marzo, 2011; Guindon and Hohmann, 2009; Kano et al., 2009; infralimbic mPFC neurons recorded extracellularly in anesthetized
Lovinger, 2008). In the rodent mPFC, CB1 is predominantly if not rats in vivo (Ji and Neugebauer, 2014). Importantly, a TRPV1 re-
exclusively expressed in GABAergic interneurons (Marsicano and ceptor antagonist (AMG9810) did not block the restored control of
Lutz, 1999; Wedzony and Chocyk, 2009) and therefore well posi- synaptic inhibition by the combination strategy, arguing against the
tioned to reduce the abnormal synaptic inhibition observed in pain involvement of TRPV1 receptors, which have been implicated in
models (see 4.2). Indeed, CB1-mediated depolarization-induced some actions of the endocannabinoid AEA.
suppression of synaptic inhibition (DSI) has been detected in
layer V infralimbic pyramidal cells in brain slices from normal rats 5.4. Role of CB1 and mGluR5 interaction in control of amygdala
(Kiritoshi et al., 2013, 2016). DSI is a form of short-term synaptic function
plasticity, involving postsynaptic calcium influx following depo-
larization, activation of DAGLa, synthesis and release of 2-AG, and A major limbic target of mPFC output is the amygdala (Gabbott
retrograde activation of CB1 receptors on the presynaptic terminal et al., 2005; McDonald, 1998). Cortical control of amygdala function
to inhibit transmitter release (Di Marzo, 2011; Kano et al., 2009; has been identified as a critical mechanism of fear extinction (Herry
Lovinger, 2008; Rivera et al., 2014). A selective CB1 agonist (ACEA) et al., 2010; Likhtik et al., 2005; Marek et al., 2013; Orsini and
decreased frequency, but not amplitude, of miniature IPSCs and Maren, 2012; Pape and Pare, 2010; Sotres-Bayon and Quirk,
increased synaptically-evoked spiking (EeS coupling, a measure of 2010). In various pain models, amygdala activity and output have
output function), suggesting that presynaptic CB1 controls inhibi- been shown to be abnormally increased due to uncontrolled
tory transmission onto mPFC pyramidal cells (Kiritoshi et al., 2013). excitatory glutamatergic and peptidergic drive onto neurons of the
In addition to DSI, mGluR5 has been linked to the production of CeA (Neugebauer, 2015). At the center of this dysfunction is
ECs and synaptic depression (Robbe et al., 2002). As previously impaired activation of an inhibitory gating mechanism centered on
discussed (see Section 4), mGluR5 can activate the phospholipase C feedforward inhibition of CeA neurons by a cluster of GABAergic
- DAGL2-AG (Gregg et al., 2012; Nyilas et al., 2009). Complementary neurons in the intercalated (ITC) cell mass, which is in turn driven
to pre-synaptically-expressed CB1, mGluR5 in the mPFC is by glutamatergic projections from BLA and mPFC (Ren et al., 2013)
expressed mostly on post-synaptic elements (Muly et al., 2003), has (Fig. 2). The infralimbic mPFC is known to target these GABAergic
excitatory effects on layer V pyramidal cells (Fontanez-Nuin et al., ITC cells (Amir et al., 2011; Berretta et al., 2005; Busti et al., 2011;
2011; Kiritoshi et al., 2013; Marek and Zhang, 2008), and plays a Pinard et al., 2012).
role in fear extinction (Fontanez-Nuin et al., 2011; Sepulveda- Co-activation of CB1 and mGluR5 in the infralimbic mPFC with
Orengo et al., 2013; Xu et al., 2009). CB1-expressing axon termi- stereotaxic application of VU0360172 and ACEA increased back-
nals synapse onto mPFC pyramidal cells expressing mGluR5 and ground and evoked activity of infralimbic pyramidal cells in a
DAGL (Lafourcade et al., 2007), and a selective mGluR5 activator model of arthritic pain (kaolin/carrageenan monoarthritis, 5e6 h
(VU0360172 - Rodriguez et al., 2010) decreased synaptic inhibition postinduction) and inhibited the pain-related increase of back-
of infralimbic pyramidal cells, in a CB1-sensitive manner (Kiritoshi ground and evoked activity in CeA neurons, but had no effect under
et al., 2013). VU0360172 increased pyramidal output (E-S coupling), normal conditions (Ji and Neugebauer, 2014). The data suggest that
which was blocked by intracellular inhibition of DAGLa with tet- infralimbic mPFC neurons are inversely linked to amygdala output
rahydrolipstatin (THL) or by the CB1 antagonist/inverse agonist and that restoring mPFC output with a combination strategy of
AM251 (Kiritoshi et al., 2013, 2016). These data suggest that mGluR5-CB1 activation can engage cortical control of abnormally
mGluR5 increases pyramidal output by controlling synaptic inhi- enhanced amygdala output in pain.
bition through 2-AG-CB1 signalling (Fig. 2).
Unexpectedly, this mGluR5-CB1-mediated effect is absent in a 5.5. Role of CB1 and mGluR5 interaction in cortical control of pain
model of arthritic pain. In brain slices from arthritic animals behaviour
(kaolin/carrageenan model, 5e6 h post-induction), DSI was
impaired, mGluR5 activation with VU0360172 had no effect on the Pain is a multidimensional disorder with sensory, emotional-
output (E-S coupling) of infralimbic pyramidal cells, and ACEA no affective, and cognitive aspects. Pain-related neuroplastic changes
longer decreased feedforward inhibition (Kiritoshi et al., 2013, in the amygdala circuitry generate emotional responses such as
2016). This breakdown of mGluR5-CB1 signalling in the mPFC vocalizations, and anxiety-like and depression-related behaviours,
leads to abnormally enhanced feedforward inhibition, and may but also contribute to increased pain sensitivity (Neugebauer,
S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120 115

2015). mPFC dysfunction has been linked to fear extinction deficits populations of patients. Because FAAH and MAGL inhibitors are not
(Fontanez-Nuin et al., 2011; Sepulveda-Orengo et al., 2013; Xu et al., specific for the endocannabinoid system, more work is also
2009) as well as to pain-related cognitive deficits such as in necessary to understand the biological roles of other lipid media-
decision-making (Apkarian et al., 2004a,b; Ji et al., 2010; Moriarty tors that are generated by these inhibitors that do not bind to
et al., 2011; Pais-Vieira et al., 2009). Restoring mPFC output cannabinoid receptors. Allosteric modulators of CB1 receptors may
would therefore be expected to mitigate the cognitive deficits as therefore be a useful strategy for amplifying effects of endo-
well as the amygdala-dependent aspects of pain. cannabinoids only at sites where they are produced and released on
Co-activation of CB1 and mGluR5 in the infralimbic mPFC via demand.
stereotaxic application of VU0360172 and ACEA increases pyrami- Nonetheless, multifunctional compounds targeting the EC sys-
dal cell output, and effectively inhibits spinal withdrawal reflexes tem allied to inhibition of COX2, antagonism of TRPV1, or in com-
and audible and ultrasonic vocalizations (reflecting sensory and bination with opioids or NSAIDs have great potential to produce a
emotional-affective responses, respectively), evoked by mechanical superior therapeutic profile that harnesses the therapeutic poten-
compression of the knee joint in the kaolin-carrageenan model of tial of the endocannabinoid signalling system while minimizing
arthritic pain (5e6 h post-induction). Importantly, stereotaxic in- unwanted cannabimimetic side effects.
jections of VU0360172 and ACEA into the anterior cingulate cortex
(area 24b) had no significant effect on hindlimb withdrawal 7. Future directions
thresholds and vocalizations of arthritic rats, suggesting that this is
a region-specific mechanism. Within the field of endocannabinoid research, significant
This rescue strategy also mitigated cognitive deficits observed in fundamental questions remain unanswered. We still do not
this arthritic pain model by measuring reward-based decision- completely understand how these hydrophobic lipid signalling
making in a rodent gambling task model (Ji et al., 2010; Sun and molecules are transported across aqueous environments such as
Neugebauer, 2011). In this task, normal animals switch from the synaptic cleft or cytoplasm to reach their target receptors. Fatty
preferring the “high-risk” lever that provides 3 chocolate coated acid binding proteins have been strongly implicated (e.g. Kaczocha
food pellets in only 3 of 10 trials to preferring the “low-risk” lever et al., 2009, 2015, 2014), but their roles have yet to be full eluci-
that provides one food reward consistently in 9 of 10 trials. Arthritic dated. Furthermore, not only are ECs promiscuous, but CB1 has been
rats persist in preferring the high-risk lever, suggesting that they demonstrated to have constitutive activity in the absence of ligand
fail to switch strategies. Stereotaxic co-application of VU0360172 signalling (Howlett et al., 2011; Lee et al., 2015), and can form
and ACEA into the infralimbic mPFC restored the ability of arthritic heterodimers with several other GPCRs (Hudson et al., 2010),
rats to switch strategies and to prefer the low-risk lever like normal further complicating interpretation of the pharmacology of EC-
rats (Kiritoshi et al., 2016). directed therapeutics. Some mystery still surrounds the role of
Consistent with these observations, optogenetic silencing of AEA in EC signalling. Anatomical evidence suggests that its pur-
parvalbumin-expressing interneurons in the mPFC decreased me- ported major synthetic enzyme, NAPE-PLD, is localized pre-
chanical and thermal hypersensitivity of mice in SNI neuropathic synaptically (Nyilas et al., 2008) whilst FAAH is found post-
pain model, while optogenetic activation of these interneurons synaptically (Gulyas et al., 2004), in stark contrast to the estab-
exacerbated pain sensitivity (Zhang et al., 2015). In the conditioned lished complementary post-synaptic position of the 2-AG synthe-
place preference test, optogenetic activation or silencing of sizing enzyme DAGLa (Gregg et al., 2012; Nyilas et al., 2009) and
parvalbumin-expressing interneurons in the mPFC decreased or pre-synaptic position of MAGL (Gulyas et al., 2004; Horva th et al.,
increased, respectively, preference of neuropathic mice for the 2014). Revealing the answers to these questions will facilitate
conditioned chamber compared to sham controls (Zhang et al., better understanding of the function of EC signalling under physi-
2015). This optogenetic silencing of inhibitory mPFC interneurons ological and pathological situations, and greatly aid the develop-
also attenuated escape behaviour in neuropathic mice (Zhang et al., ment of future therapeutic interventions.
2015). Whilst much has been achieved in the past few decades, more
These studies collectively suggest that pain is associated with work is necessary to characterize both efficacy and safety profiles of
the disruption of mGluR5-endocannabinoid signalling in the mPFC, existing EC-directed therapeutic strategies, and to answer these
resulting in decreased output due to uncontrolled feedforward fundamental questions about EC function, so that the clinical po-
inhibition driven by amygdala (BLA) inputs. Restoring mPFC output tential of modulating the endocannabinoid system for analgesia
by co-activation of mGluR5 and CB1 inhibits abnormally enhanced can be realized.
amygdala activity, hypersensitivity and emotional-affective pain
responses and also restores cognitive functions in a decision- Acknowledgements
making task, suggesting that this approach may have utility in
treating the non-sensory aspects of chronic pain states. DPF is supported by a grant from Science Foundation Ireland
(10/IN.1/B2976). AGH is supported by NIDA (DA041229).
6. Conclusions
References
The EC system is a major endogenous pain control system,
running in parallel to the opioid system and playing crucial roles Adamson Barnes, N.S., Mitchell, V.A., Kazantzis, N.P., Vaughan, C.W., 2016. Actions of
the dual FAAH/MAGL inhibitor JZL195 in a murine neuropathic pain model. Br. J.
the development and resolution of pain states, and the affective Pharmacol. 173, 77e87.
and cognitive aspects of pain. The initial promise of augmenting EC Agarwal, N., Pacher, P., Tegeder, I., Amaya, F., Constantin, C.E., Brenner, G.J.,
signalling via specific enzyme inhibitors has been diminished by Rubino, T., Michalski, C.W., Marsicano, G., Monory, K., Mackie, K., Marian, C.,
Batkai, S., Parolaro, D., Fischer, M.J., Reeh, P., Kunos, G., Kress, M., Lutz, B.,
recent clinical failures. However, greater understanding of the role
Woolf, C.J., Kuner, R., 2007. Cannabinoids mediate analgesia largely via pe-
of the EC system in non-opioid and opioid-dependent forms of ripheral type 1 cannabinoid receptors in nociceptors. Nat. Neurosci. 10,
endogenous pain suppression and exacerbation in response to 870e879.
stress, and the dysfunction of forebrain-limbic circuitry in pain Ahn, K., Johnson, D.S., Mileni, M., Beidler, D., Long, J.Z., McKinney, M.K.,
Weerapana, E., Sadagopan, N., Liimatta, M., Smith, S.E., Lazerwith, S., Stiff, C.,
states in humans will aid the development of future analgesic Kamtekar, S., Bhattacharya, K., Zhang, Y., Swaney, S., Van Becelaere, K.,
strategies, especially with respect to targeting particular Stevens, R.C., Cravatt, B.F., 2009. Discovery and characterization of a highly
116 S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120

selective FAAH inhibitor that reduces inflammatory pain. Chem. Biol. 16, Chaplan, S.R., Webb, M., 2006. Inhibition of fatty acid amide hydrolase produces
411e420. analgesia by multiple mechanisms. Br. J. Pharmacol. 148, 102e113.
Ahn, K., Smith, S.E., Liimatta, M.B., Beidler, D., Sadagopan, N., Dudley, D.T., Young, T., Cheriyan, J., Kaushik, M.K., Ferreira, A.N., Sheets, P.L., 2016. Specific sTargeting of the
Wren, P., Zhang, Y., Swaney, S., Van Becelaere, K., Blankman, J.L., Nomura, D.K., basolateral amygdala to projectionally defined pyramidal neurons in prelimbic
Bhattachar, S.N., Stiff, C., Nomanbhoy, T.K., Weerapana, E., Johnson, D.S., and infralimbic cortex. eNeuro 3.
Cravatt, B.F., 2011. Mechanistic and pharmacological characterization of PF- Clapper, J.R., Moreno-Sanz, G., Russo, R., Guijarro, A., Vacondio, F., Duranti, A.,
04457845: a highly potent and selective fatty acid amide hydrolase inhibitor Tontini, A., Sanchini, S., Sciolino, N.R., Spradley, J.M., Hohmann, A.G.,
that reduces inflammatory and noninflammatory pain. J. Pharmacol. Exp. Ther. Calignano, A., Mor, M., Tarzia, G., Piomelli, D., 2010. Anandamide suppresses
338, 114e124. pain initiation through a peripheral endocannabinoid mechanism. Nat. Neu-
Alexander, S.P.H., Kendall, D.A., 2007. The complications of promiscuity: endo- rosci. 13, 6.
cannabinoid action and metabolism. Br. J. Pharmacol. 152, 602e623. Connell, K., Bolton, N., Olsen, D., Piomelli, D., Hohmann, A.G., 2006. Role of the
Alhouayek, M., Muccioli, G.G., 2014. Harnessing the anti-inflammatory potential of basolateral nucleus of the amygdala in endocannabinoid-mediated stress-
palmitoylethanolamide. Drug Discov. Today 19, 1632e1639. induced analgesia. Neurosci. Lett. 397, 180e184.
Alkaitis, M.S., Solorzano, C., Landry, R.P., Piomelli, D., DeLeo, J.A., Romero- Costa, B., Bettoni, I., Petrosino, S., Comelli, F., Giagnoni, G., Di Marzo, V., 2010. The
Sandoval, E.A., 2010. Evidence for a role of endocannabinoids, astrocytes and dual fatty acid amide hydrolase/TRPV1 blocker, N-arachidonoyl-serotonin, re-
p38 phosphorylation in the resolution of postoperative pain. PLoS One 5, lieves carrageenan-induced inflammation and hyperalgesia in mice. Pharmacol.
e10891. Res. 61, 537e546.
Ameri, A., 1999. The effects of cannabinoids on the brain. Prog. Neurobiol. 58, Cravatt, B.F., Demarest, K., Patricelli, M.P., Bracey, M.H., Giang, D.K., Martin, B.R.,
315e348. Lichtman, A.H., 2001. Supersensitivity to anandamide and enhanced endoge-
Amir, A., Amano, T., Pare, D., 2011. Physiological identification and infralimbic nous cannabinoid signaling in mice lacking fatty acid amide hydrolase. Proc.
responsiveness of rat intercalated amygdala neurons. J. Neurophysiol. 105, Natl. Acad. Sci. U. S. A. 98, 9371e9376.
3054e3066. Cravatt, B.F., Giang, D.K., Mayfield, S.P., Boger, D.L., Lerner, R.A., Gilula, N.B., 1996.
Amit, Z., Galina, Z.H., 1986. Stress-induced analgesia: adaptive pain suppression. Molecular characterization of an enzyme that degrades neuromodulatory fatty-
Physiol. Rev. 66, 1091e1120. acid amides. Nature 384, 83e87.
Apkarian, A.V., Baliki, M.N., Geha, P.Y., 2009. Towards a theory of chronic pain. Prog. de Novellis, V., Palazzo, E., Rossi, F., de Petrocellis, L., Petrosino, S., Guida, F.,
Neurobiol. 87, 81e97. Luongo, L., Migliozzi, A., Cristino, L., Marabese, I., Starowicz, K., Di Marzo, V.,
Apkarian, A.V., Sosa, Y., Krauss, B.R., Thomas, P.S., Fredrickson, B.E., Levy, R.E., Maione, S., 2008. The analgesic effect of N-arachidonoyl-serotonin, a FAAH
Harden, R.N., Chialvo, D.R., 2004a. Chronic pain patients are impaired on an inhibitor and TRPV1 receptor antagonist, associated with changes in rostral
emotional decision-making task. Pain 108, 129e136. ventromedial medulla and locus coeruleus cell activity in rats. Neuropharma-
Apkarian, A.V., Sosa, Y., Sonty, S., Levy, R.M., Harden, R.N., Parrish, T.B., cology 55, 1105e1113.
Gitelman, D.R., 2004b. Chronic back pain is associated with decreased pre- de Novellis, V., Vita, D., Gatta, L., Luongo, L., Bellini, G., De Chiaro, M., Marabese, I.,
frontal and thalamic gray matter density. J. Neurosci. 24, 10410e10415. Siniscalco, D., Boccella, S., Piscitelli, F., Di Marzo, V., Palazzo, E., Rossi, F.,
Asmundson, G.J.G., Katz, J., 2009. Understanding the co-occurrence of anxiety dis- Maione, S., 2011. The blockade of the transient receptor potential vanilloid type
orders and chronic pain: state-of-the-art. Depress. Anxiety 26, 888e901. 1 and fatty acid amide hydrolase decreases symptoms and central sequelae in
gaud, B., Bousser, M.G., Cohen, P., Diquet, B., Duprat, P., Janssens, W., Mallaret, M.,
Be the medial prefrontal cortex of neuropathic rats. Mol. Pain 7, 1e19.
Mazue , G., Micalle, J., Monneret, C., Montastruc, J.L., Venance, L., 2016. Report by de Wied, M., Verbaten, M.N., 2001. Affective pictures processing, attention, and pain
the Temporary Specialist Scientific Committee (TSSC), “FAAH (Fatty Acid Amide tolerance. Pain 90, 163e172.
Hydrolase)”, on the Causes of the Accident during a Phase 1 Clinical Trial in Desroches, J., Charron, S., Bouchard, J.-F., Beaulieu, P., 2014. Endocannabinoids
Rennes in January 2016. decrease neuropathic pain-related behavior in mice through the activation of
Berretta, S., Pantazopoulos, H., Caldera, M., Pantazopoulos, P., Pare, D., 2005. one or both peripheral CB1 and CB2 receptors. Neuropharmacology 77,
Infralimbic cortex activation increases c-Fos expression in intercalated neurons 441e452.
of the amygdala. Neuroscience 132, 943e953. Devane, W.A., Dysarz, F.A., Johnson, M.R., Melvin, L.S., Howlett, A.C., 1988. Deter-
Blankman, J.L., Simon, G.M., Cravatt, B.F., 2007. A comprehensive profile of brain mination and characterization of a cannabinoid receptor in rat-brain. Mol.
enzymes that hydrolyze the endocannabinoid 2-arachidonoylglycerol. Chem. Pharmacol. 34, 605e613.
Biol. 14, 1347e1356. Devane, W.A., Hanus, L., Breuer, A., Pertwee, R.G., Stevenson, L.A., Griffin, G.,
Booker, L., Kinsey, S.G., Abdullah, R.A., Blankman, J.L., Long, J.Z., Ezzili, C., Boger, D.L., Gibson, D., Mandelbaum, A., Etinger, A., Mechoulam, R., 1992. Isolation and
Cravatt, B.F., Lichtman, A.H., 2012. The fatty acid amide hydrolase (FAAH) in- structure of a brain constituent that binds to the cannabinoid receptor. Science
hibitor PF-3845 acts in the nervous system to reverse LPS-induced tactile 258, 1946e1949.
allodynia in mice. Br. J. Pharmacol. 165, 2485e2496. Di Marzo, V., 2011. Endocannabinoid signaling in the brain: biosynthetic mecha-
Burke, N.N., Hayes, E., Calpin, P., Kerr, D.M., Moriarty, O., Finn, D.P., Roche, M., 2010. nisms in the limelight. Nat. Neurosci. 14, 9e15.
Enhanced nociceptive responding in two rat models of depression is associated Di Marzo, V., Fontana, A., Cadas, H., Schinelli, S., Cimino, G., Schwartz, J.-C.,
with alterations in monoamine levels in discrete brain regions. Neuroscience Piomelli, D., 1994. Formation and inactivation of endogenous cannabinoid
171, 1300e1313. anandamide in central neurons. Nature 372, 686e691.
Burke, N.N., Finn, D.P., Roche, M., 2015. Neuroinflammatory mechanisms linking Dinh, T.P., Carpenter, D., Leslie, F.M., Freund, T.F., Katona, I., Sensi, S.L., Kathuria, S.,
pain and depression. Mod. Trends Pharmacopsychiatri. 30, 36e50. Piomelli, D., 2002. Brain monoglyceride lipase participating in endocannabinoid
Busquets-Garcia, A., Puighermanal, E., Pastor, A., de la Torre, R., Maldonado, R., inactivation. Proc. Natl. Acad. Sci. U. S. A. 99, 10819e10824.
Ozaita, A. s., 2011. Differential role of anandamide and 2-arachidonoylglycerol in Dinh, T.P., Kathuria, S., Piomelli, D., 2004. RNA interference suggests a primary role
memory and anxiety-like responses. Biol. Psychiatry 70, 479e486. for monoacylglycerol lipase in the degradation of the endocannabinoid 2-
Busti, D., Geracitano, R., Whittle, N., Dalezios, Y., Manko, M., Kaufmann, W., arachidonoylglycerol. Mol. Pharmacol. 66, 1260e1264.
Satzler, K., Singewald, N., Capogna, M., Ferraguti, F., 2011. Different fear states Dougher, M.J., 1979. Sensory decision theory analysis of the effects of anxiety and
engage distinct networks within the intercalated cell clusters of the amygdala. experimental instructions on pain. J. Abnorm Psychol. 88, 137e144.
J. Neurosci. 31, 5131e5144. Egertova, M., Elphick, M.R., 2000. Localisation of cannabinoid receptors in the rat
Butler, R.K., Finn, D.P., 2009. Stress-induced analgesia. Prog. Neurobiol. 88, 184e202. brain using antibodies to the intracellular C-terminal tail of CB1. J. Comp.
Butler, R.K., Rea, K., Lang, Y., Gavin, A.M., Finn, D.P., 2008. Endocannabinoid-medi- Neurol. 422, 159e171.
ated enhancement of fear-conditioned analgesia in rats: opioid receptor de- Egertova, M., Cravatt, B.F., Elphick, M.R., 2003. Comparative analysis of fatty acid
pendency and molecular correlates. Pain 140, 491e500. amide hydrolase and CB1 cannabinoid receptor expression in the mouse brain:
Calignano, A., La Rana, G., Giuffrida, A., Piomelli, D., 1998. Control of pain initiation evidence of a widespread role for fatty acid amide hydrolase in regulation of
by endogenous cannabinoids. Lett. Nat. 394, 277e281. endocannabinoid signaling. Neuroscience 119, 481e496.
Cardoso-Cruz, H., Sousa, M., Vieira, J.B., Lima, D., Galhardo, V., 2013. Prefrontal Finn, D.P., 2010. Endocannabinoid-mediated modulation of stress responses:
cortex and mediodorsal thalamus reduced connectivity is associated with physiological and pathophysiological significance. Immunobiology 215,
spatial working memory impairment in rats with inflammatory pain. Pain 154, 629e646.
2397e2406. Finn, D.P., Beckett, S.R.G., Richardson, D., Kendall, D.A., Marsden, C.A., Chapman, V.,
Carey, L.M., Slivicki, R.A., Leishman, E., Cornett, B., Mackie, K., Bradshaw, H., 2004. Evidence for differential modulation of conditioned aversion and fear-
Hohmann, A.G., 2016. A pro-nociceptive phenotype unmasked in mice lacking conditioned analgesia by CB1 receptors. Eur. J. Neurosci. 20, 848e852.
fatty-acid amide hydrolase. Mol. Pain 12. Fitzgibbon, M., Finn, D.P., Roche, M., 2016. High times for painful blues: the endo-
Carrier, E.J., Patel, S., Hillard, C.J., 2005. Endocannabinoids in neuroimmunology and cannabinoid system in pain-depression comorbidity. Int. J. Neuro-
stress. Curr. Drug Targets CNS Neurol. Disord. 4, 657e665. psychopharmacol. 19.
Chanda, P.K., Gao, Y., Mark, L., Btesh, J., Strassle, B.W., Lu, P., Piesla, M.J., Zhang, M.-Y., Fontanez-Nuin, D.E., Santini, E., Quirk, G.J., Porter, J.T., 2011. Memory for fear
Bingham, B., Uveges, A., Kowal, D., Garbe, D., Kouranova, E.V., Ring, R.H., extinction requires mGluR5-mediated activation of infralimbic neurons. Cereb.
Bates, B., Pangalos, M.N., Kennedy, J.D., Whiteside, G.T., Samad, T.A., 2010. Cortex 21, 727e735.
Monoacylglycerol lipase activity is a critical modulator of the tone and integrity Ford, G.K., Finn, D.P., 2008. Clinical correlates of stress-induced analgesia: evidence
of the endocannabinoid system. Mol. Pharmacol. 78, 996e1003. from pharmacological studies. Pain 140, 3e7.
Chang, C.H., Maren, S., 2010. Strain difference in the effect of infralimbic cortex Ford, G.K., Kieran, S., Dolan, K., Harhen, B., Finn, D.P., 2011. A role for the ventral
lesions on fear extinction in rats. Behav. Neurosci. 124, 391e397. hippocampal endocannabinoid system in fear-conditioned analgesia and fear
Chang, L., Luo, L., Palmer, J.A., Sutton, S., Wilson, S.J., Barbier, A.J., Breitenbucher, J.G., responding in the presence of nociceptive tone in rats. Pain 152, 2495e2504.
S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120 117

Fowler, C.J., 2007. The contribution of cyclooxygenase-2 to endocannabinoid Bussey, T.J., Singewald, N., Holmes, A., 2008. Impaired fear extinction learning
metabolism and action. Br. J. Pharmacol. 152, 594e601. and cortico-amygdala circuit abnormalities in a common genetic mouse strain.
Fowler, C.J., Janson, U., Johnson, R.M., Wahlstro € m, G., Stenstro
€m, A., Norstro €m, Å., J. Neurosci. 28, 8074e8085.
Tiger, G., 1999. Inhibition of anandamide hydrolysis by the enantiomers of Hegyi, Z., Kis, G., Hollo, K., Ledent, C., Antal, M., 2009. Neuronal and glial localization
ibuprofen, ketorolac, and flurbiprofen. Arch. Biochem. Biophys. 362, 191e196. of the cannabinoid-1 receptor in the superficial spinal dorsal horn of the rodent
Fowler, C.J., Naidu, P.S., Lichtman, A., Onnis, V., 2009. The case for the development spinal cord. Eur. J. Neurosci. 30, 251e262.
of novel analgesic agents targeting both fatty acid amide hydrolase and either Herkenham, M., Lynn, A.B., Johnson, M.R., Melvin, L.S., Decosta, B.R., Rice, K.C., 1991.
cyclooxygenase or TRPV1. Br. J. Pharmacol. 156, 412e419. Characterization and localization of cannabinoid receptors in rat-brain - a
Gabbott, P.L., Warner, T.A., Jays, P.R., Salway, P., Busby, S.J., 2005. Prefrontal cortex in quantitative invitro autoradiographic study. J. Neurosci. 11, 563e583.
the rat: projections to subcortical autonomic, motor, and limbic centers. Herkenham, M., Lynn, A.B., Little, M.D., Johnson, M.R., Melvin, L.S., Decosta, B.R.,
J. Comp. Neurol. 492, 145e177. Rice, K.C., 1990. Cannabinoid receptor localization in brain. Proc. Natl. Acad. Sci.
Gamble-George, J.C., Baldi, R., Halladay, L., Kocharian, A., Hartley, N., Silva, C.G., U. S. A. 87, 1932e1936.
Roberts, H., Haymer, A., Marnett, L.J., Holmes, A., Patel, S., 2016. Cyclooxygenase- Hermanson, D.J., Gamble-George, J.C., Marnett, L.J., Patel, S., 2014. Substrate-selec-
2 inhibition reduces stress-induced affective pathology. eLife 5, e14137. tive COX-2 inhibition as a novel strategy for therapeutic endocannabinoid
Gaskin, D.J., Richard, P., 2012. The economic costs of pain in the United States. J. Pain augmentation. Trends Pharmacol. Sci. 35, 358e367.
13, 715e724. Hermanson, D.J., Hartley, N.D., Gamble-George, J., Brown, N., Shonesy, B.C.,
Gavva, N.R., 2009. Setbacks in the clinical development of TRPV1 antagonists: what Kingsley, P.J., Colbran, R.J., Reese, J., Marnett, L.J., Patel, S., 2013. Substrate-se-
next? Open Drug Discov. J. 1, 35. lective COX-2 inhibition decreases anxiety via endocannabinoid activation. Nat.
Gavva, N.R., Treanor, J.J., Garami, A., Fang, L., Surapaneni, S., Akrami, A., Alvarez, F., Neurosci. 16, 1291e1298.
Bak, A., Darling, M., Gore, A., Jang, G.R., Kesslak, J.P., Ni, L., Norman, M.H., Herry, C., Ferraguti, F., Singewald, N., Letzkus, J.J., Ehrlich, I., Luthi, A., 2010. Neuronal
Palluconi, G., Rose, M.J., Salfi, M., Tan, E., Romanovsky, A.A., Banfield, C., circuits of fear extinction. Eur. J. Neurosci. 31, 599e612.
Davar, G., 2008. Pharmacological blockade of the vanilloid receptor TRPV1 Hill, M.N., Kumar, S.A., Filipski, S.B., Iverson, M., Stuhr, K.L., Keith, J.M., Cravatt, B.F.,
elicits marked hyperthermia in humans. Pain 136, 202e210. Hillard, C.J., Chattarji, S., McEwen, B.S., 2013. Disruption of fatty acid amide
Ghilardi, J.R., Rohrich, H., Lindsay, T.H., Sevcik, M.A., Schwei, M.J., Kubota, K., hydrolase activity prevents the effects of chronic stress on anxiety and amyg-
Halvorson, K.G., Poblete, J., Chaplan, S.R., Dubin, A.E., Carruthers, N.I., dalar microstructure. Mol. Psychiatry 18, 1125e1135.
Swanson, D., Kuskowski, M., Flores, C.M., Julius, D., Mantyh, P.W., 2005. Selec- Hill, M.N., Patel, S., Carrier, E.J., Rademacher, D.J., Ormerod, B.K., Hillard, C.J.,
tive blockade of the capsaicin receptor TRPV1 attenuates bone cancer pain. Gorzalka, B.B., 2005. Downregulation of endocannabinoid signaling in the
J. Neurosci. 25, 3126e3131. Hippocampus following chronic unpredictable stress. Neuro-
Ghosh, S., Kinsey, S.G., Liu, Q.-s., Hruba, L., McMahon, L.R., Grim, T.W., Merritt, C.R., psychopharmacology 30, 508e515.
Wise, L.E., Abdulla, R.A., Selley, D.E., Sim-Selley, L., Cravatt, B.F., Lichtman, A.H., Hohmann, A.G., 2002. Spinal and peripheral mechanisms of cannabinoid anti-
2015. Full FAAH inhibition combined with partial monoacylglycerol lipase in- nociception: behavioral, neurophysiological and neuroanatomical perspectives.
hibition: augmented and sustained antinociceptive effects with negligible Chem. Phys. Lipids 121, 173e190.
cannabimimetic side effects in mice. J. Pharmacol. Exp. Ther. 354, 111e120. Hohmann, A.G., Suplita, R.L., Bolton, N.M., Neely, M.H., Fegley, D., Mangieri, R.,
Ghosh, S., Wise, L.E., Chen, Y., Gujjar, R., Mahadevan, A., Cravatt, B.F., Lichtman, A.H., Krey, J.F., Walker, J.M., Holmes, P.V., Crystal, J.D., Duranti, A., Tontini, A., Mor, M.,
2013. The monoacylglycerol lipase inhibitor JZL184 suppresses inflammatory Tarzia, G., Piomelli, D., 2005. An endocannabinoid mechanism for stress-
pain in the mouse carrageenan model. Life Sci. 92, 498e505. induced analgesia. Nature 435, 1108e1112.
Gong, J.P., Onaivi, E.S., Ishiguro, H., Liu, Q.R., Tagliaferro, P.A., Brusco, A., Uhl, G.R., Holden, J.E., Jeong, Y., Forrest, J.M., 2005. The endogenous opioid system and clinical
2006. Cannabinoid CB2 receptors: immunohistochemical localization in rat pain management. AACN Clin. Issues 16, 291e301.
brain. Brain Res. 1071, 10e23. Holt, S., Comelli, F., Costa, B., Fowler, C.J., 2005. Inhibitors of fatty acid amide hy-
Gonsiorek, W., Lunn, C., Fan, X., Narula, S., Lundell, D., Hipkin, R.W., 2000. Endo- drolase reduce carrageenan-induced hind paw inflammation in pentobarbital-
cannabinoid 2-arachidonyl glycerol is a full agonist through human type 2 treated mice: comparison with indomethacin and possible involvement of
cannabinoid receptor: antagonism by anandamide. Mol. Pharmacol. 57, cannabinoid receptors. Br. J. Pharmacol. 146, 467e476.
1045e1050. Hong, S., Fan, J., Kemmerer, E.S., Evans, S., Li, Y., Wiley, J.W., 2009. Reciprocal
Gregg, L.C., Jung, K.-M., Spradley, J.M., Nyilas, R., Suplita, R.L., Zimmer, A., changes in vanilloid (TRPV1) and endocannabinoid (CB1) receptors contribute
Watanabe, M., Mackie, K., Katona, I., Piomelli, D., Hohmann, A.G., 2012. Acti- to visceral hyperalgesia in the water avoidance stressed rat. Gut 58, 202e210.
vation of type 5 metabotropic glutamate receptors and diacylglycerol lipase- Horv ath, E., Woodhams, S.G., Nyilas, R., Henstridge, C.M., Kano, M., Sakimura, K.,
alpha initiates 2-arachidonoylglycerol formation and endocannabinoid- Watanabe, M., Katona, I., 2014. Heterogeneous presynaptic distribution of
mediated analgesia. J. Neurosci. 32, 9457e9468. monoacylglycerol lipase, a multipotent regulator of nociceptive circuits in the
Grim, T.W., Ghosh, S., Hsu, K.-L., Cravatt, B.F., Kinsey, S.G., Lichtman, A.H., 2014. mouse spinal cord. Eur. J. Neurosci. 39, 419e434.
Combined inhibition of FAAH and COX produces enhanced anti-allodynic ef- Howlett, A.C., Reggio, P.H., Childers, S.R., Hampson, R.E., Ulloa, N.M., Deutsch, D.G.,
fects in mouse neuropathic and inflammatory pain models. Pharmacol. Bio- 2011. Endocannabinoid tone versus constitutive activity of cannabinoid re-
chem. Behav. 124, 405e411. ceptors. Br. J. Pharmacol. 163, 1329e1343.
Guindon, G., Hohmann, A., 2009. The endocannabinoid system and pain. CNS Hudson, B.D., He bert, T.E., Kelly, M.E.M., 2010. Ligand- and heterodimer-directed
Neurol. Disord. Drug Targets 8, 18. signaling of the CB1 cannabinoid receptor. Mol. Pharmacol. 77, 1e9.
Guindon, J., Beaulieu, P., 2006. Antihyperalgesic effects of local injections of anan- Huggins, J.P., Smart, T.S., Langman, S., Taylor, L., Young, T., 2012. An efficient rand-
damide, ibuprofen, rofecoxib and their combinations in a model of neuropathic omised, placebo-controlled clinical trial with the irreversible fatty acid amide
pain. Neuropharmacology 50, 814e823. hydrolase-1 inhibitor PF-04457845, which modulates endocannabinoids but
Guindon, J., Beaulieu, P., 2009. The role of the endogenous cannabinoid system in fails to induce effective analgesia in patients with pain due to osteoarthritis of
peripheral analgesia. Curr. Mol. Pharmacol. 2, 5. the knee. Pain 153, 1837e1846.
Guindon, J., Desroches, J., Beaulieu, P., 2007a. The antinociceptive effects of intra- Hunt, S.P., Mantyh, P.W., 2001. The molecular dynamics of pain control. Nat. Rev.
plantar injections of 2-arachidonoyl glycerol are mediated by cannabinoid CB2 Neurosci. 2, 83e91.
receptors. Br. J. Pharmacol. 150, 693e701. Ignatowska-Jankowska, B.M., Baillie, G.L., Kinsey, S., Crowe, M., Ghosh, S.,
Guindon, J., Guijarro, A., Piomelli, D., Hohmann, A.G., 2011. Peripheral anti- Owens, R.A., Damaj, I.M., Poklis, J., Wiley, J.L., Zanda, M., Zanato, C., Greig, I.R.,
nociceptive effects of inhibitors of monoacylglycerol lipase in a rat model of Lichtman, A.H., Ross, R.A., 2015a. A cannabinoid CB1 receptor-positive allosteric
inflammatory pain. Br. J. Pharmacol. 163, 1464e1478. modulator reduces neuropathic pain in the mouse with No psychoactive effects.
Guindon, J., Hohmann, A.G., 2008. Cannabinoid CB2 receptors: a therapeutic target Neuropsychopharmacology 40, 2948e2959.
for the treatment of inflammatory and neuropathic pain. Br. J. Pharmacol. 153, Ignatowska-Jankowska, B.M., Ghosh, S., Crowe, M.S., Kinsey, S.G., Niphakis, M.J.,
319e334. Abdullah, R.A., Tao, Q., O'Neal, S.T., Walentiny, D.M., Wiley, J.L., Cravatt, B.F.,
Guindon, J., Lai, Y., Takacs, S.M., Bradshaw, H.B., Hohmann, A.G., 2013. Alterations in Lichtman, A.H., 2014. In vivo characterization of the highly selective mono-
endocannabinoid tone following chemotherapy-induced peripheral neuropa- acylglycerol lipase inhibitor KML29: antinociceptive activity without cannabi-
thy: effects of endocannabinoid deactivation inhibitors targeting fatty-acid mimetic side effects. Br. J. Pharmacol. 1392e1407.
amide hydrolase and monoacylglycerol lipase in comparison to reference an- Ignatowska-Jankowska, B.M., Wilkerson, J.L., Mustafa, M., Abdullah, R., Niphakis, M.,
algesics following cisplatin treatment. Pharmacol. Res. 67, 94e109. Wiley, J.L., Cravatt, B.F., Lichtman, A.H., 2015b. Selective monoacylglycerol lipase
Guindon, J., LoVerme, J., De Lean, A., Piomelli, D., Beaulieu, P., 2006. Synergistic inhibitors: antinociceptive vs. cannabimimetic effects in mice. J. Pharmacol.
antinociceptive effects of anandamide, an endocannabinoid, and nonsteroidal Exp. Ther. 353, 424e432.
anti-inflammatory drugs in peripheral tissue: a role for endogenous fatty-acid Imperatore, R., Morello, G., Luongo, L., Taschler, U., Romano, R., De Gregorio, D.,
ethanolamides? Eur. J. Pharmacol. 550, 68e77. Belardo, C., Maione, S., Di Marzo, V., Cristino, L., 2015. Genetic deletion of
Guindon, J., Walczak, J.S., Beaulieu, P., 2007b. Recent advances in the pharmaco- monoacylglycerol lipase leads to impaired cannabinoid receptor CB1R signaling
logical management of pain. Drugs 67, 2121e2133. and anxiety-like behavior. J. Neurochem. 135, 799e813.
Gulyas, A.I., Cravatt, B.F., Bracey, M.H., Dinh, T.P., Piomelli, D., Boscia, F., Freund, T.F., Jayamanne, A., Greenwood, R., Mitchell, V.A., Aslan, S., Piomelli, D., Vaughan, C.W.,
2004. Segregation of two endocannabinoid-hydrolyzing enzymes into pre- and 2006. Actions of the FAAH inhibitor URB597 in neuropathic and inflammatory
postsynaptic compartments in the rat hippocampus, cerebellum and amygdala. chronic pain models. Br. J. Pharmacol. 147, 281e288.
Eur. J. Neurosci. 20, 441e458. Jennings, E.M., Okine, B.N., Olango, W.M., Roche, M., Finn, D.P., 2016. Repeated
Hall, W., Solowij, N., 1998. Adverse effects of cannabis. Lancet 352, 1611e1616. forced swim stress differentially affects formalin-evoked nociceptive behaviour
Hefner, K., Whittle, N., Juhasz, J., Norcross, M., Karlsson, R.M., Saksida, L.M., and the endocannabinoid system in stress normo-responsive and stress hyper-
118 S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120

responsive rat strains. Prog. Neuro-Psychopharmacology Biol. Psychiatry 64, evidence for interaction between endocannabinoids and cholecystokinin. Eur. J.
181e189. Neurosci. 27, 8.
Jennings, E.M., Okine, B.N., Roche, M., Finn, D.P., 2014. Stress-induced hyperalgesia. Lafourcade, M., Elezgarai, I., Mato, S., Bakiri, Y., Grandes, P., Manzoni, O.J., 2007.
Prog. Neurobiol. 121, 1e18. Molecular components and functions of the endocannabinoid system in mouse
Jhaveri, M.D., Richardson, D., Kendall, D.A., Barrett, D.A., Chapman, V., 2006. Anal- prefrontal cortex. PLoS One 2, e709.
gesic effects of fatty acid amide hydrolase inhibition in a rat model of neuro- Lambert, D.M., Vandevoorde, S., Jonsson, K.O., Fowler, C.J., 2002. The palmitoyle-
pathic pain. J. Neurosci. 26, 13318e13327. thanolamide family: a new class of anti-inflammatory agents? Curr. Med. Chem.
Jhaveri, M.D., Richardson, D., Robinson, I., Garle, M.J., Patel, A., Sun, Y., Sagar, D.R., 9, 663e674.
Bennett, A.J., Alexander, S.P.H., Kendall, D.A., Barrett, D.A., Chapman, V., 2008. Laprairie, R.B., Kulkarni, P.M., Deschamps, J.R., Kelly, M.E.M., Janero, D.R.,
Inhibition of fatty acid amide hydrolase and cyclooxygenase-2 increases levels Cascio, M.G., Stevenson, L.A., Pertwee, R.G., Kenakin, T.P., Denovan-Wright, E.M.,
of endocannabinoid related molecules and produces analgesia via peroxisome Thakur, G.A., 2017. Enantiospecific allosteric modulation of cannabinoid 1 re-
proliferator-activated receptor-alpha in a model of inflammatory pain. Neuro- ceptor. ACS Chem. Neurosci. https://doi.org/10.1021/acschemneuro.6b00310.
pharmacology 55, 85e93. Latremoliere, A., Woolf, C.J., 2009. Central sensitization: a generator of pain hy-
Ji, G., Neugebauer, V., 2011. Pain-related deactivation of medial prefrontal cortical persensitivity by central neural plasticity. J. Pain 10, 895e926.
neurons involves mGluR1 and GABA(A) receptors. J. Neurophysiol. 106, Lee, S.-H., Ledri, M., To th, B., Marchionni, I., Henstridge, C.M., Dudok, B., Kenesei, K.,
2642e2652. Barna, L., Szabo , S.I., Renkecz, T., Oberoi, M., Watanabe, M., Limoli, C.L.,
Ji, G., Neugebauer, V., 2014. CB1 augments mGluR5 function in medial prefrontal Horvai, G., Soltesz, I., Katona, I., 2015. Multiple forms of endocannabinoid and
cortical neurons to inhibit amygdala hyperactivity in an arthritis pain model. endovanilloid signaling regulate the tonic control of GABA release. J. Neurosci.
Eur. J. Neurosci. 39, 455e466. 35, 10039e10057.
Ji, G., Sun, H., Fu, Y., Li, Z., Pais-Vieira, M., Galhardo, V., Neugebauer, V., 2010. Li, G.L., Winter, H., Arends, R., Jay, G.W., Le, V., Young, T., Huggins, J.P., 2012.
Cognitive impairment in pain through amygdala-driven prefrontal cortical Assessment of the pharmacology and tolerability of PF-04457845, an irre-
deactivation. J. Neurosci. 30, 5451e5464. versible inhibitor of fatty acid amide hydrolase-1, in healthy subjects. Br. J. Clin.
Kaczocha, M., Glaser, S.T., Deutsch, D.G., 2009. Identification of intracellular carriers Pharmacol. 73, 706e716.
for the endocannabinoid anandamide. Proc. Natl. Acad. Sci. 106, 6375e6380. Lichtman, A.H., Chapman, V., 2011. A FAAH-fetched approach to treat osteoarthritis
Kaczocha, M., Glaser, S.T., Maher, T., Clavin, B., Hamilton, J., O'Rourke, J., pain. Pain 152, 959e960.
Rebecchi, M., Puopolo, M., Owada, Y., Thanos, P.K., 2015. Fatty acid binding Lichtman, A.H., Martin, B.R., 2005. Cannabinoid tolerance and dependence. Handb.
protein deletion suppresses inflammatory pain through endocannabinoid/N- Exp. Pharmacol. 691e717.
acylethanolamine-dependent mechanisms. Mol. Pain 11, 52. , D., 2005. Prefrontal control of the amygdala.
Likhtik, E., Pelletier, J.G., Paz, R., Pare
Kaczocha, M., Rebecchi, M.J., Ralph, B.P., Teng, Y.H., Berger, W.T., Galbavy, W., J. Neurosci. 25, 7429e7437.
Elmes, M.W., Glaser, S.T., Wang, L., Rizzo, R.C., Deutsch, D.G., Ojima, I., 2014. Lin, H.C., Mao, S.C., Su, C.L., Gean, P.W., 2009. The role of prefrontal cortex CB1 re-
Inhibition of Fatty Acid binding proteins elevates brain anandamide levels and ceptors in the modulation of fear memory. Cereb. Cortex 19, 165e175.
produces analgesia. PLoS One 9, e94200. Little, J.P., Carter, A.G., 2013. Synaptic mechanisms underlying strong reciprocal
Kano, M., Ohno-Shosaku, T., Hashimotodani, Y., Uchigashima, M., Watanabe, M., connectivity between the medial prefrontal cortex and basolateral amygdala.
2009. Endocannabinoid-mediated control of synaptic transmission. Physiol. J. Neurosci. 33, 15333e15342.
Rev. 89, 309e380. Loeser, J.D., Treede, R.-D., 2008. The kyoto protocol of IASP basic pain terminology.
Karbarz, M.J., Luo, L., Chang, L., Tham, C.S., Palmer, J.A., Wilson, S.J., Pain 137, 473e477.
Wennerholm, M.L., Brown, S.M., Scott, B.P., Apodaca, R.L., Keith, J.M., Wu, J.J., Lomazzo, E., Bindila, L., Remmers, F., Lerner, R., Schwitter, C., Hoheisel, U., Lutz, B.,
Breitenbucher, J.G., Chaplan, S.R., Webb, M., 2009. Biochemical and biological 2015. Therapeutic potential of inhibitors of endocannabinoid degradation for
properties of 4-(3-phenyl-[1,2,4]thiadiazol-5-yl)-piperazine-1-carboxylic acid the treatment of stress-related hyperalgesia in an animal model of chronic pain.
phenylamide, a mechanism-based inhibitor of fatty acid amide hydrolase. Neuropsychopharmacology 40, 488e501.
Anesth. Analg. 108, 316e329. Long, J.Z., Li, W.W., Booker, L., Burston, J.J., Kinsey, S.G., Schlosburg, J.E., Pavon, F.J.,
Kathuria, S., Gaetani, S., Fegley, D., Valino, F., Duranti, A., Tontini, A., Mor, M., Serrano, A.M., Selley, D.E., Parsons, L.H., Lichtman, A.H., Cravatt, B.F., 2009a.
Tarzia, G., La Rana, G., Calignano, A., Giustino, A., Tattoli, M., Palmery, M., Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid
Cuomo, V., Piomelli, D., 2003. Modulation of anxiety through blockade of behavioral effects. Nat. Chem. Biol. 5, 37e44.
anandamide hydrolysis. Nat. Med. 9, 76e81. Long, J.Z., Nomura, D.K., Vann, R.E., Walentiny, D.M., Booker, L., Jin, X., Burston, J.J.,
Katona, I., Freund, T., 2012. Multiple functions of endocannabinoid signaling in the Sim-Selley, L.J., Lichtman, A.H., Wiley, J.L., Cravatt, B.F., 2009b. Dual blockade of
brain. Annu. Rev. Neurosci. 35, 529e558. FAAH and MAGL identifies behavioral processes regulated by endocannabinoid
Katona, I., Freund, T.F., 2008. Endocannabinoid signaling as a synaptic circuit crosstalk in vivo. Proc. Natl. Acad. Sci. U. S. A. 106, 20270e20275.
breaker in neurological disease. Nat. Med. 14, 923e930. LoVerme, J., Russo, R., La Rana, G., Fu, J., Farthing, J., Mattace-Raso, G., Meli, R.,
Kaur, R., Sidhu, P., Singh, S., 2016. What failed BIA 10e2474 Phase I clinical trial? Hohmann, A., Calignano, A., Piomelli, D., 2006. Rapid broad-spectrum analgesia
Global speculations and recommendations for future Phase I trials. J. Pharmacol. through activation of peroxisome proliferator-activated receptor-alpha.
Pharmacother. 7, 120e126. J. Pharmacol. Exp. Ther. 319, 1051e1061.
Khasabova, I.A., Chandiramani, A., Harding-Rose, C., Simone, D.A., Seybold, V.S., Lovinger, D.M., 2008. Presynaptic modulation by endocannabinoids. Handb. Exp.
2011. Increasing 2-arachidonoyl glycerol signaling in the periphery attenuates Pharmacol. 435e477.
mechanical hyperalgesia in a model of bone cancer pain. Pharmacol. Res. 64, Madasu, M.K., Okine, B.N., Olango, W.M., Rea, K., Lenihan, R., Roche, M., Finn, D.P.,
60e67. 2016. Genotype-dependent responsivity to inflammatory pain: a role for TRPV1
Khurana, L., Mackie, K., Piomelli, D., Kendall, D.A., 2017. Modulation of CB1 canna- in the periaqueductal grey. Pharmacol. Res. 113 (Part A), 44e54.
binoid receptor by allosteric ligands: pharmacology and therapeutic opportu- Maione, S., Bisogno, T., de Novellis, V., Palazzo, E., Cristino, L., Valenti, M.,
nities. Neuropharmacology. https://doi.org/10.1016/j.neuropharm.2017.05.018. Petrosino, S., Guglielmotti, V., Rossi, F., Di Marzo, V., 2006. Elevation of endo-
Kim, S.C., Jo, Y.S., Kim, I.H., Kim, H., Choi, J.S., 2010. Lack of medial prefrontal cortex cannabinoid levels in the ventrolateral periaqueductal grey through inhibition
activation underlies the immediate extinction deficit. J. Neurosci. 30, 832e837. of fatty acid amide hydrolase affects descending nociceptive pathways via both
King, A.R., Duranti, A., Tontini, A., Rivara, S., Rosengarth, A., Clapper, J.R., Astarita, G., cannabinoid receptor type 1 and transient receptor potential vanilloid type-1
Geaga, J.A., Luecke, H., Mor, M., Tarzia, G., Piomelli, D., 2007. URB602 inhibits receptors. J. Pharmacol. Exp. Ther. 316, 969e982.
monoacylglycerol lipase and selectively blocks 2-arachidonoylglycerol degra- Maione, S., Costa, B., Piscitelli, F., Morera, E., De Chiaro, M., Comelli, F., Boccella, S.,
dation in intact brain slices. Chem. Biol. 14, 1357e1365. Guida, F., Verde, R., Ortar, G., Di Marzo, V., 2013. Piperazinyl carbamate fatty acid
Kinsey, S.G., Long, J.Z., Cravatt, B.F., Lichtman, A.H., 2010. Fatty acid amide hydrolase amide hydrolase inhibitors and transient receptor potential channel modulators
and monoacylglycerol lipase inhibitors produce anti-allodynic effects in mice as “dual-target” analgesics. Pharmacol. Res. 76, 98e105.
through distinct cannabinoid receptor mechanisms. J. Pain 11, 1420e1428. Maione, S., De Petrocellis, L., de Novellis, V., Moriello, A.S., Petrosino, S., Palazzo, E.,
Kinsey, S.G., Long, J.Z., O'Neal, S.T., Abdullah, R.A., Poklis, J.L., Boger, D.L., Cravatt, B.F., Rossi, F.S., Woodward, D.F., Di Marzo, V., 2007. Analgesic actions of N-arach-
Lichtman, A.H., 2009. Blockade of endocannabinoid-degrading enzymes at- idonoyl-serotonin, a fatty acid amide hydrolase inhibitor with antagonistic
tenuates neuropathic pain. J. Pharmacol. Exp. Ther. 330, 902e910. activity at vanilloid TRPV1 receptors. Br. J. Pharmacol. 150, 766e781.
Kinsey, S.G., Nomura, D.K., O'Neal, S.T., Long, J.Z., Mahadevan, A., Cravatt, B.F., Malek, N., Kostrzewa, M., Makuch, W., Pajak, A., Kucharczyk, M., Piscitelli, F.,
Grider, J.R., Lichtman, A.H., 2011. Inhibition of monoacylglycerol lipase attenu- Przewlocka, B., Di Marzo, V., Starowicz, K., 2016. The multiplicity of spinal AA-5-
ates nonsteroidal anti-inflammatory drug-induced gastric hemorrhages in HT anti-nociceptive action in a rat model of neuropathic pain. Pharmacol. Res.
mice. J. Pharmacol. Exp. Ther. 338, 795e802. 111, 251e263.
Kiritoshi, T., Ji, G., Neugebauer, V., 2016. Rescue of impaired mGluR5-driven endo- Malek, N., Mrugala, M., Makuch, W., Kolosowska, N., Przewlocka, B., Binkowski, M.,
cannabinoid signaling restores prefrontal cortical output to inhibit pain in Czaja, M., Morera, E., Di Marzo, V., Starowicz, K., 2015. A multi-target approach
arthritic rats. J. Neurosci. 36, 837e850. for pain treatment - dual inhibition of fatty acid amide hydrolase and TRPV1 in
Kiritoshi, T., Sun, H., Ren, W., Stauffer, S.R., Lindsley, C.W., Conn, P.J., Neugebauer, V., a rat model of osteoarthritis. Pain 156, 890e903.
2013. Modulation of pyramidal cell output in the medial prefrontal cortex by Marek, G.J., Zhang, C., 2008. Activation of metabotropic glutamate 5 (mGlu5) re-
mGluR5 interacting with CB1. Neuropharmacology 66, 170e178. ceptors induces spontaneous excitatory synaptic currents in layer V pyramidal
Kolodny, A., Courtwright, D.T., Hwang, C.S., Kreiner, P., Eadie, J.L., Clark, T.W., cells of the rat prefrontal cortex. Neurosci. Lett. 442, 239e243.
Alexander, G.C., 2015. The prescription opioid and heroin crisis: a public health Marek, R., Strobel, C., Bredy, T.W., Sah, P., 2013. The amygdala and medial prefrontal
approach to an epidemic of addiction. Annu. Rev. public health 36, 559e574. cortex: partners in the fear circuit. J. Physiol. 591, 2381e2391.
Kurrikoff, k., Inno, J., Matsui, T., Vasar, E., 2008. Stress-induced analgesia in mice: Marrs, W.R., Blankman, J.L., Horne, E.A., Thomazeau, A., Lin, Y.H., Coy, J., Bodor, A.L.,
S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120 119

Muccioli, G.G., Hu, S.S.-J., Woodruff, G., Fung, S., Lafourcade, M., Alexander, J.P., tone. Br. J. Pharmacol. 165, 2549e2560.
Long, J.Z., Li, W., Xu, C., Moller, T., Mackie, K., Manzoni, O.J., Cravatt, B.F., Orsini, C.A., Maren, S., 2012. Neural and cellular mechanisms of fear and extinction
Stella, N., 2010. The serine hydrolase ABHD6 controls the accumulation and memory formation. Neurosci. Biobehav Rev. 36, 1773e1802.
efficacy of 2-AG at cannabinoid receptors. Nat. Neurosci. 13, 951e957. Pais-Vieira, M., Mendes-Pinto, M.M., Lima, D., Galhardo, V., 2009. Cognitive
Marsicano, G., Lutz, B., 1999. Expression of the cannabinoid receptor CB1 in distinct impairment of prefrontal-dependent decision-making in rats after the onset of
neuronal subpopulations in the adult mouse forebrain. Eur. J. Neurosci. 11, chronic pain. Neuroscience 161, 671e679.
4213e4225. Pamplona, F.A., Ferreira, J., Menezes de Lima, O., Duarte, F.S., Bento, A.F., Forner, S.,
Marsicano, G., Wotjak, C.T., Azad, S.C., Bisogno, T., Rammes, G., Cascio, M.G., Villarinho, J.G., Bellocchio, L., Wotjak, C.T., Lerner, R., Monory, K., Lutz, B.,
Hermann, H., Tang, J., Hofmann, C., Zieglgansberger, W., Di Marzo, V., Lutz, B., Canetti, C., Matias, I., Calixto, J.B., Marsicano, G., Guimar~ aes, M.Z.P.,
2002. The endogenous cannabinoid system controls extinction of aversive Takahashi, R.N., 2012. Anti-inflammatory lipoxin A4 is an endogenous allosteric
memories. Nature 418, 530e534. enhancer of CB1 cannabinoid receptor. Proc. Natl. Acad. Sci. 109, 21134e21139.
Martin, W.J., Coffin, P.O., Attias, E., Balinsky, M., Tsou, K., Walker, J.M., 1999. Pape, H.C., Pare, D., 2010. Plastic synaptic networks of the amygdala for the
Anatomical basis for cannabinoid-induced antinociception as revealed by acquisition, expression, and extinction of conditioned fear. Physiol. Rev. 90,
intracerebral microinjections. Brain Res. 822, 237e242. 419e463.
Mayer, E.A., Berman, S., Suyenobu, B., Labus, J., Mandelkern, M.A., Naliboff, B.D., Patel, S., Cravatt, B.F., Hillard, C.J., 2005. Synergistic interactions between cannabi-
Chang, L., 2005. Differences in brain responses to visceral pain between patients noids and environmental stress in the activation of the central amygdala.
with irritable bowel syndrome and ulcerative colitis. Pain 115, 398e409. Neuropsychopharmacology 30, 497e507.
McDonald, A.J., 1998. Cortical pathways to the mammalian amygdala. Prog. Neu- Pattij, T., Wiskerke, J., Schoffelmeer, A.N.M., 2008. Cannabinoid modulation of ex-
robiol. 55, 257e332. ecutive functions. Eur. J. Pharmacol. 585, 458e463.
Meagher, M.W., Arnau, R.C., Rhudy, J.L., 2001. Pain and emotion: effects of affective Pawsey, S., Wood, M., Browne, H., Donaldson, K., Christie, M., Warrington, S., 2016.
picture modulation. Psychosom. Med. 63, 79e90. Safety, tolerability and pharmacokinetics of FAAH inhibitor V158866: a double-
Mechoulam, R., Gaoni, Y., 1967. The absolute configuration of delta-1- blind, randomised, placebo-controlled Phase I study in healthy volunteers.
tetrahydrocannabinol, the major active constituent of hashish. Tetrahedron Drugs R&D 16, 181e191.
Lett. 12, 1109e1111. Pernia-Andrade, A.J., Kato, A., Witschi, R., Nyilas, R., Katona, I., Freund, T.F.,
Metz, A.E., Yau, H.J., Centeno, M.V., Apkarian, A.V., Martina, M., 2009. Morphological Watanabe, M., Filitz, J., Koppert, W., Schuttler, J., Ji, G., Neugebauer, V.,
and functional reorganization of rat medial prefrontal cortex in neuropathic Marsicano, G., Lutz, B., Vanegas, H., Zeilhofer, H.U., 2009. Spinal endocannabi-
pain. Proc. Natl. Acad. Sci. U. S. A. 106, 2423e2428. noids and CB1 receptors mediate C-Fiber-Induced heterosynaptic pain sensiti-
Millan, M.J., 1999. The induction of pain: an integrative review. Prog. Neurobiol. 57, zation. Science 325, 760e764.
1e164. Pinard, C.R., Mascagni, F., McDonald, A.J., 2012. Medial prefrontal cortical innerva-
Moore, N., 2016. Lessons from the fatal French study BIA-10-2474. BMJ 353. tion of the intercalated nuclear region of the amygdala. Neuroscience 205,
Morena, M., Patel, S., Bains, J.S., Hill, M.N., 2015. Neurobiological interactions be- 112e124.
tween stress and the endocannabinoid system. Neuropsychopharmacology 41, Price, M.R., Baillie, G.L., Thomas, A., Stevenson, L.A., Easson, M., Goodwin, R.,
80e102. McLean, A., McIntosh, L., Goodwin, G., Walker, G., Westwood, P., Marrs, J.,
Moriarty, O., McGuire, B.E., Finn, D.P., 2011. The effect of pain on cognitive function: Thomson, F., Cowley, P., Christopoulos, A., Pertwee, R.G., Ross, R.A., 2005.
a review of clinical and preclinical research. Prog. Neurobiol. 93, 385e404. Allosteric modulation of the cannabinoid CB1 receptor. Mol. Pharmacol. 68,
Muly, E.C., Maddox, M., Smith, Y., 2003. Distribution of mGluR1alpha and mGluR5 1484e1495.
immunolabeling in primate prefrontal cortex. J. Comp. Neurol. 467, 521e535. Rademacher, D.J., Meier, S.E., Shi, L., Vanessa Ho, W.S., Jarrahian, A., Hillard, C.J.,
Munro, S., Thomas, K.L., Abushaar, M., 1993. Molecular characterization of a pe- 2008. Effects of acute and repeated restraint stress on endocannabinoid content
ripheral receptor for cannabinoids. Nature 365, 61e65. in the amygdala, ventral striatum, and medial prefrontal cortex in mice.
Nahin, R.L., 2015. Estimates of pain prevalence and severity in adults: United States, Neuropharmacology 54, 108e116.
2012. J. Pain 16, 769e780. Rea, K., Olango, W.M., Harhen, B., Kerr, D.M., Galligan, R., Fitzgerald, S., Moore, M.,
Navia-Paldanius, D., Aaltonen, N., Lehtonen, M., Savinainen, J.R., Taschler, U., Roche, M., Finn, D.P., 2013. Evidence for a role of GABAergic and glutamatergic
Radner, F.P.W., Zimmermann, R., Laitinen, J.T., 2015. Increased tonic cannabinoid signalling in the basolateral amygdala in endocannabinoid-mediated fear-
CB1R activity and brain region-specific desensitization of CB1R Gi/o signaling conditioned analgesia in rats. Pain 154, 576e585.
axis in mice with global genetic knockout of monoacylglycerol lipase. Eur. J. Rea, K., Olango, W.M., Okine, B.N., Madasu, M.K., McGuire, I.C., Coyle, K., Harhen, B.,
Pharm. Sci. 77, 180e188. Roche, M., Finn, D.P., 2014. Impaired endocannabinoid signalling in the rostral
Neugebauer, V., 2015. Amygdala pain mechanisms. Handb. Exp. Pharmacol. 227, ventromedial medulla underpins genotype-dependent hyper-responsivity to
261e284. noxious stimuli. Pain 155, 69e79.
Neugebauer, V., Han, J.S., Adwanikar, H., Fu, Y., Ji, G., 2007. Techniques for assessing Ren, W., Kiritoshi, T., Gregoire, S., Ji, G., Guerrini, R., Calo, G., Neugebauer, V., 2013.
knee joint pain in arthritis. Mol. Pain 3, 8e8. Neuropeptide S: a novel regulator of pain-related amygdala plasticity and be-
Niphakis, M.J., Cognetta, A.B., Chang, J.W., Buczynski, M.W., Parsons, L.H., Byrne, F., haviors. J. Neurophysiol. 110, 1765e1781.
Burston, J.J., Chapman, V., Cravatt, B.F., 2013a. Evaluation of NHS carbamates as a Rhudy, J.L., Meagher, M.W., 2000. Fear and anxiety: divergent effects on human pain
potent and selective class of endocannabinoid hydrolase inhibitors. ACS Chem. thresholds. Pain 84, 65e75.
Neurosci. http://dx.doi.org/10.1021/cn400116z. Rhudy, J.L., Meagher, M.W., 2001. Noise stress and human pain thresholds: diver-
Niphakis, M.J., Johnson, D.S., Ballard, T.E., Stiff, C., Cravatt, B.F., 2013b. O-hydrox- gent effects in men and women. J. Pain 2, 57e64.
yacetamide carbamates as a highly potent and selective class of endocannabi- Rhudy, J.L., Meagher, M.W., 2003. Individual differences in the emotional reaction to
noid hydrolase inhibitors. ACS Chem. Neurosci. 3, 418e426. shock determine whether hypoalgesia is observed. Pain Med. 4, 244e256.
Nyilas, R., Dudok, B., Urban, G.M., Mackie, K., Watanabe, M., Cravatt, B.F., Freund, T.F., Richardson, J.D., Aanonsen, L., Hargreaves, K.M., 1998. Antihyperalgesic effects of
Katona, I., 2008. Enzymatic machinery for endocannabinoid biosynthesis spinal cannabinoids. Eur. J. Pharmacol. 345, 145e153.
associated with calcium stores in glutamatergic axon terminals. J. Neurosci. 28, Rivera, P., Arrabal, S., Cifuentes, M., Grondona, J.M., Perez-Martin, M., Rubio, L.,
1058e1063. Vargas, A., Serrano, A., Pavon, F.J., Suarez, J., Rodriguez de Fonseca, F., 2014.
Nyilas, R., Gregg, L.C., Mackie, K., Watanabe, M., Zimmer, A., Hohmann, A.G., Localization of the cannabinoid CB1 receptor and the 2-AG synthesizing
Katona, I., 2009. Molecular architecture of endocannabinoid signaling at noci- (DAGLalpha) and degrading (MAGL, FAAH) enzymes in cells expressing the
ceptive synapses mediating analgesia. Eur. J. Neurosci. 29, 1964e1978. Ca(2þ)-binding proteins calbindin, calretinin, and parvalbumin in the adult rat
O'Mahony, C.M., Clarke, G., Gibney, S., Dinan, T.G., Cryan, J.F., 2011. Strain differences hippocampus. Front. Neuroanat. 8, 56.
in the neurochemical response to chronic restraint stress in the rat: relevance to Robbe, D., Kopf, M., Remaury, A., Bockaert, J., Manzoni, O.J., 2002. Endogenous
depression. Pharmacol. Biochem. Behav. 97, 690e699. cannabinoids mediate long-term synaptic depression in the nucleus accum-
O'Sullivan, S.E., 2007. Cannabinoids go nuclear: evidence for activation of peroxi- bens. Proc. Natl. Acad. Sci. U. S. A. 99, 8384e8388.
some proliferator-activated receptors. Br. J. Pharmacol. 152, 576e582. Roche, M., Johnston, P., Mhuircheartaigh, O.N., Olango, W.M., Mackie, K., Finn, D.P.,
O'Sullivan, S.E., 2016. An update on PPAR activation by cannabinoids. Br. J. Phar- 2010. Effects of intra-basolateral amygdala administration of rimonabant on
macol. 173, 1899e1910. nociceptive behaviour and neuronal activity in the presence or absence of
Okine, B.N., Gaspar, J.C., Madasu, M.K., Olango, W.M., Harhen, B., Roche, M., contextual fear. Eur. J. Pain 14, 487e495.
Finn, D.P., 2017. Characterisation of peroxisome proliferator-activated receptor Roche, M., O'Connor, E., Diskin, C., Finn, D.P., 2007. The effect of CB1 receptor
signalling in the midbrain periaqueductal grey of rats genetically prone to antagonism in the right basolateral amygdala on conditioned fear and associ-
heightened stress, negative affect and hyperalgesia. Brain Res. 1657, 185e192. ated analgesia in rats. Eur. J. Neurosci. 26, 2643e2653.
Okine, B.N., Norris, L.M., Woodhams, S., Burston, J., Patel, A., Alexander, S.P., Rodriguez, A.L., Grier, M.D., Jones, C.K., Herman, E.J., Kane, A.S., Smith, R.L.,
Barrett, D.A., Kendall, D.A., Bennett, A.J., Chapman, V., 2012. Lack of effect of Williams, R., Zhou, Y., Marlo, J.E., Days, E.L., Blatt, T.N., Jadhav, S., Menon, U.N.,
chronic pre-treatment with the FAAH inhibitor URB597 on inflammatory pain Vinson, P.N., Rook, J.M., Stauffer, S.R., Niswender, C.M., Lindsley, C.W.,
behaviour: evidence for plastic changes in the endocannabinoid system. Br. J. Weaver, C.D., Conn, P.J., 2010. Discovery of novel allosteric modulators of
Pharmacol. 167, 627e640. metabotropic glutamate receptor subtype 5 reveals chemical and functional
Olango, W.M., Finn, D.P., 2014. Neurobiology of stress-induced hyperalgesia. Curr. diversity and in vivo activity in rat behavioral models of anxiolytic and anti-
Top. Behav. Neurosci. 20, 251e280. psychotic activity. Mol. Pharmacol. 78, 1105e1123.
Olango, W.M., Roche, M., Ford, G.K., Harhen, B., Finn, D.P., 2012. The endocannabi- Ross, R.A., 2003. Anandamide and vanilloid TRPV1 receptors. Br. J. Pharmacol. 140,
noid system in the rat dorsolateral periaqueductal grey mediates fear-condi- 790e801.
tioned analgesia and controls fear expression in the presence of nocicpetive Rubino, T., Zamberletti, E., Parolaro, D., 2015. Endocannabinoids and mental
120 S.G. Woodhams et al. / Neuropharmacology 124 (2017) 105e120

disorders. In: Pertwee, R.G. (Ed.), Endocannabinoids. Springer International medulla. Neuropharmacology 49, 1201e1209.
Publishing, Cham, pp. 261e283. Suplita, R.L., Gutierrez, T., Fegley, D., Piomelli, D., Hohmann, A.G., 2006. Endo-
Russo, R., LoVerme, J., La Rana, G., Compton, T.R., Parrott, J.A., Duranti, A., Tontini, A., cannabinoids at the spinal level regulate, but do not mediate, nonopioid stress-
Mor, M., Tarzia, G., Calignano, A., Piomelli, D., 2007. The fatty acid amide hy- induced analgesia. Neuropharmacology 50, 372e379.
drolase inhibitor URB597 (cyclohexylcarbamic acid 3'-Carbamoylbiphenyl-3-yl Swanson, D.M., Dubin, A.E., Shah, C., Nasser, N., Chang, L., Dax, S.L., Jetter, M.,
ester) reduces neuropathic pain after oral administration in mice. Breitenbucher, J.G., Liu, C., Mazur, C., Lord, B., Gonzales, L., Hoey, K., Rizzolio, M.,
J. Pharmacol. Exp. Ther. 322, 236e242. Bogenstaetter, M., Codd, E.E., Lee, D.H., Zhang, S.-P., Chaplan, S.R.,
Sagar, D., Gaw, A.G., Okine, B., Woodhams, S., Wong, A., Kendall, D., Chapman, V., Carruthers, N.I., 2004. Identification and biological evaluation of 4-(3-
2009. Dynamic regulation of the endocannabinoid system: implications for Trifluoromethylpyridin-2-yl)piperazine-1-carboxylic acid (5-
analgesia. Mol. Pain 5, 59. trifluoromethylpyridin-2-yl)amide, a high affinity TRPV1 (VR1) vanilloid re-
Sagar, D.R., Burston, J.J., Woodhams, S.G., Chapman, V., 2012. Dynamic changes to ceptor antagonist. J. Med. Chem. 48, 1857e1872.
the endocannabinoid system in models of chronic pain. Philos. Trans. R. Soc. B Tappe-Theodor, A., Agarwal, N., Katona, I., Rubino, T., Martini, L., Swiercz, J.,
Biol. Sci. 367, 3300e3311. Mackie, K., Monyer, H., Parolaro, D., Whistler, J., Kuner, T., Kuner, R., 2007.
Sagar, D.R., Jhaveri, M., Richardson, D., Gray, R.A., De Lago, E., Fernandez-Ruiz, J., A molecular basis of analgesic tolerance to cannabinoids. J. Neurosci. 27,
Barrett, D., Kendall, D., Chapman, V., 2010. Endocannabinoid regulation of spinal 4165e4177.
nociceptive processing in a model of neuropathic pain. Eur. J. Neurosci. 31, 8. Tsou, K., Brown, S., Sanudo-Pena, M.C., Mackie, K., Walker, J.M., 1998. Immunohis-
Sagar, D.R., Kendall, D.A., Chapman, V., 2008. Inhibition of fatty acid amide hydro- tochemical distribution of cannabinoid CB1 receptors in the rat central nervous
lase produces PPAR-alpha-mediated analgesia in a rat model of inflammatory system. Neuroscience 83, 393e411.
pain. Br. J. Pharmacol. Valverde, O., Ledent, C., Beslot, F., Parmentier, M., Roques, B.P., 2000. Reduction of
Sasso, O., Migliore, M., Habrant, D., Armirotti, A., Albani, C., Summa, M., Moreno- stress-induced analgesia but not of exogenous opioid effects in mice lacking
Sanz, G., Scarpelli, R., Piomelli, D., 2015a. Multitarget fatty acid amide hydrolase/ CB1 receptors. Eur. J. Neurosci. 12, 533e539.
cyclooxygenase blockade suppresses intestinal inflammation and protects Vandevoorde, S., Jonsson, K.O., Labar, G., Persson, E., Lambert, D.M., Fowler, C.J.,
against nonsteroidal anti-inflammatory drug-dependent gastrointestinal dam- 2007. Lack of selectivity of URB602 for 2-oleoylglycerol compared to ananda-
age. FASEB J. 29, 2616e2627. mide hydrolysis in vitro. Br. J. Pharmacol. 150, 186e191.
Sasso, O., Wagner, K., Morisseau, C., Inceoglu, B., Hammock, B.D., Piomelli, D., 2015b. Vardeh, D., Mannion, R.J., Woolf, C.J., 2016. Toward a mechanism-based approach to
Peripheral FAAH and soluble epoxide hydrolase inhibitors are synergistically pain diagnosis. J. Pain 17, T50eT69.
antinociceptive. Pharmacol. Res. 97, 7e15. Vardeh, D., Wang, D., Costigan, M., Lazarus, M., Saper, C.B., Woolf, C.J.,
Schlosburg, J., Carlson, B., Ramesh, D., Abdullah, R., Long, J., Cravatt, B., Lichtman, A., Fitzgerald, G.A., Samad, T.A., 2009. COX2 in CNS neural cells mediates me-
2009. Inhibitors of endocannabinoid-metabolizing enzymes reduce precipi- chanical inflammatory pain hypersensitivity in mice. J. Clin. Investig. 119,
tated withdrawal responses in THC-dependent mice. AAPS J. 11, 342e352. 287e294.
Schlosburg, J.E., Blankman, J.L., Long, J.Z., Nomura, D.K., Pan, B., Kinsey, S.G., Walker, J.M., Hohmann, A.G., 2005. Cannabinoid mechanisms of pain suppression.
Nguyen, P.T., Ramesh, D., Booker, L., Burston, J.J., Thomas, E.A., Selley, D.E., Sim- Handb. Exp. Pharmacol. 509e554.
Selley, L.J., Liu, Q.-s., Lichtman, A.H., Cravatt, B.F., 2010. Chronic mono- Walker, J.M., Huang, S.M., Strangman, N.M., Tsou, K., Sanudo-Pena, M.C., 1999. Pain
acylglycerol lipase blockade causes functional antagonism of the endocanna- modulation by release of the endogenous cannabinoid anandamide. Proc. Natl.
binoid system. Nat. Neurosci. 13, 1113e1119. Acad. Sci. U. S. A. 96, 12198e12203.
Seidel, K., Hamza, M., Ates, M., Gühring, H., 2003. Flurbiprofen inhibits capsaicin Walker, M., Huang, S., 2002. Cannabinoid analgesia. Pharmacol. Ther. 95, 127e135.
induced calcitonin gene related peptide release from rat spinal cord via an Wang, G.Q., Cen, C., Li, C., Cao, S., Wang, N., Zhou, Z., Liu, X.M., Xu, Y., Tian, N.X.,
endocannabinoid dependent mechanism. Neurosci. Lett. 338, 99e102. Zhang, Y., Wang, J., Wang, L.P., Wang, Y., 2015. Deactivation of excitatory neu-
Senn, V., Wolff, S.B., Herry, C., Grenier, F., Ehrlich, I., Grundemann, J., Fadok, J.P., rons in the prelimbic cortex via Cdk5 promotes pain sensation and anxiety. Nat.
Muller, C., Letzkus, J.J., Luthi, A., 2014. Long-range connectivity defines behav- Commun. 6, 7660.
ioral specificity of amygdala neurons. Neuron 81, 428e437. Wedzony, K., Chocyk, A., 2009. Cannabinoid CB1 receptors in rat medial prefrontal
Sepulveda-Orengo, M.T., Lopez, A.V., Soler-Cedeno, O., Porter, J.T., 2013. Fear cortex are colocalized with calbindin- but not parvalbumin- and calretinin-
extinction induces mGluR5-mediated synaptic and intrinsic plasticity in infra- positive GABA-ergic neurons. Pharmacol. Rep. 61, 1000e1007.
limbic neurons. J. Neurosci. 33, 7184e7193. Wiech, K., Tracey, I., 2009. The influence of negative emotions on pain: behavioral
Shen, L., Yang, X.J., Qian, W., Hou, X.H., 2010. The role of peripheral cannabinoid effects and neural mechanisms. NeuroImage 47, 987e994.
receptors type 1 in rats with visceral hypersensitivity induced by chronic re- Wilkerson, J.L., Ghosh, S., Mustafa, M., Abdullah, R.A., Niphakis, M.J., Cabrera, R.,
straint stress. J. Neurogastroenterol. Motil. 16, 281e290. Maldonado, R.L., Cravatt, B.F., Lichtman, A.H., 2016a. The endocannabinoid hy-
Shi, M., Qi, W.J., Gao, G., Wang, J.Y., Luo, F., 2010. Increased thermal and mechanical drolysis inhibitor SA-57: intrinsic antinociceptive effects, augmented
nociceptive thresholds in rats with depressive-like behaviors. Brain Res. 1353, morphine-induced antinociception, and attenuated heroin seeking behavior in
225e233. mice. Neuropharmacology 114, 156e167.
Sierra-Mercado, D., Padilla-Coreano, N., Quirk, G.J., 2011. Dissociable roles of pre- Wilkerson, J.L., Niphakis, M.J., Grim, T.W., Mustafa, M.A., Abdullah, R.A., Poklis, J.L.,
limbic and infralimbic cortices, ventral hippocampus, and basolateral amygdala Dewey, W.L., Akbarali, H.I., Banks, M.L., Wise, L.E., Cravatt, B.F., Lichtman, A.H.,
in the expression and extinction of conditioned fear. Neuro- 2016b. The selective monoacylglycerol lipase inhibitor MJN110 produces opioid
psychopharmacology 36, 529e538. sparing effects in a mouse neuropathic pain model. J. Pharmacol. Exp. Ther. 357,
Slivicki, R., Xu, Z., Kulkarni, P.M., Pertwee, R.G., Mackie, K., Thakur, G.A., Hohmann, 145e156.
A.G. Positive allosteric modulation of CB1 suppresses pathological pain without Wilson, A. W., Clayton, N. M., Medhurst, S. J., Bountra, C., Chessell, I. P., 2005. The
producing tolerance or dependence. Biol. Psychiatr., (in press). FAAH inhibitor URB597 reverses inflammatory pain through a CB1 receptor
Sotres-Bayon, F., Quirk, G.J., 2010. Prefrontal control of fear: more than just mediated mechanism. British Pharmacological Society Winter Meeting,
extinction. Curr. Opin. Neurobiol. 20, 231e235. Newcastle.
Starowicz, K., Di Marzo, V., 2013. Non-psychotropic analgesic drugs from the Woodhams, S.G., Sagar, D.R., Burston, J.J., Chapman, V., 2015. The role of the
endocannabinoid system: “Magic bullet” or “multiple-target” strategies? Eur. J. endocannabinoid system in pain. Handb. Exp. Pharmacol. 227, 119e143.
Pharmacol. 716, 41e53. Woodhams, S.G., Wong, A., Barrett, D.A., Bennett, A.J., Chapman, V.,
Stella, N., Schweitzer, P., Piomelli, D., 1997. A second endogenous cannabinoid that Alexander, S.P.H., 2012. Spinal administration of the monoacylglycerol lipase
modulates long-term potentiation. Nature 388, 773e778. inhibitor JZL184 produces robust inhibitory effects on nociceptive processing
Sun, H., Neugebauer, V., 2011. mGluR1, but not mGluR5, activates feed-forward and the development of central sensitization in the rat. Br. J. Pharmacol. 167,
inhibition in the medial prefrontal cortex to impair decision making. 1609e1619.
J. Neurophysiol. 106, 960e973. Xu, J., Zhu, Y., Contractor, A., Heinemann, S.F., 2009. mGluR5 has a critical role in
Suplita, R.L., Eisenstein, S.A., Neely, M.H., Moise, A.M., Hohmann, A.G., 2008. Cross- inhibitory learning. J. Neurosci. 29, 3676e3684.
sensitization and cross-tolerance between exogenous cannabinoid anti- Zeilhofer, H.U., 2010. Spinal cannabinoidsea double-edged sword? (Commentary
nociception and endocannabinoid-mediated stress-induced analgesia. Neuro- on Zhang et al.). Eur. J. Neurosci. 31, 223e224.
pharmacology 54, 161e171. Zhang, Z., Gadotti, V.M., Chen, L., Souza, I.A., Stemkowski, P.L., Zamponi, G.W., 2015.
Suplita, R.L., Farthing, J.N., Gutierrez, T., Hohmann, A.G., 2005. Inhibition of fatty- Role of prelimbic GABAergic circuits in sensory and emotional aspects of
acid amide hydrolase enhances cannabinoid stress-induced analgesia: sites of neuropathic pain. Cell Rep. 12, 752e759.
action in the dorsolateral periaqueductal gray and rostral ventromedial

You might also like