Department of Neural and Behavioral Sciences

154
The Pennsylvania State University The Graduate School Department of Neural and Behavioral Sciences FUNCTIONAL INTERACTIONS BETWEEN OPIOIDS AND A CANNABINOID RECEPTOR 2 AGONIST IN INFLAMMATORY PAIN. A Dissertation in Neuroscience by Matthew B. Yuill 2018 Matthew B. Yuill Submitted in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy May 2018

Transcript of Department of Neural and Behavioral Sciences

The Pennsylvania State University

The Graduate School

Department of Neural and Behavioral Sciences

FUNCTIONAL INTERACTIONS BETWEEN OPIOIDS AND A CANNABINOID

RECEPTOR 2 AGONIST IN INFLAMMATORY PAIN.

A Dissertation in

Neuroscience by

Matthew B. Yuill

2018 Matthew B. Yuill

Submitted in Partial Fulfillment of the Requirements

for the Degree of

Doctor of Philosophy

May 2018

The dissertation of Matthew B. Yuill was reviewed and approved* by the following:

Daniel Morgan Professor of Neuroscience Dissertation Adviser Chair of Committee

Patricia S. Grigson Professor of Neural and Behavioral Sciences

Robert Levenson Distinguished Professor of Pharmacology

John Ellis Professor of Psychiatry and Pharmacology

Jossee Guindon Assistant Professor of Pharmacology Neuroscience Texas Tech University Health Sciences Center Special Member Colin J. Barnstable Chair of Department of Neural and Behavioral Sciences

*Signatures are on file in the Graduate School

iii

ABSTRACT

The goal of this study was to test the hypothesis that the Cannabinoid 2

Receptor (CB2R) functionally interacts with the opioid system to modulate

inflammatory pain. Additionally, we tested mechanisms mediating tolerance to

multiple opioids, and to the prototypical cannabinoid Δ9-THC. CB2R agonists

produce low levels of side effects and no tolerance relative to other opioid and

cannabinoid agonists, making them an attractive pharmacotherapeutic target.

This study assessed the anti-nociceptive effects of a selective CB2R agonist

(JWH-133) in pathological pain using mice subjected to inflammatory pain using

the formalin test. Furthermore, we examined several ways in which JWH-133

may interact with the activity of opioids in this model.

JWH-133 produces dose-dependent anti-nociception during both the

acute pain and inflammatory pain phases of the formalin test. This was observed

in both male and female mice. However, a maximally efficacious dose of JWH-

133 (1 mg/kg) was not associated with somatic withdrawal symptoms, motor

impairment, or hypothermia. The efficacy of JWH-133 was blocked by application

of a CB2R selective antagonist (SR144528).

After eleven once-daily injections of 1 mg/kg JWH-133, no tolerance was

observed in the formalin test. Conversely, wild-type mice become tolerant to Δ9-

THC, morphine, and fentanyl within eleven days. Cross-tolerance for the anti-

nociceptive effects of JWH-133 and morphine were assessed to gain insight into

physiologically relevant CB2R and Mu opioid receptor (MOR) interaction. Mice

iv

made tolerant to the effects of morphine exhibited a lower JWH-133 response in

both phases of the formalin test compared to vehicle treated morphine-naïve

animals. However, repeated daily JWH-133 administration did not cause cross-

tolerance for morphine. Similar results were found for cross-tolerance between

JWH-133 and fentanyl, suggesting opioid and CB2R cross-tolerance is

unidirectional in this model. However, preliminary data suggests co-

administration of JWH-133 with morphine modestly attenuates morphine

tolerance in the formalin model. Furthermore, isobolographic analysis revealed

that co-administration of a fixed-ratio combination of JWH-133 and morphine has

an additive effect on anti-nociception in the formalin test. Overall these findings

show that CB2R may functionally interact with MOR to modulate anti-nociception

and tolerance in inflammatory pain, which suggests clinical utility.

v

TABLE OF CONTENTS

List of Figures .......................................................................................................... vii

List of Tables ........................................................................................................... viii

List of Common Abbreviations ................................................................................. ix

Chapter 1 - Introduction and literature review................................................................... 1

Pain .................................................................................................................................. 1 Anatomical pain circuitry....................................................................................... 1 Pathological pain ................................................................................................... 3

The endogenous opioid system .................................................................................. 4 Overview ................................................................................................................. 4 Mu Opioid Receptor and pain .............................................................................. 5 Clinical use of opioid drugs .................................................................................. 6

The endogenous cannabinoid system ....................................................................... 7 Overview ................................................................................................................. 7 Cannabinoid Receptor 1 signaling and localization ......................................... 9 Cannabinoid Receptor 2 signaling and localization ......................................... 10 Cannabinoids in pain............................................................................................. 12 Cannabinoid Receptor 2 in pain .......................................................................... 14 Clinical use of Cannabinoids ............................................................................... 16 Interactions between the cannabinoid system and opioid system ................. 18 Cannabinoid Receptor 2 and opioid interactions. ............................................. 20

Chapter 2 - General methods .............................................................................................. 22

Subjects .......................................................................................................................... 22 Drugs ............................................................................................................................... 22 Procedures ..................................................................................................................... 23

Tail-flick and hotplate anti-nociception ............................................................... 23 Formalin test ........................................................................................................... 24

Physical side-effects ..................................................................................................... 25 Body temperature .................................................................................................. 25 Rotarod test ............................................................................................................ 26 Precipitated Withdrawal ........................................................................................ 26

Data analysis .................................................................................................................. 27 Isobolographic analysis ........................................................................................ 28

Chapter 3 ................................................................................................................................ 30

Tolerance to the anti-nociceptive and hypothermic effects of morphine are mediated by multiple isoforms of c-Jun N-terminal Kinase ............................. 30

Chapter 4 ................................................................................................................................ 46

vi

Anti-nociceptive interactions between opioids and a cannabinoid receptor 2 agonist in inflammatory pain. ............................................................................... 46

Unpublished Experiment 1: Efficacy of JWH-133 in hotplate and tail-flick assays ..................................................................................................................... 88 Rationale ................................................................................................................. 88 Procedure ............................................................................................................... 89 Results .................................................................................................................... 89 Discussion .............................................................................................................. 91

Unpublished Experiment 2: JWH-133 and fentanyl cross-tolerance. .................... 92 Rationale ................................................................................................................. 92 Procedure ............................................................................................................... 92 Results .................................................................................................................... 93 Discussion .............................................................................................................. 95

Chapter 5 – Mechanisms of Cannabinoid Tolerance through the CB1 Receptor ....... 98

Results .................................................................................................................... 99

Chapter 6 - General Discussion and Conclusion ............................................................. 102

Future directions .................................................................................................... 109 Conclusion .............................................................................................................. 110

Appendix ................................................................................................................................. 112

Supplementary Data and Figures ............................................................................... 112

Works Cited ............................................................................................................................ 114

vii

LIST OF FIGURES

Figure 1. Cannabinoid Pain Circuitry. ................................................................................ 14

Figure 2. Tolerance to morphine in the tail-flick test. ....................................................... 39

Figure 3. Tolerance to morphine in the hotplate test. ...................................................... 40

Figure 4. Tolerance to morphine-induced hypothermia. ................................................. 42

Figure 5. Anti-nociceptive efficacy of JWH-133. ............................................................... 61

Figure 6. Comparison of morphine and JWH-133 in the formalin test. ......................... 62

Figure 7. JWH-133 acts through the CB2 Receptor. ....................................................... 63

Figure 8. Lack of JWH-113 adverse effects. ..................................................................... 68

Figure 9. Lack of observed tolerance to JWH-133. ......................................................... 70

Figure 10. Cross-tolerance between JWH-133 and morphine. ...................................... 73

Figure 11. JWH-133 co-administration modestly protects against morphine tolerance. ........................................................................................................................ 76

Figure 12. JNK signaling is partially responsible for morphine-induced cross-tolerance to JWH-133 ................................................................................................... 78

Figure 13. Non-linear isobolographic analysis of 1:10 fixed ratio JWH-133 and morphine in the formalin test. ...................................................................................... 80

Figure 14. JWH-133 dose responses in acute thermal pain .......................................... 90

Figure 15. JWH-133 and fentanyl cross tolerance. .......................................................... 94

Figure 16. Tolerance to the anti-nociceptive effects of fentanyl is not blocked by SP6. ................................................................................................................................. 97

Figure 17. Δ9-THC tolerance in the formalin test. ............................................................ 100

Figure 18. Δ9-THC and SP6 in the formalin test. .............................................................. 101

Figure 19. Morphine with a fixed-dose of JWH-133 in the formalin test. ...................... 112

viii

LIST OF TABLES

Table 1. Opioid Receptors ................................................................................................... 5

Table 2. CB2R-agonist induced anti-nociception in various pain models. ................... 16

Table 3. ED50 Values in Formalin. ...................................................................................... 113

Table 4. Paw edema following formalin. ............................................................................ 113

ix

List of Common Abbreviations

GPCR G-protein coupled receptor

2-AG 2-arachidonylglycerol

CB1R Cannabinoid Receptor 1

CB2R Cannabinoid Receptor 2

CNS central nervous system

eCB endocannabinoid

GRK G protein-coupled receptor kinase

JNK c-Jun N-terminal kinase

MAPK mitogen-activated protein kinase

MOR mu-opioid receptor

KOR Kappa opioid receptor

DOR Delta opioid receptor

PAG Periaqueductal gray

VTA Ventral tegmental area

CPP Conditioned Place Preference

SR2 SR-144,528

SP6 SP600125

i.p. intraperitoneal

%MPE Percent of Maximum possible effect

AUC Area under the curve

SEM Standard error of the mean

x

CNS Central Nervous System

RVM rostral ventral medulla

NGF Nerve Growth Factor

cAMP cyclic adenosine monophosphate

AEA N-arachidonoylethanolamide

2-AG 2-arachidonylglycerol

MAPK mitogen-activated protein kinase

PKA protein kinase A

PEA palmitoylethanolamide

GRK G-protein coupled receptor kinase

JNK c-Jun N-terminal Kinase

CPS Composite Pain Score

1

Chapter 1 - Introduction and literature review

Pain

Pain can be broadly characterized as an unpleasant sensation resulting

from intense or damaging stimuli [Basbaum 2009]. The propagation of pain is

initiated with the activation of physiological receptors, called nociceptors.

Normally, this painful sensation results from specific activation of the nociceptors

by mechanical, thermal, or chemical stimulus and is short-lived. However,

chronic or persistent pain in the absence of injury is a serious clinical challenge.

Pathologically, the modality of pain encompasses more than a simple physical

sensation. Pain can better be defined as a combination of sensory, cognitive, and

emotional aspects associated with real or potential injuries. Knowing that pain

represents a complex sensory modality accompanied by affective, motivational

and cognitive aspects highlights some of the challenges related to its treatment.

Anatomical pain circuitry

Pain is initiated primarily by stimulation of nociceptors, specialized sensory

receptors widely distributed throughout the periphery. There are multiple classes

of nociceptors which detect specific stimulus modalities, such as thermal,

mechanical, and chemical stimuli. Activated nociceptors then signal through

peripheral afferent fibers which terminate in the spinal cord. These first-order

2

afferent fibers are categorized according to structure, diameter, and conduction

velocity. C-type fibers are unmyelinated, under 2 μm in diameter and have a

conduction velocity of 0.5–2.0 m/s [Beasou 1969; Almeida 2004]. Aσ fibers are

lightly myelinated, are 2-6 μm and have a conduction velocity of 12–30

m/s[Burgess 1967]. The myelinated Aβ fibers have a diameter of more than 10

μm and a velocity of 30–100 m/s [Perl 1968].

After information from nociceptors reaches the dorsal horn of the spinal

cord, pain is signaled by the release of glutamate from primary afferent fibers,

generating excitatory post-synaptic currents (EPSCs) to second order dorsal

horn neurons. Second order neurons transmit signals among multiple ascending

pathways to different areas of the central nervous system (CNS) depending on

the type of information [Wall 1967; Fields 2004]. The spinothalamic tract

transmits information to the somatosensory cortex via the thalamus, providing

information about the location and intensity of the painful stimulus [Halliday

1972]. Other projection neurons along the spinomesencephalic tract engage the

cingulate and insular cortices via the parabrachial nucleus and amygdala,

contributing to the affective component of pain [Bernard 1994]. In addition to pain

signals ascending from the spine, signals engage neurons of the Rostral Ventral

Medulla (RVM) and Periaqueductal Gray (PAG) to engage descending feedback

tracts to regulate the output from the spinal cord and provide endogenous pain

control [Wang 1990].

3

Pathological pain

Pathological pain states can result from direct nerve injury (neuropathic

pain), or from persistent stimulation of nociceptors due to injury and

inflammation. In addition to activation of nociceptors, inflammatory processes

also sensitize nociceptors to generate pain a response to milder stimuli

(hyperalgesia). Peripheral sensitization results from the numerous chemical

changes that accompany inflammation. Bradykinins, prostaglandins, and Nerve

Growth Factor (NGF) are among the inflammatory molecules released which

can directly bind to and sensitize nociceptors [Raja 1984; Coderre TJ 1997]. In

animal models of inflammation, primary afferent fibers are made more sensitive,

and normally silent mechanoreceptors are activated due to release of

inflammatory mediators (prostaglandins, histamine, and others) from mast cells

[Di Rosa 1971; Friedman 1990; Leon 1994].

Chronic pain is one of the most pervasive clinical challenges facing

medicine today, afflicting over an estimated 100 million people in the U.S. alone

[Gaskin 2012]. As life expectancy and survivability of conditions like cancer and

HIV increase, the prevalence of chronic pain is expected to steadily increase.

Chronic pain is comorbid for serious conditions such as depression, anxiety, and

suicidal ideation [Braden 2008]. As a result, chronic pain costs the U.S. an

estimated $635 billion, annually [Gaskin and Richard 2012]. Currently available

agents (antidepressants, anticonvulsants, opioids and nonsteroidal anti-

inflammatory drugs) are either unable to completely mitigate the symptoms or, as

4

will be described below, carry significant adverse effects in doing so [Lynch

2015]. There is a critical need for new treatments.

Opioids are extensively used in treatment of both acute and chronic pain

[CDC 2011]. While drugs in the opioid class have remarkable anti-nociceptive

efficacy, there are severe adverse consequences of prolonged use. High doses

of opioid drugs result in a rapid development of tolerance, and carry a high

potential for physical dependence and use disorder. This has generated great

interest in investigating the mechanisms of opioid drug action, tolerance, and

dependence.

The endogenous opioid system

Overview

The endogenous opioid system contains three putative receptor subtypes

(mu, delta, and kappa) and three major groups of endogenous opioid peptides

(endorphins, enkephalins, and dynorphins) [Corbett 2006]. Opioid receptors

belong to the superfamily of seven transmembrane receptors and produce their

cellular effects via coupling with Gi/Go GTP-binding proteins [Ueda 1988; Wong

1988] [Waldhoer 2004]. The primary pathway involves stimulation of inwardly

rectifying potassium conductance, inhibition of adenylyl cyclase and cyclic

adenosine monophosphate. Opioid receptor activation also inactivates voltage

gated calcium channels. The net of these signals is the reduction of

5

neurotransmitter release. Activation of the opioid receptor subtypes produces

similar cellular responses, so the differences in physiological effects result

primarily from different anatomical distribution of the receptor subtypes, and

different ligand affinities (Table 1 ; [Waldhoer 2004]).

Table 1. Opioid Receptors

Receptor subtype Location Function

Highest endogenous ligand affinity

Hippocampus, PAG, cerebral cortex, dorsal horn, thalamus, primary afferent terminals

Analgesia, euphoria, miosis, constipation, respiratory depression

-endorphin

Dorsal horn, hypothalamus, PAG, NRM

Analgesia, sedation, hallucinations, dysphoria

Dynorphin

Olfactory system, dorsal horn, primary afferent terminals, limbic nuclei

Analgesia, constipation

Methionine, Enkephalin

Mu Opioid Receptor and pain

The Mu opioid receptor (MOR) is highly concentrated in the dorsal horn of

the spinal cord, and is expressed in all major supraspinal regions that modulate

pain, including the periaqueductal gray (PAG), the rostral ventromedial medulla

(RVM), amygdala and hypothalamus. MOR agonists modulate pain at both the

6

spinal and supraspinal level. In the spinal cord, Opioids regulate nociceptive

transmission both post-synaptically by hyperpolarization of dorsal horn neurons

(potassium) and by binding to presynaptic sensory neurons to inhibit

neurotransmitter release.[Wall 1967; Corbett 2006]. Among supraspinal pain

circuitry, MOR is located pre-synaptically on GABAergic interneurons. Thus,

activation of MOR leads to disinhibition, and increased descending inhibition of

pain pathways. This was first demonstrated in the RVM of rats, which have

distinct neuronal populations that are directly inhibited by opioid agonists [Pan

1990]. Similarly, analgesia resulting from direct electrical stimulation of the PAG

is blocked by naloxone [Akil 1976].

Clinical use of opioid drugs

The majority of opioid drugs in clinical use specifically target MOR as this

produces the strongest analgesic effect, in addition to significant side effects;

euphoria, constipation, respiratory depression and others. [Yaksh 1985].

Activation of MOR also causes euphoria, which plays a role in the high addictive

liability of opiates [Koob 2006a; Savage 2009]. Opioid dependence is

compounded by the development of tolerance, which can cause an escalation of

dose and development of dependence [Williams 2013]. The rate of prescription

opioid overdose has more than tripled since the 1990s, and is still on the rise

[CDC 2011]. Due to the diminishing efficacy of repeated opioid use, and the

resulting prevalence and severity of opioid drug dependence, there is great

7

interest in alternatives. As such, some attempts have been made to target other

opioid receptors.

Kappa opioid receptors (KOR) are activated by endogenous dynorphins.

Systemic KOR agonists also produce robust analgesia [Kolesnikov 1996].

However, activation of KOR negatively modulates mood and is aversive

[Wadenberg 2003]. The negative effects of Kappa agonists have limited their

clinical utility [Williams 2013].

Delta opioid receptors (DOR) are activated by endogenous enkephalins.

Generally, activation of DOR produces minimal analgesia. However, DOR

appear to be up-regulated in rodent models of chronic pain, where they may

become slightly effective. [Holdridge 2007; Kabli 2007].

The endogenous cannabinoid system

Overview

Increasing interest in the potential therapeutic value of cannabis has given

rise to a growing number of states legalizing cannabis for medical use. The

increasing public interest is accompanied by recent studies demonstrating that

states allowing use of medical cannabis show lower than predicated rates of

opiate overdose [Bachhuber 2014; Hayes 2014].

The endocannabinoid (eCB) system is a neuromodulatory system

comprised of two receptors, (CB1R and CB2R) ligands for those receptors, and

8

enzymes involved in the synthesis and degradation of those components. The.

The eCB system is of increasing interest medically, as it has been shown to play

a role in a wide variety of neuronal and immunological processes including:

analgesia, memory, neurogenesis, appetite, metabolism, stress/anxiety,

thermoregulation sleep and immune cell function [Howlett 2004].

The drugs targeting the eCB system can be divided in to several primary

categories; exogenous compounds from the cannabis plant (phytocannabinoids),

synthetic cannabinoids, and endogenous cannabinoids (endocannabinoids). The

major psychoactive component of marijuana, D9-tetrahydrocannabinol (Δ9-THC),

was isolated more than 50 years ago [Gaoni 1964]. The Major endocannabinoids

were not identified until decades later. The two major endocannabinoids,

anandamide (N-arachidonoylethanolamide: AEA, [Devane 1992]) and 2-

arachidonylglycerol (2-AG, [Mechoulam 1995]) are the most extensively studied.

Despite interest in cannabinoid drugs, our understanding of how they function

remains under investigation. In particular, the receptors upon which these

cannabinoids act are of significant interest.

Both CB1R and CB2R are 7-transmembrane GPCRs that couple primarily

to the pertussis (PTX) toxin sensitive Gi/Go subfamily of proteins to modulate a

variety of similar signaling pathways [Howlett 1986; Howlett 2004]. Human CB1R

and CB2R share 44% amino acid sequence identity throughout the total protein,

and differ in terms of localization and levels of expression in the body [Munro

1993]. This warrants that they are examined individually.

9

Cannabinoid Receptor 1 signaling and localization

CB1Rs are located primarily on presynaptic terminals both of

glutamatergic and GABAergic neurons in the CNS [Katona 1999]. Activation of

CB1R is occurs in response to increased neuronal activity, and leads to the

inhibition of neurotransmitter release. Activation of synaptic CB1R by

endogenous and exogenous agonists triggers a canonical G-protein pathway and

inhibits neurotransmitter release directly, via inhibitory coupling to voltage-

dependent calcium channels [Howlett 1989; Mackie 1992];[Sullivan 1999], or

through activation of potassium channels, which shortens action potential

duration and lessens the amount of neurotransmitter released per action

potential. CB1R can also act through secondary messenger systems, and

activate intracellular mediators such as mitogen-activated protein kinase (MAPK;

[Korzh 2008]).

CB1R is the most abundantly expressed GPCR in the CNS, and exerts a

wide variety of effects according to location. [Howlett 2004; Gong 2006]. The

high density of CB1R in hippocampus impacts memory [Herkenham 1991a].

Control over motor function results from the high occurrence of CB1R in the

basal ganglia [Herkenham 1991b], and loss of CB1R is associated with

Parkinson’s Disease [Sañudo-Peña 1998]. The presence of CB1R in cerebellum

further explains the motor effects of cannabinoids [Matsuda 1990]. Metabolism

and food intake are impacted by expression of hypothalamic CB1R, which

interacts with neuropeptides controlling energetic homeostasis and lipogenesis

10

[Cota 2003]. CB1R also impacts reward and motivational circuits, and is present

both on glutamatergic and GABAergic neurons in the Ventral Tegmental Area

(VTA) and Nucleus accumbens (NAc). This is further supported by behavior of

the genes encoding cannabinoid receptors. Polymorphisms of the CNR1 gene

encoding CB1R are correlated with increased dependence of multiple drugs of

abuse [Lopez-Moreno 2012]. Moreover, administration of morphine, cocaine, or

ethanol increases CB1R mRNA expression in limbic and striatal regions

[Gonzalez S 2002].

Cannabinoid Receptor 2 signaling and localization

CB2R is usually coupled to a pertussis toxin-sensitive Gi/Go protein that

triggers the same canonical signaling pathway as CB1R; with inhibition of

adenylyl cyclase activity leading to reduced cAMP levels and lower activation of

protein kinase A (PKA). CB2R also initiates secondary signaling pathways

through protein kinase B and β-arrestin. Unlike CB1R, CB2R appears to poorly

modulate calcium channels or inwardly rectifying potassium channels [Felder

1995; McAllister 1999]. However, because CB2R is principally located on

immune cells, the end results differ significantly.

The CB2 receptor was first cloned and discovered in 1993 and was

reported to be expressed in macrophages and to a lesser extent in the spleen

[Munro 1993]. The expression profile of the CB2 receptor is currently well

established. The ubiquitous presence of the CB2R in immune cells was initially

11

reported in 1995, and has since been extensively characterized [Galiegue 1995].

CB2R expression in the immune cells follows: B-cells > natural killer cells >>

monocytes > neutrophil cells > T8 cells > T4 cells. [Derocq 2000; Carlisle 2002].

As evidenced by CB2R’s ubiquity, it plays a significant role in immune

modulation. CB2R deficient mice lose all immune modulation in response to

cannabinoids [Buckley 2000]. Moreover, both the mRNA and CB2R protein levels

in immune cells correlate with the level of cellular activity. Many believe that

CB2R is activated in response to immune conditions to direct cell activity towards

appropriate response [Carayon 1998]. Furthermore, the ability of the

endocannabinoid 2-AG to induce immune cell migration is blocked by a CB2R-

selective antagonist [Jorda 2002; Tanikawa 2007].

Despite being initially described as an immune cell cannabinoid receptor,

CB2R has been identified in numerous other peripheral cell types. CB2R has

been found in pulmonary endothelial cells [Zoratti 2003], and can also be found

in bone (in osteocytes, osteoblasts and osteoclasts) where it controls bone

formation [Ofek 2006]. The gastrointestinal system also contains CB2R [Storr

2002; Duncan 2008].

The role of the CB2R “peripheral cannabinoid receptor” within the central

nervous system has been largely overlooked due to the belief that it was not

expressed in the CNS [Munro 1993; Atwood 2010]. However, recent discoveries

have suggested that expression of CB2R in the CNS is highly inducible under

pathological conditions. Increase in the expression of CB2 receptors in microglia

and astrocytes occurs in animal models of pain [Beltramo 2006], inflammation,

12

chronic constriction injury. [Brownjohn 2012; JC 2102], ischemia-induced hypoxia

[Ashton 2007], Alzheimer’s disease , and multiple sclerosis [Yiangou 2006].

CB2R activation has been shown to modulate multiple aspects of

neuroinflammation, including the attenuation of pro-inflammatory factors in

microglia and astrocytes [Stella 2004; Maresz 2005]. Moreover, it has been

shown that a selective CB2 agonist, JWH-015 modulates glial marker expression

[Ehrhart 2005].

Far more controversial is the assertions some have made that CB2R is

also expressed neuronally in several brain regions, including the cerebellum

[Ashton 2006], brainstem [Van Sickle 2005], PAG, thalamus, striatum, cortex,

amygdala and hippocampus [Gong 2006]. The exact nature of expression and

distribution of CB2R in the CNS is still a matter of some controversy due to

questions about antibody specificity [Baek 2013], choice of PCR probes, lack of

controls [Marchalant 2014], and species differences [Liu 2009].

Cannabinoids in pain

Systemic administration of nonselective cannabinoid agonists produces

Anti-nociception in animal models of acute and tonic pain [Pertwee 2001]

For example, the nonselective cannabinoid agonist CP55940 is anti-nociceptive

in the tail-flick test [Pugh 1997] and WIN55212-2 inhibits inflammation-induced

nociceptive behavior [Martin 1999b].

13

In order to determine where cannabinoids may produce their anti-

nociceptive effects, the first experiments involved stereotaxic administration of

cannabinoid agonists into specific regions along rat pain circuits. The regions

(see Figure 1) that produced anti-nociception included the Periaqueductal Gray

(PAG; [Lichtman 1996] ), the Rostral Ventromedial Medulla (RVM; [Martin 1998]

), and the lateral posterior nuclei of the thalamus [Martin 1999a]. It should also be

noted that this anti-nociceptive effect is ablated by administration of the CB1R-

selective antagonist, SR141716A [Lichtman 1997]. However, in preclinical rodent

models, repeated administration of cannabinoids can cause tolerance to effects

such as anti-nociception, hypothermia, and catalepsy [Nguyen 2012].

Additionally, the development of dependence is observed to Δ9-THC [Tsou

1995].

14

Figure 1. Cannabinoid Pain Circuitry.

Regions in rat pain circuitry in which injection of a cannabinoid agonist resulted in thermal anti-nocieption. Triangles indicate injections that caused the response. (PAG: Periaqueductal gray), (RVM: Rostral Ventromedial Medulla), (DH: Dorsal Horn), (+: excitatory, -: inhibitory).

Cannabinoid Receptor 2 in pain

Traditionally, the anti-nociceptive effects of Δ9-THC were thought to be a

result exclusively of CB1R activation. However, there has been increased

interest in finding alternatives to CB1R, due to adverse effects. CB2R agonists

are known to generate fewer of the psychotomimetic and sedative effects seen

15

from CB1R agonists [Fernandez-Ruiz 2009], while potentially providing

peripheral anti-nociception [Malan TP Jr 2001]. The anti-nociceptive potential of

CB2 receptor agonists was first indicated by studies using the endogenous fatty

acid derivative palmitoylethanolamide (PEA). PEA was demonstrated to have

marked anti-inflammatory, as well as anti-nociceptive effects when administered

in vivo. [Facci 1995; Calignano 1998] Although PEA is not an agonist at CB1 or

CB2 receptors, its anti-nociceptive effects were blocked by the CB2 receptor

selective antagonist, SR144528, which implicates an indirect role of CB2

receptors in the effects of PEA, or the involvement of a CB2-like receptor.

Following this, it was also observed that, mice lacking CB1R still

demonstrate Δ9-THC -induced anti-nociception in acute thermal pain tests

[Zimmer 1999]. Furthermore, using three different assays of nociception, it was

demonstrated that CB2R knockout mice have a lower anti-nociceptive response

to cannabinoids relative to wild-type mice [Ibrahim 2006]. While this alone does

not prove CB2R activity, it is intriguing. This information, in addition to the

development of a number of CB2R selective agonists has encouraged numerous

labs to investigate (see Table 2). While studies such as these show promise

individually, there remains significant discord in the field as a whole. Results

often cannot be replicated across species, different agonists, or pain modalities.

Furthermore, there are no putative mechanistic explanations for the anti-

nociceptive effects in many cases.

16

Table 2. CB2R-agonist induced anti-nociception in various pain models.

CB2R agonist Pain Model Result Reference

HU308 Formalin 2nd phase anti-nociception

[Hanus L 1999]

HU308 Post-operative pain Anti-allodynia [LaBuda 2005]

JWH133 Sciatic nerve ligation Anti-allodynia [Yamamoto 2008]

A796260 Post-operative pain Anti-allodynia [Yao 2008]

A796260 Chronic constriction injury

Anti-allodynia Yao 2008]

GW842166X Chronic constriction injury

Anti-allodynia [Clayton 2004]

AM-1241 Formalin inflammation Anti-nociception [Quartilho 2003]

AM-1241 λ-carrageenan inflammation

Anti-nociception [Nackley 2003]

AM-1241 Thermal Paw withdrawal

Anti-nociception [Malan TP Jr 2001]

GW405833 Complete Freund’s adjuvant inflammation

Anti-nociception [Whiteside 2005]

A-836339 Complete Freund’s adjuvant inflammation

Anti-nociception [Nackley 2003]

Clinical use of Cannabinoids

Preparations of the hemp plant (Cannabis sativa) have been used for the

treatment of pain for more than 4,000 years. However, there has been a

reluctance to use cannabis-based products over the past century [Jhaveri 2007].

However renewed interest in their development began in the 1960s when the

most psychoactive component of cannabis extract, Δ9-tetrahydrocannabinol (Δ9-

THC), was isolated and partially synthesized [Gaoni 1964].

17

The increased academic interest has been complemented by the growing

number of states legalizing the Cannabis sativa plant for both medical and

recreational use. For example, the drug Sativex (50% Δ9-THC and 50%

cannabidiol), has been approved in Canada and several other countries for the

treatment of neuropathic pain and cancer pain [Pacher 2013]. Δ9-

tetrahydrocannabinol (Δ9-THC;dronabinol; marinol) and its synthetic analogue,

Nabilone (Cesamet), were FDA approved over 25 years ago as medicines for

suppressing nausea and vomiting produced by chemotherapy. Subsequently, the

use of dronabinol was approved as an appetite stimulant for example in AIDS

[Pertwee 2012]. However, the FDA has not approved and cannabinoid drug for

treatment of pain. While showing potential, current attempts to target the eCB

system for pharmacotherapy have yielded inconsistent and often unexpected

results, usually resulting from the intensity of psychotropic side effects [Tanda

2003; Pacher 2006; Pertwee 2012; Pacher and Kunos 2013]. A recent

systematic review and meta-analysis of cannabinoids for medical use found that

among 28 clinical trials, cannabis was associated with greater pain reduction but

also significant side effects relative to placebo [Whiting 2015]. This highlights the

current need to more fully explore the nature of cannabinoid drugs and how they

may interact with current clinical approaches to develop more viable approaches.

In the case of pain management, this means opioids.

18

Interactions between the cannabinoid system and opioid system

It has been established that cannabinoids and endocannabinoids enhance

the anti-nociceptive effects of opiates, in addition to promoting peripheral anti-

nociception by themselves [Malan TP Jr 2001; Cichewicz 2004]. This is

correlated with studies reporting a similar distribution of CB1R and MOR in areas

that are involved in pain modulation, including the periaqueductal gray and the

dorsal horn of the spinal cord [Vigano 2005; Svizenska 2008]. Not only are the

two receptors expressed in similar brain areas, but they are co-expressed in

individual neurons in rat striatum and dorsal horn [Canals 2008].

In addition, opioid and cannabinoid receptors also share similar signal

transduction properties. Both are GPCRs that couple to Gi, and when activated

under similar circumstances will initiate similar signaling responses. For example,

both receptor types are generally found on presynaptic terminals, and will cause

inhibition of neurotransmitter release through the same mechanisms [Vigano

2005]: (1) By blocking cAMP production; (2) via activation of MAP kinases

through other second messenger systems; and (3) by inhibition of

neurotransmitter release via inhibition of calcium channels and activation of

potassium channels. As a result of these similarities, it is unsurprising that

activation of opioid or cannabinoid receptors can produce similar behavioral

effects, including anti-nociception, hypothermia, sedation, hypotension, inhibition

of intestinal motility, and motor depression.

19

Like opioids, Cannabinoid 1 receptor agonists and dual cannabinoid

agonists (including Δ9-THC) are also associated with the development of

tolerance [Tanda and Goldberg 2003; Pacher and Kunos 2013]. Moreover, opioid

and cannabinoid tolerance appear to be related. Chronic exposure to opioid

agonists induced tolerance to the anti-nociceptive effect of Δ9-THC [Bloom 1978;

Hine 1985]. Similarly, chronic Δ9-THC induced tolerance to the anti-nociceptive

effect of opioids [Smith 1994; Welch 1997].

While the exact mechanisms of opioid and cannabinoid tolerance are still

under investigation, there are definite similarities. There are multiple mechanisms

that may act to mediate opioid tolerance [Savage 2009; Williams 2013]. One

process termed desensitization occurs with the loss of MOR-effector coupling

following opioid administration, and appears to be mediated through

phosphorylation of the receptor by G-protein coupled receptor kinases and/or

second messenger regulated protein kinases. Several previous studies have

demonstrated the loss of MOR-effector coupling following agonist treatment

[Bailey 2009; Williams 2013]. This phenomenon appears to result from

phosphorylation of MORs at 3 specific C-terminal residues. [Doll 2011]. These

phosphorylation events are thought to be the result of G protein-coupled receptor

kinase (GRK) and PKC activity, and cause β-arrestin 2 recruitment [Koch 2008].

β-arrestin 2 recruitment causes both the uncoupling of MOR from its associated

G proteins and endocytosis of the receptor [Zhang 1998; Williams 2013].

Desensitization of CB1R shares similar features. Upon agonist stimulation, CB1R

recruits G-protein coupled receptor kinase 3 (GRK3; [Hsieh 1999; Jin 1999] or G-

20

protein coupled receptor kinase 2 (GRK2; [Kouznetsova 2002; Rubino 2006]) to

phosphorylate serine 426 and/or 430 in the CB1R C terminal tail.

However, this sequence of events occurs in an agonist-specific manner,

with certain ligands not causing phosphorylation sites. Some studies suggest that

internalization of MOR does not occur efficiently in response to morphine [Zhang

1998]. Several studies have demonstrated that GRK is responsible for tolerance

to fentanyl (which is strongly internalizing [Imai 2006]) but not to morphine

[Terman 2004; Melief 2010]. Previous work has demonstrated that tolerance to

the anti-nociceptive and anti-allodynic effects of morphine require JNK signaling

[Melief EJ 2010; Hervera 2012; Marcus 2015]. The agonist-selective mechanisms

of opioid tolerance, as well as their interaction with other GPCR systems is of

significant interest for this project.

Cannabinoid Receptor 2 and opioid interactions.

While there is a great deal of work with CB1R and opioids, there is much

less information on how the CB2R receptor might interact with the opioid system

in a clinically significant manner. CB2R receptor has several advantages over

CB1R as a target for the treatment of pathological pain including fewer adverse

side effects, strong anti-inflammatory properties, and a low potential of tolerance

and dependence for CB2R-directed agonists. These advantages make CB2R a

better option for many types of chronic pathological pain including inflammatory

and neuropathic pain CB2R-MOR interactions have also been suggested by

21

studies indicating that the two receptors influence the signaling of one-another

[Cichewicz 2004; Vigano 2005; Paldyova 2008; Merighi 2012]. CB2R agonists

result in decreases to MOR mRNA expression and G-protein activation by MOR

agonists in forebrain of both wild-type and CB1-Knock-out mice [Paldyova 2008].

Additionally, rates of CB2R mRNA expression are altered by drug use.

Administration of heroin or cocaine increases CB2R mRNA levels in the CNS.

[Onaivi 2008; Paldyova 2008].

Overall the current literature suggests CB2R agonists may be useful for

pain management, and some of the potential efficacy may be due to interaction

with the opioid system. This suggests CB2R activation may be a useful way to

enhance concurrent opiate anti-nociception, particularly in cases of inflammatory

pain, while also minimizing tolerance and potential side effects associated with

CB1R activation. If CB2R agonists are to be used clinically, it is essential to know

how their use impacts, and is impacted by opiate tolerance. This information will

provide context for the circumstances under which CB2R agonists may prove

beneficial, as well as limitations to their use.

22

Chapter 2 - General methods

Subjects

Experiments were carried out with wild-type C57BL6/J mice obtained from

Jackson Laboratories (Bar Harbor, Maine). All mice were group-housed, and kept

on a standard 12:12h light-dark cycle with ad libitum access to standard rodent

chow (Teklad 18% Protein Diet, Harlan Teklad, Indianapolis, IN) and water. Mice

were tested between 8-14 weeks of age, and each animal was only exposed to a

single drug or drug combination. All animal care and procedures conformed to

the guidelines of the National Institutes of Health on the Care and Use of

Animals, and were approved by the Institutional Animal Care and Use Committee

of the Penn State University College of Medicine.

Drugs

Opioid agonists morphine sulfate and fentanyl were obtained from the

National Institute on Drug Abuse Drug Supply (Bethesda, MD). JWH-133 (a CB2

receptor agonist with a Ki of 3.4 nm and approximately 200 fold selectivity for

CB2R over CB1R [Huffman 1999]), SR-144,528 (SR2, CB2R antagonist with a

Ki = 0.6 nM and over 700 fold selectivity for CB2R over CB1R [Rinaldi-Carmona

1998] ), naloxone (MOR antagonist), and rimonabant (SR1, CB1R antagonist)

were obtained from Cayman Chemical (Ann Arbor, MI.) JNK inhibitor SP600125

(SP6) was obtained from Sigma-Aldrich (St. Louis, MO [Bennett 2001].)

23

Drugs were prepared daily through dissolution either in isotonic 0.9%

saline, or saline with 5% Cremaphor and 5% ethanol, and administered via

intraperitoneal (i.p.) injection in a volume of 10 mL/kg body weight. When

animals were given multiple i.p. injections in one day, the second injection was

administered on the opposite side of the body cavity. When testing the effects of

co-administered agonists (JWH-133 and morphine), each drug was injected at

the same time. When testing for agonist selectivity, antagonists were

administered via i.p. injection 30 min prior to agonist treatment.

Procedures

Tail-flick and hotplate anti-nociception

Tail-flick anti-nociception was assessed with a Columbus Instruments TF-

1 analgesia meter (Columbus, OH). The apparatus was calibrated to elicit an

average tail-flick latency of 3-4 s in wild-type mice. A cutoff time of 10 seconds

was used to prevent tissue damage. Mice were restrained, allowing their tail to

be exposed to the radiant heat source. The latency until reflexive tail withdrawal

from the heat source was recorded.

Hotplate anti-nociception was measured with a Columbus Instruments

hotplate set to 55o C (Columbus, OH). A 30s cutoff was used to avoid paw

damage. The latency between an animal being placed on the hotplate and

withdrawal from the heated surface (jumping, shaking or licking of paws) was

24

recorded. Both the hotplate and tail-flick responses were used to calculate the

percentage of maximal possible effect (%MPE). This value was calculated using

the formula below as described previously [Morgan 2014; Marcus 2015].

(%MPE) =(post drug latency)– (pre drug latency)

(cutoff time − pre drug latency)× 100

Formalin test

The formalin test is an extensively used model of acute inflammatory pain

[Tjolsen 1992]. This method utilizes injection of formalin into an animal’s paw to

elicit a biphasic pattern of pain behavior, with a phase of acute pain followed by a

phase of inflammatory pain. The early (acute) phase is generated by the activation

of C and Aδ fibers as a result of needle penetration into the hind paw. The late

phase involves an inflammatory reaction due to intraplantar formalin in the paw

[Tjolsen 1992], Mice were subjected to the formalin test to assess basal differences

in inflammatory pain response and the anti-nociceptive effect of morphine and

JWH-133 on this type of pathological pain. Prior to testing, mice were acclimated

for 20 min in a Plexiglas (5”x5”x5”) observation chamber placed on a transparent

elevated platform. A mirror angled at 45° was placed below the platform to allow

for constant observation of the animal’s paws. Following acclimation, mice were

administered 10μL of a 2.5% formalin solution into the plantar surface of a single

hind paw using a 28 ½ gauge needle (Becton Dickinson, Franklin Lakes, NJ).

25

Immediately after the formalin injection, mice were returned to the Plexiglas

observation unit and nociceptive behavior was continuously measured in 12 five-

min intervals for a total testing time of 60 min. During each five-min time bin, the

duration spent performing pain-response behaviors was recorded. The nociceptive

behaviors were separated into three categories: (0) the injected paw has little

weight placed on it; (1) the injected paw is raised off of the ground; (2) the injected

paw is licked, shaken, or bitten. The amount of time spent in each category was

quantified and weighted with the composite pain score-weighted scores technique

(CPS-WST0,1,2), resulting in a Composite Pain Score (CPS) for each five-min

interval between 0 (no pain behaviors) to 2 (maximal pain behavior; [G. Stennis

Watsona 1997]). The Area Under the Curve (AUC, CPS x time (min)) was

calculated using the trapezoidal rule for the acute phase (0-15 min; phase I) and

the inflammatory phase (15-60 min; phase 2). To assess the anti-nociceptive

effects of drugs, mice were injected (i.p.) 60 min prior to the formalin injection.

Physical side-effects

Body temperature

Body temperature was measured using a mouse rectal thermometer

probe (Physitemp, Clifton, NJ). Temperature was measured immediately prior to,

and 60 min following drug administration. Hypothermia was reported as a percent

26

change in body temperature between pre-drug and 60 min post-drug

measurements (°C), as described by the formula:

(%∆BT) =(post − drug temperature)– (pre − drug temperature)

pre − drug temperature× 100

Rotarod test

Motor impairment was measured using a Med Associates ENV-577-M

Rotarod apparatus (Fairfax, VA). Animals were trained by undergoing two

consecutive days of six 300 second training trials. Mice were placed on a rotating

drum (3 cm in diameter), which accelerated at a constant rate from 4 to 40 rpm

over a 5 minute period. The time spent walking on top of the rod until the mouse

either fell off the rod, or slipped and held onto the rod to ride completely around

was recorded. Impairment was determined through comparison of performance

prior to, and 60 minutes following drug administration.

Precipitated Withdrawal

Physical dependence was induced using a series of 20 injections that

were given twice-daily for 10 days (5 mg/kg morphine, i.p.; 1 mg/kg JWH-133,

i.p.). Following 10 days of daily drug administration, withdrawal was precipitated

using an i.p. injection of vehicle, 10 mg/kg naloxone (to counter morphine), or 10

mg/kg SR2 (to counter JWH-133) 30 min after the final drug injection on the 11th

27

day. Somatic withdrawal symptoms (paw tremors, body tremors, diarrhea, and

jumps) were video recorded for 60 min after injection of naloxone, SR2, or

vehicle. Withdrawal symptoms were scored in alternating 5 min time intervals (5–

10, 15–20, 25–30, 35–40, 45–50, and 55–60 min, as described previously

[Morgan 2014]).

Data analysis

Values for anti-nociception, hypothermia, motor coordination, and

precipitated withdrawal were expressed as the mean ± standard error of the

mean (SEM). Data was analyzed using either one-way or two-way ANOVA,

followed by Bonferroni or Dunnett post-hoc testing as appropriate. Additional

analyses were performed using SPSS statistical software (SPSS Incorporated,

Chicago, IL). P<0.05 was considered significant.

Dose response curves and related values (ED50) were calculated using

the curve fitting functions in GraphPad PRISM. Experimental data (mean+SEM of

individual dose points) were fitted to a sigmoidal curve with variable slope,

according to:

Y=Bottom + (Top-Bottom)/(1+10^((LogEC50-X)*HillSlope)).

28

Isobolographic analysis

This analysis was performed to determine whether the combined anti-

nociceptive effects of morphine and JWH-133 were sub-additive, additive, or

synergistic (super-additive).

Full dose response curves were generated in the formalin test (as

described above) for JWH-133, morphine, and then a combination that was co-

administered in a fixed 1:10 dose ratio (see [Grabovsky 2004; Tallarida 2010;

Kazantzis 2016] for detailed explanation and formulas.) ED50 values for this

combination were determined and compared to a theoretically calculated ED50

value [Tallarida 2002]. This theoretical value was determined using the dose-

response curves of JWH-133 and morphine, alone, to generate a predicted

additive curve using the formula below [Tallarida and Raffa 2010; Kazantzis

2016].

𝐸(𝑎, 𝑏) = 𝐸𝐵

(𝑏 + 𝑏𝑒𝑞(𝑎) )𝑝

(𝑏 + 𝑏𝑒𝑞(𝑎) )𝑝 + 𝐶𝑏𝑝

Where the effect (E) of specific doses of two drugs (a,b) in combination is

estimated using the dose of drug b (beq(a)) that gives and equivalent response to

a specific dose of drug a (a), the ED50 of drug b (Cb), and the Hill slope of drug b

(p). If the experimentally determined ED50 of the combination is significantly lower

than the predicted value (according to a t-test), the combination is deemed

synergistic. If the two ED50 values are equal, the combination has only an

29

additive effect. The variance for the theoretical ED50 value is calculated by

combination of the variances of both JWH-133 and morphine according to the

formula below [Miranda 2014].

Var ED50 (combination) = (0.5)2Var ED50 Morphine + (0.5)2Var ED50 JWH-133

Isobolograms also allow visual comparison of experimental and theoretical

values. The ED50 of the first drug is plotted on the abscissa and the ED50 of the

second drug is plotted on the ordinate. A straight line is drawn connecting the two

values and is termed the line of additivity. ED50s of drug combinations falling

below this line demonstrate synergism, while values on the line demonstrate

additivity.

30

Chapter 3

Tolerance to the anti-nociceptive and hypothermic effects of morphine are mediated by multiple isoforms of c-Jun N-terminal Kinase

Matthew B Yuill1,2,3,†, Michael L Zee1,†, David Marcus1, Daniel J Morgan1,2,3*

1Department of Anesthesiology, Penn State University College of Medicine,

Hershey, PA 17033; 2Department of Pharmacology, Penn State University College

of Medicine, Hershey, PA 17033; 3Department of Neural and Behavioral Sciences,

Penn State University College of Medicine, Hershey PA 17033;

† These authors contributed equally

* To whom correspondence should be sent: [email protected]

Acknowledgements

This work has been supported by NIH grants DA036385 (DJM), DA037355

(DJM), and is also funded, in part, under a grant from the Pennsylvania

Department of Health using Tobacco CURE Funds (DJM).

Conflicts of interest

There are no conflicts of interest

31

Abstract

The abuse and overdose of opioid drugs is a growing public health

problem, globally. While progress has been made towards understanding the

mechanisms governing tolerance to opioids, the exact cellular machinery

involved remains unclear. However, there is growing evidence to suggest that c-

Jun N-terminal Kinases (JNKs) play a major role in mu opioid receptor regulation

and morphine tolerance. In this study, we aimed to determine the potential role

of different isoforms of JNK in tolerance to the anti-nociceptive and hypothermic

effects of morphine. We used the hotplate and tail-flick tests for thermal pain to

measure tolerance to the anti-nociceptive effects of once daily sub-cutaneous

injections with 10 mg/kg morphine. Body temperature was also measured to

determine tolerance to the hypothermic effects of morphine. Tolerance to

morphine was assessed in wild-type mice and compared to single knockout (KO)

mice lacking each of the three c-jun N-terminal kinase (JNK) isoforms (JNK 1,

JNK2, or JNK3). We found that loss of each individual JNK isoform causes

impairment in tolerance for the anti-nociceptive and hypothermic effects of daily

morphine. However, disruption of JNK2 seems to have the most profound effect

on morphine tolerance. These results demonstrate a clear role for c-jun N-

terminal kinase (JNK) signaling pathways in morphine tolerance. This

complements previous studies suggesting that the JNK2 isoform is required for

morphine tolerance, but presents additional novel data suggesting that additional

JNK isoforms also contribute to this process.

32

Keywords: tolerance, morphine, JNK, anti-nociception, opioids, mu opioid

receptor, desensitization, GPCR

Introduction

Opioid drugs, such as morphine, fentanyl and oxycodone, remain a

preferred and commonly prescribed class of drug for pain management [CDC

2011]. While they demonstrate remarkable efficacy for treating acute pain, there

are several limitations to their use. Opioid drugs are associated with rapid

development of tolerance and also high abuse potential. Despite this, they

remain the default approach to treatment of many chronic pain conditions. As a

result, abuse and overdose of opioids are the fastest growing issues for narcotic

drugs in the US. This is punctuated by a tripling in the rate of prescription opioid

overdose in just two decades [CDC 2011].

The opioid system is comprised of multiple opioid receptors, each with a

unique distribution and function. The anti-nociceptive effects of many opioid

drugs are mediated through the mu-opioid receptor (MOR, [Waldhoer 2004]);

making this receptor one of the most extensively studied G protein-coupled

receptors (GPCRs,[Corbett 2006]). MOR is expressed in numerous regions of

the central nervous system, among them: the dorsal horn of the spinal cord, the

periaqueductal gray, and the cortex. As a result, it is a crucial receptor in the

modulation of pain circuitry at both the supra-spinal and spinal level. Activation of

MOR also causes euphoria, which plays a role in the high addictive liability of

opiates [Koob 2006a; Savage 2009]. Opioid dependence is compounded by the

33

development of tolerance, which can cause an escalation of dose and a

progression to dependence [Williams 2013]. Due to the diminishing efficacy of

repeated opioid use, and the resulting prevalence and severity of opioid drug

dependence, understanding the mechanisms behind tolerance to these drugs is

of significant interest.

There are multiple mechanisms that may act to mediate opioid tolerance

[Savage 2009; Williams 2013]. One process termed desensitization occurs with

the loss of MOR-effector coupling following opioid administration, and appears to

be mediated through phosphorylation of the receptor and recruitment of β-

arrestin proteins [Koch and Hollt 2008]. Several previous studies have

demonstrated the loss of MOR-effector coupling resulting from the use of

agonists [Bailey 2009; Williams 2013]. This phenomena appears to result from

phosphorylation of MORs at C-terminal threonine 370 and/or serine 375 [Doll

2011]. These phosphorylation events are thought to be the result of G-protein

coupled receptor kinase (GRK)2 and/or GRK3 activity, and cause β-arrestin 2

recruitment [Koch and Hollt 2008]. β -arrestin 2 causes both the uncoupling of

MOR from its associated G proteins, and also results in endocytosis [Zhang

1998; Williams 2013]. This sequence of events occurs in an agonist-specific

manner, and some studies suggest that internalization of MOR does not occur

efficiently in response to morphine [Zhang 1998].

Several studies have demonstrated that GRK is responsible for fentanyl

but not morphine tolerance [Terman 2004; Melief 2010]. Recent work has shown

that tolerance to morphine is attenuated through the use of the JNK inhibitor,

34

SP600125 [Chen 2008; Guo 2009; Hervera 2012; Marcus 2015]. SP600125 is

an anthrapyrazolone capable of inhibiting JNK1, JNK2, and JNK3 with high

affinity [Zhuang 2006]. It has been demonstrated that this compound prevents

phosphorylation of JNK in the spinal cord, resulting in attenuation of tolerance to

the anti-nociceptive and anti-allodynic effects of morphine [Guo 2009; Hervera

2012]. However, use of this JNK inhibitor is non-selective for the different JNK

isoforms and does not allow determination of which one(s) are responsible for

morphine tolerance. It has been suggested by recent studies that JNK2 is

required for tolerance to the anti-nociceptive effects of morphine [Kuhar 2015].

However, no studies thus far have characterized morphine tolerance in JNK1,

JNK2, and JNK3 Knock-Out (KO) mice.

Therefore, this novel work examined tolerance to the anti-nociceptive

effects of morphine in JNK1, JNK2, and JNK3 KO mice. We tested the

hypothesis that tolerance to morphine would be disrupted in JNK 2 KO mice, and

found that, while this was indeed the case, JNK1, (and to a lesser extent) JNK3

also contribute to morphine tolerance. This finding represents a novel and

significant addition to recent work demonstrating the role of JNK signaling in

morphine tolerance and MOR regulation.

35

Methods

Subjects

Experiments were carried out with wild-type C57BL6/J mice obtained from

Jackson Laboratories (Bar Harbor, Maine), and three strains of JNK mutant mice.

Mice lacking either JNK1, JNK2, or JNK3 were generously provided by Dr.

Charles Chavkin at the University of Washington School of Medicine. The

generation of JNK1 KO [Dong 1998], JNK2 KO [Yang 1998], and JNK3 KO mice

[Yang 1997] has been described previously. All mice were kept on a standard

12:12h light-dark cycle with ad libitum access to standard mouse chow and

water. All animal care and procedures conformed to the guidelines of the

National Institutes of Health on the Care and Use of Animals, and were approved

by the Institutional Animal Care and Use Committee of the Penn State University

College of Medicine.

Drugs

Morphine sulfate was obtained from the National Institute on Drug Abuse

Drug Supply (Bethesda, MD). Morphine was dissolved in isotonic 0.9% saline

and administered sub-cutaneously in an injection volume of 10 mL/kg body

weight.

36

Tail-flick and hotplate anti-nociception

Tail-flick anti-nociception was assessed with a Columbus Instruments TF-

1 analgesia meter (Columbus, OH). The apparatus was calibrated to elicit an

average tail-flick latency of 3-4 s in wild-type mice. A cutoff time of 10 seconds

was used to prevent tissue damage. Mice were restrained, allowing their tail to

be exposed to the radiant heat source. The latency until reflexive tail withdrawal

from the heat source was recorded.

Hotplate anti-nociception was measured with a Columbus Instruments

hotplate set to 55o C (Columbus, OH). A 30s cutoff was used to avoid paw

damage. The latency between an animal being placed on the hotplate and

withdrawal from the heated surface (jumping, shaking of paws) was recorded.

Both the hotplate and tail-flick responses were used to calculate the percentage

of maximal possible effect (%MPE). This value was calculated using the formula

%MPE = (post-drug latency – pre-drug latency)/(cutoff time- pre-drug latency) x

100. These procedures and calculations have been described previously

[Morgan 2014; Marcus 2015].

Measurement of body temperature

Body temperature was measured using a mouse rectal thermometer

probe (Physitemp, Clifton, NJ). Hypothermia was reported as a % change in

body temperature between pre-drug and post-drug measurements, as

demonstrated by the formula:

37

(%∆BT)=[(pre-morphine temperature)–(post-morphine temperature)]/[pre-

morphine temperature] x 100.

Procedures

Anti-nociception and hypothermia were measured in groups of mice [wild-

type (n=17), JNK1 KO (n=15), JNK2 KO (n=14), and JNK3 KO (n=20)] receiving

daily sub-cutaneous (s.c.) injections of morphine (10 mg/kg x 10 days). Mice

were tested for body temperature, tail-flick and hotplate latency immediately prior

to, and one hour after morphine injection on each day.

Data analysis

Anti-nociception and hypothermia values were expressed as mean ±

SEM. Values were analyzed using two-way mixed factorial ANOVA (genotype x

day) followed by Bonferroni post-hoc testing. Differences in baseline tail-flick and

hotplate latencies and basal body temperatures were analyze by one-way

ANOVA. Analyses of the initial responses to the first injection of morphine were

also analyzed by one-way ANOVA. Analyses were performed using PRISM6

statistical software (Graphpad, La Jolla, CA). P<0.05 was considered significant.

38

Results

Tolerance to the anti-nociceptive effects of 10 mg/kg morphine

Baseline tail-flick latencies were different between WT (3.35±0.09 sec)

and JNK 1 KO (4.39±0.28 sec; p<0.001), JNK 2 KO (4.04±0.20 sec; p<0.001),

and JNK 3 KO (4.34±0.16 sec; p<0.0001) mice. However, the response to the

first injection of morphine was not different (F3,70 = 1.164, P=0.33) between WT

(90.9±4.6% MPE), JNK 1 KO (90.0±3.4% MPE), JNK 2 KO (98.7±1.2% MPE),

and JNK 3 KO (94.4±2.4% MPE) mice.

Wild-type mice rapidly developed tolerance to the anti-nociceptive effects

of daily s.c. morphine (10 mg/kg) injections in the tail-flick test (Figure 2).

Tolerance to the anti-nociceptive effect of 10 mg/kg morphine, in the tail-flick test,

developed in a time-dependent manner (F9,637 = 12.77, P<0.0001) that was also

dependent on genotype (F3,637 = 105.6, P<0.0001). There was also a significant

day x genotype interaction effect (F27,637 = 3.26, P<0.0001). Bonferroni post-hoc

tests show that tolerance to morphine, in the tail-flick test, was different between

wild-type mice and JNK 1 KO (p<0.0001), JNK 2 KO (p<0.0001), and JNK 3 KO

mice (p<0.0001). However, post-hoc testing also indicated that JNK2 KO mice

also developed less tolerance than either JNK1 KO (p<0.0001) or JNK3 KO

(p<0.001) mice. There was no difference in morphine tolerance between JNK1

KO and JNK3 KO mice.

Baseline hotplate latencies were also different between WT (7.88 ± 0.57

sec.), JNK 1 KO (11.48±0.77 sec; p<0.001), JNK 2 KO (6.84±0.57 sec; p<0.001),

and JNK 3 KO (5.96±0.26 sec; p<0.0001) mice. However, responses to the first

39

injection of morphine were not different (F3,64 = 1.747, P=0.17) between WT

(67.7±6.1% MPE), JNK 1 KO (83.9±5.7% MPE), JNK 2 KO (67.1±8.9% MPE),

and JNK 3 KO (65.3±2.4% MPE) mice.

Figure 2. Tolerance to morphine in the tail-flick test.

Tolerance to the anti-nociceptive effects of morphine in the tail-flick test is altered in mutant mice lacking JNK 1, JNK 2, or JNK 3. Wild-type (WT; black squares and line), JNK1 KO (red circles and line), JNK2 KO (orange circles and line), and JNK3 KO (magenta circles and line) mice were injected (s.c.) with 10 mg/kg morphine once daily for ten days. All three knockout mouse lines showed impaired tolerance to the anti-nociceptive effects of morphine, in the tail-flick test, relative to wild-type animals (p<0.0001). Data are expressed as mean ±SEM (n=15-20 per group).

Similar results were observed for tolerance to the anti-nociceptive effects

of 10 mg/kg morphine, in the hotplate test (Figure 3). However, all mutant mouse

groups showed delayed onset of tolerance to the anti-nociceptive effects of

morphine, relative to wild-type mice. Two-way ANOVA analysis reveals main

40

effects of genotype (F3,577 = 39.39, P<0.0001) and time (F9,577 = 32.2, P<0.0001).

However, no significant day x interaction effect was detected (F27,577 = 1.28,

P=0.155).

Figure 3. Tolerance to morphine in the hotplate test.

Tolerance to morphine-induced anti-nociception, in the hotplate test, is altered in mutant mice lacking JNK 1, JNK 2, or JNK 3. Wild-type (WT; black squares and line), JNK1 KO (red circles and line), JNK2 KO (orange circles and line), and JNK3 KO (magenta circles and line) mice were injected (s.c.) with 10 mg/kg morphine once daily for ten days. All three knockout mouse lines showed impaired tolerance to the anti-nociceptive effects of morphine, in the hotplate test, relative to wild-type animals (p<0.0001). Data are expressed as mean ±SEM (n=15-20 per group). Tolerance to morphine-induced hypothermia

Basal body temperature was also different between WT (38.6 ± 0.1 °C),

and JNK 1 KO (37.7±0.1 °C; p<0.001), JNK 2 KO (36.8±0.1 °C; p<0.0001), and

JNK 3 KO (37.1±0.1 °C; p<0.0001) mice. There were also differences in basal

41

body temperature between JNK 1 and JNK 2 KO mice (p<0.01) as well as JNK 1

KO and JNK 3 KO mice (p<0.5). There were also genotype differences in the

hypothermic response to morphine (F3,68 = 6.537, P=0.0006), with Bonferroni

post-tests revealing that the main effect of genotype was due to differences

between WT (-4.2 ± 0.7 % change) and JNK 3 KO (-1.6 ± 0.5 % change) mice

(p<0.1) and between JNK 1 (-4.6 ± 0.4 % change) and JNK 3 KO mice (p<0.01).

There were no differences between JNK 2 KO mice (-2.5 ± 0.7 % change) and

the other genotypes.

Rapid tolerance developed to the hypothermic effects of once daily 10

mg/kg morphine in wild-type mice (Figure 4). Tolerance to the hypothermic effect

of 10 mg/kg morphine developed in a time-dependent manner (F9,607 = 18.44,

P<0.0001) that also depended on genotype (F3,607 = 31.24, P<0.0001). There

was also a significant day x genotype interaction effect (F27,607 = 3.33, P<0.0001).

Bonferroni post-hoc tests show that tolerance to morphine was different between

wild-type mice and JNK 1 KO (p<0.001), JNK 2 KO (p<0.0001), and JNK 3 KO

mice (p<0.05). However, post-hoc testing also indicated that JNK 2 KO mice

also developed less tolerance to morphine hypothermia than JNK3 KO (p<0.05)

mice. There was also a difference in tolerance to the hypothermic effects of

morphine between JNK1 KO and JNK 3 KO mice (p<0.0001). However, there

was no difference in morphine tolerance between JNK1 KO and JNK2 KO mice.

42

Figure 4. Tolerance to morphine-induced hypothermia.

Tolerance to morphine-induced hypothermia is altered in mutant mice lacking JNK 1, JNK 2, or JNK 3. Wild-type (WT; black squares and line), JNK1 KO (red circles and line), JNK2 KO (orange circles and line), and JNK3 KO (magenta circles and line) mice were injected (s.c.) with 10 mg/kg morphine once daily for ten days. All three knockout mouse lines showed impaired tolerance to the hypothermic effect of morphine relative to wild-type animals (p<0.0001). Data are expressed as mean ±SEM (n=15-20 per group).

43

Discussion

The primary finding of this study is that all three isoforms of JNK (JNK1,

JNK2, and JNK3) are involved in tolerance to the anti-nociceptive and

hypothermic effects of morphine. Although deletion of JNK2 had the greatest

impact on morphine tolerance, the loss of JNK1 or JNK 3 in KO mice also

attenuated morphine tolerance. Our results suggest that JNK3 plays the least

prominent role in morphine tolerance. Our results are novel in the finding that all

three forms of JNK contribute to morphine tolerance, and they are consistent with

previous work demonstrating that JNK2 can mediate tolerance to morphine

[Melief 2010; Kuhar 2015].

Interestingly, disruption of JNK2 appears to have a greater impact on

tolerance than the other isoforms in the tail-flick but not the hotplate test. This

may be due to the mediation of tail-flick response primarily by spinal circuitry,

versus the mediation of hotplate response by both spinal and supraspinal

circuitry [Langerman 1995]. It should also be noted that hotplate responses are

not reflexive, and thus the possibility of a learning effect among the mice altered

their responses. While the JNK3 KO mice showed a significant diminishment in

hypothermic tolerance relative to wild-type mice, it is important to note that JNK 3

KO mice exhibit profound differences in the hypothermic response to the first

morphine injection. Further investigation is warranted to determine the cause for

reduced morphine-induced hypothermia in JNK 3 KO mice.

Morphine appears to be different from many other opioids, as it does not

produce MOR desensitization through the common GRK/β-arrestin pathway

44

[Terman 2004; Melief 2010]. As such, the question of how JNK is mechanistically

involved in morphine tolerance is of significant interest. Application of morphine

results in an increase in spinal JNK phosphorylation through a protein kinase C

(PKC)-dependent process [Melief 2010]. Furthermore, this effect was abolished

in JNK2 KO, but not JNK1 KO or JNK3 KO mice [Kuhar 2015]. Both of these

studies also demonstrated that JNK2 KO mice have a reduction in acute

morphine tolerance. Despite these results, it should not be concluded that

morphine tolerance is strictly the result of JNK2 phosphorylation by PKC. While

other opioids such as fentanyl also cause JNK2 phosphorylation, their tolerance

is largely JNK-independent [Kuhar 2015; Marcus 2015]. Moreover, while spinal

MOR-desensitization by morphine requires JNK2, the desensitization of MOR in

the locus ceruleus appears to be JNK-independent [Levitt 2012].

Our finding that all three JNK isoforms appear to impact morphine

tolerance further suggests that morphine tolerance is not always mediated by

JNK2 alone. This raises the question of how the other two JNK isoforms might be

involved. While all three JNK isoforms are expressed in parts of the CNS, they

have distinct localization patterns and levels of expression in vivo [Bogoyevitch

2006]. It is an intriguing possibility that development of tolerance to distinct

effects of morphine are mediated to varying degrees by each JNK isoform.

Continued investigation of JNK pathways are thus essential to integrate the

currently disparate findings in the literature to develop a comprehensive

understanding of MOR desensitization and tolerance.

45

Conclusions

Our findings suggest a possible role for multiple isoforms of JNK on the

development of tolerance to morphine. We found a diminishment of tolerance to

the anti-nociceptive and hypothermic effects of morphine not only in JNK 2 KO

mice, but also JNK 1 KO and JNK 3 KO mice. This may be a significant factor in

the developing picture regarding the mechanisms behind morphine tolerance.

Determining the nature through which these signaling pathways act on MOR will

be an important task if JNKs become a therapeutic target in pain management

and opioid abuse.

46

Chapter 4

Anti-nociceptive interactions between opioids and a cannabinoid receptor

2 agonist in inflammatory pain.

Matthew B Yuill1,2,3, David E Hale1, Josée Guindon4,†,* and Daniel J Morgan1,2,3,†,*

Abstract

The cannabinoid 1 receptor (CB1R) and cannabinoid 2 receptor (CB2R) can both

be targeted in the treatment of pain, yet they have some important differences.

CB1R is expressed at high levels in the central nervous system (CNS), whereas

CB2R is found predominantly, although not exclusively, outside the CNS. The

objective of this study was to investigate potential interactions between CB2R

and the mu opioid receptor (MOR) in pathological pain. The low level of adverse

side effects and lack of tolerance for CB2R agonists are attractive

pharmacotherapeutic traits. This study assessed the anti-nociceptive effects of a

selective CB2R agonist (JWH-133) in pathological pain using mice subjected to

inflammatory pain using the formalin test. Furthermore, we examined several

ways in which JWH-133 may interact with morphine. JWH-133 produces dose-

dependent anti-nociception during both the acute and inflammatory phases of the

formalin test. This was observed in both male and female mice. However, a

maximally efficacious dose of JWH-133 (1 mg/kg) was not associated with

47

somatic withdrawal symptoms, motor impairment, or hypothermia. After eleven

once-daily injections of 1 mg/ JWH-133, no tolerance was observed in the

formalin test. Cross-tolerance for the anti-nociceptive effects of JWH-133 and

morphine were assessed to gain insight into physiologically relevant CB2R and

MOR interaction. Mice made tolerant to the effects of morphine exhibited a lower

JWH-133 response in both phases of the formalin test compared to vehicle

treated morphine-naïve animals. However, repeated daily JWH-133

administration did not cause cross-tolerance for morphine, suggesting opioid and

CB2R cross-tolerance is unidirectional. However, preliminary data suggests co-

administration of JWH-133 with morphine modestly attenuates morphine

tolerance. Isobolographic analysis revealed that co-administration of JWH-133

and morphine has an additive effect on anti-nociception in the formalin test.

Overall these findings show that CB2R may functionally interact with MOR to

modulate anti-nociception in the formalin test.

Keywords:

CB2R, morphine, pain, tolerance, formalin, JWH-133, opioid, cannabinoid

agonist

1Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine,

Hershey, PA

2Department of Pharmacology, Penn State University College of Medicine, Hershey, PA

3Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey PA

48

4Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock,

TX

† These authors contributed equally

Corresponding authors:

Daniel J Morgan. Department of Anesthesiology and Perioperative Medicine, Penn State University College

of Medicine, Hershey, PA

Email: [email protected]

Josée Guindon. Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences

Center, Lubbock, TX

Email: [email protected]

49

Introduction

Pain is one of the most widespread and costly clinical challenges facing

medicine today, afflicting an estimated 120 million Americans and costing $600

billion annually in medical expenses, loss of work productivity, and long-term

insurance disability [Gaskin and Richard 2012; Nahin 2015]. Long term pain is

also characterized by the high occurrence of comorbid side effects such as

depression, anxiety, and suicidal ideation [Braden and Sullivan 2008]. Opioid

drugs, which exert analgesic activity through MOR, are the current gold standard

for the treatment of acute and long term chronic pain [CDC 2011]. While opioid

drugs have remarkable anti-nociceptive efficacy for certain types of pain, there

are severe adverse consequences that can occur with prolonged use. For

example, chronic use of opioids causes tolerance and a high potential risk for

physical dependence and abuse [Koob 2006b; Savage 2009; Williams 2013].

The rate of overdose from prescription opioids has more than tripled since the

early 1990s, and is still on the rise [CDC 2011]. More than 33,000 deaths in the

United States were attributed to opioid overdose in 2015 [CDC 2017].

Consequently, there is a current unmet medical need to find possible alternatives

or adjuvants to opioids for the treatment of chronic pain. Therefore, an interest in

understanding the potential interactions of the opioid system with other pathways

involved in alleviating pain is crucial.

The evidence is mounting in terms of the use of cannabinoids for the

treatment of unalleviated pain. They constitute a new class of agents that can be

added to the pharmaceutical toolbox for the management of chronic pain

50

[Eisenberg 2014; Lynch and Ware 2015]. In human clinical trials and case

studies, drugs targeting the endocannabinoid (eCB) system have shown promise

for treatment of numerous pathologies, including chronic and acute pain [Pacher

2006]. For example, the drug Sativex (50% Δ9-THC and 50% cannabidiol), has

been approved in Canada and several other countries for the treatment of

neuropathic pain and cancer pain [Pacher and Kunos 2013]. While CB1R

agonists and dual cannabinoid receptor agonists (including Δ9-THC) demonstrate

potent analgesic effects in rodent models and in human use, they also have

several disadvantages. Use of these drugs is associated with the development of

tolerance, psychotomimetic effects, and numerous other physical side effects

[Tanda and Goldberg 2003; Pacher and Kunos 2013].

However, selective activation of CB2R in rats and mice does not produce

these psychotropic adverse side effects [Guindon 2008; Kinsey 2011; Deng

2015b], making CB2R an attractive target for the treatment of pain and other

pathologies. Indeed, CB2R agonists have been shown to alleviate acute,

inflammatory, and chronic pain causing them to garner increased attention as a

potential alternative to the use of opioids for treatment of pain [Guindon and

Hohmann 2008].

In recent years, mounting evidence of the importance for CB2R in

pathological pain has increased interest as demonstrated by the synthesis of a

variety of CB2R-selective cannabinoid agonists [Atwood 2010]. CB2R agonists

have been shown to have efficacy in multiple models of pathological pain in

preclinical rodent models including post-operative pain [Romero-Sandoval 2007],

51

inflammatory pain [Guindon and Hohmann 2008], chemotherapeutic pain [Deng

2015b], and cancer-induced pain [Curto-Reyes 2010]. However, the mechanisms

through which the anti-nociceptive and analgesic effects of CB2R agonists are

mediated are not completely well characterized.

The CB2R agonists have anti-inflammatory properties, and many pain

studies have suggested a mechanism of action through the regulation of

inflammation (including neuroinflammation, see [Ehrhart 2005; Benito 2008]).

CB2R activation attenuates the release of pro-inflammatory agents (Tumor

Necrosis Factor-α, Nitric Oxide) and increases release of anti-inflammatory

agents (Interleukin 10) from microglia and astrocytes [Ehrhart 2005; Romero-

Sandoval 2009; Correa 2010]. The release of these pro-inflammatory factors is

stimulated by activation of MOR on microglia, and this inflammatory response is

thought to facilitate opioid tolerance [Watkins 2009; Merighi 2012]. As such, the

use of CB2R agonists could both relieve pain and help mitigate the

consequences of opioid treatment. This possibility is supported more generally

by previous work both in animals and humans showing inhaled cannabis and Δ9-

THC enhance the pain relief of opioids [Malan TP Jr 2001; Cichewicz 2004].

However, the interaction between opioids and CB2R selective agonists must be

more extensively investigated.

Therefore, the objectives of this study were to investigate the efficacy of a

CB2R agonist on inflammatory pain and to examine potential interactions

between CB2R and MOR in this pathological pain model. First, we determined

the anti-nociceptive efficacy of a CB2R agonist (JWH-133) in an inflammatory

52

pain model. Second, we evaluated potential side effects and tolerance to JWH-

133. Co-administration of JWH-133 and morphine was also done to assess

potential functional interactions between CB2R and MOR agonists. Together, the

results of this study suggest that co-administration of a CB2R agonist with an

opioid could result in opioid-sparing effects in the treatment of inflammatory pain,

thus minimizing adverse effects and potentially protecting against the

development of opioid tolerance.

Materials and Methods

Subjects

Experiments were carried out with wild-type C57BL6/J mice obtained from

Jackson Laboratories (Bar Harbor, Maine). Mice used were male unless

specifically noted otherwise, and were tested between 8-14 weeks of age. All

mice were group-housed, and were kept on a standard 12:12h light-dark cycle

with ad libitum access to standard mouse chow and water. All animal care and

procedures conformed to the guidelines of the National Institutes of Health on the

Care and Use of Animals, and were approved by the Institutional Animal Care

and Use Committee of the Penn State University College of Medicine.

Drugs

Morphine sulfate was obtained from the National Institute on Drug Abuse

Drug Supply (Bethesda, MD). JWH-133 (CB2R agonist), SR144528 (SR2, CB2R

53

antagonist), naloxone (MOR antagonist), and Rimonabant (SR1, CB1R

antagonist) were obtained from Cayman Chemical (Ann Arbor, MI.). SP600125

(SP6), an inhibitor of c-Jun N-terminal kinases (JNK), was obtained from Sigma-

Aldrich (St. Louis, MO [Bennett 2001].) Drugs were dissolved in isotonic 0.9%

saline (90 %) with Cremaphor (5 %) and ethanol (5 %), and administered via

intraperitoneal (i.p.) injection in a volume of 10 mL/kg body weight.

General Experimental Protocols

JWH-133 dose–response curves. To assess the anti-nociceptive efficacy of JWH-

133, groups of age- and sex-matched drug-naïve mice (n=4-7 per dose) were

given log-scale doses of JWH-133 (0.01, 0.03, 0.1, 0.3, 1, 3, 10 mg/kg) via i.p.

injection. Anti-nociception was measured using the formalin test 60 min after JWH-

133 injection. A similar dose-response analysis was performed for morphine (0.01-

10 mg/kg, i.p.). The data from these experiments were fitted to standard sigmoidal

curves with variable slope to determine the ED50 values and maximal doses for

both drugs. For subsequent experiments the minimum dose of JWH-133 (1 mg/kg)

required to produce a maximal anti-nociceptive response in the formalin test was

used. To determine which receptor was responsible for mediating the anti-

nociceptive effect of 1 mg/kg JWH-133 in the formalin test, it was co-administered

with CB2R antagonist SR2 (10 mg/kg), CB1R antagonist SR1 (10 mg/kg) or MOR

antagonist naloxone (10 mg/kg).

54

To assess tolerance, mice were injected (i.p.) once-daily with either 10

mg/kg morphine or 1 mg/kg JWH-133 for up to eleven consecutive days. This

duration of repeated dosing is sufficient to cause complete tolerance to the anti-

nociceptive effects of morphine. The mice were assessed in the formalin model 60

min following drug administration on the final day. To assess cross-tolerance

between morphine and JWH-133, some groups were given a challenge dose of

the alternate drug (i.e., mice given 10 days of morphine were given a JWH-133

challenge and vice versa) on the eleventh day. Some groups were co-

administered 10 mg/kg morphine and 1 mg/kg JWH-133 for ten days and then

tested for anti-nociception using morphine alone on day 11 to assess the potential

protective effects of JWH-133 co-administration on morphine tolerance.

Since previous published work from our laboratory has demonstrated that

JNK signaling is required for morphine tolerance, we examined whether JNK

signaling was required for morphine-induced cross-tolerance to JWH-133

[Marcus 2015; Yuill 2016]. This experiment was done by examining the efficacy

of JWH-133 (1 mg/kg) anti-nociception in the formalin model using mice that

received either five days of repeated morphine alone (10 mg/kg), mice that

received daily pre-treatment with SP6 (3 mg/kg) prior to morphine (10 mg/kg) and

mice receiving daily vehicle injections. Pre-treatment with SP6 was given 60 min

before injection of morphine.

55

Formalin Test

The formalin test is an extensively used model of inflammatory pain [Tjolsen

1992]. This method elicits a biphasic pattern of pain behavior, with a phase of acute

pain followed by a phase of inflammatory pain. The early (acute) phase is

generated by the activation of C and Aδ fibers as a result of needle penetration

into the hind paw. The late phase involves an inflammatory reaction due to

presence of formalin in peripheral tissue [Tjolsen 1992], the development of central

sensitization and additionally the activation of primary afferent nociceptors [Puig

1996; Coderre TJ 1997]. Mice were subjected to the formalin test to assess basal

differences in inflammatory pain response and the anti-nociceptive effect of

morphine and JWH-133 on pathological pain. Prior to testing, mice were

acclimated for 20 min in a Plexiglas (5”x5”x5”) observation chamber placed on a

transparent elevated platform. A mirror angled at 45° was placed below the

platform to allow for constant observation of the animal’s paws. Following

acclimation, mice were administered 10μL of a 2.5% formalin solution into the

plantar surface of a single hind paw using a 28 ½ gauge needle (Becton Dickinson,

Franklin Lakes, NJ). Immediately after the formalin injection, mice were returned

to the Plexiglas observation unit and nociceptive behavior was continuously

measured in 12 five-min intervals for a total testing time of 60 min. During each

five-min time bin, the duration spent performing pain-response behaviors was

recorded. The nociceptive behaviors were separated into three categories: (0) the

injected paw has little weight placed on it; (1) the injected paw is raised off of the

ground; (2) the injected paw is licked, shaken, or bitten. The amount of time spent

56

in each category was quantified and weighted with the composite pain score-

weighted scores technique (CPS-WST0,1,2), resulting in a Composite Pain Score

(CPS) for each five-min interval between 0 (no pain behaviors) to 2 (maximal pain

behavior; [G. Stennis Watsona 1997]). The Area Under the Curve (AUC, CPS x

time(min)) was calculated using the trapezoidal rule for the acute phase (0-15 min;

phase I) and the inflammatory phase (15-60 min; phase 2). To assess the anti-

nociceptive effects of drugs, mice were injected (i.p.) 60 min prior to the formalin

injection.

Measurement of body temperature

Body temperature was measured using a mouse rectal thermometer probe

(Physitemp, Clifton, NJ). Mice were measured immediately prior to, and 60 min

following drug administration. Hypothermia was reported as a percent change in

body temperature between pre-drug and 60 min post-drug measurements, as

described by the formula:

(%∆BT) =(post − drug temperature)– (pre − drug temperature)

pre − drug temperature× 100

Rotarod test

Motor impairment was measured using a Med Associates ENV-577-M

rotarod apparatus (St. Albans, VT). Animals were trained by undergoing six

57

training trials per day over two consecutive days. The maximum cut-off for

training trails was limited to 300s. Mice were placed on a rotating rod (3 cm in

diameter), which accelerated at a constant rate from 4 to 40 rpm over the 5 min

testing period. The time spent walking on top of the rod until the mouse either fell

off the rod, or slipped and held onto the rod to ride completely around was

recorded. Motor impairment was determined by calculating the change in

performance between the pre-test and post-test given 60 min after JWH-133,

morphine or vehicle injection.

Precipitated withdrawal

Physical dependence was induced using a series of 20 injections that

were given twice-daily for 10 days (5 mg/kg morphine, i.p.; 1 mg/kg JWH-133,

i.p.). Following 10 days of daily drug administration, withdrawal was precipitated

using an i.p. injection of vehicle, 10 mg/kg naloxone (to counter morphine), or 10

mg/kg SR2 (to counter JWH-133) 30 min after the final drug injection on the 11th

day. Somatic withdrawal symptoms (paw tremors, body tremors, diarrhea, and

jumps) were video recorded for 60 min after injection of naloxone, SR2, or

vehicle. Withdrawal symptoms were scored in alternating 5 min time intervals (5–

10, 15–20, 25–30, 35–40, 45–50, and 55–60 min, as described previously

[Morgan 2014]).

58

Co-administration

When animals were given multiple i.p. injections, the second injection was

administered on the opposing side of the body cavity. When testing the effects of

co-administered agonists (JWH-133 and morphine), each drug was injected at

the same time. When testing for agonist selectivity, antagonists were

administered via i.p. injection 30 min prior to agonist treatment.

Isobolographic analysis

This analysis was performed to determine whether the combined anti-

nociceptive effects of morphine and JWH-133 were sub-additive, additive, or

synergistic (super-additive). Full dose response curves were generated in the

formalin test (as described above) for JWH-133, morphine, and then a

combination that was co-administered in a fixed 1:10 dose ratio (see [Grabovsky

and Tallarida 2004; Tallarida and Raffa 2010; Kazantzis 2016] for detailed

explanation and formulas.) ED50 values for this combination were determined and

compared to a theoretically calculated ED50 value [Tallarida 2002]. This

theoretical value was determined using the dose-response curves of JWH-133

and morphine, alone, to generate a predicted additive curve using the formula

below [Tallarida and Raffa 2010; Kazantzis 2016].

59

𝐸(𝑎, 𝑏) = 𝐸𝐵

(𝑏 + 𝑏𝑒𝑞(𝑎) )𝑝

(𝑏 + 𝑏𝑒𝑞(𝑎) )𝑝 + 𝐶𝑏𝑝

Where the effect (E) of specific doses of two drugs (a,b) in combination is

estimated using the dose of drug b (beq(a)) that gives and equivalent response to

a specific dose of drug a (a), the ED50 of drug b (Cb), and the Hill slope of drug b

(p). If the experimentally determined ED50 of the combination is significantly lower

than the predicted value (according to a t-test), the combination is deemed

synergistic. If the two ED50 values are equal, the combination has only an

additive effect.

Data analysis

Values for anti-nociception, hypothermia, motor coordination, and

precipitated withdrawal were expressed as the mean ± standard error of the

mean (SEM). Data was analyzed using either one-way (Figure 3, Figure 4(c),

Figure 6(d) or two-way ANOVA (all other figures), followed by Bonferroni or

Dunnett post-hoc testing as appropriate. Analyses were performed using SPSS

statistical software (SPSS Incorporated, Chicago, IL). P<0.05 was considered

significant.

60

Results

Anti-nociceptive effect of JWH-133 in the formalin test

The anti-nociceptive effect of JWH-133 was assessed by performing a

dose-response analysis in the formalin test using male (Figure 1(a), N=32) and

female (Figure 1(b), N=28) mice. In males, JWH-133 diminished both acute (0-15

min post-formalin, ED50=0.23 mg/kg, [F(7,72)=9.72, p<0.0001]) and inflammatory

pain (15-60 min post-formalin, ED50=0.23 mg/kg, [F(7,216) = 8.42, p<0.00001]) in a

dose-dependent manner (Figure 5(a)). There was also a significant dose-

dependent effect in the levels of both acute pain (ED50=0.24 mg/kg, [F(7,60)= 4.62,

p=0.0004]) and inflammatory pain (ED50= 0.20 mg/kg, [F(7,180)=3.69, p=0.0014]) in

female mice (Figure 5(b)). Both male and female mice displayed maximal anti-

nociceptive effects with 1 mg/kg JWH-133.

Comparison of the anti-nociceptive effect of JWH-133 and morphine in the

formalin test

The anti-nociceptive effect of a maximal dose of JWH-133 (1 mg/kg) was

compared to a maximal dose of morphine in groups of male mice (10 mg/kg,

Figure 6, N=4 per group). Analysis of the AUC of pain behavior revealed that

both morphine and JWH-133 reduced pain behavior relative to the vehicle group

in the acute (F(2,9)= 32.28, P < 0.0001, Figure 6(a)) and inflammatory phases

(F(2,9)= 132.47, P < 0.0001, Figure 6(b)) of the formalin test. Moreover, morphine

produced a greater anti-nociceptive effect than JWH-133 in both phases (P <

0.037 acute; P < 0.0001 inflammatory).

61

Figure 5. Anti-nociceptive efficacy of JWH-133.

Wild-type male (a) and female (b) mice (N=4-7 per dose) were tested using the formalin model. Testing was conducted 60 min after i.p. injection of JWH-133 (0.01, 0.03, 0.1, 0.3, 1, 3, 10 mg/kg). The area under the curve (AUC) represents the pain behavior obtained from a composite pain score. Treatment with JWH-133 reduced both acute (F(6,35)= 16.02, p<0.0001) and inflammatory (F(6,35)= 21.53, p<0.0001) pain in a dose-dependent fashion, with a maximal effect occurring at a dose of 1 mg/kg. The calculated ED50 values for acute and

inflammatory phases were 0.23 mg/kg and 0.23 mg/kg for males. For females the ED50 values were 0.24 mg/kg and 0.20 mg/kg.

62

Figure 6. Comparison of morphine and JWH-133 in the formalin test.

Mice (n=4–6 per dose) were tested in the formalin model 60 min after i.p. administration of maximal doses of either JWH-133 (1 mg/kg) or morphine (10 mg/kg). The area under the curve (AUC) represents the pain behavior obtained from the composite pain score. Both morphine and JWH-133 reduced pain behavior relative to the vehicle group in the acute (a) and inflammatory (b) phases. Morphine produced a greater anti-nociceptive effect than JWH-133 in both phases. *p<0.0001 for JWH-133 and morphine versus vehicle group (ANOVA with Bonferroni post hoc) and *+p<0.037 for morphine versus JWH-133 group (ANOVA with Bonferroni post hoc). ANOVA: analysis of variance.

63

Figure 7. JWH-133 acts through the CB2 Receptor.

The efficacy of JWH-133 (1 mg/kg, i.p.) in the formalin test was examined in the presence of multiple antagonists administered 30 minutes prior to JWH-133 treatment. (n=4-5 per treatment) The effect of JWH-133 was blocked by CB2R antagonist SR2 (10 mg/kg, i.p.), in both acute (p<0.05) and inflammatory pain (p<0.001) According to a one-way ANOVA with Bonferonni post-hoc. JWH-133 was not impacted by CB1R antagonist rimonabant (SR1, 10 mg/kg) nor by MOR antagonist naloxone (10 mg/kg).

64

JWH-133 acts through the CB2 Receptor

To establish that JWH-133 was selective for the CB2R, it was co-

administered with antagonists for CB1R (SR1, 10 mg/kg) or CB2R (SR2, 10

mg/kg). The impact of these antagonists on the anti-nociceptive effects of JWH-

133 was then measured using male mice in the formalin test (Figure 7, N=4-5 per

group). We found that JWH-133 alleviates formalin-induced pain behaviors in

both the acute (Figure 7(a), F(5,18)= 9.21, P < 0.0001) and inflammatory (Figure

5(b), F5,18= 18.69, P < 0.0001) phases. The anti-nociceptive effect of JWH-133

was blocked by SR2 (P < 0.006, acute; P < 0.0001, inflammatory), but not by

SR1 (P = 1.00, acute; P = 0.83 inflammatory). Indeed, the effect of JWH-133 is

not mediated through CB1R since its combination with a CB1R antagonist (SR1)

produced anti-nociceptive effects when compared to SR1 alone for the acute (P

< 0.012) and inflammatory (P < 0.03) phases. There were no difference in pain

values between JWH-133 alone or JWH-133 combined with SR1 (P = 1.00 acute

and P = 0.833 inflammatory).

JWH-133 does not act through mu-opioid receptor

JWH-133 suppressed formalin-induced pain scores in both the acute (F(3,12)

= 10.39, P < 0.001) and inflammatory (F(3,12) = 58.09, P < 0.0001) phases. This

anti-nociceptive effect is not blocked by the MOR antagonist naloxone (p = 0.688,

acute; P = 0.751, inflammatory). Formalin-induced pain behavior was similar in

groups receiving JWH-133 alone or co-administered naloxone (p = 0.69, acute; P

= 0.75, inflammatory (Figure 7(c) and (d)).

65

Evaluation of side effects

Male mice were injected daily with JWH-133 (1 mg/kg) or morphine (10

mg/kg) for seven days to examine the possibility of either acute or cumulative

negative side effects such as hypothermia. JWH-133 had no impact on body

temperature (F(1,27)=2.78, p=0.11), and the effects of JWH-133 on body

temperature were not significantly different from vehicle after one (p=0.530), four

(p=0.7359), or seven (p=0.8917) days of repeated administration according to

Dunnett’s post-hoc testing (Figure 8(a)). By contrast, morphine does have a

significant impact on hypothermia (F(1,27)=9.88, p<0.004), and was significantly

different from vehicle on day one (P<0.0001) but not on days 4 (p=0.1334) or 7

(p=0.1673, Figure 8(a)).

Motor impairment was measured using a rotarod apparatus because this

is a common side effect for both opiates and many cannabinoids. Similarly to the

hypothermia test, JWH-133 treatment did not produce motor impairment

(F(1,27)=0.17, p=0.68), and did not differ from vehicle on days 1 (p=0.999), 4

(p=0.7774), or 7 (p=0.9998, Figure 8(b)). Comparatively, morphine treatment had

a significant effect on motor impairment (F(1,27)=21.32, p<0.0001) and was

significantly different from vehicle on day 1 (p=0.0001), but not on days 4

(p=0.2120), or 7 (p=0.8429, Figure 8(b)).

66

Precipitated somatic withdrawal

Following ten days of twice daily injections of either JWH-133 or morphine,

antagonists were given to determine whether somatic withdrawal symptoms

could be precipitated. Precipitation of physical withdrawal from JWH-133 using

the CB2R antagonist SR2 did not result in any detectable somatic withdrawal

events following repeated JWH-133. Similarly, SR2 did not elicit somatic

withdrawal symptoms in mice treated with daily saline (Figure 8(c), N=4 per

group). However, treatment of morphine-tolerant mice with naloxone elicited an

increase in paw tremors (p<0.001), body tremors (p<0.001), and jumping

behavior (p<0.001) compared to vehicle treated mice.

67

68

Lack of observed tolerance to 11 once-daily injections of JWH-133

In order to investigate the development of tolerance, male mice (N= 3-5

per group) were assessed using the formalin model following 1, 6, or 11 days of

JWH-133 (1 mg/kg; Figure 9(a)) or morphine (10 mg/kg; Figure 9(b)) treatment.

Analysis of the AUC of pain behavior revealed that JWH-133 reduced pain

behavior relative to the vehicle group in the acute (F(3,15) = 15.01, P < 0.0001)

and inflammatory phases (F(3,15) = 41.21, P < 0.0001) of the formalin test after 1,

6 or 11 days of repeated administration ( P <0.0002 acute and P < 0.0001

inflammatory). The magnitude of JWH-133’s anti-nociceptive efficacy did not

diminish following 1, 6 or 11 days of continuous administration (P = 1.000). In the

acute phase, morphine reduced pain behavior in comparison to the vehicle group

(F(3,15) = 47.05, P < 0.0001) after 1 and 6 days of repeated administration (P <

0.0001), but not after 11 days (P = 0.149). In the inflammatory phase, morphine

Figure 8. Lack of JWH-113 adverse effects.

Mice (n=5-7 per group) were chronically injected with a maximal dose of JWH-133 (1 mg/kg), or morphine (10 mg/kg) for seven days to look for either acute or cumulative physical side effects. The hypothermic effects of JWH-133 were not significantly different from vehicle according to a two-way ANOVA with Dunnett’s post-hoc test (A). The same trends were observed when measuring rota-rod motor impairment of JWH-133 (B). Comparatively, morphine injected mice showed significant hypothermic effects on day 1(p=0.0001), but not 4(p=0.1334), or 7(p=0.1673). Morphine also showed significant motor impairment on day 1(p=0.0001), but not 4(p=0.2120), or 7(p=0.8429, Fig 3B). Following ten days of chronic drug administration, antagonists and frequency of withdrawal behaviors were recorded for60 minutes (C). While SR2 administration following chronic JWH-133 did not result in withdrawal behavior, morphine-treated mice challenged with naloxone showed significant amounts of paw tremors (p<0.001). Body tremor (p<0.001), and jumping (p<0.001) relative to vehicle.

69

reduced pain behavior relative to the vehicle group (F(3,15) = 55.03, P < 0.0001)

after 1, 6 or 11 days of repeated administration (P <0.031). However, this anti-

nociceptive effect of morphine was different in a time-dependent manner since

greater anti-nociceptive effect is observed at day 1 relative to day 6 (P < 0.001)

and from day 6 relative to day 11 (P < 0.039).

70

Figure 9. Lack of observed tolerance to JWH-133.

Mice (n=3-5 per group) were measured in the formalin test following 1, 6, or 11 days of JWH-133 (1 mg/kg, a) or morphine (10 mg/kg, b) administration. Data are expressed as mean ± S.E.M. * P < 0.0001 for JWH-133 or morphine vs vehicle group (ANOVA with Bonferroni post-hoc); + P < 0.001 for morphine day 1 vs morphine day 6 group (ANOVA with Bonferroni post-hoc); # P < 0.039 for morphine day 6 vs morphine day 11 group (ANOVA with Bonferroni post-hoc).

71

JWH-133 and morphine cross-tolerance

Male mice (N= 5-8 per group) were injected for ten days with either vehicle

or JWH-133 (1 mg/kg), and then given a challenge dose of morphine (10 mg/kg)

on day 11 to assess cross-tolerance in the formalin test (Figure 10(a)). The

repeated (10 days) administration of vehicle or JWH-133 and a challenge dose (on

day 11) of morphine (F(2,13) = 325.04, p < 0.0001) suppressed CPS relative to

control group in a time-dependent manner ( F(22,143) = 11.46, p < 0.0001, Figure

10(a)). This suppression was observed from 5 to 15 min (acute phase; F(2,13)

=22.24 (5), 5.86 (10), 4.57 (15); p < 0.0001 for all time points) and from 20 to 50

min (inflammatory phase 2; F(2,13) = 25.92 (20), 37.56 (25), 71.57 (30), 71.67 (35),

57.10 (40), 12.30 (45) and 4.37 (50); p < 0.035 for all time points) post-formalin

injection compared to the control group.

Analysis of the AUC of pain behavior revealed that both the repeated

vehicle group and the repeated JWH-133 group showed anti-nociceptive efficacy

of morphine challenge relative to the vehicle-only control group in both phase 1

(F(2,13) = 63.58, p < 0.0001; Figure 6(b)) and phase 2 (F(2,13) = 356.04, p < 0.0001;

Figure 10(c)) of the formalin test.

Male mice were also injected for ten days with either vehicle or morphine

and then given a challenge dose of the JWH-133 drug on day 11 to assess cross-

tolerance in the opposing direction. Only the repeated (10 days) administration of

vehicle and a challenge dose (on day 11) of JWH-133 (F(2,14) = 17.61, p < 0.0001)

suppressed CPS relative to control group in a time-dependent manner (F(22,154) =

6.30, p < 0.0001, Figure 10(d)). This suppression was observed at 5 (acute phase;

72

F(2,14) =5.65; p < 0.001) min and from 30 to 50 min (inflammatory phase 2; F(2,14) =

5.65 (30), 24.97 (35), 24.93 (40), 13.84 (45); p < 0.016 for all time points) post-

formalin injection compared to the control or repeated morphine + JWH-133

challenge groups. There was no difference in formalin-induced pain values

between the control and repeated morphine + JWH-133 challenge groups from 5

to 60 min (P > 0.064 for all time points) except at 20 (P < 0.001) and 45 (P < 0.004)

min.

In these mice, analysis of the AUC of pain behavior revealed that only the

repeated vehicle with JWH-133 challenge group produced anti-nociception relative

to the control or repeated morphine + JWH-133 challenge groups in both Phase 1

(F(2,14) = 22.39, p < 0.0001; Figure 10(e)) and Phase 2 (F(2,14) = 6.57, p < 0.01;

Figure 10(f)) of the formalin test. Similar formalin-induced pain values are observed

in the control and repeated morphine + JWH-133 challenge groups in both phases

(P=1.00 acute and inflammatory).

In groups of female mice (N= 4-7), analysis of the AUC of pain behavior

revealed that only the repeated vehicle with JWH-133 challenge group produced

anti-nociception relative to the control or repeated morphine + JWH-133 challenge

groups in both Phase 1 (F(2,8) = 13.72, p < 0.003; Figure 10(g)) and Phase 2 (F(2,8)

= 9.71, p < 0.01; Figure 10(h)) of the formalin test. Similar formalin-induced pain

values were observed in the control and repeated morphine + JWH-133 challenge

groups in both phases (P = 0.164 acute; 1.00 inflammatory).

73

Figure 10. Cross-tolerance between JWH-133 and morphine.

The formalin model of inflammatory pain was utilized to examine cross-tolerance for the anti-nociceptive effects of a challenge dose of morphine (10 mg/kg, i.p.) after 10 days of chronic JWH-133 (1 mg/kg i.p.) administration (A), and cross-tolerance for the anti-nociceptive effects of JWH-133 after 10 days of chronic morphine administration (B, n=5-8 per group). A two-way ANOVA failed to reveal an effect of chronic JWH-133 administration on morphine anti-nociceptive efficacy in either acute pain (p=0.8583) or inflammatory pain (p=0.2992). However, chronic injection of morphine significantly reduced the anti-nociceptive efficacy of JWH-133 both in acute [F(2,20)=16.63, p<0.0001] and inflammatory pain [F(2,20=28.13, p<0.001].

74

JWH-133 co-administration modestly attenuates morphine tolerance

Male mice (N=4 per group) were co-administered 10 mg/kg morphine and

1 mg/kg JWH-133, given morphine alone, or given vehicle for up to ten days. All

groups were then tested for morphine-induced anti-nociception using a challenge

dose morphine alone on (days 2, 6, 11) to assess potential protective effects of

JWH-133 on morphine tolerance.

On day 2, analysis of the AUC of pain behavior revealed that

vehicle+morphine challenge, morphine+morphine challenge or combination

(morphine+JWH-133)+morphine challenge groups showed lower pain values

relative to the control group for both phases (F(3,12) = 60.26 acute and 360.55

inflammatory, p < 0.0001, Figure 11). In the inflammatory phase, the vehicle +

morphine challenge group shows lower pain values relative to morphine +

morphine challenge group (P < 0.004).

On day 6, the AUC of pain behavior also revealed that vehicle+morphine

challenge, morphine+morphine challenge or combination (morphine+JWH-

133)+morphine challenge groups showed lower pain values relative to the control

group for both phases (F(3,12) = 117.69 acute and 165.98 inflammatory, p <

0.0001). In the acute and inflammatory phases, the vehicle +morphine challenge

or the combination+morphine challenge groups showed lower pain values

relative to morphine+morphine challenge group (P < 0.002, acute and P < 0.047,

inflammatory). These results show the protective effect of JWH-133 co-

administration with repeated administration of morphine on both phases of the

75

formalin test. In the inflammatory phase only, the vehicle+morphine challenge

group showed the lowest pain values (P < 0.004).

On day 11, the AUC of pain behavior revealed that vehicle+morphine

challenge and combination (morphine+JWH-133)+morphine challenge groups

showed lower pain values relative to the control group for both phases (F(3,13) =

74.44 acute and 179.36 inflammatory, p < 0.0001). The control group showed

similar values to the morphine + morphine challenge group for both phases (P =

1.00 acute and P = 0.05 inflammatory). In the acute and inflammatory phases,

the vehicle+morphine challenge or the combination + morphine challenge groups

show lower pain values relative to morphine + morphine challenge group (P <

0.019, acute and P < 0.037, inflammatory). In both phases, the vehicle+morphine

challenge group shows the lowest pain values (P < 0.0001 acute and

inflammatory phases).

76

Figure 11. JWH-133 co-administration modestly protects against morphine

tolerance.

The efficacy of morphine (10 mg/kg) phase I (a) and phase 2 (b) anti-nociception in the formalin model compared between mice receiving daily morphine alone (10 mg/kg), and mice receiving daily morphine (10 mg/kg) and daily JWH-133 (1 mg/kg). Data are expressed as mean ± S.E.M. (n = 4 per group). * P < 0.004 vs vehicle group (ANOVA with Bonferroni post-hoc); + P < 0.047 vs morphine + morphine challenge group (ANOVA with Bonferroni post-hoc); # P < 0.003 vs combination + morphine challenge group (ANOVA with Bonferroni post-hoc).

77

JNK signaling is partially responsible for morphine-induced cross-tolerance to

JWH-133

The efficacy of a challenge dose of JWH-133 (1 mg/kg) in the formalin

model was compared between groups of male mice that received either five days

of morphine alone (10 mg/kg), five days of the JNK inhibitor SP600125 (3

mg/kg) and morphine (10 mg/kg), or five days of vehicle (Figure 12, N=4 per

group). Pre-treatment with SP600125 prior to morphine caused a significant

increase in JWH-133 efficacy (F(2,9)=91.86 acute, and F(2,9)=55.37 inflammatory,

p<0.0001 for both phases). Therefore, repeated co-administration of SP600125

(3 mg/kg) with morphine appears to reduce observed cross-tolerance to JWH-

133. In both phases of the formalin test, the vehicle and JWH-133 challenge

group shows the lowest pain values (P < 0.0001 acute and inflammatory

phases).

78

Figure 12. JNK signaling is partially responsible for morphine-induced

cross-tolerance to JWH-133

The efficacy of a challenge dose of JWH-133 (1 mg/kg) in the formalin model was compared between mice that received five days of daily morphine alone (10 mg/kg), mice receiving both daily morphine (10 mg/kg) and SP6 (3 mg/kg), and mice receiving daily vehicle. Co-administration of SP6 with morphine resulted in a significant increase of JWH-133 efficacy both in acute and inflammatory pain. Data are expressed as mean ± SEM (n = 3-5 per group). * P < 0.0001 vs chronic morphine group (ANOVA with Bonferroni post-hoc); + P < 0.001 vs chronic morphine + SP600125 group (ANOVA with Bonferroni post-hoc).

79

Isobolographic analysis

To examine the potential synergy between JWH-133 and morphine,

isobolographic analysis was used to compare the theoretical and experimental

dose-response curves for a 1:10 fixed ratio dose combination of JWH-133 and

morphine in the formalin test (Figure 13(a) and (b)). In this analysis, there is a

substantial difference in the maximal efficacies of JWH-133 and morphine.

Therefore, a non-linear isobolographic analysis [Tallarida and Raffa 2010] was

used. In phase I of the formalin test, the theoretical ED50 of 1:10 JWH-

133/morphine combination (0.78 mg/kg) was found to overlap with the

experimentally determined value (0.72 mg/kg, Figure 13(c)). This indicates that

the 1:10 fixed ratio combination is likely to be additive, but not synergistic. In

phase II of the formalin test, the experimentally determined ED50 (0.62 mg/kg)

appears to be lower than the predicted ED50 (0.93 mg/kg, Figure 13(d)).

However, the 95% confidence intervals of the two values are overlapping. Thus

our results demonstrate that the combined anti-nociceptive effects of JWH-133

and morphine are additive, but likely not synergistic.

80

Figure 13. Non-linear isobolographic analysis of 1:10 fixed ratio JWH-133

and morphine in the formalin test.

We compared experimentally determined ED50±SEM values with theoretical ED50 values calculated if the combination were exactly additive. In acute pain (c) the theoretical ED50 (0.78±0.18 mg/kg) was found to overlap with the experimentally determined value (0.72±0.11 mg/kg), suggesting additivity. In inflammatory pain (d), the experimentally determined ED50 (0.62±0.06 mg/kg) appears to be lower than the predicted ED50 (0.93±0.12 mg/kg).

81

Discussion

The primary goal of this study was to test the hypothesis that CB2R

functionally interacts with the opioid system to modulate inflammatory pain. First,

we examined the effects of CB2R activation alone and found that JWH-133

causes dose-dependent anti-nociception in both the acute and inflammatory

phases of the formalin test (Figure 4). Using the CB2R-seletive antagonist SR2,

we demonstrated that the anti-nociceptive effects of JWH-133 occur through

activation of CB2R (Figure 6). The efficacy of JWH-133 is consistent with

previous work demonstrating that CB2R agonists can produce robust anti-

nociception in other inflammatory and neuropathic pain models [Gutierrez 2011;

Hsieh 2011; Brownjohn and Ashton 2012]. For example, systemic administration

of the CB2R agonist GW405833 to mice and rats was able to reverse

hyperalgesia in a model of inflammatory pain involving complete Freund’s

adjuvant (CFA, [Whiteside 2005]). In addition, two other CB2R agonists A-

836339 and AM-1241 were anti-nociceptive in a rat CFA model when

administered spinally (intrathecally) or via direct intraplantar injection to the

inflamed paw [Hsieh 2011]. Previous studies report that AM-1710 was able to

completely reverse paclitaxel-induced neuropathic pain in mice [Deng 2015b].

Interestingly, continuous administration of AM-1710 before and after paclitaxel

treatment can prevent the development of neuropathic pain for several weeks

[Rahn 2014].

Additionally, we sought to examine potential sex differences in JWH-133

anti-nociception between males and females using the formalin test (Fig 5(a),

82

5(b)). Females displayed higher levels of pain behavior at every JWH-133 dose.

However this difference appears to be due to higher basal levels of pain in

female mice rather than reduced sensitivity to the anti-nociceptive effects of

JWH-133. The finding that females generally exhibit more pain behaviors is

consistent with animal and human studies of pain [Cepeda 2003; Fillingim 2009;

Bergeson 2016; Blanton 2016; Henderson-Redmond 2016]. Our study did not

suggest that female mice were more sensitive to JWH-133, as the calculated

ED50 values and maximally efficacious doses did not differ between male and

female mice. However, there is evidence for sex differences in the response to

cannabinoids. Previous studies have shown that female rats are more sensitive

to Δ9-THC, and develop tolerance to it more quickly [Wakley 2014]. It is possible

that this discrepancy is due to the fact that many previously reported sex

differences in cannabinoid response involved compounds that also activate

CB1R. It is also possible that previously reported sex differences in rats are

partially species-dependent.

We also sought to assess potential negative side effects of JWH-133. Our

results closely aligned with previous studies that failed to demonstrate adverse

effects of other CB2R agonists (AM-1241, AM-1710 and HU-308) in rodents

[Hanus L 1999; Kinsey 2011; Deng 2015b]. We did not observe negative side

effects typically associated with agonists for the CB1R and/or opioid receptors

which includes; hypothermia, motor incoordination, or antagonist-induced

somatic withdrawal symptoms (tremors, jumping, diarrhea) for JWH-133 (Figure

8). However, it is important to recognize that this list of potential adverse effects

83

is not exclusive, and more research studies are needed to assess the impact of

these adverse side effects and possibly sex-specific differences and their impact

on pain pathways and alleviation of pain. In particular, the short duration of

repeated JWH-133 administration and limited number of observed somatic

withdrawal symptoms here are not sufficient to fully assess the possibility that

subtle somatic symptoms might occur. Furthermore, since we only examined

possible somatic symptoms, it is possible that precipitated JWH-133 withdrawal

could cause affective and behavioral withdrawal symptoms such as anxiety,

depression, and anhedonia. Lastly, sex-differences could also be found.

Regardless of the pain model used, multiple studies observe a lack of

tolerance to repeated administration of CB2R agonists. For example, daily

systemic administration of AM-1710 attenuated chemotherapy-evoked

neuropathic pain for up to 8 days with no evidence of tolerance [11]. Similarly,

the CB2R agonist, JWH015 (intrathecal, once daily), reversed surgery-induced

allodynia for up to 5 days without tolerance [Romero-Sandoval 2008]. In this

study we report a lack of tolerance to the anti-nociceptive effects of JWH-133

after 11 consecutive days of administration (Figure 9). However, it is possible

that tolerance to JWH-133 may occur with longer periods of chronic dosing

and/or the affective component of pain (anxiety, depression, and anhedonia)

could be dysregulated. Previous studies evaluating the absence of tolerance of

CB2 agonists failed to evaluate the affective component of pain.

To fully understand why CB2R agonists do not cause obvious tolerance, it

is important to increase our understanding of the mechanisms through which

84

they produce anti-nociception and possible role of changes in the affective

component of pain and/or sex-differences may shed light into this lack of

tolerance to sensory stimulation. First, the anti-nociceptive effect of JWH-133

might be caused by the ability of CB2R activation to suppress inflammation at the

site of injury [Ehrhart 2005; Benito 2008]. For example, paclitaxel-induced

neuropathic pain in rats and mice can be prevented if the CB2R agonist, MDA7 is

co-administered during treatment with paclitaxel [Naguib 2012]. Rats receiving

MDA7 did not display the expected increase of markers for microglial and

astrocyte activation in the spinal cord that are associated with inflammatory

response during paclitaxel treatment. One possible explanation for this finding is

that CB2R activation blocked the normal inflammatory response to paclitaxel,

thus preventing inflammation, nerve damage, and pain.

Beyond mediating inflammation, a second proposed mechanism for this

crosstalk between cannabinoid and the opioid systems is agonist-stimulated

release of endogenous ligands. We have directly examined the possibility that

JWH-133-mediated anti-nociception might be mediated through endogenous

opioid release. Our results indicate that anti-nociceptive efficacy of JWH-133 was

not prevented by antagonism of MOR with naloxone (Figure 6). This is consistent

with previous studies using CB2R agonist GW405833 to reduce CFA-induced

inflammatory allodynia in the presence of naltrexone, another MOR antagonist

[Whiteside 2005]. These findings suggest that certain CB2R agonists (including

JWH-133) can attenuate inflammatory pain through a MOR-independent

mechanism. Differences in the ability of CB2R agonists to stimulate endogenous

85

opioid release might be due to signaling bias. For example, JWH-133 appears to

be strongly biased towards G protein dependent mechanisms and biased against

signaling through β-arrestin recruitment [Atwood 2012; Dhopeshwarkar 2016;

Soethoudt 2017]. However, there are other mechanisms of action that could

explain these results including morphine-stimulated endocannabinoid release

and/or morphine-stimulated down-regulation of CB2R.

Despite the lack of tolerance to the anti-nociceptive effects of JWH-133,

we observe morphine-induced cross-tolerance for JWH-133 following repeated

daily injections of morphine (Figure 9). However, this effect is unidirectional since

repeated daily injections of JWH-133 do not cause cross-tolerance to a challenge

dose of morphine. Previous work has demonstrated that tolerance to the anti-

nociceptive and anti-allodynic effects of morphine require JNK signaling [Melief

EJ 2010; Hervera 2012; Marcus 2015]. Therefore, examined the possibility that

morphine-induced cross-tolerance to JWH-133 was also mediated through a JNK

signaling mechanism. We found that this cross-tolerance to JWH-133 is only

partially attenuated by SP600125, a broad-spectrum inhibitor of all three JNK

isoforms (Figure 11). However, there are other mechanisms of action that could

explain these results including morphine-stimulated endocannabinoid release

and/or morphine-stimulated down-regulation of CB2R. Thus, the specific

mechanism of action for opioid-induced cross-tolerance to JWH-133 is not

understood and requires additional study.

Interestingly, our results suggest that JWH-133 co-administration may be

protective against the development of morphine tolerance (Figure 10). This

86

finding is in agreement with a previous study of cancer pain that showed a delay

in morphine tolerance when morphine was co-administered with a sub-analgesic

dose of AM-1241 [Zhang 2016]. While intriguing, the magnitude of the JWH-133

effect on morphine tolerance is quite modest in our study.

Co-administration of CB2R agonists with opioids may also result in

increased anti-nociceptive efficacy due to drug synergism. One potential benefit

of a synergistic interaction between JWH-133 is that it could allow effective anti-

nociception with considerably lower morphine doses. Such an opioid-sparing

effect for JWH-133 could help minimize the adverse effects of opioids. Human

studies have shown increased pain relief in patients when inhaled cannabis use

is combined with opioids [Abrams 2011]. Therefore, we performed isobolographic

analysis of morphine and JWH-133 in combination to test for potential synergy

(Figure 12). We were able to demonstrate an additive effect of this combination,

but were not able to demonstrate a greater-than-additive (synergistic) effect.

However, only one fixed ratio (10:1) drug combination was used. Nonetheless,

even an additive effect of CB2R agonists could be beneficial due to the lack of

adverse effects [Stone 2014].

Conclusion

Our study demonstrates unidirectional morphine-induced cross-tolerance

to JWH-133. This finding raises the possibility that CB2R may functionally

interact with the opioid system to modulate inflammatory pain. Interestingly, we

also find that co-administration of JWH-133 with morphine produces an additive

87

anti-nociceptive effect in the formalin test while also potentially protecting against

morphine tolerance. Taken together, these findings highlight the potential

therapeutic applications for different CB2R ligands in pathological pain states

without tolerance or adverse effects associated with currently available

treatments. In particular, further investigation into the use of CB2R agonist as

adjuvant treatments to opioid therapy should be investigated.

88

Unpublished Experiment 1: Efficacy of JWH-133 in hotplate and tail-flick assays

Rationale

Used together, the hotplate and tail flick assays serve to measure acute

thermal nociceptive thresholds in subjects tested. While both utilize heat as a

noxious stimuli, tail flick elicits a more reflexive response at the level of the spinal

cord and hotplate involves more cognitive and emotional components [Espejo

1993]. These assays are beneficial as preliminary measurements of a drug’s

anti-nociceptive efficacy for several reasons. These assays can be performed

quickly and without any injury or lasting effects on the subjects, and are simple to

perform. However, acute pain as a result of noxious stimuli is entirely different

from clinically relevant chronic and pathological pain modalities. Furthermore, the

possibility of a learning effect often makes interpretation of long-term testing in

these assays difficult. They could still be valuable in the case of JWH-133 as the

literature surrounding this agonist in pain models is so limited.

There is considerable clinical evidence that opiate analgesics differentially

affect responses to pain produced by brief noxious stimulation and those

associated with persistent pain states [Fields 2004]. Acute pain is usually highly

sensitive to opiate therapy yet high doses of opiates are required to relieve

inflammatory and neuropathic pain states because they are longer term and

involve sensitization [Basbaum 2009]. Similar results are observed with CB1R-

selective agonists [Liu 2006; da Fonseca Pacheco 2008]. However, CB2R may

89

offer a more effective target in pathological pain relative to acute pain due to its

role in inflammatory modulation. Therefore, comparison of JWH-133 effect in

thermal and inflammatory models may be informative.

Procedure

The anti-nociceptive effect of JWH-133 was assessed in male mice by

performing dose-response analyses in the hotplate and tail flick tests as

described previously. Animals (n=6 per dose) were given log-scale doses of the

CB2R agonist JWH-133 ( 0.01, 0.1, 0.3, 1, 3 mg/kg) via i.p. injection. Post-drug

measurements were taken 60 min following JWH-133 administration in both

assays. Values are reported as the percentage of the maximal possible effect

(%MPE).

Results

In the hotplate test, JWH-133 increased %MPE in a dose-dependent

fashion [F(4,29)=7.1, p=0.0006] until a maximum value of 20±4 %MPE was

reached (Figure 14). Similarly, JWH-133 had a dose-dependent effect in the tail

flick test [F(4,29)=4.9, (Figure 14)} up to a maximum of 15±4 %MPE.

90

Figure 14. JWH-133 dose responses in acute thermal pain

Animals were given log-scale doses of JWH-133 ( 0.01, 0.1, 0.3, 1, 3 mg/kg) via i.p. injection. Data are expressed as mean±SEM, and p<0.05 (*) is considered significant. (ANOVA with Bonferonni post-hoc, JWH-133 vs. vehicle).

91

Discussion

While JWH-133 demonstrated anti-nociceptive efficacy in both models of

acute pain, the results and possible analyses are limited. Even at maximal doses,

the highest effect was between 15-20% MPE. Comparatively, opioids such as

morphine and fentanyl can demonstrate 100% MPE (Figures 2, 3, 15).

Moreover, the shift from almost no effect to maximal effect occurred over only 1

order of magnitude increase in dose. The low effect for JWH-133 suggests that

using these assays to measure subtle changes or interactions with other

compounds would not be worthwhile.

92

Unpublished Experiment 2: JWH-133 and fentanyl cross-tolerance.

Rationale

Tolerance to fentanyl, another mu opioid receptor agonist, is not affected

by pre-treatment with SP600125 (figure 16, [Marcus 2015]). Interestingly,

tolerance to fentanyl is absent in G protein-coupled receptor kinase (GRK) 3 KO

mice suggesting that tolerance for this agonist is mediated exclusively through a

classic mechanism involving GRK 3 phosphorylation and β-arrestin2 recruitment

[Melief 2010]. Because cross-tolerance with morphine appeared to be partially

dependent upon JNK signaling, we wanted to examine interaction with an opiate

that desensitizes through non-JNK mechanisms. Therefore, we examined

whether daily fentanyl injections could cause cross-tolerance to JWH-133.

Procedure

Following the procedures for morphine tolerance, male mice were injected

(i.p.) once-daily with either 0.3 mg/kg fentanyl or 1 mg/kg JWH-133 for up to eleven

consecutive days. The mice were assessed in the formalin model 60 min following

drug administration on the final day. To assess cross-tolerance between fentanyl

and JWH-133, some groups were given a challenge dose of the alternate drug

(i.e., mice given 10 days of fentanyl were given a JWH-133 challenge and vice

versa) on the eleventh day. Some groups were co-administered 0.3 mg/kg

fentanyl and 1 mg/kg JWH-133 for ten days and then tested for anti-nociception

93

using morphine alone on day 11 to assess the potential protective effects of JWH-

133 co-administration on morphine tolerance.

Results

We found that fentanyl also causes cross-tolerance to JWH-133, and that

it is similar in magnitude to what we observe for morphine. In these mice,

analysis of the AUC of pain behavior revealed that only the repeated vehicle with

JWH-133 challenge group produced anti-nociception relative to the control or

repeated morphine + JWH-133 challenge groups in both Phase 1 (F(2,14) = 22.39,

p < 0.0001; Figure 15) and Phase 2 (F(2,14) = 6.57, p < 0.01; Figure 15) of the

formalin test.

94

Figure 15. JWH-133 and fentanyl cross tolerance.

The formalin model of inflammatory pain was utilized to examine cross-tolerance between fentanyl and JWH-133. The AUCs for acute and inflammatory pain are shown for male mice that received a challenge dose of JWH-133 after 10 days of repeated fentanyl administration Data are expressed as mean ± S.E.M. (n = 5-8 per group). *** P < 0.0001 vs control group (ANOVA with Bonferroni post-hoc); ** P < 0.01 vs control (ANOVA with Bonferroni post-hoc).

95

Discussion

While chronic morphine administration diminishes CB2R agonist efficacy,

the reverse effect is not observed (Figure 9). Similar trends were observed with

JWH-133 and fentanyl (Figure 15). The results of these experiments suggest that

there is unidirectional cross-tolerance between CB2R agonists and opioids. Our

lab and others have demonstrated that the mechanisms governing tolerance to

morphine and fentanyl occur through different cellular mechanisms [Melief EJ

2010; Hervera 2012; Marcus 2015]. Furthermore, we found that the cross

tolerance of JWH-133 due to chronic morphine was partially attenuated by the

addition of the JNK inhibitor SP6 (Fig 11.) This agrees with previous findings

showing that JNKs mediate morphine tolerance, and also suggests that they are

also involved in the mechanism causing this tolerance to cross over to JWH-133

(Figure 16 below, [Marcus 2015]). Thus the specific mechanism of action for

opioid-induced cross-tolerance to JWH-133 is not understood and requires

additional study.

96

97

Figure 16. Tolerance to the anti-nociceptive effects of fentanyl is not

blocked by SP6.

JNK inhibition attenuates chronic tolerance to the hypothermic but not the anti-nociceptive effects of repeated treatment with fentanyl (0.3 mg/kg). Wild-type mice were treated with vehicle (black line with triangles), 3 mg/kg SP600125 (dashed black line with diamonds), or 10 mg/kg SP600125 (black line with diamonds) 60 min prior to administration of fentanyl (0.3 mg/kg) for ten consecutive days. Tail-flick anti-nociception (a), hotplate nociception (b), and body temperature (c) were measured 60 min later. Treatment with only SP600125 (3 mg/kg) attenuated tolerance to the hypothermic effects of chronically administered 0.3 mg/kg fentanyl alone. Data are expressed as mean ± S.E.M. (n = 10–24 per group). *p < 0.0001 for SP600125 (3 mg/kg) vs. vehicle or SP600125 (10 mg/kg) groups (ANOVA, Bonferroni post hoc).

98

Chapter 5 – Mechanisms of Cannabinoid Tolerance through the CB1 Receptor

Δ9-THC produces potent anti-nociceptive effects but, like opiates, is

subject to the onset of tolerance [Morgan 2014]. As discussed, there is also

overlap in the mechanisms of CB1R and opioid desensitization. The objective of

this study was to examine potential mechanisms of tolerance from daily

administration of Δ9-THC using a model of pathological pain. As with MOR,

CB1R desensitization could involve GRK or JNK phosphorylation. S426A/S430A

mutant mice express two serine to alanine point mutations on the CB1R sites

phosphorylated by GRK, and exhibit enhanced response to Δ9-THC [Morgan

2014]. Therefore, we used the formalin test to compare the development of Δ9-

THC tolerance in these animals and in wild-type mice. Additionally, The impact of

c-Jun N-terminal kinase (JNK) inhibitor SP600125 (SP6) on the development of

Δ9-THC tolerance was also assessed in wild-type mice, as previous research has

suggested it is significant in the development of tolerance to morphine [Marcus

2015].

99

Results

Δ9-THC Tolerance in S426A/S430A mice

Male wild-type C57BL/6 mice, or S426A/S430A mutant mice were

subjected to the formalin test after receiving daily injections of Δ9-THC (6 mg/kg)

ranging from zero to twelve days (Figure 17). Wild-type mice exhibited complete

tolerance to the anti-nociceptive effects of Δ9-THC in the formalin test after eight

days of daily Δ9-THC. These results are consistent with the findings of studies

assessing Δ9-THC tolerance in other models of pain, which often show onset of

tolerance by three days of chronic administration. Interestingly, we find that Wild-

type mice show development of tolerance in inflammatory pain, however,

S426A/S430A mice showed reduced tolerance to Δ9-THC specifically for

inflammatory pain.

SP6 impairs Δ9-THC tolerance in the formalin test

In a second experiment, groups of wild-type mice were also tested for up

to twelve days receiving SP6 (3 mg/kg), Δ9-THC (6 mg/kg), or SP6 and Δ9-THC

(Figure 18). Anti-nociception was then measured in the formalin test to determine

the role of JNK signaling in the acquisition of tolerance. Administration of SP6

alone provides effective anti-nociception in both pain types with no observed

tolerance within 12 days of chronic administration. Moreover, co-administration of

SP6 with Δ9-THC reduces the tolerance developed to Δ9-THC in both acute (A),

and inflammatory (B) pain.

100

Figure 17. Δ9-THC tolerance in the formalin test.

Wild-type (n=34, black) and S426A/S430A mutant (n=27, red) mice were given chronic daily doses of Δ9-THC (6 mg/kg, i.p). The anti-nociceptive efficacy of Δ9-THC was then assessed using the formalin model, which allows measurement of both acute (A) and inflammatory (B) pain.

101

Figure 18. Δ9-THC and SP6 in the formalin test.

Male wild-type mice (n=23) were given chronic daily injection of either vehicle (black), SP600125 (SP6) alone (3 mg/kg, red), Δ9-THC (6 mg/kg, blue), or SP6 in combination with Δ9-THC (6 mg/kg, white). The formalin model was used to assess acute (A) and inflammatory (B) pain.

102

Chapter 6 - General Discussion and Conclusion

The primary goal of this study was to test the hypothesis that CB2R

functionally interacts with the opioid system to modulate inflammatory pain.

Additionally, we tested mechanisms mediating tolerance to multiple opioids, and

to the prototypical cannabinoid Δ9-THC.

First, we examined the effects of CB2R activation alone both in acute and

inflammatory pain. In the case of our study, we found mild anti-nociceptive effect

in acute thermal pain (Figure 4). Moreover, we found that JWH-133 causes

considerable dose-dependent anti-nociception in both the acute and

inflammatory phases of the formalin test (Figures 5&6). As discussed, there are

considerable discrepancies among the literature investigating the potential

mechanisms and location of action for the anti-nociceptive effects of CB2R

agonists [Malan TP Jr 2001; Cichewicz 2004]. In the case of our study, the first

possibility is inhibition of inflammation at the site of formalin injection. The

efficacy of JWH-133 in inflammatory pain compared to the limited efficacy in the

tail-flick and hotplate tests (Figure 4) suggests that the anti-nociceptive effect of

JWH-133 is related to reduction of inflammation [Ehrhart 2005; Benito 2008].

This is further supported by the literature. For example, paclitaxel-induced

neuropathic pain in rats and mice can be prevented if the CB2R agonist, MDA7 is

co-administered during treatment with the chemotherapeutic agent paclitaxel

[Naguib 2012]. Rats receiving MDA7 did not display the expected increase of

markers for microglial and astrocyte activation in the spinal cord that are

103

associated with inflammatory response during paclitaxel treatment. One possible

explanation for this finding is that CB2R activation blocked the normal

inflammatory response to paclitaxel, thus preventing inflammation, nerve

damage, and pain. Rats subjected to the spared nerve injury procedure were

found to self-administer AM-1241 for its anti-allodynic effects [Gutierrez 2011].

The dual CB1/CB2 agonist CP55,940 was found to dose-dependently reduce

paclitaxel-induced allodynia in CB1 KO mice [Deng 2015a]. Another CB2R-

selective agonist, AM-1710, was found to completely reverse paclitaxel-induced

neuropathic pain in mice [Deng 2015b]. Neuropathic allodynia can not only be

reduced, but also reversed by a CB2R agonist (MDA7) through reduction of

microglial activity and cytokine release [Naguib 2012]. Similarly, CB2R agonist O-

3223 reduces LPS-induced paw edema [Kinsey 2011]. Conversely, we did not

see a difference in formalin-induced paw edema between animals given JWH-

133 or vehicle as measured by microcaliper (appendix, Table 4), which agrees

with literature showing that JWH-133 also does not diminish λ-carrageenan-

induced inflammation [Elmes 2004]. However, this does not rule out mediation of

inflammation as a possibility, as paw edema is a very gross measure of

inflammation. More quantitative measurements, such as cytokine levels in tissue

of treated animals would be a more informative test to address mechanism.

A second potential mechanism is mediation of spinal inflammatory

response and sensitization. CB2R agonists could activate CB2R on mast or other

immune cells in the spine, thereby decreasing the inflammation-evoked release

104

of sensitizing molecules such as NGF, cytokines, or histamine. This could

decrease the sensitivity of primary afferent neurons and inhibit pain response.

It has been shown that Microglial CBR2 activation inhibits pERK in spinal cord,

which results in a reduction of TNF expression and microglial motility. Overall,

this would result in a reduction in pro-algesic factors that would create

hyperalgesia or allodynia in response to pathology.

An alternative explanation for CB2 receptor-mediated attenuation

of acute nociceptive responses is the putative presence of CB2R on primary

afferent fibers. Currently, there is conflicting evidence for the expression of CB2R

receptors on dorsal root ganglia [Hohmann 1999; Ross 2001]. However, it has

been demonstrated that intraplantar injection of JWH-133 directly inhibited

mechanically evoked responses of rat dorsal horn neurons [Elmes 2004].

Some studies suggest that CB2R activation triggers endogenous opioid

release in the spinal cord [Ibrahim 2005; Curto-Reyes 2010]. In our case, the

anti-nociceptive efficacy of JWH-133 was not prevented by antagonism of MOR

with naloxone (Figure 7). Similarly, the CB2R agonist GW405833 reduces CFA-

induced inflammatory allodynia even with co-administration of naltrexone,

another MOR antagonist [Whiteside 2005]. These findings suggest that certain

CB2R agonists (including JWH-133) can attenuate inflammatory pain through a

MOR-independent mechanism. Similarly, the inability of a CB1R antagonist to

block the effect of JWH-133 in our study reinforces that the mechanism occurs

directly through CB2R.

105

Differences between these studies may be attributable to signaling biases

of specific ligands. In recent years, mounting evidence of the importance for

CB2R in pathological pain has increased interest as demonstrated by the

synthesis of a variety of CB2R-selective cannabinoid agonists [Atwood 2010].

JWH-133 appears to be strongly biased towards G protein dependent

mechanisms and biased against signaling through β-arrestin recruitment;

meaning that this agonist also very weakly desensitizes and internalizes CB2R

[Dhopeshwarkar and Mackie 2016]. JWH-133 is among the most CB2R-selective

cannabinoids, and recent analysis found that it has no activity at any off-target

proteins [Soethoudt 2017].

In addition to mechanism, there is also the question of location of effect.

Numerous studies have suggested ability of CB2R agonists to reduce allodynia

and hyperalgesia is spinally mediated, and that intrathecal administration is

effective [Romero-Sandoval 2008; Yamamoto 2008; Curto-Reyes 2010; Deng

2015b]. Conversely, some studies report CB2R agonists show efficacy from

systemic but not intrathecal administration [Brownjohn and Ashton 2012]. As

such, caution should be taken to assume the same mechanism of action across

different CB2R ligands or pain modalities. This further suggests that more

localized administration of JWH-133 should be performed in the future.

Additionally, we sought to examine potential sex differences in JWH-133

anti-nociception between males and females using the formalin test. The finding

that females generally exhibit more pain behaviors is consistent with animal and

human studies of pain [Cepeda and Carr 2003; Fillingim 2009; Bergeson 2016;

106

Blanton 2016; Henderson-Redmond 2016]. Our study did not suggest that female

mice were more sensitive to JWH-133, as the calculated ED50 values and

maximally efficacious doses did not differ between male and female mice.

However, there is evidence for sex differences in the response to cannabinoids.

Previous studies have shown that female rats are more sensitive to Δ9-THC, and

develop tolerance to it more quickly [Wakley 2014]. Female rats also exhibit more

severe adverse effects (sedation, catalepsy) and precipitated withdrawal from Δ9-

THC [Marusich 2014]. In human studies, the same trends (faster onset of

tolerance, more severe adverse effects) are reported for female cannabis

smokers (for review, see [Craft 2013]). It is frequently suggested that this is due

to a higher percentage of adipose tissue in females. Cannabinoids are highly

lipophilic and stored in adipose cells.

Regardless of the pain model used, multiple studies have observed a lack

of tolerance to repeated administration of CB2R agonists. For example, daily

systemic administration of AM-1710 attenuated chemotherapy-evoked

neuropathic pain for up to 8 days with no evidence of tolerance [Deng 2015b].

Similarly, the CB2R agonist, JWH015 (intrathecal, once daily), reversed surgery-

induced allodynia for up to 5 days without tolerance [Romero-Sandoval 2008]. In

this study we report a lack of tolerance to the anti-nociceptive effects of JWH-133

after 11 consecutive days of administration (Figure 9). This duration of dosing in

the formalin model was sufficient to generate complete tolerance to morphine,

fentanyl, and Δ9-THC. However, it is possible that tolerance to JWH-133 may

occur with longer periods of chronic dosing and/or the affective component of

107

pain (anxiety, depression, and anhedonia) could be dysregulated. Previous

studies evaluating the absence of tolerance of CB2R agonists failed to evaluate

the affective component of pain.

Previous work has demonstrated that tolerance to the anti-nociceptive and

anti-allodynic effects of morphine require JNK signaling [Melief EJ 2010; Hervera

2012; Marcus 2015]. Recent work has also shown that tolerance to morphine is

attenuated through the use of the JNK inhibitor SP600125 [Chen 2008; Guo

2009; Hervera 2012; Marcus 2015]. SP600125 is an anthrapyrazolone capable of

inhibiting JNK1, JNK2, and JNK3 with high affinity [Zhuang 2006]. It has been

demonstrated that systemic administration of SP600125 prevents

phosphorylation of JNK in the spinal cord, resulting in attenuation of tolerance to

the anti-nociceptive and anti-allodynic effects of morphine [Guo 2009; Hervera

2012]. We have also previously demonstrated that multiple JNK isoforms

mediate morphine tolerance [Yuill 2016]. We also demonstrated that inhibition of

JNK attenuates the onset of tolerance to Δ9-THC. Therefore, we wanted to

examine the possibility that morphine-induced cross-tolerance to JWH-133 was

also mediated through a JNK signaling mechanism. We found that this cross-

tolerance to JWH-133 is only partially attenuated by SP600125, a broad-

spectrum inhibitor of all three JNK isoforms (Figure 12). Moreover, there was

cross-tolerance between JWH-133 and fentanyl, despite fentanyl tolerance being

JNK-independent (Figure 15). However, there are other mechanisms of action

that could explain these results including morphine-stimulated endocannabinoid

release and/or morphine-stimulated down-regulation of CB2R.

108

Interestingly, our results suggest that JWH-133 co-administration may be

protective against the development of morphine tolerance (Figure 11). This

finding is in agreement with a previous study of cancer pain that showed a delay

in morphine tolerance when morphine was co-administered with a sub-analgesic

dose of AM-1241 [Zhang 2016]. While intriguing, the magnitude of the JWH-133

effect on morphine tolerance is quite modest in our study. Furthermore, our

results do not suggest any specific mechanisms to explain the effect. However,

one possibility is that CB2R activation might suppress the release of microglial

cytokines involved in the development of opioid tolerance [Watkins 2009]. JWH-

133 blocks JNK phosphorylation on microglia [Correa 2010]. In vitro models

suggest that opioid mediated pro-inflammatory cytokine release is attenuated

through concurrent CB2R activation [Merighi 2012]. This protective effect of

CB2R against opioid tolerance raises the possibility that they might be useful as

an adjuvant to traditional opiate treatment. It should also be noted that

Cannabioids do impact cytochrome-mediated metabolism, which could alter the

pharmacokinetic profile of opioids [Abrams 2011].

Co-administration of CB2R agonists with opioids may also result in

increased anti-nociceptive efficacy due to drug synergism. One potential benefit

of a synergistic interaction between JWH-133 is that it could allow effective anti-

nociception with considerably lower morphine doses. Therefore, we performed

isobolographic analysis of morphine and JWH-133 in combination to test for

potential synergy (Figure 13). We were able to demonstrate an additive effect of

this combination, but were not able to demonstrate a greater-than-additive

109

(synergistic) effect. However, only one fixed ratio (10:1) drug combination was

used. Nonetheless, even an additive effect of CB2R agonists could be beneficial

due to the lack of adverse effects [Stone 2014].

Future directions

While both inflammatory pain and neuropathy are types of pathological

pain, literature suggests that CB2R may impact the two differently [Zhang 2003].

Rats subjected to the spared nerve injury procedure were found to self-

administer AM-1241 for its anti-allodynic effects [Gutierrez 2011]. The dual

CB1R/CB2R agonist CP55,940 was found to dose-dependently reduce

paclitaxel-induced allodynia in CB1RKO mice [Deng 2015a]. Another CB2R-

selective agonist, AM-1710, was found to completely reverse paclitaxel-induced

neuropathic pain in mice [Deng 2015b]. Interestingly, continuous administration

of AM-1710 before and after paclitaxel treatment can prevent the development of

neuropathic pain for several weeks [Rahn 2014]. Due to the marked differences

in physiology between neuropathic and inflammatory pain our results from the

formalin test should be replicated in models of neuropathy.

From a scientific perspective, it is also important to investigate the

mechanisms governing CB2R/MOR interactions. Measuring in vivo changes to

CB2R expression and activity may explain any observed cross-tolerance

between CB2R and MOR agonists. Tissue from mice given chronic morphine

could be analyzed to measure changes in CB2R mRNA and protein expression,

110

as well as CB2R binding and G-protein coupling. Previous studies have

demonstrated that changes in CB2R expression during pathological conditions

are measurable in the dorsal root ganglia (DRG [Svizenska 2013]). As CB2R

functions by coupling to Gi/o protein [Izzo 2001], measurement of G protein

activity using agonist-stimulated [35S]GTPγS binding assays would provide a

direct measurement of CB2R desensitization. This technique has been used to

measure CB2R coupling and desensitization previously [Bouaboula 1999; Marini

2013].

Conclusion

The results of these experiments suggest that there is unidirectional cross-

tolerance between CB2R agonists and multiple different opioids. While chronic

morphine administration diminishes CB2R agonist efficacy, the reverse effect is

not observed. Overall these findings suggest that CB2R may functionally interact

with the opioid system to modulate anti-nociception in the formalin test in

response to inflammatory pain. This information will provide context for the

circumstances under which CB2R agonists may prove beneficial clinically, as

well as limitations to their use. Diminished efficacy of CB2R agonists due to

opiate tolerance demonstrates the importance of applying them proactively as an

adjuvant to opiates rather than as a reactionary measure after opiates have been

administered. Furthermore, co-administration of a CB2R agonist with morphine

111

suggests both anti-nociceptive synergy and protection against tolerance. Further

investigation of this combination in treatment of pathological pain is warranted.

112

Appendix

Supplementary Data and Figures

Figure 19. Morphine with a fixed-dose of JWH-133 in the formalin test.

Dose-response of Morphine with a fixed-dose of JWH-133 in the formalin test. Fixed dose drug combinations were analyzed by comparing parallelism of the linear regression of the two curves [Suhnel 1998]. There was no significant shift of the curve or ED50 value for morphine due to 0.1 mg/kg JWH-133 (Table 3).

113

Table 3. ED50 Values in Formalin.

Drug Combination Phase I ED50 ( mg/kg + 95% CI)

Phase II ED50 ( mg/kg + 95% CI)

Morphine Alone 0.98 (0.89 to 1.08)

0.79 (0.54 to 0.98)

JWH-133 Alone 0.23 (0.89 to 1.08)

0.25 (0.89 to 1.08)

Morphine + 0.1 mg/kg fixed dose JWH-133

1.45 (1.20 to 1.70)

1.77 (1.51 to 2.04)

Experimental 1:10 JWH-133 and morphine fixed ratio

0.78 (0.61 to 0.90)

0.62 (0.54 to 0.72)

Theoretical 1:10 JWH-133 and morphine fixed ratio

0.72 (0.54 to 0.98)

0.93 (0.80 to 1.05)

Table 4. Paw edema following formalin.

Treatment Paw edema (% increase in thickness)

JWH-133 (1 mg/kg) 39.9 ± 6.6

JWH-133 (10 mg/kg) 36.1 ± 5.4

Morphine (10 mg/kg) 42.2 ± 5.8

Vehicle 38.9 ± 3.8

Paw edema (Table 4) was compared in mice (n=4-5 per group) receiving vehicle, JWH-133 (1 mg/kg), and morphine (10 mg/kg) to see if JWH-133 measurably reduced formalin inflammation. Thickness of the base of the right hind paw was measured immediately before and following testing using a digital micrometer.

114

Works Cited

Abrams, D. I., P. Couey, S. B. Shade, M. E. Kelly and N. L. Benowitz (2011).

"Cannabinoid-opioid interaction in chronic pain." Clin Pharmacol Ther

90(6): 844-851.

Akil, H., Mayer, DJ, Liebeskind, JC. (1976). "Antagonism of stimulation-produced

analgesia by naloxone, a narcotic antagonist." Science 5(191): 961-962.

Almeida, T. F., S. Roizenblatt and S. Tufik (2004). "Afferent pain pathways: a

neuroanatomical review." Brain Res 1000(1-2): 40-56.

Ashton, J. C., Friberg D, Darlington CL, Smith PF (2006). "Expression of the

cannabinoid CB2 receptor in the rat cerebellum: an immunohistochemical

study." Neurosci Lett(396): 113-116.

Ashton, J. C., R. M. Rahman, S. M. Nair, B. A. Sutherland, M. Glass and I.

Appleton (2007). "Cerebral hypoxia-ischemia and middle cerebral artery

occlusion induce expression of the cannabinoid CB2 receptor in the brain."

Neurosci Lett 412(2): 114-117.

Atwood, B. K., & Mackie, K. (2010). "CB2: a cannabinoid receptor with an identity

crisis." Br J Pharmacol 160: 467-479.

Atwood, B. K., J. Wager-Miller, C. Haskins, A. Straiker and K. Mackie (2012).

"Functional selectivity in CB(2) cannabinoid receptor signaling and

regulation: implications for the therapeutic potential of CB(2) ligands." Mol

Pharmacol 81(2): 250-263.

115

Bachhuber, M. A., B. Saloner, C. O. Cunningham and C. L. Barry (2014).

"Medical cannabis laws and opioid analgesic overdose mortality in the

United States, 1999-2010." JAMA Intern Med 174(10): 1668-1673.

Baek, J. H., C. L. Darlington, P. F. Smith and J. C. Ashton (2013). "Antibody

testing for brain immunohistochemistry: brain immunolabeling for the

cannabinoid CB(2) receptor." J Neurosci Methods 216(2): 87-95.

Bailey, C., J. Llorente, B. Gabra, F. Smith, W. Dewey, E. Kelly and G. Henderson

(2009). "Role of protein kinase C and mu-opioid receptor (MOPr)

desensitization in tolerance to morphine in rat locus coeruleus neurons."

Eur J Neurosci 29(2): 307-318.

Basbaum, A. I., D. M. Bautista, G. Scherrer and D. Julius (2009). "Cellular and

molecular mechanisms of pain." Cell 139(2): 267-284.

Beasou, P. (1969). "Response of cutaneous sensory units with unmyelinated

fibers to noxious stimuli." J Neurophysiol 32: 1025-1043.

Beltramo, M., N. Bernardini, R. Bertorelli, M. Campanella, E. Nicolussi, S.

Fredduzzi and A. Reggiani (2006). "CB2 receptor-mediated

antihyperalgesia: possible direct involvement of neural mechanisms." Eur

J Neurosci 23(6): 1530-1538.

Benito, C., R. M. Tolon, M. R. Pazos, E. Nunez, A. I. Castillo and J. Romero

(2008). "Cannabinoid CB2 receptors in human brain inflammation." Br J

Pharmacol 153(2): 277-285.

Bennett, B. L., D. T. Sasaki, B. W. Murray, E. C. O'Leary, S. T. Sakata, W. Xu, J.

C. Leisten, A. Motiwala, S. Pierce, Y. Satoh, S. S. Bhagwat, A. M.

116

Manning and D. W. Anderson (2001). "SP600125, an anthrapyrazolone

inhibitor of Jun N-terminal kinase." Proc Natl Acad Sci U S A 98(24):

13681-13686.

Bergeson, S. E., H. Blanton, J. M. Martinez, D. C. Curtis, C. Sherfey, B.

Seegmiller, P. C. Marquardt, J. A. Groot, C. L. Allison, C. Bezboruah and

J. Guindon (2016). "Binge Ethanol Consumption Increases Inflammatory

Pain Responses and Mechanical and Cold Sensitivity: Tigecycline

Treatment Efficacy Shows Sex Differences." Alcohol Clin Exp Res 40(12):

2506-2515.

Bernard, J., Huang, G., Besson, J. (1994). "The parabrachial area:

electrophysiological evidence for an involvement in visceral nociceptive

processes." J Neurophysiol 71: 1646-1660.

Blanton, H. e. a. (2016). "Gender-specific pain responses in the formalin test

using a synthetic tetracycline compound." J Pain 17(4): 50-51.

Bloom, A. S. and W. L. Dewey (1978). "A comparison of some pharmacological

actions of morphine and delta9-tetrahydrocannabinol in the mouse."

Psychopharmacology (Berl) 57(3): 243-248.

Bogoyevitch, M. and B. Kobe (2006). "Uses for JNK: the many and varied

substrates of the c-Jun N-terminal kinases." Microbiol Mol Biol Rev 70(4):

1061-1095.

Bouaboula, M., Desnoyer, N., Carayon, P., Combes, T., and Casellas, P. (1999).

"Gi Protein Modulation Induced by a Selective Inverse Agonist for the

Peripheral Cannabinoid Receptor CB2: Implication for

Intracellular Signalization Cross-Regulation." Mol Pharmacol 55: 473-480.

117

Braden, J. B. and M. D. Sullivan (2008). "Suicidal thoughts and behavior among

adults with self-reported pain conditions in the national comorbidity survey

replication." J Pain 9(12): 1106-1115.

Brownjohn, P. W. and J. C. Ashton (2012). "Spinal cannabinoid CB2 receptors as

a target for neuropathic pain: an investigation using chronic constriction

injury." Neuroscience 203: 180-193.

Buckley, N. E., McCoy,K.L., Mezey,E., Bonner,T., Zimmer,A., Felder,C.C.,

Glass,M., and Zimmer,A. (2000). "Immunomodulation by cannabinoids is

absent in mice deficient for the cannabinoid CB(2) receptor." Eur J

Pharmacol(396): 141-149.

Burgess, P. R. a. P., E.R. (1967). "Myeiinated afferent fibres responding

specifically to noxious stimulation on the skin." J. Physiol. (Lond.)(190):

541-562. .

Calignano, A., La Rana G, Giuffrida A, Piomelli D (1998). "Control of pain

initiation by endogenous cannabinoids." Nature(394): 277-281.

Canals, M. and G. Milligan (2008). "Constitutive activity of the cannabinoid CB1

receptor regulates the function of co-expressed Mu opioid receptors." J

Biol Chem 283(17): 11424-11434.

Carayon, P., J. Marchand, D. Dussossoy, J. M. Derocq, O. Jbilo, A. Bord, M.

Bouaboula, S. Galiegue, P. Mondiere, G. Penarier, G. L. Fur, T. Defrance

and P. Casellas (1998). "Modulation and functional involvement of CB2

peripheral cannabinoid receptors during B-cell differentiation." Blood

92(10): 3605-3615.

118

Carlisle, S. J., F. Marciano-Cabral, A. Staab, C. Ludwick, G.A. Cabral (2002).

"Differential expression of the CB cannabinoid receptor by 2 rodent

macrophages and macrophage-like cells in relation to cell activation." Int

Immunopharm(2): 69-82.

CDC (2011). "Vital signs: Overdoses of prescription opioid pain relievers –United

States, 1999-2008." Morbidity and Mortality Weekly Report 60 1487-1492.

CDC (2017). Wide-ranging online data for epidemiologic research (WONDER).

Atlanta, GA: CDC,, National Center for Health Statistics.

Cepeda, M. S. and D. B. Carr (2003). "Women Experience More Pain and

Require More Morphine Than Men to Achieve a Similar Degree of

Analgesia." Anesthesia & Analgesia: 1464-1468.

Chen, Y., C. Geis and C. Sommer (2008). "Activation of TRPV1 contributes to

morphine tolerance: involvement of the mitogen-activated protein kinase

signaling pathway." J Neurosci 28(22): 5836-5845.

Cichewicz, D. L. (2004). "Synergistic interactions between cannabinoid and

opioid analgesics." Life Sci 74(11): 1317-1324.

Clayton, N. M. W., A.W.; Collins, S.D.; Giblin, G.M.; Mitchell, B.L.; Goldsmith, P.;

Chessell, I.P.; O'Shaughnessy, C.; Bountra, C. (2004). "Anti-

hypersensitive and anti-inflammatory activity of the potent and selective

CB2 agonist GW842166X." Proc Brit Pharm Soc.

Coderre TJ, K. J. (1997). "Peripheral and central hyperexcitability: differential

signs and symptoms in persistent pain." Behav Brain Sci 20: 404-419.

119

Corbett, A., G. Henderson, A. McKnight and S. Paterson (2006). "75 years of

opioid research: the exciting but vain quest for the Holy Grail." Br J

Pharmacol 147 Suppl 1: S153-162.

Correa, F., M. Hernangomez, L. Mestre, F. Loria, A. Spagnolo, F. Docagne, V. Di

Marzo and C. Guaza (2010). "Anandamide enhances IL-10 production in

activated microglia by targeting CB(2) receptors: roles of ERK1/2, JNK,

and NF-kappaB." Glia 58(2): 135-147.

Cota, D., Marsicano G, Tschop M, et al (2003). "The endogenous cannabinoid

system affects energy balance via central orexigenic drive and peripheral

lipogenesis." J Clin Invest(112): 423-431.

Craft, R. M., J. A. Marusich and J. L. Wiley (2013). "Sex differences in

cannabinoid pharmacology: a reflection of differences in the

endocannabinoid system?" Life Sci 92(8-9): 476-481.

Curto-Reyes, V., Llames, S., Hidalgo, A., Menendez, L., and Baamonde, A.

(2010). "Spinal and peripheral analgesic effects of the CB2 cannabinoid

receptor agonist AM1241 in two models of bone cancer-induced pain." Br

J Pharmacol 160: 561-573.

da Fonseca Pacheco, D., A. Klein, A. de Castro Perez, C. M. da Fonseca

Pacheco, J. N. de Francischi and I. D. Duarte (2008). "The mu-opioid

receptor agonist morphine, but not agonists at delta- or kappa-opioid

receptors, induces peripheral antinociception mediated by cannabinoid

receptors." Br J Pharmacol 154(5): 1143-1149.

120

Deng, L., B. L. Cornett, K. Mackie and A. G. Hohmann (2015a). "CB1 Knockout

Mice Unveil Sustained CB2-Mediated Antiallodynic Effects of the Mixed

CB1/CB2 Agonist CP55,940 in a Mouse Model of Paclitaxel-Induced

Neuropathic Pain." Mol Pharmacol 88(1): 64-74.

Deng, L., J. Guindon, B. L. Cornett, A. Makriyannis, K. Mackie and A. G.

Hohmann (2015b). "Chronic cannabinoid receptor 2 activation reverses

paclitaxel neuropathy without tolerance or cannabinoid receptor 1-

dependent withdrawal." Biol Psychiatry 77(5): 475-487.

Derocq, J. M., Jbilo,O., Bouaboula,M., Segui,M., Clere,C., and Casellas,P.

(2000). "Genomic and functional changes induced by the activation of the

peripheral cannabinoid receptor CB2 in the promyelocytic cells HL-60.

Possible involvement of the CB2 receptor in cell differentiation." J Biol

Chem(275): 15621-15628.

Devane, W. A., Hanus, L., Breuer, A., Pertwee, R.G., Stevenson, L.A., Griffin, G.,

Gibson, D., Mandelbaum ,A., Etinger, A., and Mechoulam, R. (1992).

"Isolation and structure of a brain constituent that binds to the cannabinoid

receptor." Science(258): 1946-1949.

Dhopeshwarkar, A. and K. Mackie (2016). "Functional Selectivity of CB2

Cannabinoid Receptor Ligands at a Canonical and Noncanonical

Pathway." J Pharmacol Exp Ther 358(2): 342-351.

Di Rosa, M., Giroud JP, Willoughby DA. (1971). "Studies on the mediators of the

acute inflammatory response induced in rats in different sites by

carrageenan and turpentine." J Pathol 104: 15-29.

121

Doll, C., J. Konietzko, F. Poll, T. Koch, V. Hollt and S. Schulz (2011). "Agonist-

selective patterns of m-opioid receptor phosphorylation revealed by

phosphosite-specific antibodies." British Journal of Pharmacology(164):

298–307.

Dong, C., D. Yang, M. Wysk, A. Whitmarsh, R. Davis and R. Flavell (1998).

"Defective T Cell Differentiation in the Absence of JNK1." Science 282:

2092-2095.

Duncan, M., A. Mouihate, K. Mackie, C. M. Keenan, N. E. Buckley, J. S. Davison,

K. D. Patel, Q. J. Pittman and K. A. Sharkey (2008). "Cannabinoid CB2

receptors in the enteric nervous system modulate gastrointestinal

contractility in lipopolysaccharide-treated rats." Am J Physiol Gastrointest

Liver Physiol 295(1): G78-g87.

Ehrhart, J., D. Obregon, T. Mori, H. Hou, N. Sun, Y. Bai, T. Klein, F. Fernandez,

J. Tan and R. D. Shytle (2005). "Stimulation of cannabinoid receptor 2

(CB2) suppresses microglial activation." J Neuroinflammation 2: 29.

Eisenberg, E., M. Ogintz and S. Almog (2014). "The pharmacokinetics, efficacy,

safety, and ease of use of a novel portable metered-dose cannabis inhaler

in patients with chronic neuropathic pain: a phase 1a study." J Pain Palliat

Care Pharmacother 28(3): 216-225.

Elmes, S. J., M. D. Jhaveri, D. Smart, D. A. Kendall and V. Chapman (2004).

"Cannabinoid CB2 receptor activation inhibits mechanically evoked

responses of wide dynamic range dorsal horn neurons in naive rats and in

rat models of inflammatory and neuropathic pain." Eur J Neurosci 20(9):

2311-2320.

122

Espejo, E. M., D. (1993). "Structure of the rat's behaviour in the hot plate test."

Behavioural Brain Research 56: 171-176.

Facci, L., Dal Toso R, Romanello S, Buriani A, Skaper SD, Leon A (1995). "Mast

cells express a peripheral cannabinoid receptor with differential sensitivity

to anandamide and palmitoylethanolamide." Proc Natl Acad Sci U S A(92):

3376-3380.

Felder, C. C., K. E. Joyce, E. M. Briley, J. Mansouri, K. Mackie, O. Blond, Y. Lai,

A. L. Ma and R. L. Mitchell (1995). "Comparison of the pharmacology and

signal transduction of the human cannabinoid CB1 and CB2 receptors."

Molecular Pharmacology 48(3): 443-450.

Fernandez-Ruiz, J. (2009). "The endocannabinoid system as a target for the

treatment of motor dysfunction." Br J Pharmacol 156(7): 1029-1040.

Fields, H. (2004). "State-dependent opioid control of pain." Nat Rev Neurosci

5(7): 565-575.

Fillingim, R. B., C. D. King, M. C. Ribeiro-Dasilva, B. Rahim-Williams and J. L.

Riley, 3rd (2009). "Sex, gender, and pain: a review of recent clinical and

experimental findings." J Pain 10(5): 447-485.

Friedman, H., S. Specter, T. Klein, C. Newton, M. Rivenbark, D. Rowlands and

D. T. Walz (1990). "Auranofin-induced suppression of autoimmune

antibody production and inflammation in genetically autoimmune-prone

mice." Inflammation 14(4): 463-470.

G. Stennis Watsona, K. J. S., b, Terence J. Coderrec (1997). "Optimal scoring

strategies and weights for the formalin test in rats." Pain 70: 53-58.

123

Galiegue, S., Mary,S., Marchand,J., Dussossoy,D., Carriere,D., Carayon,P.,

Bouaboula,M., Shire,D., Le Fur,G., and Casellas,P. (1995). "Expression of

central and peripheral cannabinoid receptors in human immune tissues

and leukocyte subpopulations." Eur J Biochem(232): 54-61.

Gaoni, Y., Mechoulam, R. (1964). "Isolation, structure and partial synthesis of the

active constituent of hashish." J Am Chem Soc 86: 1646-1647.

Gaskin, D. J. and P. Richard (2012). "The economic costs of pain in the United

States." J Pain 13(8): 715-724.

Gong, J. P., E. S. Onaivi, H. Ishiguro, Q. R. Liu, P. A. Tagliaferro, A. Brusco and

G. R. Uhl (2006). "Cannabinoid CB2 receptors: immunohistochemical

localization in rat brain." Brain Res 1071(1): 10-23.

Gonzalez S, F.-R. J., Sparpaglione V, Parolaro D, Ramos JA. (2002). "Chronic

exposure to morphine, cocaine or ethanol in rats produced different effects

in brain cannabinoid CB(1) receptor binding and mRNA levels." Drug

Alcohol Depend 66(1): 77-84.

Grabovsky, Y. and R. J. Tallarida (2004). "Isobolographic analysis for

combinations of a full and partial agonist: curved isoboles." J Pharmacol

Exp Ther 310(3): 981-986.

Guindon, J. and A. G. Hohmann (2008). "Cannabinoid CB2 receptors: a

therapeutic target for the treatment of inflammatory and neuropathic pain."

Br J Pharmacol 153(2): 319-334.

124

Guo, R. X., M. Zhang, W. Liu, C. M. Zhao, Y. Cui, C. H. Wang, J. Q. Feng and P.

X. Chen (2009). "NMDA receptors are involved in upstream of the spinal

JNK activation in morphine antinociceptive tolerance." Neurosci Lett

467(2): 95-99.

Gutierrez, T., J. D. Crystal, A. M. Zvonok, A. Makriyannis and A. G. Hohmann

(2011). "Self-medication of a cannabinoid CB2 agonist in an animal model

of neuropathic pain." Pain 152(9): 1976-1987.

Halliday, A., Logue, V. (1972). "Painful sensations evoked by electrical

stimulation in the thalamus." Neurophysiology: 221-230.

Hanus L, B. A., Tchilibon S, Shiloah S, Goldenberg D, Horowitz M et al. (1999).

"HU-308: A specific agonist for CB2, a peripheral cannabinoid receptor."

Proc Natl Acad Sci U S A 96: 14228-14233.

Hayes, M. J. and M. S. Brown (2014). "Legalization of medical marijuana and

incidence of opioid mortality." JAMA Intern Med 174(10): 1673-1674.

Henderson-Redmond, A. N., M. B. Yuill, T. E. Lowe, A. M. Kline, M. L. Zee, J.

Guindon and D. J. Morgan (2016). "Morphine-induced antinociception and

reward in "humanized" mice expressing the mu opioid receptor A118G

polymorphism." Brain Res Bull 123: 5-12.

Herkenham, M., Lynn, A.B., Johnson, M.R., Melvin, L.S., de Costa, B.R., Rice,

K.C. (1991a). "Characterization and localization of cannabinoid receptors

in rat brain: a quantitative in vitro autoradiographic study." J Neurosci 11:

563-583.

125

Herkenham, M., Lynn, AB, de Costa, BR, Richfield, EK. (1991b). "Neuronal

localization of cannabinoid receptors in basal ganglia of the rat." Brain Res

Bull(547): 267-274.

Hervera, A., S. Leanez and O. Pol (2012). "The inhibition of the nitric oxide-

cGMP-PKG-JNK signaling pathway avoids the development of tolerance

to the local antiallodynic effects produced by morphine during neuropathic

pain." Eur J Pharmacol 685(1-3): 42-51.

Hine, B. (1985). "Morphine and delta 9-tetrahydrocannabinol: two-way cross

tolerance for antinociceptive and heart-rate responses in the rat."

Psychopharmacology (Berl) 87(1): 34-38.

Hohmann, A. G. and M. Herkenham (1999). "Cannabinoid receptors undergo

axonal flow in sensory nerves." Neuroscience 92(4): 1171-1175.

Holdridge, S. V. and C. M. Cahill (2007). "Spinal administration of a delta opioid

receptor agonist attenuates hyperalgesia and allodynia in a rat model of

neuropathic pain." Eur J Pain 11(6): 685-693.

Howlett, A. C., C. S. Breivogel, S. R. Childers, S. A. Deadwyler, R. E. Hampson

and L. J. Porrino (2004). "Cannabinoid physiology and pharmacology: 30

years of progress." Neuropharmacology 47 Suppl 1: 345-358.

Howlett, A. C., J. M. Qualy and L. L. Khachatrian (1986). "Involvement of Gi in

the inhibition of adenylate cyclase by cannabimimetic drugs." Mol

Pharmacol 29(3): 307-313.

126

Howlett, A. C., D. K. Scott and G. H. Wilken (1989). "Regulation of adenylate

cyclase by cannabinoid drugs. Insights based on thermodynamic studies."

Biochem Pharmacol 38(19): 3297-3304.

Hsieh, C., S. Brown, C. Derleth and K. Mackie (1999). "Internalization and

recycling of the CB1 cannabinoid receptor." J Neurochem 73(2): 493-501.

Hsieh, G. C., M. Pai, P. Chandran, B. A. Hooker, C. Z. Zhu, A. K. Salyers, E. J.

Wensink, C. Zhan, W. A. Carroll, M. J. Dart, B. B. Yao, P. Honore and M.

D. Meyer (2011). "Central and peripheral sites of action for CB(2) receptor

mediated analgesic activity in chronic inflammatory and neuropathic pain

models in rats." Br J Pharmacol 162(2): 428-440.

Huffman , J. W. e. a. (1999). "3-(10,10-Dimethylbutyl)-1-deoxy-D8-THC and

Related Compounds: Synthesis of Selective Ligands for the CB2

Receptor." Bioorganic & Medicinal Chemistry 7: 2905-2914.

Ibrahim, M., Rude ML, Stagg NJ, Mata HP, Lai J, Vanderah TW (2006). "CB2

cannabinoid receptor mediation of antinociception." pain 122: 36-42.

Ibrahim, M. M., F. Porreca, J. Lai, P. J. Albrecht, F. L. Rice, A. Khodorova, G.

Davar, A. Makriyannis, T. W. Vanderah, H. P. Mata and T. P. Malan, Jr.

(2005). "CB2 cannabinoid receptor activation produces antinociception by

stimulating peripheral release of endogenous opioids." Proc Natl Acad Sci

U S A 102(8): 3093-3098.

Imai, S., M. Narita, S. Hashimoto, A. Nakamura, K. Miyoshi, H. Nozaki, N.

Hareyama, T. Takagi, M. Suzuki, M. Narita and T. Suzuki (2006).

"Differences in tolerance to anti-hyperalgesic effects between chronic

127

treatment with morphine and fentanyl under a state of pain." Nihon Shinkei

Seishin Yakurigaku Zasshi 26(5-6): 183-192.

Izzo, A., Mascolo, N., and Capasso, F. (2001). "The gastrointestinal

pharmacology of cannabinoids." Curr Opin Pharmacol 1: 597-603.

JC, A. (2102). "The use of knockout mice to test the specificity of

antibodies for cannabinoid receptors." Hippocampus 22: 643-644.

Jhaveri, M. D., D. R. Sagar, S. J. Elmes, D. A. Kendall and V. Chapman (2007).

"Cannabinoid CB2 receptor-mediated anti-nociception in models of acute

and chronic pain." Mol Neurobiol 36(1): 26-35.

Jin, W., S. Brown, J. P. Roche, C. Hsieh, J. P. Celver, A. Kovoor, C. Chavkin and

K. Mackie (1999). "Distinct domains of the CB1 cannabinoid receptor

mediate desensitization and internalization." J Neurosci 19(10): 3773-

3780.

Jorda, M. A., S. E. Verbakel, P. J. Valk, Y. V. Vankan-Berkhoudt, M. Maccarrone,

A. Finazzi-Agro, B. Lowenberg and R. Delwel (2002). "Hematopoietic cells

expressing the peripheral cannabinoid receptor migrate in response to the

endocannabinoid 2-arachidonoylglycerol." Blood 99(8): 2786-2793.

Kabli, N. and C. M. Cahill (2007). "Anti-allodynic effects of peripheral delta opioid

receptors in neuropathic pain." Pain 127(1-2): 84-93.

Katona, I., Sperlagh B, Sik A, Käfalvi A, Vizi ES, Mackie K (1999).

"Presynaptically located CB1 cannabinoid receptors regulate GABA

release from axon terminals of specific hippocampal interneurons." J

Neurosci(19): 4544-4558.

128

Kazantzis, N. P., S. L. Casey, P. W. Seow, V. A. Mitchell and C. W. Vaughan

(2016). "Opioid and cannabinoid synergy in a mouse neuropathic pain

model." Br J Pharmacol 173(16): 2521-2531.

Kinsey, S. G., A. Mahadevan, B. Zhao, H. Sun, P. S. Naidu, R. K. Razdan, D. E.

Selley, M. Imad Damaj and A. H. Lichtman (2011). "The CB2 cannabinoid

receptor-selective agonist O-3223 reduces pain and inflammation without

apparent cannabinoid behavioral effects." Neuropharmacology 60(2-3):

244-251.

Koch, T. and V. Hollt (2008). "Role of receptor internalization in opioid tolerance

and dependence." Pharmacol Ther 117(2): 199-206.

Kolesnikov, Y., S. Jain, R. Wilson and G. W. Pasternak (1996). "Peripheral kappa

1-opioid receptor-mediated analgesia in mice." Eur J Pharmacol 310(2-3):

141-143.

Koob, G. (2006a). "Drug addiction: The yin and yang of hedonic homeostasis."

Neuron 16: 893-896.

Koob, G. (2006b). "The neurobiology of addiction: a neuroadaptational view

relevantfor diagnosis." Addiction 101: 23-30.

Korzh, A., O. Keren, M. Gafni, H. Bar-Josef and Y. Sarne (2008). "Modulation of

extracellular signal-regulated kinase (ERK) by opioid and cannabinoid

receptors that are expressed in the same cell." Brain Res 1189: 23-32.

Kouznetsova, M., B. Kelley, M. Shen and S. A. Thayer (2002). "Desensitization of

cannabinoid-mediated presynaptic inhibition of neurotransmission

129

between rat hippocampal neurons in culture." Mol Pharmacol 61(3): 477-

485.

Kuhar, J. R., A. Bedini, E. J. Melief, Y. C. Chiu, H. N. Striegel and C. Chavkin

(2015). "Mu opioid receptor stimulation activates c-Jun N-terminal kinase 2

by distinct arrestin-dependent and independent mechanisms." Cell Signal

27(9): 1799-1806.

LaBuda, C. J. K., M.; Little, P.J. (2005). "Cannabinoid CB2 receptor agonist

activity in the hindpaw incision model of postoperative pain." Eur J

Pharmacol 527: 172-174.

Langerman, L., M. I. Zakowski, B. Piskoun and G. J. Grant (1995). "Hot plate

versus tail flick: evaluation of acute tolerance to continuous morphine

infusion in the rat model." J Pharmacol Toxicol Methods. 34: 23-27.

Leon, A., A. Buriani, R. Dal Toso, M. Fabris, S. Romanello, L. Aloe and R. Levi-

Montalcini (1994). "Mast cells synthesize, store, and release nerve growth

factor." Proc Natl Acad Sci U S A 91(9): 3739-3743.

Levitt, E. S. and J. T. Williams (2012). "Morphine desensitization and cellular

tolerance are distinguished in rat locus ceruleus neurons." Mol Pharmacol

82(5): 983-992.

Lichtman, A. H., Cook, S.A., Martin, B.R (1996). "Investigation of brain sites

mediating cannabinoid-induced antinociception in rats: evidence

supporting periaqueductal gray involvement." Pharmacol Biochem

Behav(57): 7-12.

130

Lichtman, A. H., Martin, B.R., (1997). "The selective cannabinoid antagonist SR

141716A blocks cannabinoid-induced antinociception in rats." Pharmacol

Biochem Behav(57): 7-12.

Liu, C. and J. M. Walker (2006). "Effects of a cannabinoid agonist on spinal

nociceptive neurons in a rodent model of neuropathic pain." J

Neurophysiol 96(6): 2984-2994.

Liu, Q. R., C. H. Pan, A. Hishimoto, C. Y. Li, Z. X. Xi, A. Llorente-Berzal, M. P.

Viveros, H. Ishiguro, T. Arinami, E. S. Onaivi and G. R. Uhl (2009).

"Species differences in cannabinoid receptor 2 (CNR2 gene): identification

of novel human and rodent CB2 isoforms, differential tissue expression

and regulation by cannabinoid receptor ligands." Genes Brain Behav 8(5):

519-530.

Lopez-Moreno, J. A., V. Echeverry-Alzate and K. M. Buhler (2012). "The genetic

basis of the endocannabinoid system and drug addiction in humans." J

Psychopharmacol 26(1): 133-143.

Lynch, M. E. and M. A. Ware (2015). "Cannabinoids for the Treatment of Chronic

Non-Cancer Pain: An Updated Systematic Review of Randomized

Controlled Trials." J Neuroimmune Pharmacol 10(2): 293-301.

Mackie, K. a. H., B. (1992). "Cannabinoids inhibit N-type calcium channels in

neuroblastoma-glioma cells." Proc Natl Acad Sci U S A(89): 3825-3829.

Malan TP Jr, I. M., Deng H, Liu Q, Mata HP, Vanderah T, Porreca F, Makriyannis

A. (2001). "CB2 cannabinoid receptor-mediated peripheral

antinociception." Pain 93(3): 239-245.

131

Marchalant, Y., P. W. Brownjohn, A. Bonnet, T. Kleffmann and J. C. Ashton

(2014). "Validating Antibodies to the Cannabinoid CB2 Receptor: Antibody

Sensitivity Is Not Evidence of Antibody Specificity." J Histochem

Cytochem 62(6): 395-404.

Marcus, D. J., M. Zee, A. Hughes, M. B. Yuill, A. G. Hohmann, K. Mackie, J.

Guindon and D. J. Morgan (2015). "Tolerance to the antinociceptive

effects of chronic morphine requires c-Jun N-terminal kinase." Mol Pain

11: 34.

Maresz, K., E. J. Carrier, E. D. Ponomarev, C. J. Hillard and B. N. Dittel (2005).

"Modulation of the cannabinoid CB2 receptor in microglial cells in

response to inflammatory stimuli." J Neurochem 95(2): 437-445.

Marini, P., M. G. Cascio, A. King, R. G. Pertwee and R. A. Ross (2013).

"Characterization of cannabinoid receptor ligands in tissues natively

expressing cannabinoid CB2 receptors." Br J Pharmacol 169(4): 887-899.

Martin, W. J., Coffin, P.O., Attias, E., Balinsky, M., Tsou, K (1999a).

"Cannabinoid receptormediated inhibition of the rat tail-flick reflex after

microinjection into the rostral ventromedial medulla." Brain Res(822): 237-

242.

Martin, W. J., C. M. Loo and A. I. Basbaum (1999b). "Spinal cannabinoids are

anti-allodynic in rats with persistent inflammation." Pain 82(2): 199-205.

Martin, W. J., Tsou, K., Walker, J.M., (1998). "Cannabinoid receptormediated

inhibition of the rat tail-flick reflex after microinjection into the rostral

ventromedial medulla." Neurosci Lett(242): 33-36.

132

Marusich, J. A., T. W. Lefever, K. R. Antonazzo, R. M. Craft and J. L. Wiley

(2014). "Evaluation of sex differences in cannabinoid dependence." Drug

Alcohol Depend 137: 20-28.

Matsuda, L. A., Lolait, S.J., Brownstein, M.J., Young, A.C., Bonner, T.I.. (1990).

"Structure of a cannabinoid receptor and functional expression of the

cloned cDNA." Nature 346: 561-564.

McAllister, S. D., Griffin G, Satin LS, Abood ME (1999). "Cannabinoid receptors

can activate and inhibit G protein-coupled inwardly rectifying potassium

channels in a xenopus oocyte expression system." J Pharmacol Exp

Ther(291): 618-626.

Mechoulam, R., Ben Shabat,S., Hanus,L., Ligumsky,M., Kaminski,N.E.,

Schatz,A.R., Gopher,A., Almog,S., Martin,B.R., Compton,D.R.. and

(1995). " Identification of an endogenous 2-monoglyceride, present in

canine gut, that binds to cannabinoid receptors." Biochem. Pharmacol(50):

83-90.

Melief, E. J., M. Miyatake, M. R. Bruchas. and C. Chavkin (2010). "Ligand-

directed c-Jun N-terminal kinase activation disrupts opioid receptor

signaling." PNAS 107: 11608-11613.

Melief EJ, M. M., Bruchas MR, Chavkin C (2010). "Ligand Directed c Jun N

Terminal Kinase Activation Disrupts Opioid Receptor Signaling." Proc Natl

Acad Sci USA 107(25): 11608-11613.

Merighi, S., S. Gessi, K. Varani, D. Fazzi, P. Mirandola and P. A. Borea (2012).

"Cannabinoid CB(2) receptor attenuates morphine-induced inflammatory

133

responses in activated microglial cells." Br J Pharmacol 166(8): 2371-

2385.

Miranda, H. F., Noriega, V., Zanetta, P., Prieto, J.C., Aranda, N., Sierralta, F.

(2014). "Isobolographic analysis of the opioid-opioid interactions in a tonic

and a phasic mouse model of induced nociceptive pain." J Biomed Sci

21(62).

Morgan, D. J., B. J. Davis, C. S. Kearn, D. Marcus, A. J. Cook, J. Wager-Miller,

A. Straiker, M. H. Myoga, J. Karduck, E. Leishman, L. J. Sim-Selley, T. A.

Czyzyk, H. B. Bradshaw, D. E. Selley and K. Mackie (2014). "Mutation of

putative GRK phosphorylation sites in the cannabinoid receptor 1 (CB1R)

confers resistance to cannabinoid tolerance and hypersensitivity to

cannabinoids in mice." J Neurosci 34(15): 5152-5163.

Munro, S., Kerrie, L., Abu-Shaar, M., and Abu-Shaar T. (1993). "Molecular

characterization of a peripheral receptor for cannabinoids." Nature 365:

61-65.

Nackley, A. G., A. Makriyannis and A. G. Hohmann (2003). "Selective activation

of cannabinoid CB2 receptors suppresses spinal fos protein expression

and pain behavior in a rat model of inflammation." Neuroscience 119(3):

747-757.

Naguib, M., J. J. Xu, P. Diaz, D. L. Brown, D. Cogdell, B. Bie, J. Hu, S. Craig and

W. N. Hittelman (2012). "Prevention of paclitaxel-induced neuropathy

through activation of the central cannabinoid type 2 receptor system."

Anesth Analg 114(5): 1104-1120.

134

Nahin, R. L. (2015). "Estimates of pain prevalence and severity in adults: United

States, 2012." J Pain 16(8): 769-780.

Nguyen, P. T., C. L. Schmid, K. M. Raehal, D. E. Selley, L. M. Bohn and L. J.

Sim-Selley (2012). "beta-arrestin2 regulates cannabinoid CB1 receptor

signaling and adaptation in a central nervous system region-dependent

manner." Biol Psychiatry 71(8): 714-724.

Ofek, O., Karsak,M., Leclerc,N., Fogel,M., Frenkel,B., Wright,K., Tam,J., Attar-

Namdar,M., Kram,V., Shohami,E., Mechoulam,R., Zimmer,A., and Bab,I.

(2006). "Peripheral cannabinoid receptor, CB2, regulates bone mass."

Proc Natl Acad Sci U S A(103): 696-701.

Onaivi, E. S., H. Ishiguro, J. P. Gong, S. Patel, P. A. Meozzi, L. Myers, A.

Perchuk, Z. Mora, P. A. Tagliaferro, E. Gardner, A. Brusco, B. E.

Akinshola, Q. R. Liu, S. S. Chirwa, B. Hope, J. Lujilde, T. Inada, S.

Iwasaki, D. Macharia, L. Teasenfitz, T. Arinami and G. R. Uhl (2008).

"Functional expression of brain neuronal CB2 cannabinoid receptors are

involved in the effects of drugs of abuse and in depression." Ann N Y Acad

Sci 1139: 434-449.

Pacher, P., S. Batkai and G. Kunos (2006). "The endocannabinoid system as an

emerging target of pharmacotherapy." Pharmacol Rev 58(3): 389-462.

Pacher, P. and G. Kunos (2013). "Modulating the endocannabinoid system in

human health and disease--successes and failures." FEBS J 280(9):

1918-1943.

Paldyova, E., E. Bereczki, M. Santha, T. Wenger, A. Borsodi and S. Benyhe

(2008). "Noladin ether, a putative endocannabinoid, inhibits mu-opioid

135

receptor activation via CB2 cannabinoid receptors." Neurochem Int 52(1-

2): 321-328.

Pan, Z. Z., Williams, J. T. & Osborne, P. B. (1990). "Opioid actions on single

nucleus raphe magnus neurons from rat and guinea pig in vitro." J.

Physiol. (Lond.) 427: 519-532.

Perl, E. (1968). "Myelinated afferent fibres innervating the primate skin and their

response to noxious stimuli." J Physiol (Lond): 593-615.

Pertwee, R. G. (2001). "Cannabinoid receptors and pain." Progress in

Neurobiology 63: 569-611.

Pertwee, R. G. (2012). "Targeting the endocannabinoid system with cannabinoid

receptor agonists: pharmacological strategies and therapeutic

possibilities." Philos Trans R Soc Lond B Biol Sci 367(1607): 3353-3363.

Pugh, G., Jr., D. J. Mason, Jr., V. Combs and S. P. Welch (1997). "Involvement

of dynorphin B in the antinociceptive effects of the cannabinoid CP55,940

in the spinal cord." J Pharmacol Exp Ther 281(2): 730-737.

Puig, S., Sorkin, L. (1996). "Formalin-evoked activity in identified primary afferent

fibers: systemic lidocaine suppresses phase-2 activity." Pain 64(2): 345-

355.

Quartilho, A., Mata HP, Ibrahim MM, Vanderah TW, Porreca F, Makriyannis A,

Malan TP Jr (2003). "Inhibition of inflammatory hyperalgesia by activation

of peripheral CB2 cannabinoid receptors." Anesthesiology(99): 955-960.

136

Rahn, E. J., L. Deng, G. A. Thakur, K. Vemuri, A. M. Zvonok, Y. Y. Lai, A.

Makriyannis and A. G. Hohmann (2014). "Prophylactic cannabinoid

administration blocks the development of paclitaxel-induced neuropathic

nociception during analgesic treatment and following cessation of drug

delivery." Mol Pain 10: 27.

Raja, S. N., Campbell, J.N., Meyer, R.A. (1984). "Evidence for different

mechanisms of primary and secondary hyperalgesia following heat injury

to the glabrous skin." Brain 107: 1179-1188.

Rinaldi-Carmona, M. E. a. (1998). "SR 144528, the first potent and selective

antagonist of the CB2 cannabinoid receptor." J Pharmacol Exp Ther 284:

644-650.

Romero-Sandoval, A., Eisenach, J. (2007). "Spinal Cannabinoid Receptor Type 2

Activation Reduces Hypersensitivity and Spinal Cord Glial Activation after

Paw Incision." Anesthesiology 106: 787-794.

Romero-Sandoval, A., N. Nutile-McMenemy and J. A. DeLeo (2008). "Spinal

microglial and perivascular cell cannabinoid receptor type 2 activation

reduces behavioral hypersensitivity without tolerance after peripheral

nerve injury." Anesthesiology 108(4): 722-734.

Romero-Sandoval, E. A., R. Horvath, R. P. Landry and J. A. DeLeo (2009).

"Cannabinoid receptor type 2 activation induces a microglial anti-

inflammatory phenotype and reduces migration via MKP induction and

ERK dephosphorylation." Mol Pain 5: 25.

Ross, R. A., A. A. Coutts, S. M. McFarlane, S. Anavi-Goffer, A. J. Irving, R. G.

Pertwee, D. J. MacEwan and R. H. Scott (2001). "Actions of cannabinoid

137

receptor ligands on rat cultured sensory neurones: implications for

antinociception." Neuropharmacology 40(2): 221-232.

Rubino, T., D. Vigano, F. Premoli, C. Castiglioni, S. Bianchessi, R. Zippel and D.

Parolaro (2006). "Changes in the expression of G protein-coupled receptor

kinases and beta-arrestins in mouse brain during cannabinoid tolerance: a

role for RAS-ERK cascade." Mol Neurobiol 33(3): 199-213.

Sañudo-Peña, M., Patrick SL, Khen S, Patrick RL, Tsou K,Walker JM (1998).

"Cannabinoid effects in basal ganglia in a rat model of Parkinson's

disease." Neurosci Lett(248): 171-174.

Savage, S. R. (2009). "Management of Opioid Medications in Patients With

Chronic Pain and Risk of Substance Misuse." Current Psychiatry Reports

11: 377-384.

Smith, P. B., S. P. Welch and B. R. Martin (1994). "Interactions between delta 9-

tetrahydrocannabinol and kappa opioids in mice." J Pharmacol Exp Ther

268(3): 1381-1387.

Soethoudt, M., U. Grether, J. Fingerle, T. W. Grim, F. Fezza, L. de Petrocellis, C.

Ullmer, B. Rothenhausler, C. Perret, N. van Gils, D. Finlay, C. MacDonald,

A. Chicca, M. D. Gens, J. Stuart, H. de Vries, N. Mastrangelo, L. Xia, G.

Alachouzos, M. P. Baggelaar, A. Martella, E. D. Mock, H. Deng, L. H.

Heitman, M. Connor, V. Di Marzo, J. Gertsch, A. H. Lichtman, M.

Maccarrone, P. Pacher, M. Glass and M. van der Stelt (2017).

"Cannabinoid CB2 receptor ligand profiling reveals biased signalling and

off-target activity." Nat Commun 8: 13958.

Stella, N. (2004). "Cannabinoid signaling in glial cells." Glia 48(4): 267-277.

138

Stone, L., Geman, J., Kitto, KF, Fairbanks, CA, Wilcox, GL (2014). "Morphine

and Clonidine Combination Therapy Improves Therapeutic Window in

Mice: Synergy in Antinociceptive but Not in Sedative or Cardiovascular

Effects." PLoS One 9(10).

Storr, M., E. Gaffal, D. Saur, V. Schusdziarra and H. D. Allescher (2002). "Effect

of cannabinoids on neural transmission in rat gastric fundus." Can J

Physiol Pharmacol 80(1): 67-76.

Suhnel, J. (1998). "Parallel Dose–Response Curves in Combination

Experiments." Bulletin of Mathematical Biology 60: 197-213.

Sullivan, J. M. (1999). "Mechanisms of cannabinoid-receptor-mediated inhibition

of synaptic transmission in cultured hippocampal pyramidal neurons." J

Neurophysiol 82(3): 1286-1294.

Svizenska, I., P. Dubovy and A. Sulcova (2008). "Cannabinoid receptors 1 and 2

(CB1 and CB2), their distribution, ligands and functional involvement in

nervous system structures--a short review." Pharmacol Biochem Behav

90(4): 501-511.

Svizenska, I. H., V. Brazda, I. Klusakova and P. Dubovy (2013). "Bilateral

changes of cannabinoid receptor type 2 protein and mRNA in the dorsal

root ganglia of a rat neuropathic pain model." J Histochem Cytochem

61(7): 529-547.

Tallarida, R. J. (2002). "The interaction index: a measure of drug synergism."

pain 98: 163-168.

139

Tallarida, R. J. and R. B. Raffa (2010). "The application of drug dose equivalence

in the quantitative analysis of receptor occupation and drug combinations."

Pharmacol Ther 127(2): 165-174.

Tanda, G. and S. R. Goldberg (2003). "Cannabinoids: reward, dependence, and

underlying neurochemical mechanisms--a review of recent preclinical

data." Psychopharmacology (Berl) 169(2): 115-134.

Tanikawa, T., K. Kurohane and Y. Imai (2007). "Induction of preferential

chemotaxis of unstimulated B-lymphocytes by 2-arachidonoylglycerol in

immunized mice." Microbiol Immunol 51(10): 1013-1019.

Terman, G. W., W. Jin, Y. P. Cheong, J. Lowe, M. G. Caron, R. J. Lefkowitz and

C. Chavkin (2004). "G-protein receptor kinase 3 (GRK3) influences opioid

analgesic tolerance but not opioid withdrawal." Br J Pharmacol 141(1): 55-

64.

Tjolsen, A., Berge, O., Hunskaar, S., Rosland, J., and Hole, K. (1992). "The

formalin test: an evaluation of the method." Pain 51: 5-17.

Tsou, K., S. L. Patrick and J. M. Walker (1995). "Physical withdrawal in rats

tolerant to delta 9-tetrahydrocannabinol precipitated by a cannabinoid

receptor antagonist." Eur J Pharmacol 280(3): R13-15.

Ueda, H., Harada, H., Nozaki, M., Katada, T., Ui, M., Satoh, M., and Takagi, H.

(1988). "Reconstitution of rat brain mu opioid receptors with purified

guanine nucleotide-binding regulatory proteins, Gi and Go." Proc Natl

Acad Sci U S A(85).

140

Van Sickle, M. D., M. Duncan, P. J. Kingsley, A. Mouihate, P. Urbani, K. Mackie,

N. Stella, A. Makriyannis, D. Piomelli, J. S. Davison, L. J. Marnett, V. Di

Marzo, Q. J. Pittman, K. D. Patel and K. A. Sharkey (2005). "Identification

and functional characterization of brainstem cannabinoid CB2 receptors."

Science 310(5746): 329-332.

Vigano, D., T. Rubino and D. Parolaro (2005). "Molecular and cellular basis of

cannabinoid and opioid interactions." Pharmacol Biochem Behav 81(2):

360-368.

Wadenberg, M. L. (2003). "A review of the properties of spiradoline: a potent and

selective kappa-opioid receptor agonist." CNS Drug Rev 9(2): 187-198.

Wakley, A. A., J. L. Wiley and R. M. Craft (2014). "Sex differences in

antinociceptive tolerance to delta-9-tetrahydrocannabinol in the rat." Drug

Alcohol Depend 143: 22-28.

Waldhoer, M., S. E. Bartlett and J. L. Whistler (2004). "Opioid receptors." Annu

Rev Biochem 73: 953-990.

Wall, P. D. (1967). "The laminar organisation of dorsal horn and effects of

descending impulses." J. Physiol. (Lond.) 188: 403-423.

Wang, Q., Mao L., Shi, Y., Han, J. (1990). "Lumbar intrathecal administration of

naloxone antagonizes analgesia produced by electrical stimulation of the

hypothalamic arcuate nucleus in pentobarbital-anesthetized rats."

Neuropharmacology 29: 1123-1129.

141

Watkins, L. R., M. R. Hutchinson, K. C. Rice and S. F. Maier (2009). "The "toll" of

opioid-induced glial activation: improving the clinical efficacy of opioids by

targeting glia." Trends Pharmacol Sci 30(11): 581-591.

Welch, S. P. (1997). "Characterization of anandamide-induced tolerance:

comparison to delta 9-THC-induced interactions with dynorphinergic

systems." Drug Alcohol Depend 45(1-2): 39-45.

Whiteside, G. T., S. L. Gottshall, J. M. Boulet, S. M. Chaffer, J. E. Harrison, M. S.

Pearson, P. I. Turchin, L. Mark, A. E. Garrison and K. J. Valenzano

(2005). "A role for cannabinoid receptors, but not endogenous opioids, in

the antinociceptive activity of the CB2-selective agonist, GW405833." Eur

J Pharmacol 528(1-3): 65-72.

Whiting, P. F., R. F. Wolff, S. Deshpande, M. Di Nisio, S. Duffy, A. V. Hernandez,

J. C. Keurentjes, S. Lang, K. Misso, S. Ryder, S. Schmidlkofer, M.

Westwood and J. Kleijnen (2015). "Cannabinoids for Medical Use: A

Systematic Review and Meta-analysis." Jama 313(24): 2456-2473.

Williams, J. T., S. L. Ingram, G. Henderson, C. Chavkin, M. von Zastrow, S.

Schulz, T. Koch, C. J. Evans and M. J. Christie (2013). "Regulation of mu-

opioid receptors: desensitization, phosphorylation, internalization, and

tolerance." Pharmacol Rev 65(1): 223-254.

Wong, Y. H., C. D. Demoliou-Mason and E. A. Barnard (1988). "ADP-ribosylation

with pertussis toxin modulates the GTP-sensitive opioid ligand binding in

digitonin-soluble extracts of rat brain membranes." J Neurochem 51(1):

114-121.

142

Yaksh, T. L. N., R. (1985). "The physiology and pharmacology of spinal opiates."

Ann Rev Phamacol Toxicol 25(433-462).

Yamamoto, W., T. Mikami and H. Iwamura (2008). "Involvement of central

cannabinoid CB2 receptor in reducing mechanical allodynia in a mouse

model of neuropathic pain." Eur J Pharmacol 583(1): 56-61.

Yang, D., D. Conze, A. Whitmarsh, T. Barrett, R. Davis, M. Rincon and R. Flavell

(1998). "Differentiation of CD4+ T Cells to Th1 Cells Requires MAP

Kinase JNK 2." Immunity 9: 575-585.

Yang, D., C. Kuan, A. Whitmarsh, M. Rincon, T. Zheng, R. Davis, P. Rakic and

R. Flavell (1997). "Absence of excitotoxicity induced apopotosis in the

hippocampus of mice lacking the Jnk3 gene." Nature 389.

Yao, B. B. H. G., Frost JM, Fan Y, Garrison TR, Daza AV, Grayson GK, Zhu CZ,

Pai M, Chandran P, Salyers AK, Wensink EJ, Honore P, Sullivan JP, Dart

MJ, Meyer MD (2008). "In vitro and in vivo characterization of A-796260: a

selective cannabinoid CB2 receptor agonist exhibiting analgesic activity in

rodent pain models." Br J Pharmacol 153: 390-401.

Yiangou, Y., Facer P, Durrenberger P, Chessell IP, Naylor A, Bountra C, Banati

RR, Anand P (2006). "COX-2, CB2 and P2X7-immunoreactivities are

increased in activated microglial cells/macrophages of multiple sclerosis

and amyotrophic lateral sclerosis spinal cord." BMC Neurol(6): 12.

Yuill, M. B., M. L. Zee, D. Marcus and D. J. Morgan (2016). "Tolerance to the

antinociceptive and hypothermic effects of morphine is mediated by

multiple isoforms of c-Jun N-terminal kinase." Neuroreport 27(6): 392-396.

143

Zhang, J., Ferguson, SSG, Barak, LS, Bodduluri, SR, Laporte, SA, Law, PY,

Caron, MG (1998). "Role for G protein-coupled receptor kinase in agonist-

specific regulation of m-opioid receptor responsiveness." Neurobiology 95:

7`157-7162.

Zhang, J., Hoffert, C., Huy, K., Groblewski, T., Ahmad, S., O'Donnell, D. (2003).

"Induction of CB2 receptor expression in the rat spinal cord of neuropathic

but not inflammatory chronic pain models." Eur J Neurosci 17: 2750-2754.

Zhang, M., K. Wang, M. Ma, S. Tian, N. Wei and G. Wang (2016). "Low-Dose

Cannabinoid Type 2 Receptor Agonist Attenuates Tolerance to Repeated

Morphine Administration via Regulating mu-Opioid Receptor Expression in

Walker 256 Tumor-Bearing Rats." Anesth Analg 122(4): 1031-1037.

Zhuang, Z. Y., Y. R. Wen, D. R. Zhang, T. Borsello, C. Bonny, G. R. Strichartz, I.

Decosterd and R. R. Ji (2006). "A peptide c-Jun N-terminal kinase (JNK)

inhibitor blocks mechanical allodynia after spinal nerve ligation: respective

roles of JNK activation in primary sensory neurons and spinal astrocytes

for neuropathic pain development and maintenance." J Neurosci 26(13):

3551-3560.

Zimmer, A., Zimmer, A.M., Hohmann, A.G., Herkenham, M., Bonner, T.I. (1999).

"Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1

receptor knockout mice." Proc Natl Acad Sci U S A 96: 5780-5785.

Zoratti, C., Kipmen-Korgun,D., Osibow,K., Malli,R., and Graier,W.F. (2003).

"Anandamide initiates Ca(2+) signaling via CB2 receptor linked to

phospholipase C in calf pulmonary endothelial cells." Br J

Pharmacol(140): 1351-1362.

144

VITA

Education

Franklin and Marshall College B.A. (Neuroscience) (2007-2011)

Penn State University Ph.D. (Neuroscience, expected) (2011-2017)

Publications Yuill, MB., Hale, DE., Guindon J, Morgan DJ. (2017). “Anti-nociceptive interactions between opioids and a cannabinoid receptor 2 agonist in inflammatory pain.” Molecular Pain.

Yuill, MB., Marcus, DJ., Zee, ML., Morgan DJ (2016). “Tolerance to the anti-nociceptive and hypothermic effects of morphine are mediated by multiple isoforms of c-Jun N-terminal Kinase.” Neuroreport. Marcus DJ, Zee M, Hughes, A., Yuill, MB., Hohmann AG, Mackie K, Guindon J, Morgan DJ (2015). Tolerance to the antinociceptive effects of chronic morphine requires c-Jun N-terminal kinase. Molecular Pain. Henderson-Redmond, AN., Yuill, MB., Lowe, TE., Kline, AM., Zee, ML., Guindon, J., Morgan, DJ., (2015) “Morphine-induced antinociception and reward in "humanized" mice expressing the mu opioid receptor A118G polymorphism” Brain Res. Bul. Nealon, C., Davis, BJ., Henderson Redmond, A., Yuill, MB., Muller, J., Haskins, CP., Marcus, DJ., Czyzyk, TA., Mackie, K., Guindon, J., Morgan, DJ.“c-Jun N terminal kinase signaling pathways mediate cannabinoid tolerance in an agonist-specific manner.” (In preparation).

Abstracts Yuill, MB., Morgan, DJ., and Guindon, J. (2016). Impact of a CB2R Cannabinoid Agonist On Inflammatory Pain And Morphine Tolerance. International Cannabinoid Research Society Annual Meeting. Bukovina, Poland. Yuill, MB., Morgan, DJ., and Guindon, J. (2015). Mu opioid receptor and cannabinoid Receptor 2 in inflammatory pain. International Cannabinoid Research Society Annual Meeting. Wolfville, Nova Scotia. Yuill, MB., Morgan, DJ., and Guindon, J. (2015). Tolerance to the antinociceptive effects of Δ9-THC in the formalin model of inflammatory pain. Society for Neuroscience Annual Meeting. Chicago, Illinois. Yuill, MB., Morgan, DJ. Levenson, R., and Guindon, J. (2015). Cross-tolerance between opioid receptor and cannabinoid receptor 2 agonists in inflammatory pain. Gill Symposium annual meeting. Bloomington, Indiana.