141
MECHANISMS OF EPHB2 MEDIATED OPIATE-DEPENDENT TOLERANCE AND LEARNING By Sofia Huroy A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Pharmaceutical Sciences Faculty of Pharmacy, University of Toronto © Copyright by Sofia Huroy 2012

MECHANISMS OF EPHB2 MEDIATED OPIATE-DEPENDENT TOLERANCE ... · Mechanisms of EphB2 Mediated Opiate-Dependent Tolerance and ... tolerance and alters behavioural ... Opiate-Dependent

  • Upload
    vandat

  • View
    220

  • Download
    1

Embed Size (px)

Citation preview

MECHANISMS OF EPHB2 MEDIATED OPIATE-DEPENDENT TOLERANCE AND LEARNING

By

Sofia Huroy

A thesis submitted in conformity with the requirements for the degree of Master of Science

Graduate Department of Pharmaceutical Sciences Faculty of Pharmacy, University of Toronto

© Copyright by Sofia Huroy 2012

ii

Mechanisms of EphB2 Mediated Opiate-Dependent Tolerance and Learning

Master of Science, 2012

Sofia Huroy

Department of Pharmaceutical Sciences, University of Toronto

ABSTRACT

The underlying mechanism of morphine tolerance remains unclear. EphB2 regulates synaptic

efficiency with respect to learning and memory. Previously, we demonstrated that loss of EphB2

significantly accelerates the rate of morphine tolerance and alters behavioural responses to

morphine following tolerance. However, EphB2 null mice exhibit no significant alteration in

their metabolism of morphine compared to littermate controls, or altered mu opioid receptor

expression levels within the spinal cord or brain compared to littermate controls. Therefore, we

investigated whether loss of EphB2 alters learned responsiveness to morphine through

modification of hippocampal function. Interestingly, results indicate that electrolytic lesions of

the dorsal hippocampus of wild-type mice display similar behavioural responses seen in EphB2

null mice compared to sham operated controls. These findings suggest that loss of EphB2

function within the hippocampus is a critical feature in mediating morphine-dependent

tolerance, and suggests a novel role for EphB2 receptor signaling in opiate-dependent learning.

  iii

ACKNOWLEDGEMENTS

What a memorable two years it has been. First and foremost, I would like to thank my

supervisor, Dr. Jeffrey Henderson, who has mentored, challenged and motivated me throughout

my graduate studies. I am grateful and appreciate all the training I have learned and acquired

throughout these past two years.

I would also like to thank my thesis advisory committee members, Dr. ZhengPing Jia and Dr.

Peter Wells for their valuable suggestions and constructive comments throughout my study. I

would also like to acknowledge the following individuals who took time to assist me with my

research:

• Dr. Derek van der Kooy for his insightful suggestions and feedback, as well as providing

us with a generous supply of morphine.

• Dr. Ryan Ting-A-Kee for his statistical assistance.

• Dr. Carolyn Cummins and Lilia Magomedova for their assistance with optimization of the

LC-MS/MS protocol (adapted from Shu Chen).

• Dr. Sandy Pang for use of her lab’s C18 column to conduct our LC-MS/MS analyses, and

for providing us with a supply of morphine-3-glucuronide.

• Dr. James Eubanks for use of the activity monitors and Dr. Richard Logan for his

assistance in troubleshooting problems related to the activity monitors.

I would also like to thank my past and present lab members: Yanshan Cao, Ashlin Kanawaty,

Maya Latif, Mary Shan, William Tang and Zoe Winterton-Perks. Your support and friendship

was very much appreciated.

  iv

Finally, I would like to thank my parents, my siblings, and my dearest friends for their

unconditional support, encouragement and love throughout these years. Thank you for all the

laughter and joy. I could not have done it without you by my side. Thank-you!

  v

TABLE OF CONTENTS

ABSTRACT __________________________________________________________ ii

ACKNOWLEDGEMENTS ______________________________________________ iii

TABLE OF CONTENTS _________________________________________________ v

LIST OF FIGURES ____________________________________________________ ix

LIST OF TABLES _____________________________________________________ xi

SUMMARY OF ABBREVIATIONS _____________________________________ xii

CHAPTER 1: INTRODUCTION __________________________________________ 1

1.1 History of Opioids __________________________________________________ 2

1.2 Opioid Receptors ____________________________________________________ 3

1.2.1 Mu Opioid Receptor Structure ______________________________________ 5

1.3 Ligands for Opioid Receptors _________________________________________ 9

1.3.1 Endogenous Ligands _____________________________________________ 9

1.3.2 Exogenous Ligands _____________________________________________ 10

1.3.2.1 Morphine _________________________________________________ 14

1.4 Mu Opioid Receptor Signaling _______________________________________ 17

1. 5 Morphine Tolerance _______________________________________________ 19

1.6 Opiates and NMDA Receptors ________________________________________ 22

1.7 Animal Model: Mu Opioid Receptor Knockout Mice ______________________ 24

1.8 Eph Receptors and Ephrins ___________________________________________ 25

1.8.1 Eph/Ephrin Structure ____________________________________________ 25

1.9 Eph-ephrin Interactions and Signaling __________________________________ 30

  vi

1.9.1 Forward Signaling ______________________________________________ 33

1.9.2 Reverse Signaling ______________________________________________ 34

1.9.3 Termination of Eph-ephrin Signaling _______________________________ 35

1.10 EphB2 Expression in the CNS _______________________________________ 35

1.11 EphB2 and Synaptic Plasticity _______________________________________ 37

1.12 EphB and Pain Modulation _________________________________________ 39

1.13 Learning and Memory: Overview ____________________________________ 41

1.13.1 Hippocampus _________________________________________________ 44

1.14 Opiate-Dependent Tolerance and Learning _____________________________ 46

1.14.1 EphB2 and Opiate Tolerance _____________________________________ 47

1.15 Thesis Rationale __________________________________________________ 54

1.15.1 Thesis Hypotheses _____________________________________________ 54

CHAPTER 2: MATERIALS AND METHODS _____________________________ 55

2.1 Animals _________________________________________________________ 56

2.2 Chemicals ________________________________________________________ 56

2.3 Morphine Tolerance Tests: EphB2 Wild-type and Null Mice _______________ 56

2.3.1 Sensory Analyses: Tail Pinch and Tail Flick Assay _____________________ 57

2.4 Pharmacokinetic Analyses of Morphine Metabolism ______________________ 57

2.4.1 Preparation of LC-MS/MS Standard Solutions ________________________ 57

2.4.2 Collection of Blood and Brain Samples ______________________________ 60

2.4.3 Purification of Blood and Brain Samples _____________________________ 60

2.5 LC-MS/MS Analyses _______________________________________________ 61

2.6 Immunohistochemistry ______________________________________________ 61

  vii

2.7 Stereotactic Surgeris ________________________________________________ 62

2.7.1 Kainic Acid Induced Lesion _______________________________________ 62

2.7.2 Electrolytic Lesion ______________________________________________ 63

2.8 Behavioral Analyses ________________________________________________ 63

2.8.1 Passive Avoidance ______________________________________________ 63

2.8.2 Activity Monitor ________________________________________________ 65

2.9 Morphine Related Behavior __________________________________________ 65

2.9.1 Morphine Induced Hyperactivity ___________________________________ 65

2.9.2 Morphine Tolerance Tests: Lesioned and Sham Operated Control Animals __ 65

2.10 Statistical Analyses _______________________________________________ 66

CHAPTER 3: RESULTS ________________________________________________ 67

3.1 Morphine related responses of EphB2 null mice __________________________ 68

3.2 Distribution of mu opioid receptors in the CNS __________________________ 70

3.3 Pharmacokinetic Analysis of Morphine in EphB2 Null Mice ________________ 75

3.4 EphB2 null mice display deficits in hippocampal learning ___________________ 81

3.4.1 Behavioral assessments of animals with bilateral electrolytic lesions in the

dorsal hippocampus __________________________________________________ 83

3.5 Morphine related responses in animals with bilateral electrolytic lesions in the

dorsal hippocampus ____________________________________________________ 91

CHAPTER 4: DISCUSSION ____________________________________________ 105

4.1 Changes in morphine responsiveness seen in EphB2-null mice are mediated via

modification of cortical influences on sensory function. ______________________ 106

  viii

4.2 EphB2 null mice exhibit deficiencies in contextual learning similar to that seen

in wild-type animals containing bilateral electrolytic lesions of the dorsal

hippocampus. ________________________________________________________ 109

4.3 Impaired opiate-dependent responses seen in EphB2 null mice arise from

hippocampal-dependent deficiencies in contextual learning. ___________________ 111

4.4 Concluding remarks and future studies _________________________________ 114

REFERENCES _______________________________________________________ 116

  ix

LIST OF FIGURES

CHAPTER 1: INTRODUCTION

Figure 1.1 Alternative splicing of mouse Oprm1 gene

Figure 1.2 Crystal structure of the mu-opioid receptor bound to morphinan antagonist β-FNA

Figure 1.3 Structures of common MOR ligands

Figure 1.4 Metabolism of morphine

Figure 1.5 Pre- and postsynaptic MOR signaling

Figure 1.6 Proposed pathways for MOR regulation

Figure 1.7 Eph ligand and receptor structure

Figure 1.8 Eph-ephrin Signaling

Figure 1.9 Morphine tolerance in EphB2 null mice on Day 1

Figure 1.10 Morphine tolerance in EphB2 null mice on Day 3

Figure 1.11 Morphine tolerance in EphB2 null mice on Day 6

Figure 1.12 Antinociceptive responses of EphB2 wild-types upon removal to novel

environment.

Figure 1.13 Antinociceptive responses of EphB2 null mice upon removal to novel environment.

CHAPTER 2: MATERIALS AND METHODS

Figure 2.1 Schedule of morphine dosing

Figure 2.2 Schedule of analysis following stereotactic surgery.

CHAPTER 3: RESULTS

Figure 3.1 Expression of EphB2 in the CNS

  x

Figure 3.2 Distribution of mu opioid receptor in dorsal spinal cord of EphB2 null mice and

controls

Figure 3.3 Distribution of mu opioid receptors in striatum and hippocampus of EphB2 null mice

and wild-type littermates

Figure 3.4 LC/MS/MS analyses of morphine and metabolites

Figure 3.5 LC/MS/MS analyses of brain morphine metabolism in EphB2 null mice and controls

Figure 3.6.LC/MS/MS analyses of blood morphine metabolism in EphB2 null mice and

controls

Figure 3.7 LC/MS/MS analyses of brain and blood morphine metabolism in EphB2 null mice

and controls

Figure 3.8 Passive avoidance responses of EphB2 mice.

Figure 3.9 Example of an electrolytic lesion of the dorsal hippocampus.

Figure 3.10 Serial views of electrolytic lesions of the dorsal hippocampus

Figure 3.11 Passive avoidance responses of lesioned wild-type animals

Figure 3.12 Active time of EphB2 null mice and lesioned wild-types

Figure 3.13 Motor activity of EphB2 null mice and lesioned wild-types

Figure 3.14 Morphine induced hyperactivity in wild-type mice.

Figure 3.15 Morphine induced hyperactivity in EphB2 null mice.

Figure 3.16 Spontaneous motor activity of saline injected animals.

Figure 3.17 Morphine induced hyperactivity in sham operated control animals.

Figure 3.18 Morphine induced hyperactivity in lesioned animals.

Figure 3.19 Antinociceptive responses of sham and lesioned animals in home versus novel

environments

  xi

LIST OF TABLES

CHAPTER 1: INTRODUCTION

Table 1.1 Overview of opioid receptors.

Table 1.2 Binding affinities of endogenous opioid peptides to Mu, Delta and Kappa opiate

receptors in nanomolar

Table 1.3 Binding affinities of ephrin-Fc’s for EphB2 ligand binding domain.

Table 1.4 Calculated binding affinities of ephrin ligands to EphA4.

CHAPTER 3: RESULTS

Table 3.1 Analyses of day 1, 3 and 6 antinociceptive responses by 3-way ANOVA

Table 3.2 Analyses of day 7 antinociceptive responses by 3-way ANOVA

  xii

SUMMARY OF ABBREVIATIONS

AC Anterior commissure

ACpa Pars anterior branch of the AC

ACpp Pars posterior branch of the AC

ADAM A-disintegrin and metalloprotease

AMPA 2-amino-3-(5-methyl-3-oxo-1,2- oxazol-4-yl)propanoic acid

ANOVA Analysis of variance

CGRP Calcitonin gene related peptide

CNS Central nervous system

DAB 3,3’-diaminobenzidine

DAMGO [D-Ala2, N-MePhe4, Gly-ol]-enkephalin

DAPI 4',6-diamidino-2-phenylindole

DOR Delta opioid receptor

DRG Dorsal root ganglion

ECL Extracellular loop

Eph receptor Erythropoietin producing hepatocellular receptor

Ephexin Eph-interacting exchange proteins

Ephrin Eph family receptor interacting protein

ERK Extracellular signal-regulated kinase

GABA γ-Aminobutyric acid

GDP Guanosine diphosphate

GEF Guanine nucleotide exchange factor

GFAP Glial fibrillary acidic protein

  xiii

GPI Glycosyl-phosphatidyl-inositol

Grb Growth factor receptor-bound protein

GIRK channel G protein-coupled inwardly-rectifying potassium channel

GPCR G protein coupled receptor

Grb4 Growth factor receptor-bound protein 4

GRIP Glutamate receptor-interacting protein1

GRK G protein-coupled receptor kinase

GTP Guanosine triphosphate

HRP Horseradish peroxidase

IB4 Isolectin B4

ICL Intracellular loop

JMR Juxtamembrane region

KD Kinase domain

KOR Kappa opioid receptor

LBD Ligand binding domain

LC-MS/MS Liquid chromatography–tandem mass spectrometry

LTD Long term depression

LTP Long term potentiation

M3G Morphine-3-glucuronide

M6G Morphine-6-glucuronide

MAPK Mitogen-activated protein kinase

MOR Mu opioid receptor

MRM Multiple Reaction monitoring

M/Z Mass-to-charge ratio

  xiv

N-WASP Neural Wiskott-Aldrich syndrome protein

NMDA N-methyl-D-aspartate

PBS Phosphate buffered saline

PDZ postsynaptic density protein (PSD95)/Drosophila disc large

tumour suppressor (Dlga)/ zonula occludens-1 protein (zo-1)

PFA Paraformaldehyde

PKA Protein kinase A

PKC Protein kinase C

RT Retention time

RTK Receptor tyrosine kinase

SAM Sterile α motif

SEM Standard error of the mean

SFK Src family kinase

SH2 Src-homology 2 domain

TLT Transfer latency time

TM Transmembrane

CHAPTER 1: INTRODUCTION

1

1.1 History of Opioids

For thousands of years, crude opium extracts from poppy seeds (Papaver somniferum)

have been used for medicinal and recreational purposes, and are among the oldest known agents

to treat pain [1]. Despite their significant worldwide usage clinically, use of opioids is

complicated by a variety of side effects including the risk of respiratory depression, sedation,

and gastrointestinal dysfunction [2, 3]. As well, repeated use of opioids increases the risks for

the development of tolerance, physical dependence and addiction [2, 3]. Opium is a mixture of

plant alkaloids comprised primarily of benzylisoquinoline and phenanthrene class-alkaloids [3].

In 1805, morphine, a phenanthrene alkaloid, became the first pure alkaloid isolated from opium

extracts. The name morphine is derived from Morpheus, the Greek god of sleep [1]. Over the

next few decades, additional alkaloids were isolated from opium extracts and as a class the drug

group became known as opiates [3].

In 1973, three independent research laboratories determined that opiates bound to

specific receptors within the brain [4-6]. Given the nature of these receptor responses,

researchers hypothesized that these receptors must normally bind endogenous forms of these

ligands. In 1975 Hughes et al., determined the first of these endogenous ligands now termed

enkephalins [5]. When introduced, this short polypeptide exhibited morphine-like properties

with binding to opiate receptors within the brain [7]. Shortly thereafter, two additional peptide

classes with morphine-like properties were discovered and classified as endorphins and

dynorphins [8, 9]. Collectively, these ligands are now termed opioids, distinguishing them from

opiates, compounds found naturally in opium [3, 10]. Historically, opioid receptors were named

and classified based their most selective agonists. As such, receptors were named mu (µ) for

morphine, kappa (κ) for ketocyclazocine, and sigma (σ) for SKF 10,047 (or N-

allylnormetazocine) [11]. However, the sigma opioid receptor is no longer considered a true

opioid receptor and has been replaced by the delta (δ) opioid receptor [12]. My thesis focuses

2

on understanding the interaction between a specific EphB-family receptor and the mu opioid

receptor.

1.2 Opioid Receptors

As indicated above, three primary types of opioid receptors exist: mu (µ), delta (δ), and

kappa (κ). In recent years, a fourth subtype of opioid receptors has also been reported, named

the Opioid Receptor Like-1 (ORL1) [13-15]. This receptor subclass, although its amino acid

sequence is similar to that of other opioid receptors, it does not bind to classical opioid ligands

[15, 16]. All opioid receptors are encoded by separate structural genes. The receptor genes for

mu, kappa, delta, and ORL1 are Oprm1, Oprk1, Oprd1 and Oprl1 respectively. Binding sites

within opiate receptors share several structural similarities. Thus, opioid receptors can exhibit

significant overlap with respect to binding of particular ligands, despite noted profiles of

selectivity. The majority of opioids and opiates can bind to multiple receptor subtypes, but may

exhibit noted selectivity toward a particular receptor subtype [1]. Also, a few variants exist for

each opioid receptor subtype: µ1/ µ2, δ1/δ2, and κ1/ κ2/ κ3 [3, 12]. Table 1.1 summarizes the

prototypic ligands and important physiological effects of each receptor subtype [3, 17-19].

Structurally, the genes of opioid receptors are highly homologous to one another, with their

coding regions divided over three exons. Exon 1 codes for the extracellular domain and for

transmembrane domain I. Exon 2 codes for transmembrane domains II–IV. Exon 3 codes for

transmembrane domains V–VII followed by the cytoplasmic C-terminal region. The only

variation currently known is for the Oprm1 gene where the last 12 codons of exon 3 are actually

found on a fourth coding exon [20], and in recent years a number of additional exons having

been reported (see below).

Research over the past several decades has confirmed all opioid receptors display classic

G-protein coupled receptor (GPCR) signaling characteristics [12, 21, 22]. Like other GPCRs,

3

OpioidReceptor Exogenous Ligands Expression within CNS Physiological

Effects

μ

MorphineHydromorphineEtonitazeneHeroin

ThalamusHypothalamusStriatum Periaqueductal gray Median raphe nucleusHippocampusSuperior colliculusOlfactory bulbsSubstantia gelatinosaLaminae I and IIouter of the dorsal spinal cordDorsal root ganglia

AnalgesiaEuphoriaPhysical -dependenceEuphoriaRespiratory-depressionMiosisReduced GI motilityProlactin release

κ

PentazocineTifluadomNalorphineBremazocineEthylketocyclazocine

HypothalamusPeriaqueductal gray ClaustrumStriatumSubstantia gelatinosa

AnalgesiaPhysical -dependenceSedationMiosisDysphoriaHallucinations

δEtorphineD-Pen2-D-Pen5-enkephalinD-Ala2- Glu4-deltorphin

Pontine nucleiAmygdalaOlfactory bulbsDeep cortexHippocampusNucleus accumbensStriatum

Weak analgesiaAnti-depressantPhysical -dependenceInhibition of smooth muscle

ORL1NNC 63-0532Ro64 – 6198

AmygdalaThalamusHypothalamusHippocampusSeptal nucleiHabenulaHypothalamusRaphe nuclei Locus coeruleus

AnxietyDepressionAppetiteDevelopment of tolerance to mu agonists

Table 1.1. Overview of opioid receptors. For each receptor subtype, prototypic ligands,

CNS expression and physiological effects are summarized.

4

opioid receptors are believed to be capable of hetero or homodimerization [21, 23]. GPCRs can

be classified into six primary groups based on their relative sequence homology and functional

similarities. Opioid receptors belong to Class A, the Rhodopsin-like receptor family. Within this

class, opioid receptors are categorized under the γ subfamily [12, 22, 24]. Like many GPCRs,

opioid receptors are composed of 7 transmembrane (TM) spanning regions [12, 25]. These

opioid receptors display high sequence homology with highly conserved intracellular loops and

transmembrane (TM) domains. In particular TM helices II, III and VII display the highest

sequence homology (75%) among the different classes of opioid receptors. Variations among

opioid receptors classes include the N-terminus, extracellular loops (especially ECL 2 and 3), C-

terminus and the number and location of surface glycosylation sites (5 on MORs, 2 on DORs

and 2 on KORs) [12, 26]. These differences are thought to explain the different affinities and

actions of opiate ligands on opioid receptors.

1.2.1 Mu Opioid Receptor Structure

Cloning and characterization of MORs [27-29] provided much insight into their

structure-function relationships. Initially, the MOR gene Oprm1 was believed to contain one

promoter and four exons, all of which encoded one protein [30]. However, pharmacological

binding studies revealed multiple isoforms of MORs existed: mu1, mu2, and morphine-6-B-

glucuronide [31]. However, given that a mouse has a single Oprm gene, researchers began to

investigate how multiple subtypes of the MOR could exist. Researchers hypothesized that

alternative pre-mRNA splicing may be playing a role [30-32]. Multiple studies have shown that

that the mouse Oprm1 gene is more complex than initially thought, and in fact comprises two

promoters generating up to 27 splice variants [30] (Figure 1.1). These splice variants have been

established to exist either through differential 3′-splicing or 5’-splicing. Variants of 3′-splicing

are typically generated through the exon1 promoter and are traditional G protein coupled

5

Figure 1.1. Alternative splicing of mouse Oprm1 gene. Two major classes of splice

variants exist, those generated by the promoter associated with exon 1 (white), and those

generated by the promoter in exon 11 (yellow).

Adapted from Majumdar et al., Proc Natl Acad Sci USA , 2011

6

receptors with 7-TM regions [32]. These variants differ in their carboxyl termini, which create

functional differences in their agonist-induced G-protein activation, adenylyl cyclase activity,

receptor internalization and agonist-induced receptor phosphorylation [30, 31]. As well, some

variants display different desensitization properties, where some MOR isoforms are more

resistant to agonist-induced desensitization [33, 34]. In contrast, 5′ splice variants which are

believed to be generated through exon 11 promoter, comprise traditional full-length receptors

but also truncated 6-TM and 1-TM variants [32]. These exon 11-associated variants are believed

to mediate drug specific actions such as analgesia without the common side effects typically

observed [32]. Specifically, it has been shown that exon 11 knockout mouse exhibited normal

morphine and methadone induced analgesia, but failed to display analgesic response following

heroin, M6G, and fentanyl administration [35]. These variants also displayed differences in

regional distributions within the CNS [36].

Until recently, the crystal structure of opioid receptors was extrapolated from closely

related GPCRS such as the β-adrenergic receptor [37]. Recently Manglik et al., (2012)

crystallized the 2.8A˚ mouse MOR complexed with the irreversible morphinan antagonist β-

FNA (β-Funaltrexamine) [38] (Figure 1.2). As expected, the structure consisted of seven TM α-

helices connected by three extracellular loops (ECL1–3) and three intracellular loops (ICL1–3).

TM3 is connected to ECL2 by a conserved disulphide bridge [38]. The intracellular surface of

the MOR closely resembled that of the related GPCR, rhodopsin, with respect to the relative

positions of TM3, TM5 and TM6. The MOR crystallized structure was found arranged in

dimmers, with the association found between the TM5 and TM6 interface. Another, dimer

structure was also found to exist between the interface of TM1 and TM2, and helix 8.

Normally, ligand binding pocket within GPCRs is buried within a helical bundle [38]. In

contrast, the binding pocket for β-FNA in the MOR was largely exposed to the extracellular

surface, and made contact with TM3, TM5, TM6 and TM7 [38]. The authors suggest that this

7

Figure 1.2. Crystal structure of the mu-opioid receptor bound to morphinan antagonist β-

Funaltrexamine. (A) Chemical structure of β-Funaltrexamine. (B) Crystal structure of mu

opiate receptor showing seven transmembrane domains typical of G-protein coupled receptors.

β-FNA ligand is indicated in green.

Adapted from Manglik et al., Nature 2012

β-Funaltrexamine

A. B.Extracellular

Intracellular

8

type of exposed ligand binding pocket may provide a basis for fast dissociation kinetics of

opiate ligands, and thus could be used to explain some of the unique pharmacologic and

physiologic properties of distinct opioid ligands. For instance, potent opioids such as

buprenorphine and etorphine have an inhibition constant (Ki) of 740pM and 270pM,

respectively; and rapid dissociation half-lives of 44 min and 1 min, respectively [38]. Thus, the

authors of the study suggested this may explain why heroin overdoses are rapidly reversible by

naloxone (highly selective competitive MOR antagonist); given that the MOR binding pocket is

largely exposed to the extracellular surface [38].

1.3 Ligands for Opioid Receptors

1.3.1 Endogenous Ligands

Opioid receptors differ in their physiological responses, tissue distribution and relative

affinity for various opioid ligands. All endogenous opioid peptides share a common NH2-

terminal Tyr-Gly-Gly-Phe sequence which interacts with the opioid receptor [1, 39]. Opioids act

both centrally and peripherally, and depending on the class of opioid receptors activated, they

mediate different physiological responses. Opioid peptides are initially synthesized as part of a

larger precursor molecule. Each opioid peptide arises from a unique precursor, which has

prepro- and pro- forms, from which the active opioid peptide and other neuroendocrine peptides

are derived from [10].

Enkephalins are short pentapeptides cleaved from the precursor pro-enkephalin A, and

was first identified in the adrenal medulla [10]. Subtypes of this peptide include: Met-

enkephalin, Leu-enkephalin, Met-enkephalin-Arg6-Phe7, Met-enkephalin-Arg6-Gly7-Leu8 and

peptide E. Met-enkephalin and Leu-enkephalin possess high selectivity for DORs [10, 39]. Met-

enkephalin-Arg6-Phe7 and Met-enkephalin-Arg6-Gly7-Leu8 display comparable affinities for

9

MORs and DORs, with lower affinities for KORs. Peptide E shows high affinity to MORs but

also for KORs [10]. β-endorphins are 31-amino acids long peptides, which are cleaved from

pro-opiomelanocortin (POMC), found in the pituitary [10]. β-endorphins are the main parent

peptide of the endorphin family, although shorter cleavage products of β-endorphins have been

reported. They display binding selectivity for MORs over DORs, with negligible affinity for

KORs [10, 40]. Dynorphins are 17-amino acids long peptides. They are cleaved from pro-

dynorphin (also known as pro-enkephalin B). This family of peptides is comprised of:

dynorphinA (1-17), dynorphinA (1-8), dynorphin B, α-neo-endorphin, and β-neo-endorphin

[10]. The dynorphin family of peptides display greater preference for KORs. However,

dynorphinA (1-8) display some binding affinities for DORs and dynorphinA (1-13) is potent at

both KORs and MORs [10, 39]. For the ORL1 receptor, the only known ligand is Orphanin FQ

(OFQ or Nociceptin). Structurally it resembles other opioid peptides particularly dynorphin A.

Similar to other peptides described, it is also derived from a preproOFQ precursor to yield a

peptide that is 17-amino acids long [14-16]. Table 1.2 summarizes the relative binding affinities

of these peptides to their respective opioid receptors [41].

1.3.2 Exogenous Ligands

Exogenous ligands for opioid receptors comprise a spectrum of drugs that includes

opiates (derived from opium) but also semi-synthetic and synthetic compounds (Figure 1.3).

Examples of opiates include morphine and codeine (3-methylmorphine) [3]. Semi-synthetic

morphine derivatives display morphine-like pharmacology and consist of a morphine-like

chemical structure. For example, heroin is a diacetylated morphine, which is twice as potent as

morphine; or oxycodone which has a methoxy substituent replacing the C3 hydroxyl group and

has better oral bioavailability than morphine [3]. Fully synthetic opioids also display morphine-

like pharmacology but do not possess a morphine-like chemical structure. For example,

10

Ligand

Ki (nM)

Mu Delta Kappa

β‐endorphin 1.0±0.1 4.0±0.1 15.6±1.5

Met‐enkephalin 22.3±0.1 5.8±1.1 1566.0±221

Leu‐enkephalin 68.2±10.8 9.4±1.8 3697.0±420

Met‐enkephalin‐Arg6‐Phe7 6.9±1.0 3.9±1.2 19.2±4.3

Met‐enkephalin‐Arg6‐Gly7‐Leu8 23.7±1.4 9.0±1.0 9.3±2.1

Peptide E 2.0±0.5 5.6±1.0 7.3±0.86

Dynorphin A (1‐8) 17.7±2.2 11.5±2.0 2.3±0.7

Dynorphin A (1‐17) 2.6±0.9 6.3±0.9 0.3±0.09

Dynorphin B (1‐13) 12.6±2.1 20.5±2.5 1.4±0.32

α‐Neo‐Endorphin 4.0±1.2 2.6±0.8 3.6±1.1

β‐Neo‐Endorphin 15.6±3.2 9.8±1.2 5.0±1.0

Table 1.2. Binding affinities of endogenous opioid peptides to Mu, Delta and Kappa

opiate receptors in nanomolar.

11

Figure 1.3. Structures of common MOR ligands. (A) Example of an endogenous MOR

agonist β-endorphin. (B) Common MOR opiates and semi-synthetic ligands. (C) Synthetic

ligands. (D) Common antagonists and partial agonist (buprenorphine).

12

β-endorphin

Morphine Codeine Heroin

Naloxone Naltrexone

Oxycodone

Buprenorphine

Fentanyl Methadone

A.

C.

D.

B.

A

C

B

3

6

13

methadone which is as potent as morphine but has a longer duration of action, and fentanyl

which has a rapid onset but a short duration of action [3]. In addition, over the years researchers

have designed several synthetic opioids demonstrating greater class specificity. Examples of

these highly selective agonists for the mu, delta and kappa receptor sites are DAMGO (D-

Ala2,N-MePhe4,Gly-ol5-enkephalin), DPDPE (D-Pen2,D-Pen5-enkephalin) and U69,593,

respectively [42-44]

Structurally, opiates can exist in either the levorotatory (-) or dextrorotatory (+) form.

The biologically active isomer of morphine is the levo isomer. The dextro isomers do not

possess analgesic properties [3]. Within morphine (Figure 1.3), the presence of the phenolic OH

groups in position 3 is considered important for opiate action [39]. Substitution of the methyl

group on the nitrogen atom on morphine determines the agonistic or antagonistic nature of the

ligand. For instance a methyl group results in agonistic activity, whereas substitutions of allyl/

cyclopropylmethyl/ or propyl groups results in antagonistic pharmacological activity [39].

Examples of opioid antagonists include naloxone, naltrexone, and buprenorphine (Figure 3).

Naloxone which has an allyl substitution has the greatest affinity for MORs [39]. Naltrexone

which has a cyclopropylmethyl is similar to naloxone, but has a longer half-life [3, 39].

Buprenorphine has a cyclopropylmethyl substituent on the N-atom like naltrexone. However,

because it also has a methoxy group on C6 similar to codeine, it displays mixed agonistic and

antagonistic properties [3].

1.3.2.1 Morphine

Morphine is a prototypical MOR opiate [3]. Its C-ring adopts a boat conformation,

which places the 6α-hydroxyl group in an equatorial position (Figure 1.3). This hydroxyl group

is believed to confer the principle component of selectivity [25]. Morphine can also bind with

lower affinities to KORs and DORs and produce diminished analgesic responses, as examined

14

through MOR knockout mice [45]. Since MORs are expressed in the CNS and peripherally,

treatment with morphine frequently provokes undesirable side effects such as respiratory

depression, constipation, nausea and vomiting [2, 3, 46].

Metabolism of morphine is primarily catalyzed by uridine 5’-diphospho-

glucuronosyltransferase (UGT) enzymes [47]. Morphine exhibits a systemic half life of

approximately two hours in humans and 50 minutes in mice [48]. In humans, morphine is

primarily metabolized by UGT2B7 to morphine-3-glucuronide (M3G) and morphine-6-

glucuronide (M6G) (Figure 1.4) [47, 49, 50]. In rodents, UGT2B1 has also been shown to

catalyze the formation of M3G [49]. Glucuronidation occurs on free hydroxyl groups. Aromatic

hydroxyl groups (position 3 of morphine) are glucuronidated more readily than alicyclic

hydroxyl groups (position 6 of morphine) [47, 50]. In humans, approximately 60% of morphine

is converted to M3G and 10% to M6G [51]. The remaining 30% of morphine is converted to

compounds such as normorphine, 3-acetylmorphine, morphine-6-sulfate, morphine-6-sulfate,

and 6-acetylmorphine [51].

A significant fraction of morphine binding occurs within the CNS. The exact

mechanism of transport of morphine and its metabolites into and out of the brain is still unclear.

However, the primary blood brain barrier transporter of morphine is believed to be the P

glycoprotein (Pgp) (also known as ABCB1 transporter), which is an ATP-dependent active

efflux pump [47, 52]. This transporter has been shown to limit the rate of morphine

accumulation within the brain [52]. However, this transporter has not been shown to transport

morphine-glucuronides across the blood brain barrier [47, 53].

In humans, both M3G and M6G are produced. Mice also metabolism morphine to M3G,

however whether they metabolize morphine to M6G is still a matter of some debate. Several

studies suggest that little to no M6G is produced in rodents [54, 55]. However, the issue is

clouded by the relative low levels of M6G compared to other morphine metabolites. M3G is

15

MORPHINE

MORPHINE-6-GLUCURONIDE

(M6G)

UGT 2B7 UGT 2B7

Figure 1.4. Metabolism of morphine. Morphine is metabolized by the liver to two major

metabolites: morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G). M3G

comprises approximately 60% of morphine metabolites, while M6G comprises 10%.

MORPHINE-3-GLUCURONIDE

(M3G)

UDP-glucuronosyltransferaseUDP-glucuronosyltransferase

16

biologically inactive and does not bind to the MORs [47]. Treatment with M3G alone does not

illicit analgesic activity [47]. However, some studies have reported it can antagonize some

pharmacological actions of morphine [56], although this is still a matter of some debate [57-59].

In contrast, M6G possesses distinct analgesic properties such as exhibiting a slower onset of

action and longer duration of action compared to morphine. M6G has also been reported to

exhibit equal or greater potency than morphine for MORs [56, 60-62]. Notably, M6G bypasses

many of the common side effects seen with morphine such as respiratory depression [47]. This

has created much interest in M6G as a possible therapeutic alternative to morphine. A downside

of this is that M6G is more hydrophilic than morphine and hence exhibits lower blood brain

barrier permeability [47, 63].

1.4 Mu Opioid Receptor Signaling

Activation of MOR signalling mediates many physiological functions such as analgesia,

respiratory depression, state of euphoria, sedation, reduced gastrointestinal mobility, nausea, and

miosis [2, 3, 46]. As with other GPCRS, opioid receptors convey their signals through activation

of heterotrimeric G-proteins via exchange of bound GDP for GTP, and interaction with the

inhibitory (Gi/Go) G-proteins [37, 64-66]. Such conformational modulation is achieved through

interaction of the MOR cytoplasmic domain with the G-protein heterotrimer. Specifically,

ligand binding causes a re-arrangement of transmembrane domains 3, 6, and 7 from the inactive

to the active conformation. In the CNS, MORs are found presynaptically and postsynaptically.

Presynaptically, MOR mediated G-protein activation results in inhibition of adenylyl cyclase,

which decreases cAMP production and inhibition of Ca2+ influx [67]. This decrease in calcium

influx ultimately results in decreased neurotransmitter release (Figure1.5). Postsynaptically,

MORs work through G-proteins to enhance cellular K+ efflux through G protein-coupled

17

Ca2+

MOR

Adenylate Cyclase

AMPARNMDAR

GIRK

Presynaptic Cell

K+

Neurotransmitter Release

MOR

Ca2+

Na2+ K+

Postsynaptic Cell

HYPERPOLARIZATION

Figure 1.5. Pre- and postsynaptic MOR signaling. Presynaptic MORs inhibit adenylate

cyclase activity and presynaptic Ca2+ channels, while promoting voltage-gated K+ channel

opening, resulting in reduced neurotransmitter release (such as GABA) from synapses.

Postsynaptic MORs induce hyperpolarization through activation of GIRK channels, thus

inhibiting NMDA and AMPA channel opening.

18

inwardly-rectifying potassium channels (GIRKs). This action hyperpolarizes the neuron,

inhibiting activity of both NMDA and AMPA receptors [68, 69] (Figure 1.5).

Activation of the dopaminergic mesolimbic system plays a role in mediating opioid

reward, and development of opioid dependence and addiction. The current accepted neuronal

circuit model suggests morphine excites and activates dopamine neurons in the ventral

tegmental area (VTA) indirectly through disinhibition of GABAergic neurons in the VTA [70-

72]. Specifically, morphine activation of MORs hyperpolarizes local GABAergic interneurons

in the VTA, thereby decreasing GABA release onto dopamine neurons [70-72]. This decreased

GABA release removes the inhibition on the VTA neurons. As a result, VTA neurons can then

project back to nucleus accumbens and increase dopamine release from dopaminergic neurons

[70-72]. This has been shown experimentally through numerous studies where opioids such as

DAMGO microinjected directly into the VTA caused an increase of dopamine release in the

nucleus accumbens following activation of MORs in the VTA [70-72]. Opioid injections into

the VTA have also been demonstrated to mediate rewarding effects through numerous self-

administration and place conditioning studies [70-72].

1. 5 Morphine Tolerance

Like many opiates, morphine exhibits significant susceptibility toward the development

of tolerance and physical dependence which limits its use in several contexts of chronic pain.

Following the development of tolerance, increasing doses of the opiate must be administered to

maintain an equivalent level of analgesia. Tolerance poses significant limitations to opiate use as

increasing morphine concentration may cause toxic effects, physical dependence and addiction

[73]. Traditionally, tolerance had been viewed as a consequence of decreased number of

functional receptors at the cell membrane. However, recent studies have shown that simple

down-regulation of opioid receptors does not occur unanimously with all opioid agonists.

19

Following activation, MORs may undergo receptor desensitization and endocytosis

(Figure 1.6). Following endocytosis, receptors can be recycled from endosomes back to the

plasma membrane, thus allowing resensitization. Alternatively, receptors can be retained

intracellularly or targeted to lysomoes/proteosomes for degradation thus down regulating

receptor numbers [74]. MOR regulation following activation is suspected to be ligand

dependent. Various ligands such as β-endorphin, fentanyl, methadone and DAMGO are

believed to promote rapid MOR internalization and recycling [75, 76]. In contrast, morphine,

pentazocine and buprenorphine promote less MOR internalization [76-80]. Thus, it is believed

that morphine has a greater tendency to induce development of tolerance and dependence

compared to other MOR agonists such as DAMGO and fentanyl, since morphine activation of

MORs does not promote removal of MORs from the plasma membrane [74, 78, 81].

The MOR specific agonist DAMGO promotes MOR internalization and induces rapid

desensitization, endocytosis and recycling (Figure 1.6) [75, 76]. DAMGO activates MORs in a

similar pattern through the heterotrimeric G-protein. Repeated exposure results in MOR

phosphorylation through the G-protein coupled receptor kinase (GRK). It is this phosphorylation

that attracts arrestin to the MOR, thereby uncoupling the MOR-G-protein complex, and

interrupting activation of downstream signaling cascades [82]. The arrestin-MOR complex

becomes endocytosed through clathrin coated pits and fuse with early endosomes [79]. It is in

the early endosomes that DAMGO unbinds MORs and the MORs become dephosphorylated

and return to the cell surface for another round of activation [79].

Unlike DAMGO, morphine does not promote MOR internalization [77, 79]. Therefore,

researchers have hypothesized that it is this feature that makes morphine administration prone to

the development of tolerance. Morphine induces weak desensitization with minimal

endocytosis. That is because morphine activated MORs fail to undergo sufficient receptor

phosphorylation by GRKs [77, 79]. Without phosphorylation, arrestin fails to be recruited to

20

Figure 1.6. Proposed pathways for MOR regulation. Regulation of MORs have been

proposed to be ligand specific. Following receptor activation, MORs may become

phosphorylated by GRKs which then recruits arrestins to the receptor. Arrestins subsequently

uncouple MORs from associated G-proteins. Following uncoupling, MORs frequently

undergo endocytosis and are internalized. Once internalized, MORs enter the degredative

pathway or are recycled. In the recycling pathway, phosphatases dephosphorylate the MOR,

thereby re-sensitizing it and allowing its return the cell surface.

Adapted from Connor et al., British Journal of Pharmacology, 2004

21

MORs thereby limiting clathrin-dependent endocytosis. Therefore, differential agonist

regulation of MORs has been suggested to occur due to differences in regulation of receptor

phosphorylation. MORs possess approximately 20 serine, threonine, and tyrosine residues on

their intracellular loops and carboxyl terminal tail [73, 83]. Following activation, these amino

acids on MORs may become phosphorylated by kinases depending on the agonist [84]. For

example, G-protein coupled receptor kinase (GRK2 or 3) may regulate the activity of MORs

through phosphorylation of its intracellular domains, and subsequently recruiting arrestin 2 or

arrestin 3 binding (depending on the agonist induced activation) to the phosphorylated residues

of MORs [82]. Thus, agonist-induced receptor phosphorylation is believed to be important for

regulation of opioid tolerance.

Few studies have shown that morphine can induce increased receptor internalization

following over expression of GRK2 [81, 85]. Overexpression of GRK-2 increased

phosphorylation and hence desensitization of MORs [85]. Similarly, overexpression of β-

arrestin (arrestin 2) has been shown to increase MOR internalization in vitro [81, 85].

Alternatively, loss of the β-arrestin-2 (arrestin 3) gene in mice strongly impaired agonist

induced desensitization of MORs, and enhanced analgesic response following morphine

treatment [86]. As well, mice lacking β-arrestin-2 displayed reduced development to tolerance

to MOR opioids such as morphine, suggesting that MOR regulation requires β-arrestin-2 [86].

Although, mice lacking β-arrestin-2 failed to develop tolerance, they were able to develop

physical dependence [87]. This is consistent with other studies that have suggested that

tolerance and dependence maybe mediated by separate mechanisms [86-88].

1.6 Opiates and NMDA Receptors

NMDA receptors are vital for synaptic plasticity. The connections between MOR

signaling and hippocampal LTP have also been examined upon opiate administration. Studies

22

have revealed that repeated exposure to morphine could impair LTP in the hippocampal CA1

region [89, 90]. However, this LTP could be restored following re-exposure to morphine [89,

90]. Thus, numerous studies have implicated a role for NMDA receptor signalling in promoting

the development of morphine tolerance [91]. Consistent with this, administration of non-

competitive NMDA antagonists such as MK-801, has been shown to attenuate the development

of morphine tolerance [92, 93]. Although studies have attempted to examine the role of NMDA

in morphine tolerance, the specific mechanisms governing these effects remain unclear. The

current accepted model of opioid dependence is that repeated opiate administration causes

adaptive increases through the influx of Ca2+ into the synapse [94]. The increased Ca2+ has been

shown to be mediated through the activation of NMDA receptors which leads to the opening of

the receptor-gated ion channels, allowing Ca2+ to enter the neuron [94]. The increased Ca2+

influx results in elevated expression of Ca2+/calmodulin-dependent protein kinase (CaMKII).

CaMKII in turn phosphorylates CREB, which increases c-Fos mRNA expression [94]. Gene

expression is thought to play an important role in many forms of neuronal plasticity. Blocking

NMDA receptors, which is known to be required for development of morphine tolerance,

inhibits the activation of CaMKII and thus negatively regulates gene expression.

The exact mechanism underlying the role of NMDA receptors in morphine tolerance

remains unclear. Provided that morphine activated MORs fail to undergo GRK phosphorylated-

arrestin mediated internalization, researchers have investigated whether there are other

mechanisms regulating MOR desensitization. A number of studies have demonstrated that

protein kinase C (PKC) is involved in opioid tolerance or desensitization. Specifically, PKC has

been shown to mediated inhibition of MOR internalization and thus play a role in the

development of acute tolerance through desensitization of MORs [95, 96]. It has been suggested

that perhaps PKC phosphorylates MORs directly or indirectly through phosphorylation of other

proteins involved in receptor desensitization [95, 96]. Treatment with PKC inhibitors has been

23

shown to induce morphine-mediated MOR internalization, and thus attenuate morphine

tolerance [95, 96]. As previously mentioned, chronic morphine treatment has been shown to

enhance NMDA activity and thus elevate intracellular levels of Ca2+. MOR activation induced

Ca2+ influx has been shown to activate protein kinases such as PKC [97, 98]. Increased

intracellular PKC has been shown to potentiate NMDA activated currents, by increasing the

probability of channel openings and by reducing the voltage-dependent Mg2+ block of NMDA-

receptor channels [97, 98]. PKC inhibitors blocked this potentiation of NMDA receptors.

1.7 Animal Model: Mu Opioid Receptor Knockout Mice

Over the years several strains of mice lacking the mu opioid receptor have been

generated. These knockouts were created through either the deletion of exon 1 [99, 100],

insertion of a Neo cassette in exon 2 [101], or deletion of exons 2 and 3 [102]. All MOR null

mutants exhibit normal growth and are fertile, indicating that MORs are not essential for

survival. No morphologic or behavioural abnormalities were detected in MOR null mice [99,

100]. Loss of MORs did not alter expression or distribution of other opioid receptors, or alter

transcription regulation of genes encoding for endogenous ligands. Moreover, administration of

morphine in MOR null mice did not induce analgesia, hyperlocomotion, reward, physical

dependence, withdrawal, induction of drug-dependent place preference activity, or other

peripheral effects (e.g. respiratory depression, inhibition of gastric motility) [99, 100]. Loss

MORs was also functionally confirmed using [3H] DAMGO binding [20]. Similar to morphine,

administration of DAMGO induced no analgesic response in these animals [20]. Although

morphine is known to bind with lower affinity to receptors other than the MORs, no major

changes in physiological responses were observed suggesting no major compensatory changes

occurred within the opiate receptors. These findings confirm that the major physiologic effects

of morphine are mediated through MORs [99, 100].

24

1.8 Eph Receptors and Ephrins

Erythropoietin-producing hepatocellular carcinoma (Eph) receptors represent the largest

known family of mammalian receptor tyrosine kinases [103]. They were discovered while

attempting to identify oncogenic kinases within the carcinoma cell line, ETL-1 [103]. Eph

receptors are classified into two major sub-groups, EphA and EphB, depending on their ligand

binding preferences [104, 105]. A total of 14 Eph receptors have been characterized: EphA1-8,

10 and EphB1-4,6 [106]. Eph receptors bind to Eph receptor interacting ligands named ephrins.

Ephrins are similarly divided into 2 major sub-classes: ephrinAs which are bound to the cell’s

outer membrane via a glycophosphatidylinositol (GPI) linkage, and ephrinBs which are

transmembrane proteins with their own intracellular signaling capabilities [104, 105]. In

mammals, 8 ephrins have been identified, ephrin-A1–5 and ephrin-B1–3 [106, 107].

This ligand-receptor system is unusual in that both the receptor and ligand are membrane

bound. This allows cell signaling to be propagated bidirectionally through Eph receptor

mediated (forward) signaling or through ephrin mediated (reverse) signalling [108, 109]. In

most instances, EphA receptors preferentially bind to ephrinA ligands, while EphB receptors

bind to ephrinB ligands. However, known cases of receptor promiscuity exist, such as the

binding of EphA4 to ephrinB2. Similarly EphB2 demonstrates significant affinity for ephrinA5

[106, 110]. Tables 1.3 and 1.4 summarize the relative binding affinities of EphB2 and EphA4

[104, 111]. Ligand binding results in receptor auto-phosphorylation. Following receptor

activation, signaling cascades are initiated (see below).

1.8.1 Eph/ Ephrin Structure

EphA and EphB share similar basic structural motifs, differing principally in the amino

acid sequences governing the ligand binding site (Figure 1.7). The ectodomain region consists

25

Ephrin-Fc Binding Affinity(Kd)

EphrinB2 2.4x10-10 M

EphrinB1 1.2x10-9 M

EphrinA5 2.8x10-8 M

EphrinA2 2.4x10-7 M

EphrinA4 5.4x10-7 M

EphrinA1 7.4x10-7 M

EphrinB3 1.0x10-6 M

Ephrin A3 2.0x10-6 M

Table 1.3. Binding affinities of ephrin-Fc’s for EphB2 ligand binding domain.

Interactions were determined by surface plasmon resonance. Note the class promiscuity

present with respect to the relative affinities of ephrins for EphB2.

26

Ephrin Calculated Affinity (Kd)

EphrinB1 8.6nM

EphrinA1 0.395nM

EphrinA3 3.0nM

EphrinA2 3.96nM

Table 1.4. Calculated binding affinities of ephrin ligands to EphA4.

27

P

P

P

P

P

P

Ephrin-B

PDZ

Ephrin-A

P

P

EphA Receptor EphB Receptor

-----------------SAM-----------------

------------Kinase------------

-------Juxtamembrane Region-------

-------------Fibronectin III-------------

-------------Fibronectin III-------------

---------Cystein-rich domain---------

-------Ephrin-binding domain-------

------------------PDZ------------------

GlycophosphatidylinositolLinkage

Figure 1.7. Eph ligand and receptor structure. Eph receptors consist of an extracellular

ligand binding domain followed by a cysteine-rich and two fibronectin type three domains.

Beyond the transmembrane domain lies the juxtamembrane tyrosine motif and kinase

domain, followed by the sterile alpha motif (SAM) and C-terminal PDZ region. Ephrin A

ligands are associated with the plasma membrane via glycophosphatidylinositol (GPI)

linkages, whereas ephrin B ligands possess both transmembrane and intracellular regions.

28

of an amino-terminal ligand-binding globular domain, a cysteine-rich region, and two

fibronectin type III repeats, followed by a single transmembrane domain [107, 111-113]. The

intracellular domain is comprised of a short juxtamembrane region with several conserved

tyrosine residues, a tyrosine kinase domain, a sterile-alpha-motif (SAM) protein domain, and a

C-terminal PDZ (postsynaptic density protein/disc large/zona occludens) binding motif [106,

107]. Between EphA and B receptors, sequence identities are approximately 30–70% in the

extracellular domains, and approximately 65–90% in the kinase domains [111].

Conserved tyrosine residues within the juxtamembrane region have been shown to

regulate the early stages of receptor activation. In the unphosphorylated state these

juxtamembrane tyrosine residues fold to inhibit kinase activity [114, 115]. Phosphorylation of

these residues creates charge repulsion which opens the kinase domain, thus allowing

subsequent phosphorylation of tyrosine residues within the kinase motif to occur, and thereby

prompting full activation [114, 115]. Subsequently, the phosphorylated juxtamembrane region

serves as docking sites for Src homology 2 (SH2)-domain containing proteins [106, 116-118].

Furthermore, the SAM domain which may homo- and hetero-oligomerize with other SAM

domains, has been suggested to aid in receptor oligomerization by serving to stabilize receptor

clustering [119-121] . As well, the SAM domain may modulate active Eph receptors at the cell

surface by regulating endocytosis and receptor degradation [122]. Similarly, the PDZ domain

aids in stabilizing Eph/ ephrin clustering [106], but may also serve as target site for many

cytoplasmic scaffolding proteins, such as the recruitment of the Ras family proteins through

interaction with PDZ domains of other intracellular proteins [123].

EphrinA and ephrinBs share 30–70% identity within their core sequence which

comprises approximately 125 amino acids, including 4 invariant cysteine residues [111].

Ephrins are membrane bound ligands (Figure 1.7). The ephrinA subclass is anchored to the cell

membrane through a GPI linkage, which is uncommon among ligand families of other types of

29

receptor tyrosine kinases [111]. Ephrin-B ligands possess a single-pass transmembrane domain

and a short cytoplasmic tail of approximately 80 amino acids long containing five conserved

tyrosine residues. This followed by a C-terminal PDZ binding motif, which is important for

ephrin signaling (see below) [124, 125].

1.9 Eph-Ephrin Interactions and Signaling

The unique feature of Eph receptors and ephrins both being membrane bound, allows for

two different types of Eph-ephrin interactions: trans or cis interactions. When Eph receptors and

ephrins are expressed on opposing cells this is known as trans interactions. This type of

interaction results in bidirectional signaling (Figure 1.8). In contrast, when Eph receptors and

ephrins are expressed in the same cell this is known as cis interactions. This type of interaction

is believed not to mediate any active signaling [108].

Eph receptors require oligomerization for biological activity. Crystal structural analysis

of EphB2-ephrinB2 complex, revealed two distinct ephrin binding sites [126]. One site is

believed to be the high affinity binding site to ephrinB ligand, while the other is a lower affinity

binding site to another ephrinB ligand following dimerization with another EphB-ephrinB

complex [126]. EphBs and ephrinBs first bind with high affinity and specificity to form

heterodimers. Then at high concentrations, two EphB-ephrinB complexes (two heterodimer

complexes) form a circular tetramer [126]. In the tetramer complex, each ligand interacts with

two receptors and similarly each receptor interacts with two ligands [126]. This has been

suggested to promote higher-order clustering and initiation of bidirectional signalling [126].

However, this tetramer complex is believed to exist only with EphB-ephrinB complexes. That is

because, studies examining EphB2 bound to ephrinA5 showed that this complex exists solely as

a dimer [110, 127].

30

Figure 1.8. Eph-ephrin Signaling. Both Eph receptors and ephrins are membrane bound,

thus allowing a variety of cell-cell interactions and the potential for bidirectional signaling.

Signaling mediated by the Eph receptors is referred to as forward signaling, while that

mediated via ephrins is termed reverse signaling. Activation of Eph receptors alters Rho-

family (RhoA, Rac, and Cdc42) GTPases activity influencing actin polymerization and

ultimately growth cone and dendritic spine morphology. In addition, Eph receptors can

signal through Ras GTPase members influencing ERK/MAPK signaling to regulate cell

proliferation. EphrinBs illicit signaling cascades following phosphorylation by Src-family

kinases (SFKs). Following activation, signaling adaptors such as Grb are recruited to these

sites of phosphorylation, regulating features such as spine and synapse formation.

Phosphatase such as PTP-BL terminate ephrinB signaling cascades.

31

Ephrin-B

P

P

P

P

P

P

P

P

Rho GEFs(intersectin, kalirin)

Rac-GTP/Cdc24-GTP

Dendritic spine morphogenesis

Rho GEFs(Ephexin,

RhoA-GDP

Growth cone collapse

RasGAP

ERK/MAPK

Proliferation

SFK

Grb CytoskeletalDynamics

PTP-BL

Rev

erse

Sig

nalin

gFo

rwar

d Si

gnal

ing

Ephrin-A Ephrin-B

EphA Receptor EphB Receptor

RhoA-GTP Rac-GDP/Cdc24-GDP

32

When an ephrin binds to an Eph receptor, receptor auto-phosphorylation occurs. Soluble

ephrins do not trigger receptor auto-phosphorylation unless artificially pre-clustered [109]. Non-

clustered forms can act as functional antagonists [108]. This limits signaling of this nature to

cell-to-cell communication. Thus, as mentioned above, Eph-ephrin signaling is bidirectional.

Signaling may mediate and proceed through the receptor-bound membrane (forward signaling)

or through the ligand bound membrane (reverse signaling). Both signaling events can happen

simultaneously, and the relative contributions of Eph forward and ephrin reverse signaling can

vary depending on cellular context [108].

1.9.1 Forward Signaling

The majority of Eph-mediated effects on repulsive axon guidance and synaptic plasticity

are mediated through forward signaling via the Rho family of GTPases. The Rho family is

comprised of RhoA, Cdc42 and Rac. Cycling between the active GTP-bound form and the

inactive GDP-bound conformation is mediated through guanine exchange factors (GEFs). GEFs

exchange bound GDP for GTP thus activating Rho GTPases. EphA and EphB receptors activate

a unique subset of GEFs [128] (Figure 1.8). For example, the Ephexin family of Rho GEFs are

EphA specific. Ephexin promotes the activation of RhoA, while it inhibits activation of Rac and

cdc42. Activation of RhoA downstream of EphA receptors mediates growth cone collapse in

neurons through shifts in actin cytoskeleton dynamic to increase contraction and reduce

extension [129, 130]. In contrast, EphB specific Rho GEFs such as Kalirin and Intersectin

activate and signal through Rac and cdc24. Intersectin activates Cdc42 (filopodia extension) and

Kalirin activates Rac (dendritic spine extension) to regulate spine morphology [131-133].

However, both Kalirin and Intersectin regulate signalling downstream of EphB receptors

differentially. Kalirin binds to activated EphB2 receptors. In contrast, Intersectin binds to the

33

kinase domain of EphB2 (which is associated with the neural Wiskott-Aldrich syndrome protein

(N-WASP)), independent of receptor activation [132].

In addition, Eph receptors also regulate the Ras family of GTPases, such as H-Ras to

regulate cell proliferation. Upon activation, H-Ras subsequently activates a cascade of

serine/threonine kinases including Raf1, Mek1, and the MAP kinases Erk1 and Erk2 [128]. Eph

receptors can act as positive and negative regulators of the MAPK pathway. For instance,

EphB1 receptors have been shown to activate the MAPK cascade through recruitment of the

Grb2/Sos complex, which acts as a Ras-specific GEF [134]. This leads to activation of Ras

followed by activation of downstream serine/threonine kinases such as Raf1, Mek1, and the

MAP kinases Erk1/2. This induces cell proliferation. In contrast, in neuronal cells, EphB2 has

been shown to negatively regulate MAPK, through recruitment of p120RasGAP, which

suppresses H-Ras [135, 136]. This inhibits Ras activation, and thus inhibits activation of Raf1,

Mek1, and Erk1/2, and suppresses proliferative response (Figure 1.8).

1.9.2 Reverse Signaling

Following EphB binding and clustering, activated ephrinB subunits recruit Src-family

kinases (SFKs) which phosphorylate the tyrosine residues on the cytoplasmic tail of ephrinB

ligands [137]. These phosphorylated tyrosine residues induce conformational changes that

disrupt the β-hairpin structure. This allows for the binding of Src-homology 2/3 (SH2/SH3)

domain-containing signaling proteins, such as Grb4 [138, 139]. EphrinB reverse signaling

through Grb4 in neurons has been shown to play a role in a diverse set of activities including

spine maturation, synaptic plasticity and synaptogenesis [140, 141]. Signaling is terminated

upon dephosphorylation of ephrins by phosphatases such as PTP-BL [137] (Figure 1.8).

Moreover, reverse signaling mediated through ephrinA is at still unclear. Unlike ephrinB,

ephrinA ligands lack a cytoplasmic tail to transduce signalling cascades. However, it has been

34

postulated that perhaps ephrinA, through interactions with lipid-raft-associated protein

complexes recruit the Src family kinases, and mediate downstream signaling [142].

1.9.3 Termination of Eph-Ephrin Signaling

Proteases such as the ADAM family of metalloproteases and γ-secretase proteases have

been reported to cleave Eph receptors and ephrins thereby disrupting oligomer complexes and

terminating signaling [143, 144]. Eph-ephin complexes may also undergo trans-endocytosis,

whereby the entire complex is internalized into either the Eph or ephrin expressing cell [145,

146]. The mechanism underlying trans-endocytosis are still unclear, however there is evidence

to suggest the endocytosis is mediated by clathrins [147]. As well, ongoing research suggests

that direction of the endocytosis is bidirectional, mediated and dependent on the type of

signalling occurring: reverse or forward signaling. That is because, studies examining EphB2

receptors lacking the cytoplasmic region, which fail to mediate forward signaling when in

complex with ephrinB1s, were found to be internalized into the ephrin expressing cell.

Similarly, an EphB2 interaction with a truncated ephrin-B1 resulted in internalization into the

receptor expressing cell, while truncation of both EphB2 and ephrin-B1 prevented

internalization [145]. Endocytosis of the EphB-ephrinB complex is necessary to mediate cell-

cell repulsion to guide migrating cells and axons.

1.10 EphB2 Expression in the CNS

Eph-ephrin interactions regulate many functions such as axon guidance, cell

proliferation, cell migration, synaptic plasticity, and oncogenesis [106, 124]. During the

development of the CNS, the expression of Eph and ephrins fluctuate throughout different

regions of the CNS [148]. EphB2 which is also known as Cek5, Nuk and Sek3 prior to

standardization of Eph nomenclature in 1997 [105], has been shown to be expressed during

35

development and adulthood in the CNS. To investigate the expression pattern and role of EphB2

in the CNS, two mutant alleles of EphB2 null mice were generated [149]. One represented a null

mutation of the EphB2/Nuk gene. This was generated through homologous recombination in

embryonic stem cells by deletion of the 5′ segment of the Nuk locus and insertion of a neomycin

resistance cassette. In the second strain, an EphB2-β-galactosidase fusion was generated just

past the juxtamembrane tyrosines, hence eliminating the kinase, SAM and C-terminal PDZ

domains [149]. The generation of this NuklacZ allele allowed localization of EphB2 to by

dynamically tracked (in heterozygotes) and allowed differentiation of forward versus reverse

signaling processes.

During development, expression of EphB2 is largely within the developing nervous

system with higher expression among axonal tracts versus neuronal dendrites [150]. The earliest

expression of EphB2 in mice has been reported to be at E8.5, in the neuroectodermal cells of the

neural plate, as well as the ventral midbrain and hindbrain rhombomeres r3 and r5 [149]. At

E9.25 EphB2 expression is observed within the hypothalamic region of the diencephalon and

the tegmental region of the midbrain [149]. During embryonic development, EphB2 expression

is highest in sensory and motor neuronal axons. By E13.5, EphB2 becomes defined in a dorsal-

ventral gradient within the retinal ganglion cells [151]. EphB2 expression increases in other

regions such as the hypothalamus and preoptic area at E14.5. In addition, regions ventral to the

anterior commissure (AC) begin to express EphB2 around this period. By E15.5, the pars

posterior AC (ACpp) tracts are observed to cross the midline. In EphB2 null mice the ACpp

axons fail to migrate and project toward the floor of the forebrain instead [149]. As the

embryonic development nears the end, EphB2 expression within the brain largely shuts down

except in areas such as the superior colliculus and ventral forebrain [150]. By postnatal day 7,

EphB2 expression in the CNS begins to remerge, but expression pattern also reverses. In

contrast to embryonic development, postnatally, EphB2 expression is high in neuronal dendrites

36

[150]. Expression of EphB2 remerges in areas such as the CA3 and dentate gyrus of the

hippocampus. By postnatal day 10, EphB2 expression pattern widens and increases into the

hippocampus, neocotex, amygdala, and thalamic centers, as well as Purkinje cells. This

expression continues into adult hood [149, 150]. Therefore, the continued expression of EphB2

in regions undergoing continual synaptic modification signifies the important role of EphB2 in

regulating and modulating synaptic function.

1.11 EphB2 and Synaptic Plasticity

The sustained expression of Eph receptors and ephrins in the adult brain, especially in

regions associated with synaptic remodeling such as the hippocampus, olfactory bulb and

cerebellum suggests that EphB-ephrin signaling may play a role in regulating synaptic plasticity.

The importance of the Eph family in learning and memory has been examined using multiple

animal models. Comparisons between different Eph/ephrin knockouts and wild-type mice have

shown that pre- and post-synaptic Eph/ephrins mediate dendritic spine formation, synapse

formation and synaptic plasticity, all of which is required for learning and memory [107, 152,

153]. For instance, EphA4 null mice display morphologically disorganized, long and

overlapping dendritic spines [107]. While EphA6 knockout mice exhibit behavioural deficits in

learning and memory. Specifically, loss of EphA6 produced impairments in the fear

conditioning paradigm where EphA6 null mice displayed less freezing; and in the Morris Water

Maze where they failed to learn quickly in the hidden platform task compared to control

littermates [154].

EphB receptors have been showed to be localized at excitatory synapses, suggestive of

their role in synapse formation and regulation [155]. The specific synaptic roles of EphB

receptors have also been extensively studied using animal models such as the

EphB1/EphB2/EphB3 triple knockout mice [156]. The EphB family of receptor tyrosine

37

kinases, which is enriched at excitatory synapses, is important during synapse and spine

formation and maintenance. Triple knockout mice lacking EphB1/B2/B3 displayed fewer

excitatory synapses and decreased number of immature dendritic spines. However, this was

absent in the single or double knockout mice [156]. As well, hippocampal neurons cultured from

EphB1/EphB2/EphB3 triple knockout mice were abnormally long and thin in morphology, and

failed to produce mature spines [156]. This suggested that EphB-ephrinB signalling is required

for spine formation and maturation [150, 156, 157].

Excitatory synapses contain both NMDA and AMPA receptors. At these synapses, EphB

receptors have been shown to associate and cluster with NMDA receptors to indirectly regulate

Ca2+ influx [158]. EphB receptors phosphorylate NR2B in a Src-dependent manner to potentiate

Ca2+ influx through NMDA receptors. Elevated levels of intracellular Ca2+ phosphorylates

Ca2+/cAMP-responsive element binding protein (CREB), which induces the immediate early

gene c-Fos. Studies have shown that EphB2 receptors which are strongly expressed in the

hippocampus specifically associate with NMDA receptors [158, 159]. EphB2 plays a role in

stabilizing NMDA-dependent synaptic plasticity and synapse formation in regions such as the

hippocampus [158, 159]. EphB2, through its extracellular domains also directly interacts with

the NR1 subunit of NMDA receptors [155]. As such, EphB2 receptors have been implicated in

LTP generation through the NMDA receptors [150, 157]. Loss of EphB2 in mice has been

shown to reduce LTP and reduce localization of NMDA receptors in the cell membrane of the

synapse [150, 157]. Such findings indicate that EphB2 signalling plays a role in modulating

NMDA-mediated signaling to regulate synaptic plasticity which is essential processing in

learning and memory. Furthermore, EphB2 receptors have also been shown to associate with

and regulate AMPA receptors localization through PDZ domain containing proteins such as

GRIP [141].

38

The role of EphB, especially EphB2 in learning and memory has been a hot area of

research in the past few years. Loss of EphB2 and EphA4 receptors has been shown to precede

memory decline in a murine model of Alzheimer’s disease [160]. As well, recent research has

suggested that amyloid-β oligmoemers bind to the fibronectin repeats domain of EphB2, which

signals EphB2 for degradation. Loss of EphB2 in an Alzheimer model was found to display

reduced NMDA currents and impaired LTP in the dentate gyrus [161]. This is consistent with

previous findings regarding the role of EphB2 in modulating NMDA receptor activity.

1.12 EphB and Pain Modulation

A new area of research has emerged over the past few years examining the role of EphB-

EphrinB signaling in pain processing. As previously mentioned, EphB receptors have been

shown to regulate synaptic plasticity through interaction with NMDA receptors. Battaglia et al.,

was first to report and investigate pain processing through EphB-ephrin interactions in the rat

spinal cord. In the rat lumbar spinal cord, exogenous activation of EphB receptors using the

dimeric chimeric molecule, ephrinB2-Fc, decreased analgesic response by about 50% and

increased thermal hyperalgesia upon exposure to noxious thermal stimuli [162]. However, this

induced hyperalgesia was blocked if pretreated with NMDA receptor antagonist, MK-801[162].

Thus, this suggested that EphB activation through interaction of NMDA receptors was

mediating neuropathic pain [162].

Given that EphB and ephrinB expression is present within the DRG and spinal cord

[163, 164], a few studies examined whether expression of EphB/ephrinB increased following

injury. One study, through immunohistochemical methods reported that ephrinB2 expression

was enhanced in the DRG and spinal cord following a spinal nerve crushing injury model

[163]. To examine the exact involvement of the Eph system following neuropathic pain, the

researchers administrated ephrinB2 siRNA to reduce ephrinB2 expression. This resulted in

39

reduced mechanical allodynia [163]. Similarly, Song et al. also reported upregulated expression

of EphB1 and ephrin in the DRG and spinal cord following a chronic constriction injury model

[164]. As well, blocking EphB-receptors, through administration of EphB1-Fc and EphB2-Fc

chimeras, inhibited induction and maintenance of nerve injury-induced thermal hyperalgesia and

mechanical allodynia [165]. These blockers also prevented and suppressed the nerve injury-

induced hyperexcitability of nociceptive small DRG neurons, and reduced LTP induction at

synapses between C fibers and postsynaptic dorsal horn neurons in the spinal cord [165]. These

findings indicated that EphB- ephrinB receptor signaling contributes to the regulation of

neuropathic pain.

Given the role of EphB receptors in synaptic plasticity, researchers began to examine the

role of EphB in the processing of neuropathic pain and alterations of morphine responses. A

study by Han and colleagues reported that peripheral nerve injury unlike in wild-type animals

did not induce thermal hyperalgesia in EphB1 null mice [166]. As well, while intrathecal

injections of EphB receptor blocking reagent EphB2-Fc diminished behavioral responses to

morphine withdrawal, EphB1 null mice failed to exhibit development of physical dependence to

morphine compared to control littermates [166]. These findings indicated that the EphB1

receptor was necessary for the development of neuropathic pain and physical dependence on

morphine [166]. Interestingly, Liu et al. examined the role of NMDA receptor subunit NR2B

following morphine exposure. They reported that chronic morphine exposure significantly

increased phosphorylation of NR2B [167]. However, intrathecal administration of EphB2-Fc

inhibited NR2B phosphorylation. Therefore, these findings suggested that EphB receptor

signaling through interaction with NMDA receptors played a role in the development of opioid

physical dependence [167]. Another recent study by Liu and colleagues examined the role of

EphB1 signaling in regulating morphine tolerance with respect to bone cancer pain. They

40

reported that blocking EphB1 receptor activation using EphB2-Fc rescued the analgesic effect of

morphine and prevented the development of morphine tolerance [168].

The aforementioned studies indicate that EphB‐family signaling plays a role in

modulating nociceptive sensory responses and morphine responses. Research over the years has

also implicated a role for NMDA receptors in regulating morphine tolerance through its

influences on opiate signaling. However, the exact mechanism through which EphB receptors

such as EphB2 interact with NMDA receptors to mediate morphine related responses and

regulate morphine dependent tolerance remains unknown.

1.13 Learning and Memory: An overview

Cases such as H.M. (Henry Gustav Molaison 1926-2008) and others have provided

valuable insights into the organization and execution of processes which mediate human

learning and memory [169]. In an attempt to cure H.M.’s epilepsy, an experimental bilateral

medial temporal-lobe resection was performed, where components of his hippocampus,

parahippocampal gyrus, and amygdala were removed [170]. As a result of this procedure the

patient experienced severe anterograde amnesia, and demonstrated deficiencies in acquisitions

of new episodic and semantic knowledge [171]. He also suffered moderate retrograde amnesia

and could not remember the majority of events 3 years prior to his surgery [170]. However, his

working and procedural memory were intact, as he could learn and retain new motor skills for

short periods of time [170, 171]. In this respect, H.M.’s short-term and procedural memories

were largely unimpaired; however his long-term memory exhibited severe deficits. This and

other similar cases lead scientists to postulate that more than one region of the brain was

involved in the learning and execution of memory, thereby supporting the concept that multiple

memory systems existed [172].

41

Over the years, research has defined several different forms of memory. Memory can be

classified into short-term and long-term memory. Short-term memory is the temporary storage

of information for a short period of time; working memory is a form of short-term [173]. Long-

term memory has been broadly categorized into declarative (explicit) and non-declarative

(implicit) components [169, 172]. Through the imposition of specific brain lesions, researchers

have been able to interrogate which regions are critical for specific forms of memory. Studies

from H.M. and of other patients highlighted the importance of structures within the medial

temporal lobe and medial thalamic regions in establishing and maintaining declarative

memories, which represent explicit descriptions of facts, events, places and objects, [174-176].

However, integration of non-declarative or implicit (sub-conscious) memory is believed to lie

largely outside of these temporal structures, in regions such as the basal ganglia, cerebellum and

components of the limbic system [169, 172]. Non-declarative memory has been defined as the

facilitation of performance on a task due to previous experience without conscious recollection

or awareness of that experience [177]. Non-declarative memory is further subdivided into

priming, procedural, non-associative learning, and associative/classical conditioning learning

[172]. Priming represents the recognition of objects/words due to prior exposure and the

neocortex has been suggested to processes these types of memories. Procedural memories are

implicit memories that become skills and habits, such that once acquired these memories are

evoked unconsciously and automatically [175]. Through localized brain lesion and

pharmacological blocking approaches, research has shown that the dorsal striatum mediates a

component of this type of procedural memory [178, 179]. The cerebellum has also been shown

to be involved in mediating procedural memory [175] and is an important region for motor

learning together with constituents of the basal ganglia [180]. Furthermore, non-associative

learning is learning to a single stimulus that evokes a reflex [172]. In contrast, associative

learning is based on responses to multiple stimuli. Classic conditioning (common examples

42

include fear conditioning or conditioned place preference) represent forms of associative

learning [172]. Lesion studies in the context of fear conditioning have revealed a role for the

amygdala together with the hippocampus [172, 179]. The conditioned place preference

paradigm is used to measure the rewarding properties of drugs such as opiates, where animals

develop a preference for the drug paired environment. Studies have shown that components of

the hippocampus and the limbic system such as the amygdala are necessary for the development

of a conditioned place preference to a drug [181, 182].

Though distinct forms of learning may be mediated throughout different regions of the

brain, the basic cellular mechanisms mediating these forms of learning are believed to be

fundamentally similar. Because of the nature of its structural organization and central role in

learning and memory, studies of synaptic plasticity have focused on the hippocampus to identify

the cellular features of learning and memory. Research has demonstrated several forms of

cellular plasticity within this structure consistent with the Hebbian properties seen in learning.

These include activity dependent models of synaptic plasticity termed long-term potentiation

(LTP) and long-term depression (LTD) [183-185]. Induction of LTP involves coincident

activation of NMDA receptors with subsequent activation of intracellular calcium-dependent

signaling. Specifically, there is a transient increase in postsynaptic calcium through the NMDA

receptor, and through voltage-gated calcium channels, or through calcium permeable AMPA

receptor channels. This increase in calcium influx leads to activation of protein kinases such as

calcium-calmodulin dependent protein kinase II (CaMKII), protein kinase A (PKA) and protein

kinase C (PKC) [183]. However, LTP has been shown not to be confined solely to synapses of

the hippocampus, but can be induced within numerous CNS regions associated with learning

[183]. Though, LTP has been a primary cellular model of learning and memory, recent evidence

suggests that LTD may also play a similar role, particularly for motor learning at sites such as

the cerebellum [175, 180, 186].

43

1.13.1 Hippocampus

The hippocampus is a component of the medial temporal lobe and consists of a largely

unidirectional neural circuit. The main input into the hippocampus is from the entorhinal cortex,

which projects to the dentate gyrus via the perforant path [187, 188]. Inputs from dentate gyrus

project to the CA3, at which point Schaffer collaterals from the CA3 project to the CA1 region

of the hippocampus. From here the outputs flow to the subiculum which serves as the main

output for the hippocampus [187, 188]. Originally the hippocampus was believed to be uniform

in function, however over the years studies have suggested functional dissociations exists

within hippocampal sub-regions, specifically along the septotemporal axis [189-191]. Numerous

studies over the past three decades have highlighted the critical role of the hippocampus in

spatial and contextual learning. Studies demonstrate that loss of hippocampal function

substantially impairs acquisition and retention of spatial learning in rodent paradigms such as

the water and radial mazes [179, 192]. Specifically, lesions confined to the dorsal hippocampus

were shown to impair spatial learning in the water and the eight-arm radial maze tasks [189,

191]. As well, recent studies also suggest the CA1 region of the dorsal hippocampus is

important for acquisition of spatial memories, as mutant mice lacking the NMDA receptors in

the CA1 synapses exhibit impaired synaptic currents and LTP and impaired spatial learning

[193]. In contrast, lesions confined to the ventral hippocampus were comparable to controls with

respect to spatial learning. Ventral hippocampal lesions instead appeared to be involved in the

processing of emotional responses to fear [189-191]. Such findings are consistent with studies

demonstrating that the dorsal hippocampus receives greater sensory input from visual, auditory

and somateosensory association areas via connections from the entorhinal cortex, while the

ventral hippocampus is more strongly connected to sub-cortical loci [194, 195]. As well, the

dorsal hippocampus contains a higher proportion of place cells [194]. Place cells are believed to

44

be hippocampal pyramidal cells that are important for processing of spatial information. Studies

done in rats reveal that a collection of place cells represent specific regions within the

environment, and when the rat is in a particular location in the environment (place field) these

cells fire in their place fields. The firing of these cells thus provides the spatial information of a

specific location within the environment [196, 197].

Moreover, lesion studies reveal a requirement for intact hippocampal function for the

processing of contextual cues [192]. Researchers have utilized tests such as the Pavlovian fear

conditioning task to monitor hippocampal involvement in contextual learning and memory

[192]. In classical fear conditioning, a conditioned stimulus is paired with an aversive

unconditioned stimulus such as an electrical foot shock. This typically elicits conditioned fear

responses, such as freezing [198]. Studies have revealed that the hippocampus is required for the

processing of the contextual cues associated with the unconditioned stimulus. Lesions to the

hippocampus, specifically, animals with lesions on the dorsal hippocampus fail to develop fear

and exhibit impairment in freezing when tested in a contextual fear conditioning task [198, 199].

Morris water and radial maze studies have been previously used to examine hippocampal

dependent spatial and contextual learning. However, in my own studies passive avoidance and

open field tests were utilized to examine hippocampal function. Passive avoidance represents an

associative/contextual task, where animals are conditioned to avoid the dark compartment paired

with the aversive stimulus (foot-shock). Thus, the passive avoidance task tests an animals’

ability to process and link a specific contextual cue (dark compartment) with an aversive

stimulus (foot-shock). It has been shown that animals with lesions to the dorsal hippocampal

CA1 and CA3 regions are impaired in this conditioned learning, and are likely to enter the dark

compartment [200]. In fact, CA1 lesioned animals performed as poorly as CA3 lesioned animals

despite less cellular damage evident in the CA1 lesioned animals versus the CA3 lesioned

animals [200]. Furthermore, open field tests allow measurement of exploratory activity, which is

45

dependent upon interaction of the animal with a novel environment [201]. Studies have shown

that hippocampal lesions, correlate with increased locomotor hyperactivity [202]. Thus, this

open field activity has been utilized as a measure of exploration, but also habituation and

learning due to the environmental cue [201].

1.14 Opiate-Dependent Tolerance and Learning

In an attempt to understand the mechanisms underlying drug tolerance, researchers have

begun to explore how environmental variables might influence drug response following

administration. Early theories of opiate tolerance suggest that tolerance was primarily due to

decreases in the sensitivity or number of active opiate receptors within a given interval [203-

205]. However, evidence subsequently emerged that the phenomena of morphine tolerance

could be viewed as a form of Pavlovian conditioning, where the environment represents the

conditioned stimulus, and the pharmacological actions of a drug represents the unconditioned

stimulus [203]. As such, associations between the systemic effects of the drug and local

environmental cues might influence experience-dependent drug response; promoting a

conditioned response. Key studies by Shepard Siegel revealed that if rats were made tolerant to

morphine in a given environment, those animals continued to display analgesic tolerance in the

environment in which they had previously been administered morphine [203, 204]. However,

tolerant rats failed to display analgesic tolerance in an alternative environment for which they

were exposed to for the first time [203, 204]. Thus, environmental cues associated with

morphine administration continued to establish tolerance, where as environmental cues not

linked to morphine attenuated tolerance [203-205]. Therefore, the phenomena of morphine

tolerance may include a component of experience-dependent learned response. However, the

exact molecular mechanisms by which such actions occur remain uncertain.

46

1.14.1 EphB2 and Opiate Tolerance

Previous work done in our laboratory, examined the basic motor and sensory intrinsic

responses between EphB2 null mice and wild-type litter mates. Motor tasks, such as the

hindlimb extension, edge performance, hindlimb grip response, grip strength, wire platform

locomotion and 90º incline climb were compared between EphB2 null and wild type animals

(Stephanie Ho, Thesis 2009). For all motor tasks with the exception of wire platform, EphB2

null mice performed identical to wild-type controls. In addition, sensory responses were

examined using Von Frey filament, tail pinch and tail flick tests. Similar to motor responses

examined, EphB2 null mice exhibited wild-type like sensory responses for the above tests.

Our laboratory examined and compared sensory responsiveness to the opiate morphine

between EphB2 null and wild-type animals. Examinations of the analgesic response of EphB2

null and wild-type littermates following morphine administration using tail pinch and tail flick

(data not shown) assays was performed by a previous graduate student (Ashlin Kanawaty,

Thesis 2011). As shown on Figure 1.9, upon initial exposure to morphine, both EphB2 null and

control littermates exhibit similar patterns of morphine-induced analgesia as measured by tail

pinch latency during the first 30 minutes. However, at time periods greater than 45 minutes

following morphine injection on day 1 EphB2null mice demonstrated significantly lower tail

pinch latencies compared to wild-type controls. Repeated morphine administration over the next

several days induced a reproducible pattern of tolerance to morphine as demonstrated by the

reduced analgesic response. On Day 3 (Figure 1.10), consistent with the effects seen on day one,

EphB2 null mice continued to display accelerated tolerance compared to wild type littermates,

as shown by significantly reduced tail pinch latencies compared to controls. By day 6 (Figure

1.11), both EphB2 null mice and control littermates are substantially tolerized to the effects of

morphine as demonstrated with similar tail pinch latencies. These results suggest that while loss

47

0

1

2

3

4

5

6

7

8

9

10

0 15 30 45 60 75 90 105 120

Mea

n ta

il pi

nch

late

ncy

(sec

)

Time post morphine injection (min)

EphB2 +/+ n=22

EphB2 +/- n=26

EphB2 -/- n=24

*

*

**

Figure 1.9. Morphine tolerance in EphB2 null mice on Day 1. Analyses of

antinociceptive responses on day 1 as a function of tail pinch latency following initial

exposure to morphine. Shown are results for wild type, heterozygous and EphB2 null

littermates. EphB2 null mice initially exhibit tail pinch latencies similar to wild type

littermates (arrow) , however by 45 minutes EphB2 null mice deviate significantly from

controls. Error bars ±SEM, *p<0.05, wild type versus EphB2 null mice.

48

0

1

2

3

4

5

6

7

8

9

10

0 15 30 45 60 75 90 105 120

Mea

n ta

il pi

nch

late

ncy

(sec

)

Time post morphine injection (min)

EphB2 +/+ n=22

EphB2 +/- n=26

EphB2 -/- n=24

** *

*

* *

Figure 1.10. Morphine tolerance in EphB2 null mice on Day 3. Analyses of

antinociceptive responses on day 3 as a function of tail pinch latency following initial

exposure to morphine. Shown are results for wild type, heterozygous and EphB2 null

littermates. EphB2 null mice consistently deviate significantly from controls. Error bars

±SEM, *p<0.05, wild type versus EphB2 null mice.

49

0

1

2

3

4

5

6

7

8

9

10

0 15 30 45 60 75 90 105 120

Mea

n ta

il pi

nch

late

ncy

(sec

)

Time post morphine injection (min)

EphB2 +/+ n=22

EphB2 +/- n=26

EphB2 -/- n=24

** * * *

Figure 1.11. Morphine tolerance in EphB2 null mice. Analyses of antinociceptive

responses on day 6 as a function of tail pinch latency following initial exposure to morphine.

Shown are results for wild type, heterozygous and EphB2 null littermates. All groups now

show evidence of tolerance, however EphB2 null mice still deviate significantly from

controls. Error bars ±SEM, * p<0.05, wild type versus EphB2 null mice.

50

of EphB2 does not alter baseline sensory responses, it significantly alters the rate of morphine

tolerance.

To determine the degree to which the differences in morphine tolerance seen in EphB2

null mice was a function of modified higher cortical input as opposed to direct changes in opiate

receptor signal transduction, the opiate-dependent hippocampal behavior of EphB2 null mice

and control littermates was examined. Previous work by ourselves and others has demonstrated

that loss of EphB2 receptors disrupts the stability of postsynaptic NMDA receptors, reducing the

efficiency of hippocampal dependent LTP [150, 157]. We therefore examined the analgesic

response of EphB2 null mice and controls following a change in environmental setting. As

described in the materials and methods, following six days of twice daily morphine exposure,

treatment groups were divided on day 7. One group remained in the home environment, while

the other was transferred to a novel environmental setting. Consistent with previously reported

results [203], wild-type mice tolerized to morphine exhibited an increase in tail pinch latency

when placed in a novel environment compared to those retained in their home environment

(Figure 1.12) (Ashlin Kanawaty, Thesis 2011). In contrast, EphB2 null mice exhibited no such

enhancement in sensory analgesia upon placement into a novel environment on day 7 (Figure

1.13). Thus, loss of EphB2 alters these morphine-dependent learned responses, suggesting that

EphB2 null mice lack the ability to integrate these sensory-dependent associative cues.

51

0

1

2

3

4

5

6

7

8

9

10

0 15 30 45 60 75 90 105 120

Mea

n ta

il pi

nch

late

ncy

(sec

)

Time post morphine injection (min)

EphB2 +/+ SAME n=7EphB2 +/+ NOV n=7

#

# ##

#

Figure 1.12. Antinociceptive responses of EphB2 wild-types upon removal to novel

environment. Following seven days of morphine treatment, half of the wild-type animals

were placed in a novel environment and tested for antinociceptive response. Similar to

previous reports, wild-type littermates exhibited a significant increase in antinociceptive

response when placed in a novel environment (dotted line) compared to animals retained in

their home environment (solid line). Error bars ±SEM, #p<0.05, home versus novel

environment.

52

0

1

2

3

4

5

6

7

8

9

10

0 15 30 45 60 75 90 105 120

Mea

n ta

il pi

nch

late

ncy

(sec

)

Time post morphine injection (min)

EphB2 -/- SAME n=8

EphB2 -/- NOV n=9

#

Figure 1.13. Antinociceptive responses of EphB2 null mice upon removal to novel

environment. Following seven days of morphine treatment, half of the EphB2 null mice

were placed in a novel environment and tested for antinociceptive response. In contrast to

wild-type littermates, EphB2 null mice exhibited no significant difference in antinociceptive

response in the novel environment (dotted line) compared to animals retained in their home

environment (solid line). Error bars ±SEM, #p<0.05, home versus novel environment.

53

1.15 Thesis Rationale

Following the development of opiate tolerance, the degree of physiologic analgesia can

be significantly modified by alterations in previously unlinked environmental stimuli. Such

effects demonstrate that there is a significant learned component in the perception and execution

of opiate dependent analgesia. With respect to such learned behaviors, numerous studies have

demonstrated the central role by which NMDA-mediated signaling plays in regulating the

development of behaviors such as LTP. Inhibition of such signaling is known to inhibit

development of morphine tolerance. Despite such progress, the molecular mechanisms linking

these features remain elusive.

Previously we and others have demonstrated that the EphB receptor, EphB2, interacts

with NMDA receptors to prolong their activity, thereby enhancing features such as LTP. As

well, EphB2 is persistently expressed within the adult CNS at sites involved in sensation and

processing of sensory stimuli, and EphB signalling has been shown to modulate sensory

responses. In these present studies, I am examining the relationship between EphB2, morphine

tolerance, and the role of hippocampal learning in modification of this process.

1.15.1 Thesis Hypotheses

The specific hypotheses to be addressed in my thesis are:

1) EphB2-mediated signaling regulates the development of morphine-dependent opiate

tolerance.

2) Such EphB2-mediated actions are mediated through a direct influence on neuronal

signaling, as opposed to secondary influences such as modifications of morphine

metabolism or changes in receptor distribution.

3) The effects of EphB2 on morphine tolerance are mediated via an influence on

hippocampal learning.

54

CHAPTER 2: MATERIAL AND METHODS

55

2.1 Animals

EphB2 wild type, heterozygous, and null littermate mice were generated as previously described

[150] and bred and maintained at University of Toronto Ramsay Wright Zoological

Laboratories. Mice used for studies were two to four months of age, weighing between 25 and

35 grams. Outbred male CD1 wild-type mice (aged two to three months, weighing between 35-

42 grams) from Charles River Laboratories (Wilmington, MA, USA) were used for

hippocampal lesion experiments. A mu opioid receptor knockout mouse (Oprm-/-) (The Jackson

Laboratory, Bar Harbor, Maine, USA) served as a negative control for immunohistological

studies.

2.2 Chemicals

Drugs: Morphine sulphate was obtained from the laboratory of Dr. Van der Kooy (Wiler PCCA,

London, ON) and was dissolved at a concentration of 1.5 mg/mL in 0.9% saline and

administered at a dose of 10 mg/kg intraperitoneally (i.p.). Morphine-3-glucuronide (M3G) was

obtained from the laboratory of Dr. Sandy Pang (National Institutes on Drug Abuse, Rockville,

MD, USA). All surgeries were done under anesthesia with 2.5% Avertin (Sigma-Aldrich)

injected i.p. at a dose of 0.2 mL/10g. Reagents used for LC-MS/MS analysis: Acetonitrile HPLC

Grade (Caledon), Methanol HPLC Grade (Caledon), Caffeine Anhydrous (Bioshop), Formic

Acid reagent grade (Sigma Aldrich), Perchloric Acid (Sigma Aldrich).

2.3 Morphine Tolerance Tests: EphB2 Wild-type and Null Mice

Animals were injected with 10 mg/kg morphine sulphate i.p. twice per day in the morning and

afternoon at intervals of 8 hours for a period of six days. Sensory tests (see below) were done

after morning injections on Day 1, 3 and 6. On day 7, animals were split into two groups. They

either remained in their home environment, or alternatively were placed in a novel environment

56

prior to receiving their daily injection of morphine and subsequent behavioral assessment

(Figure 2.1). This was previously performed on EphB2 wild-type and null mice by a previous

graduate student, Ashlin Kanawaty. These studies were repeated with lesioned and sham

operated control animals (see below).

2.3.1 Sensory Analyses: Tail Pinch and Tail Flick Assay

To test the analgesic response of morphine on animals, mechanoceptive (tail pinch) and

thermoceptive (tail flick) tests following morphine injection were conducted. Tail pinch assay

was performed using a flat forcep, and briefly the closing force was applied to the proximal

third of the animal’s tail. Nociceptive responses were indicated by the latency required for the

animal to respond to the pressure. To avoid tissue damage, a cut-off time of 10 sec was set. The

forcep was applied every 15, 30, 45, 60, 90 and 120 minutes following morphine administration.

Tail flick assay was performed at 55ºC. Animals were placed in a Plexiglas mouse retainer

allowing free tail movement. The distal third of a mouse tail was immersed in the warm water

bath and nociceptive responses were measured by the latency required for the animal to respond

to the temperature. To avoid tissue damage, a cut-off time of 15 sec was set. The tail was

immersed 30 min following morphine administration.

2.4 Pharmacokinetic Analyses of Morphine Metabolism

2.4.1 Preparation of LC-MS/MS Standard Solutions. Stock solutions of morphine and M3G

were prepared in 0.9% saline. All subsequent working standard solutions were prepared using

serial dilutions in acetonitrile, and stored at -20ºC. Whole blood and brain homogenates were

obtained from non-injected morphine naive mice. Standard solutions of morphine and M3G

were used to spike blank blood and blank brain homogenates with known concentrations to

57

Figure 2.1. Schedule of morphine dosing. Animals were injected i.p. with 10mg/kg

morphine twice per day at 8 hour intervals. Analgesic tests were conducted following the

morning injection on day 1, 3 and 6. Experiments on days 1-6 were conducted in the same

environmental setting (purple). For tests performed on day 7, animals were divided into 2

groups, with half remaining in the home environment (purple) and the other half placed in a

novel environment (pink).

58

Day 0

Day 1

Day 2

Day 3

Day 4

Day 5

Day 6

Day 7

TEST

10 am

TEST

6 pm

10 am

6 pm

10 am

TEST

6 pm

10 am

6 pm

10 am

6 pm

10 am

TEST

6 pm

10 am

TEST

6 pm

10 am

TEST

6 pm

Day 7

59

create a desired concentration range. Caffeine was used as an internal standard and prepared in

water at a stock concentration of 3 μg/mL.

2.4.2 Collection of Blood and Brain Samples. EphB2 wild-type and null animals were given a

bolus injection of 10 mg/kg morphine sulphate. Blood was then collected terminally by heart

puncture at 30, 60, and 90 min after injection. Blood samples were frozen immediately and

stored at -80ºC until analyzed. Brain samples (excluding the cerebellum) were homogenized in

0.1N perchloric acid, to a final concentration of 0.33g tissue/mL homogenate, vortexed and then

stored at -80ºC. Following one freeze-thaw cycle, brain homogenates were sonicated using the

Misonix 3000 water bath sonicator for 10-12 min at high speed in ice cold water. Samples were

then centrifuged at 15,000 rpm for 10 min. The supernatants were collected in new

microcentrifuge tubes and neutralized with 2M NaOH and stored at -80ºC until analyzed.

2.4.3 Purification of Blood and Brain Samples. A 10 μl and 20 μl aliquot of the internal

standard caffeine (3 μg/ml) was added to each 100 μL blood sample or to each 200 μL brain

sample, respectively. Blood and brain samples were vortexed for 60 seconds with 400 μL and

800 μL of an equal mixture of methanol and acetonitrile, respectively. Samples were then

centrifuged at 13,000×g for 10 min to precipitate the proteins. The supernatants were transferred

into Sep-Pak Vac C18 3 cc cartridges (200 mg; Waters, Milford, MA, USA). Each cartridge was

pre-conditioned with 2 mL of acetonitrile followed by 2 mL of Millipore water. After loading of

the sample, the samples were eluted with 2 mL of acetonitrile. The eluent was pooled and dried

under a stream of nitrogen at room temperature.

60

2.5 LC-MS/MS Analyses

The residue from blood and brain samples was reconstituted with 200 μL and 100 μL,

respectively, of the mobile phase (70% water with 0.1%v/v formic acid and 30% acetonitrile

with 0.1%v/v formic acid). For blood samples 1 μL of the reconstituted sample, and for brain

samples 5 μL of the reconstituted sample, was injected into the LC–MS/MS system for analysis.

Samples were analyzed by LC-MS/MS using 6410 Triple Quad LC/MS instrument (Agilent

Technologies) with ESI source in positive ion modes. Samples were separated on a C18 column

at 1 mL/min. The mobile phase consisted of HPLC grade water (A) and acetonitrile (B) both

containing 0.1% formic acid. The following gradient was run: 0-1 min, 4% (B); 4-5 min, 4%

(B); 5-9 min 4% to 100% (B); 9-10 min, 100% (B); 10-11min 100% to 4% B; 11-16 min, 4%

(B). MS parameters were as follows: gas temperature 350°C, nebulizer pressure 50 psi, drying

gas (nitrogen) 11 L/min and VCap 3500V. Using MRM monitoring (in positive-ion mode) the

following transitions were observed: morphine-3-glucoronide (m/z 462 286, RT 6.7 min),

morphine (m/z 286.1 165, RT 7.8 min) and caffeine (m/z 195 138, RT 11.4 min). Fragmentor

voltage settings used for morphine, M3G and caffeine were 155V, 160V and 85V, respectively.

Collision energy settings used for morphine, M3G and caffeine were 42V, 32V and 20V,

respectively.

2.6 Immunohistochemistry

Brain and spinal sections from wild-type, heterozygous, EphB2 null, and MOR knockout mice

(Oprm-/-) were prepared following intracardial perfusion of saline followed by 4%

paraformaldehyde in 0.1 M phosphate buffered saline pH 7.4 (PBS). Following overnight

fixation, samples were embedded in paraffin wax and 7 μm thick sections were sliced.

To probe sections with MOR antibody, antigen retrieval was performed. Following dewaxing,

sections were immersed in 10 mM sodium citrate (pH 6.0) and heated in a pressure cooker for

61

10 min to bring the water to a boil, followed by an additional two minutes at a boil. Following

cooling, sections were then pre-incubated in blocking solution (5% goat serum, 0.25% Tween-

20 dissolved in PBS) for 30min. Sections were then incubated with primary antibody, anti-MOR

(1:1000,Immunostar, rabbit, polyclonal) diluted in blocking solution overnight at 4ºC. Next day,

slides were washed (3x5 minutes in blocking buffer), then incubated in biotinylated goat anti-

rabbit secondary antibody (1:200, Vector Labs) for two hours at room temperature, followed by

tertiary reagent (avidin-horseradish peroxidase, Vector Labs) for 45 minutes, and visualized

using 3,3-diaminobenzidine (DAB).

2.7 Stereotactic Surgeries

Bilateral lesions of the dorsal hippocampus were performed as follows:

2.7.1 Kainic Acid Induced Lesion. Adult male CD1s mice (2-3 months of age) were

anaesthetized with 2.5% Avertin (0.2 mL/kg), and the scalp was incised along the sagittal

suture. At a position located 60% from the bregma, and 1.7 mm lateral of the sagittal suture,

burr holes were made in the skull using a carbon dioxide laser. The needle was inserted 1.15

mm below the dura and the kainic acid (Tocris Biosciences) was injected bilaterally into the

hippocampus at a concentration of 20 mM and a total volume of 300 nL. Thus, each mouse

received 6 nmoles of kainic acid per hippocampus. Following surgery, scalp was closed and the

animals were left to recover for 72 hours prior to start of behavioural assessments. Since

previous attempts at this experimental procedure resulted in seizures, a dose of 10mg/kg of

Diazepam immediately prior to surgery was administered. However, due to the continued high

mortality rate observed with kainic acid injections, this procedure was discontinued and

electrolytic lesions were performed as an alternative method.

62

2.7.2 Electrolytic Lesion. Adult male CD1s mice (2-3 months of age) were anaesthetized with

2.5% Avertin (0.2 mL/kg), and the scalp was incised along the sagittal suture. At the midpoint

between the lambda and bregma sutures, two 0.2 mm burr holes using a carbon dioxide laser

were drilled on either side of the sagittal midline at a displacement of 1.8 mm. A 0.1 mm

diameter platinum-iridium electrode with an exposed tip of 0.5 mm was then inserted to a depth

of 1.5 mm at each site. Electrolytic lesions were produced with a single constant direct current

discharge of 3 mA for 3 sec at each site of the hippocampus (Figure 2.2). Following surgery,

scalp was closed and the animals were left to recover for 72 hours prior to start of behavioural

assessments (Figure 2.2). Sham operated controls underwent the same exact procedure minus

the electrode insertion. After the completion of all behavioural testing, all mice were perfused

transcardially with 4% PFA. Brain sections were embedded in paraffin wax and 7 μm thick

sections were collected. Sections were stained with thionin dye to examine the full extent of the

lesion. Paraffin sections we also stained with GFAP (1:400, Dako) and DAPI.

2.8 Behavioral Analyses

2.8.1 Passive Avoidance. The passive avoidance apparatus consists of light and dark chambers.

Mice were initially placed in the light chamber for 30 seconds. Following this, the door to the

dark chamber is opened and the time required to enter the dark chamber was recorded as the

transfer latency time (TLT). This was recorded as the TLT-acquisition. Following 10 seconds in

the dark chamber, EphB2 wild-type and null mice were subjected to a one time shock of 0.5 mA

for 5 seconds (or 0.7 mA for 5 seconds for CD1 animals). After a total time of 30 sec in the dark

chamber, the animal was free to return to the light chamber, and subsequently returned to its

home cage. Twenty-four hours following acquisition, mice were re-introduced into the light

chamber. The time required for the animal to enter the dark chamber was recorded as TLT-

63

Lesion Sites

Day 0

Perform Stereotactic Surgeries

Post-SurgeryDay 3

72hrs

Passive Avoidance-Acquisition

Post-SurgeryDay 4

Passive Avoidance-Retention

Open Field Test

24hrs

A.

B.

Figure 2.2. Schedule of analysis following stereotactic surgery. (A) Bilateral lesions in the

dorsal hippocampus were performed at the midpoint between the lambda and bregma

sutures, 1.8 mm lateral from the midline and at a depth of 1.5 mm below the dura. Platinum

electrodes with an outer diameter of 100 μm were then placed to the proper depth following

laser drilling of the skull. Electrolytic lesions were induced using a current of 3 mA for 3

seconds. (B) Schedule of surgical recovery and behavioural testing following establishment

of lesions.

64

retention. A cut-off time of 300 sec was used if the animal failed to enter the dark chamber. This

test was previously examined on wild-type and EphB2 null animals by a previous graduate

student, Ashlin Kanawaty. Studies were repeated with lesioned and sham operated control

animals.

2.8.2 Activity Monitor. The activity monitor was used to conduct open field tests. EphB2 wild-

type and null mice were examined in a 25 cm by 42 cm open field for a period of one hour.

Spontaneous motor activity was recorded using an automated movement detection system

(AM1053 activity monitors; Linton Instrumentation, UK). The apparatus consisted of 24 (16 by

8) infrared beams forming a grid. Animal movement resulted in beam breakage and the resulting

activity and type of movement was registered. Studies were repeated with lesioned and sham

operated control animals.

2.9 Morphine Related Behaviour

2.9.1 Morphine Induced Hyperactivity

To determine morphine naïve performance, EphB2 wild-type and null animals were examined

on the activity monitor for a period of one hour. Twenty-four hours later they were injected with

a single dose of 10 mg/kg of morphine and analyzed for activity for a period of 60 minutes

as described above. Animals injected with 0.9% saline served as control. This was repeated with

lesioned and sham operated control animals.

2.9.2 Morphine Tolerance Tests: for Lesioned and Sham Operated Control Animals

Lesioned and sham operated control animals following the 72 hour recovery period and

assessment of behavioural functions were investigated for their response to morphine tolerance.

65

As described above, animals were injected with 10 mg/kg morphine sulphate i.p. twice per day

in the morning and afternoon at intervals of 8 hours for a period of six days. On day 7, mice

were split into two groups. They either remained in their home environment, or alternatively

were placed in a novel environment prior to receiving their injection of morphine and

subsequent behavioral assessment via tail flick. The tail flick assay (as described above) was

performed on day 7 at 30, 60, 90 and 120 min following the morning morphine injection.

2.10 Statistical Analyses

Two-way ANOVA with repeated measurement (time) using GraphPad Prism software V5.0

(GraphPad Software Inc, La Jolla, CA, USA) was used to assess latency and concentration

differences among animal groups overtime. Three-way ANOVA was conducted to compare

latency differences among animal groups overtime and over days using SPSS 16 (SPSS Inc.,

Chicago, IL, USA). Simple t-test was used to compare differences between each test group and

its corresponding control group using Microsoft Excel. All data are presented as mean±SEM.

Statistical results are considered significant if P ≤0.05.

66

CHAPTER 3: RESULTS

67

3.1 Morphine related responses of EphB2 null mice

Previously, our laboratory examined the baseline motor and sensory responses between

EphB2 null mice and wild-type littermates (Stephanie Ho, Thesis 2009). Similar to motor

responses examined, EphB2 null mice exhibited wild-type like sensory responses. Interestingly,

temporal and spatial distribution of EphB2 exists within the sensory nervous system (Figure

3.1). However, given that EphB2 null mice demonstrate no differences in baseline motor and

sensory responses from that seen in littermate controls, we began to examine whether other

elements of sensory response might be altered in EphB2 null animals. Thus, we examined

sensory responsiveness to the opiate morphine. Examinations of the analgesic response of

EphB2 null and wild-type littermates following morphine administration using tail pinch assay

was performed by a previous graduate student (Ashlin Kanawaty, Thesis 2011). As shown on

Figures 1.9-1.11, EphB2 null displayed accelerated morphine tolerance compared to control

littermates. These results suggested that while loss of EphB2 does not alter baseline sensory

responses, it significantly alters the rate of morphine tolerance. Consistent with this, analysis of

day 1 data using two-way ANOVA (genotype versus time with repeated measurement)

demonstrated a significant effect of both genotype [F(1,40)=6.85, p<0.05], and time

[F(6,240)=50.81, p<0.05], with a significant interaction between these factors [F(6,240)=2.91,

p<0.05]. Analysis of Day 3 data using two-way ANOVA also revealed a significant effect of

both genotype [F(1,40)=21.91, p<0.05], and time [F(6,240)=18.98, p<0.05], with significant

interaction between these factors [F(6,240)=6.79, p<0.05]. Similarly, analysis of Day 6 data by

two-way ANOVA again demonstrated a significant effect of both genotype [F(1,40)=11.39,

p<0.05], and time [F(6,240)=15.97, p<0.05], with significant interaction between these factors

[F(6,240)=7.34, p<0.05]. Analysis of day 1, 3 and 6 data using three-way ANOVA examining

genotype (wild-type versus EphB2 null) as between-subject factors, with time (0, 15, 30, 45, 60,

68

Figure 3.1. Expression of

EphB2 in the CNS.

Localization of EphB2 was

unequivocally determined

using animals heterozygous

for the EphB2lacZ allele in

which the intracellular kinase

domain is replaced by beta-

galactosidase. Arrows

indicate relative sites of

EphB2 expression: (A)

Periaqueductal gray, (B)

dorsal root ganglia, (C)

dorsal spinal laminae 1-3 of

the lumbar spinal cord, (D.1)

hippocampus , (D.2)

cingulate cortex, (E.1)

amygdala, (E.2) thalamus,

(E.3) hypothalamus.

B.

C.

A.

D.

E.

1

2

1 3

2

69

90 and 120 minutes) and day of analysis (1, 3 and 6) as the within-subjects factors, found all

effects and interactions to be significant (Table 3.1).

Our lab also previously performed Shepard Siegel’s experiment [203], to investigate

whther EphB2 null mice exhibited any alterations in learning/ behavioural responses to

morphine following the development of tolerance (Ashlin Kanawaty, Thesis 2011). Consistent

with previously reported results [203], wild type mice tolerized to morphine exhibited an

increased analgesic response when placed in a novel environment compared to those retained in

their home environment (Figure 1.12). In contrast, EphB2 null mice failed to display enhanced

analgesic response upon placement into a novel environment on day 7 (Figure 1.13). This

suggested that Eph B2 null mice lack the ability to integrate these sensory-dependent associative

cues. Analysis of this context-dependent learning using three-ANOVA using genotype (EphB2

null or control) and environment (novel or same) as between-subjects factors, and time (0, 15,

30, 45, 60, 90 and 120 minutes) as with within-subjects factors, demonstrated all effects and

interactions to be significant with the exception of the interaction between genotype and

environment (Table 3.2).

3.2 Distribution of mu opioid receptors in the CNS

We examined whether the accelerated development of morphine tolerance was attributed

to the direct loss of EphB2 receptors and its direct affect on MOR expression levels or

distribution pattern in the CNS. We examined MOR expression and distribution at several key

CNS sites using immunohistochemistry (Figures 3.2-3.3). EphB2 null mutants and control

littermates exhibited similar distributions of MORs within the dorsal spinal cord, which is

known to be enriched with MORs (Figure 3.2). A MOR null mouse (Oprm-/- mouse) served as a

negative control. As well, EphB2 wild-types, heterozygous and null mutants exhibited similar

patterns of MOR distribution within regions of the striatum and the hippocampus (Figure 3.3).

70

Factors ANOVA Results Significant?

time [F(6,240)=62.44, p<0.001] Yes

day [F(2,80)=51.66, p<0.001] Yes

genotype [F(1,40)=17.54, p<0.001] Yes

Interactions ANOVA Results Significant?

genotype and time [F(6,240)=9.00, p<0.001] Yes

genotype and day [F(2,80)=3.88, p=0.025] Yes

time and day [F(12,480)=9.14, p<0.001] Yes

genotype, time and day [F(12,480)=2.27, p=0.008] Yes

Table 3.1 Analyses of day 1, 3 and 6 antinociceptive responses by three-way ANOVA.

For each of the comparisons indicated, wild-type and EphB2 null (genotype) data were

compared as between-subject factors with time (0-120 minutes) and day (1, 3 and 6) as with

within-subjects factors. EphB2 null mice were found to differ significantly from wild-type

littermates for each of the parameters indicated.

71

Factors ANOVA Results Significant?

time [F(6,162)=11.19, p<0.001] Yes

environment [F(1,27)=51.66, p<0.001] Yes

genotype [F(1,27)= 4.97, p<0.001] Yes

Interactions ANOVA Results Significant?

genotype and time [F(6,162)= 19.64, p=0.034] Yes

genotype and environment [F(1,27)= 2.72, p>0.05] No

time and environment [F(6,162)= 7.48 p= 0.011] Yes

genotype, time andenvironment [F(6,162)= 5.99, p=0.038] Yes

Table 3.2. Analyses of day 7 antinociceptive responses by three-way ANOVA. For the

comparisons indicated, genotype (wild-type or EphB2 null) and environment (home or

novel) were utilized as the between-subject factors, with time (0 -120 minutes) as the within-

subject factors. EphB2 null mice were found to differ significantly from wild-type

littermates for each of the parameters indicated, with the exception of the interaction

between genotype and the environment.

72

A. B.

C. D.

400µm 400µm

400µm 400µm

Figure 3.2. Distribution of mu opioid receptors in the dorsal spinal cord of EphB2 null

mice and controls. Compared to wild-type littermates (A), loss of EphB2 does not alter the

laminar distribution of MORs within the dorsal spinal cord (B). Shown below are

comparable lumbar spinal sections from MOR null mice (C), and wild-type sections with no

addition of primary antibody (D).

73

Eph

B2-/-

Eph

B2+/

+E

phB

2+/-

400µm 400µm

400µm 400µm

400µm 400µm

A. Striatum B. Hippocampus

Eph

B2-/-

Eph

B2+/

+E

phB

2+/-

Figure 3.3. Distribution of mu opioid receptors in the striatum and hippocampus of

EphB2 null mice and wild-type littermates. Loss of EphB2 does not alter the pattern or

distribution of MORs in any of the target structures examined including the striatum (A) or

hippocampus (B), compared to controls.

74

Therefore, loss of EphB2 does not significantly alter the distribution of MORs in the spinal cord

and brain regions examined, compared to wild-type littermates.

3.3 Pharmacokinetic analysis of morphine in EphB2 null mice

One possible explanation for the accelerated morphine tolerance observed in EphB2 null

mice compared to wild-type littermates is that these animals may possess differences in their

relative level of morphine metabolism. For instance, if EphB2 null mice metabolized morphine

at a higher rate compared to wild-type controls, they might exhibit lower relative levels of

morphine analgesia at specific periods post-injection. Therefore, we investigated whether loss of

EphB2 significantly altered the kinetics of morphine metabolism. Initially, levels of morphine

and its main metabolite M3G were examined in a time-dependent manner in brain and whole

blood following first time exposure to morphine using LC-MS/MS. Concentrations of morphine

and M3G were determined in brain and whole blood at 30, 60 and 90 min following a single

injection of morphine. LC-MS/MS analyses revealed sufficient separation of primary

compounds within 15 min (Figure 3.4). Examination of morphine and M3G levels in the brain

demonstrated no significant differences between EphB2 null and wild-type animals at all time

points measured (Figure 3.5). Examination of morphine and M3G concentration in whole blood

was also found to be not significantly different between EphB2 null and wild-type animals at 60

and 90 min, but did differ at 30 min (Figure 3.6). In addition, I examined brain and whole blood

levels of morphine and M3G following repeated morphine exposure for 7 days. Consistent with

the above results, EphB2 null and wild-type animals display no significant differences in brain

or blood levels of morphine or M3G following chronic morphine exposure (Figure 3.7). Thus,

loss of EphB2 therefore does not induce major alterations in morphine metabolism.

Analysis of morphine brain levels using two-way ANOVA (genotype versus time with

repeated measurement) revealed no significant effect of both genotype [F(1,10)=0.64, p>0.05],

75

Figure 3.4. LC/MS/MS analyses of morphine and M3G. Elution profiles of morphine

and M3G compared to caffeine (internal calibration standard) in whole blood and brain

extracts. Using multiple reaction monitoring (MRM) the following transitions were

observed: M3G (m/z 462 286, RT 6.7 min), morphine (m/z 286.1 165, RT 7.8 min) and

caffeine (m/z 195 138, RT 11.4 min).

76

Morphine‐3‐Glucuronide

Morphine‐3‐Glucuronide

77

0.00

0.01

0.02

0.03

0.04

0.05

0.06

0.07

30 60 90

Mor

phin

e L

evel

s (ug

/g)

Time (min) of Brain Collection

EphB2+/+ (n=6)

EphB2-/- (n=6)

Figure 3.5. LC/MS/MS analyses of brain morphine metabolism in EphB2 null mice

and controls. No significant differences in brain morphine (A) or M3G (B) levels are

observed between EphB2 null and wild-type littermates following initial exposure to

morphine. Error bars: ±SEM.

A.

0.00

0.01

0.02

0.03

0.04

0.05

0.06

30 60 90

Con

cent

ratio

n (u

g/g)

Time (min) of Brain Collection

EphB2+/+ (n=6)

EphB2-/- (n=6)

B.

78

0.00

0.05

0.10

0.15

0.20

0.25

0.30

30 60 90

Con

cent

ratio

n (µ

g/m

L)

Minutes Post Morphine Injection

EphB2+/+ (n=6)

EphB2-/- (n=6)

*

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

30 60 90

Con

cent

ratio

n (µ

g/m

L)

Minutes Post Morphine Injection

EphB2+/+ (n=6)

EphB2-/- (n=6)

*

A.

B.

Figure 3.6. LC/MS/MS analyses of blood morphine metabolism in EphB2 null mice and

controls. No significant differences in morphine (A) or M3G (B) levels in whole blood are

observed between EphB2 null and wild-type littermates following initial exposure to

morphine at the 60 or 90 minute time points. A significant difference was observed at the 30

minutes. Error bars: ±SEM, *p<0.05, wild-type versus EphB2 null mice.

79

0.00

0.01

0.02

0.03

0.04

0.05

0.06

Morphine M3G

Con

cent

ratio

n (u

g/g)

Drug

EphB2+/+ (n=4)

EphB2-/- (n=4)

0.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

Morphine M3G

Con

cent

ratio

n (u

g/m

L)

Drug

EphB2+/+ (n=4)

EphB2-/- (n=4)

A.

B.

Figure 3.7. LC/MS/MS analyses of brain and blood morphine metabolism in EphB2

null mice and controls. No significant differences in (A) brain or (B) blood morphine and

M3G levels are observed in EphB2 null versus wild-type littermates following repeated

exposure to morphine (analysis 60 minutes following injection on day 7). Error bars: ±SEM.

80

and time [F(2,20)=2.48, p>0.05], with no significant interaction between these factors

[F(2,20)=1.79, p>0.05]. Analysis of M3G brain levels using two-way ANOVA revealed no

significant effect of both genotype [F(1,10)=0.40, p>0.05], and time [F(2,20)=6.10, p>0.05],

with no significant interaction between these factors [F(2,20)=0.18, p>0.05]. Analysis of

morphine blood levels using two-way ANOVA revealed no significant effect of genotype

[F(1,10)=2.83, p>0.05], but a significant effect of time [F(2,20)=14.18, p<0.05], however no

significant interaction between these factors [F(2,20)=4.06, p>0.05]. Analysis of M3G blood

levels using two-way ANOVA also revealed no significant effect of genotype [F(1,10)=4.14,

p>0.05], but a significant effect of time [F(2,20)=14.42, p<0.05], however no significant

interaction between these factors [F(2,20)=3.21, p>0.05].

3.4 EphB2 null mice display deficits in hippocampal learning.

Previous work in the lab (Ashlin Kanawaty, Thesis 2011) demonstrated that EphB2 null

mice exhibit significantly reduced transfer latency times compared to wild type controls in the

task of learned passive avoidance. In this task, animals experience single-pass training to an

aversive stimulus in a darkened chamber. Wild-type animals exhibited similar average transfer

latency times during acquisition as EphB2 null mice (Figure 3.8). However, following single

pass training, wild-type animals exhibited an expected increase in transfer latency times

indicating a level of learning had occurred. In contrast, EphB2 null mice exhibited significantly

lower transfer latency times into the dark chamber following single‐pass training relative to

littermate controls. These data support the contention that EphB2 null mice exhibit a deficit in

hippocampal-dependent tasks.

As previously mentioned, EphB2 null animals display deficits in NMDA-dependent

synaptic plasticity in the hippocampus as a result of reduced LTP in CA1 neurons of the

hippocampus. Therefore, to determine if the deficiencies in hippocampal learning observed in

81

0

50

100

150

200

250

300

EphB2+/+ EphB2-/-

Mea

n tr

ansf

er L

aten

cy (s

ec)

Genotype

Acquisition Retention

*

Figure 3.8. Passive avoidance responses of EphB2 mice. During initial acquisition,

transfer latency times were 56 ± 18 seconds for wild-type animals versus 32 ± 6 seconds for

EphB2 null mice. Following single pass training against an aversive stimulus, wild-type

animals (n=14) exhibited an increase in transfer latency to 242 ± 28 seconds. In contrast

EphB2 null mice (n=10) exhibited a significantly lower transfer latency period of 138 ± 9

seconds. Error bars: ±SEM, *p<0.05 for wild-type versus EphB2 null mice.

82

EphB2 null mice are directly related to the changes in morphine-dependent behavior seen in

these animals, I performed focal bilateral lesions of the dorsal hippocampus in wild-type mice in

an attempt to duplicate the effects seen following the ablation of EphB2. An example of such

lesion is shown in Figure 3.9. Lesions were localized to the dorsal hippocampus and serial

thionin stained sections showing the extent of these lesions is shown in Figure 3.10. Following

surgical recovery, animals were examined in a series of behavioral tests and compared with the

behavioural responses seen in EphB2 null mice.

3.4.1 Behavioural assessments of animals with bilateral electrolytic lesions in the dorsal

hippocampus

We investigated whether lesioned animals displayed altered responsiveness to learned

behaviours using the learned passive avoidance test. The passive avoidance task tests an

animal’s ability to learn the contextual cue (dark compartment) is paired with an aversive

stimulus (foot-shock). As shown in Figure 3.11, sham animals exhibited average transfer latency

times of 9.5±1.3 seconds in the acquisition phase, while lesioned animals had a latency time of

15±1 seconds. Following single pass training sham animals exhibited an expected increase in

transfer latency times to 84±23 seconds, indicating a level of learning had occurred. In contrast,

lesioned animals exhibited significantly lower transfer latency times into the dark chamber

following single‐pass training relative to sham animals (39.5±9 seconds). This is the same

pattern that was observed between EphB2 null and wild-type mice.

EphB2 null mice due to impaired hippocampal learning may be unable to habituate

quickly upon exposure to a novel environment. Thus, we also measured spontaneous

exploratory activity of animals upon placement into a novel environment using the Activity

Monitor. As shown in Figures 3.12-3.13, both EphB2 null and wild-type animals display

elevated levels of exploratory activity that decreased overtime throughout continuous exposure

83

Figure 3.9. Example of an electrolytic lesion of the dorsal hippocampus. Thionin (A),

GFAP (B), and DAPI staining (C) of regions proximal to electrolytic lesion are shown,

demonstrating destruction of CA1 pyramidal cells and overlying cortex.

400µm

500μm

500μm

A.

B.

C.

84

Figure 3.10. Serial views of electrolytic lesions of the dorsal hippocampus. Shown are

serial thionin stained sections taken at 200 µm intervals from (A) a non-stimulated control

hippocampus, and (B) a hippocampus containing a typical electrolytic lesion. Lesions were

generated bilaterally.

1mm

1mm

A.

B.

85

0

50

100

150

Sham Lesioned

Mea

n tr

ansf

er L

aten

cy (s

ec)

Animal Group

Accquisition Retention

*

Figure 3.11. Passive avoidance responses of lesioned wild-type animals. Following

induction of bilateral hippocampal lesions, animals underwent single pass training against an

aversive stimulus. Sham operated controls (n=17) and lesioned animals (n=23) demonstrated

similar transfer latency times during acquisition. However, when tested for retention,

lesioned animals display significantly lower transfer latency times compared sham operated

control animals. Error bars: ±SEM, *p<0.05 sham versus lesioned animals.

86

Figure 3.12. Active time of EphB2 null mice and lesioned wild-types. Spontaneous

activity as measured by mean active time per five minute period, was monitored for 60

minutes following introduction to a novel environment. (A) EphB2 null mice versus wild-

type littermates, (B) lesioned animals versus sham operated controls. Both EphB2 null mice

and lesioned wild-types exhibit higher levels of activity compared to wild-type controls.

Error bars: ±SEM, *p<0.05, comparison of control versus experimental animals.

87

0

40

80

120

160

200

5 10 15 20 25 30 35 40 45 50 55 60

Mea

n A

ctiv

e Ti

me

(sec

)

Time (min)

EphB2+/+ (n=18)

EphB2-/- (n=14)

*

** * *

*

**

**

0

40

80

120

160

200

240

280

320

5 10 15 20 25 30 35 40 45 50 55 60

Mea

n A

ctiv

e Ti

me

(sec

)

Time (min)

Sham (n=16)

Lesioned (n=19)*

**

**

*

A.

B.

88

Figure 3.13. Motor activity of EphB2 null mice and lesioned wild-types. Motor activity

as defined by mean distance traveled per five minute period, was monitored for 60 minutes

following introduction to a novel environment. (A) EphB2 null mice versus wild-type

littermates, (B) lesioned animals versus sham operated controls. Both EphB2 null mice and

lesioned wild-types exhibit greater levels of distance traveled compared to wild-type

controls. Error bars: ±SEM, *p<0.05, comparison of control versus experimental animals.

89

0

5

10

15

20

25

5 10 15 20 25 30 35 40 45 50 55 60

Mea

n D

ista

nce

Trav

elle

d (m

)

Time (min)

EphB2+/+ (n=24)

EphB2-/- (n=20)*

** * *

** *

**

**

0

5

10

15

20

25

5 10 15 20 25 30 35 40 45 50 55 60

Mea

n D

ista

nce

Trav

eled

(m)

Time (min)

Sham (n=16)

Lesioned (n=19)

*

**

* ** *

*

A.

B.

90

of the novel environment. However, EphB2 null mice display significantly elevated exploratory

activity following introduction into the novel environment compared to wild type controls.

Therefore, we investigated wither lesioned animals also displayed the same pattern of

exploratory activity as EphB2 null mice. As shown in Figures 3.12-3.13 both lesioned and sham

operated control animals display elevated exploratory activity that decreased overtime.

However, lesioned animals like the EphB2 null animals displayed elevated spontaneous

exploratory activity compared to sham operated controls. These results indicate that bilateral

electrolytic lesions of the dorsal hippocampus of wild-type animals display strikingly similar

behavioural responses seen in EphB2 null mice compared to sham operated controls.

3.5 Morphine related responses in animals with bilateral electrolytic lesions in the dorsal

hippocampus

We have shown that both EphB2 null mice and animals with bilateral electrolytic lesions

in the dorsal hippocampus exhibit similar behavioural responses. Thus, this suggests EphB2 null

possess deficits in hippocampal learning. EphB2 null mice also displayed accelerated morphine

tolerance and failed to display enhanced analgesic response in a novel environment following

development of morphine tolerance. Thus, we were interested in examining whether

impairments in hippocampal learning had an effect on morphine-specific learning. Multiple

studies have reported that morphine induces hyperactivity known as a running fit in multiple

strains of mice [206, 207]. Therefore, we examined the spontaneous motor activity of EphB2

null and wild-type mice upon first time exposure to morphine. We observed that both EphB2

null and wild-type mice both display morphine induced hyperactivity about 25min post

morphine injection. The key difference in this observable running fit is the duration. EphB2

wild-type mice display sustained morphine induced hyperactivity throughout the duration of the

Activity Monitor recording (Figure 3.14), while EphB2 null mice display morphine induced

91

Figure 3.14. Morphine induced hyperactivity in wild-type mice. Shown are measures of

spontaneous activity 24 hours prior to (left panel) and immediately following (right panel)

morphine injection. Arrow represents time of morphine injection. Dashed red line represents

averaged morphine-induced activity during first 15 minutes of observation. Dashed black

line represents average of motor activity during final 15 minutes of observation period 24

hours prior to morphine injection. Note morphine-induced motor hyperactivity is maintained

throughout the 60 minute observation period. N≥15, Error bars +SEM, *p≤0.05 compared to

baseline values (dashed black line).

92

020406080100

120

140

160

-5-1

0-1

5-2

0-2

5-3

0-3

5-4

0-4

5-5

0-5

5-6

05

1015

2025

3035

4045

5055

60

Mean Active Time (sec)

Tim

e (m

in)

**

**

**

**

**

**

93

hyperactivity for about 10min but then the elevated spontaneous motor activity decays overtime

as seen in the non-morphine state (Figure 3.15). As a control, we injected animals with saline

and this did not illicit the morphine induced hyperactivity behaviour, thus confirming the effect

we observed was due to morphine (Figure 3.16).

We also examined the morphine induced hyperactivity for our lesioned and sham

operated control animals. As shown in Figures 3.17-3.18, we observed that both our sham and

lesioned animals display morphine induced hyperactivity about 20 and 25min post morphine

injection, respectively. The difference once again is the duration. Sham animals like wild-type

animals display relatively sustained morphine induced hyperactivity throughout the duration of

the Activity Monitor recording (Figure 3.17). In contrast, lesioned animals like the EphB2 null

mice display morphine induced hyperactivity for about 15min and then the elevated spontaneous

motor activity starts to decline overtime as seen in the non-morphine state (Figure 3.18).

As indicated above, unlike wild-type animals EphB2 null mice do not display enhanced

morphine dependent analgesic response following transfer to a novel environment following

development of morphine tolerization on day 7. To determine whether these findings were

hippocampal dependent, and a result of impairment in learning and memory, we performed the

same study in our lesioned and sham operated control animals. Following twice daily injection

of morphine for 6 days, on day 7 animals were divided and injected in either the same

environment or a novel environment and tested for their antinociceptive response using tail

flick. Although, antinociceptive responses of EphB2 animals were tested using tail pinch and

tail flick (data not shown) assays, lesioned and sham operated control animals were only tested

using tail flick assay. Perhaps due to differences in the genetic background between EphB2 null

mice and lesioned animals, we were unable to obtain robust response measurements using the

tail pinch assay, thus we only measured analgesic responses using the tail flick assay. Consistent

with previous results, lesioned animals like EphB2 null mice, failed to display enhanced

94

Figure 3.15. Morphine induced hyperactivity in EphB2 null mice. Shown are measures

of spontaneous activity 24 hours prior to (left panel) and immediately following (right panel)

morphine injection. Arrow represents time of morphine injection. Dashed blue line

represents averaged morphine-induced activity during first 15 minutes of observation.

Dashed black line represents average of motor activity during final 15 minutes of

observation period 24 hours prior to morphine injection. Morphine-induced motor

hyperactivity is only maintained transiently during the observation period. N≥13, Error bars

+SEM,*p≤0.05 compared to baseline values (dashed black line).

95

020406080100

120

140

160

180

-5-1

0-1

5-2

0-2

5-3

0-3

5-4

0-4

5-5

0-5

5-6

05

1015

2025

3035

4045

5055

60

Mean Active Time (sec)

Tim

e (m

in)

**

**

**

**

*

96

Figure 3.16. Spontaneous motor activity of saline injected animals. Shown are measures

of spontaneous activity 24 hours prior to (left panel) and immediately following (right panel)

saline injection. Arrow represents time of saline injection. Dashed yellow line represents

averaged activity during first 15 minutes of observation. Dashed black line represents

average of motor activity during final 15 minutes of observation period 24 hours prior to

saline injection. Standard depression in exploratory activity is observed. N≥11, Error bars

+SEM, *p≤0.05 compared to baseline values (dashed black line).

97

020406080100

120

140

160

180

200

-5-1

0-1

5-2

0-2

5-3

0-3

5-4

0-4

5-5

0-5

5-6

05

1015

2025

3035

4045

5055

60

Mean Active Time (sec)

Tim

e (m

in)*

**

*

98

Figure 3.17. Morphine induced hyperactivity in sham operated control animals. Shown

are measures of spontaneous activity 24 hours prior to (left panel) and immediately

following (right panel) morphine injection. Arrow represents time of morphine injection.

Dashed red line represents averaged morphine-induced activity during first 15 minutes of

observation. Dashed black line represents average of motor activity during final 15 minutes

of observation period 24 hours prior to morphine injection. Note morphine-induced motor

hyperactivity is maintained throughout the 60 minute observation period. N≥7, Error bars

+SEM, *p≤0.05 compared to baseline values (dashed black line).

99

0306090120

150

180

210

-5-1

0-1

5-2

0-2

5-3

0-3

5-4

0-4

5-5

0-5

5-6

05

1015

2025

3035

4045

5055

60

Mean Active Time (sec)

Tim

e (m

in)

**

**

**

*

*

100

Figure 3.18 . Morphine induced hyperactivity in lesioned animals. Shown are measures

of spontaneous activity 24 hours prior to (left panel) and immediately following (right panel)

morphine injection. Arrow represents time of morphine injection. Dashed blue line

represents averaged morphine-induced activity during first 15 minutes of observation.

Dashed black line represents average of motor activity during final 15 minutes of

observation period 24 hours prior to morphine injection. Morphine-induced motor

hyperactivity is only maintained transiently during the observation period. N≥8, Error bars

+SEM,*p≤0.05 compared to baseline values (dashed black line).

101

0306090120

150

180

210

240

270

-5-1

0-1

5-2

0-2

5-3

0-3

5-4

0-4

5-5

0-5

5-6

05

1015

2025

3035

4045

5055

60

Mean Active Time (sec)

Tim

e (m

in)

*

102

analgesic response upon transfer to novel environment following development of morphine

tolerization on day 7 (Figure 3.19). In contrast, sham operated controls like EphB2 wild-type

animals displayed some enhanced analgesic response upon transfer to a novel environment

(Figure 3.19). Thus, animals with hippocampal lesions localized to the dorsal hippocampus

mimicked EphB2 null morphine related responses. This suggests that EphB2 null mice as a

result of hippocampal learning deficits, lack the ability to integrate these sensory-dependent

environmental cues associated with morphine exposure.

103

Figure 3.19. Antinociceptive responses of sham and lesioned animals in home versus

novel environments. Following 7 days of morphine treatment lesioned (n=3) and sham

operated control (n=5) animals were either retained in their home environment or moved to a

novel environment and tested for antinociceptive response using the tail flick assay. Similar

to previous reports, sham operated control animals exhibited a significant increase in

antinociceptive response when placed in a novel environment (red dotted line) compared to

those retained in their home environment (red solid line). However, lesioned animals

exhibited no significant difference in antinociceptive response in the novel environment

(blue dotted line) compared to those retained in their home environment (blue solid line).

Error bars: ±SEM, $p<0.05 sham novel environment versus sham same environment;

*p<0.05 sham versus lesioned animals in novel environment; #p<0.05 sham versus lesioned

animals in home environment.

0

2

4

6

8

10

12

14

30 60 90 120

Mea

n Ta

il Fl

ick

Lat

ency

(sec

)

Time

Sham SAME Sham NOVEL Lesioned SAME Lesioned NOVEL

$

*

*

*

## #

104

CHAPTER 4: DISCUSSION

105

4.1 Changes in morphine responsiveness seen in EphB2-null mice are mediated via

modification of cortical influences on sensory function.

EphB2 receptors are persistently expressed within the adult CNS at sites involved in

sensation and processing of sensory stimuli. Although the expression of EphB2 typically

diminishes at a given site following the establishment of primary synaptic connections, EphB2

expression does remerge at several locations following late embryogenesis [150]. Specifically,

EphB2 expression remerges at sites which undergo significant activity-dependent modification

including the hippocampus, amygdala, thalamus, and cingulate cortex [150]. Within the murine

spinal cord, EphB2 expression also undergoes a significant shift during the late embryonic/early

postnatal period, becoming prominently expressed within sensory ganglia, dorsal rexed laminae

and ascending sensory pathways [150]. Despite this, EphB2 null mice display no significant

differences in baseline motor or sensory function compared to control littermates, and EphB2

null mutants displayed similar analgesic responses to littermate controls upon initial exposure to

morphine. However over time, EphB2 null mutants exhibit a substantially accelerated pattern of

morphine tolerance compared to wild-type controls. This suggests that loss of EphB2 may alter

the rate of morphine metabolism, or alter the availability of morphine binding to MORs, or alter

the extent of MOR signaling, or modify some central response regulating this form of sensory

input.

The primary site of morphine metabolism is the liver. However, EphB2 is never

expressed in the liver during developmental or adulthood. Nevertheless some degree of

morphine metabolism is known to occur locally within the CNS. Thus to investigate and

confirm loss of EphB2 does not alter morphine metabolism, we examined levels of morphine

and its metabolite M3G in the blood and brain of EphB2 null and control littermates using LC-

MS/MS. In depth pharmacokinetic studies of morphine in mice are few, and few studies have

used LC-MS/MS for quantification. However, our values are within range of what has

106

previously been reported [208, 209]. The results of both naïve and morphine-tolerized mice

demonstrated that the rates of morphine metabolism are not significantly altered in the absence

of EphB2. The metabolite M3G is inactive and not believed to bind to MORs or illicit analgesic

response [56]. M3G does however represent the main metabolite of morphine, with about 60%

of morphine metabolized to this compound. In the brain, morphine and M3G levels were found

not to differ significantly between EphB2 null and control littermates at all time points

measured. Similarly in whole blood, morphine and M3G concentrations were found not to differ

significantly between EphB2 null and control animals at all time points; excluding at 30

minutes. At this time point, EphB2 null mutants appear to exhibit significantly higher

concentrations of morphine and M3G (differences within the brain at this time point were not

statistically significant). These differences at 30min may be attributed to higher variability

observed following the initial distribution of morphine injected i.p. As well, it appears unrelated

to the previous functional findings observed in EphB2 null mice. EphB2 null mice would if

anything be expected to exhibit lower levels of morphine in the brain and whole blood since

they display significantly lower tail pinch latencies compared to wild-type mice. As well, two-

way ANOVA analysis of the data indicated no statistical effect of genotype is present. Morphine

and M3G concentrations were also quantified following repeated exposure of morphine for 7

days. As expected, EphB2 null mice displayed no significant difference in morphine or M3G

concentration in the blood and brain compared to wild-type controls. This indicates morphine

tolerance data observed for day 7 in EphB2 null mice is independent of differences in morphine

metabolism, but perhaps due to modification of central response. All together, loss of EphB2

receptors does not appear to significantly alter morphine metabolism in mice.

With respect to loss of EphB2 altering the extent of mu opiate receptor binding or

capacity, previous studies performed in the laboratory have determined that affinity and total

binding capacity of mu opioid receptors is unaltered in EphB2 null mice within the tissues

107

examined (spinal cord, and superior colliculus) (Ashlin Kanawaty, Thesis 2011).

Immunohistological studies have previously examined MOR expression and distribution in

rodents, within various regions of the brain such as the cortex, caudate-putamen, hippocampus,

superior/inferior colliculus, and dorsal horn of the spinal cord [210, 211]. We also examined and

demonstrated that the distribution of MORs within the nervous system (lumbar dorsal root

ganglia, spinal cord, superior colliculus, and striatum) is unaltered in EphB2 null mice compared

to littermate controls. Within the spinal cord, EphB2 null mutants and control littermates exhibit

similar laminar distributions of MORs within the dorsal laminae I-III. These positions were

verified by previous work in the lab using known markers such as IB4 or CGRP (Ashlin

Kanawaty, Thesis 2011). We also examined the striatum; since EphB2 receptors are not

expressed in the striatum this region of the brain served as a good control. Within the striatum,

the distribution of mu opioid receptors within the striatal and matrix compartments examined,

demonstrated no significant morphologic differences between EphB2 null and control

littermates. We hypothesized that our morphine tolerance observations may be hippocampal

dependent (see below). Thus we examined MOR distribution within the hippocampus, where no

difference in distribution was observed between EphB2 null and control littermates. Overall, no

morphological difference was observed in the distribution of MORs between EphB2 null and

wild-types at all CNS regions examined. Thus, loss of EphB2 receptors did not significantly

alter MOR distribution and expression in the CNS.

Together, these aforementioned findings indicate loss of EphB2 does not alter rate of

morphine metabolism or affect MOR distribution in the CNS. Thus the action of EphB2 to alter

morphine-dependent signaling may be mediated through higher-order influences that modify

morphine dependent contextual learning (see below).

108

4.2 EphB2 null mice exhibit deficiencies in contextual learning similar to that seen in wild-

type animals containing bilateral electrolytic lesions of the dorsal hippocampus.

The hippocampus plays a central role in learning and memory. Numerous lesion studies

in a variety of mammalian systems have demonstrated the important role of the hippocampus in

formation of new episodic memories, short-term memory consolidation, and cognitive features

involved in learning to novel environmental stimuli [170, 192, 200, 212-215]. However, the

hippocampus is not a uniform structure [187, 189, 194]. Localized lesions have shown that

different sub-regions of the hippocampus mediate different forms of learning and memory.

Studies comparing dorsal hippocampal lesions to ventral hippocampal lesions have shown that

the dorsal hippocampus is important for processing of spatial memory and learning of

conceptual cues; whereas the ventral hippocampus is involved with stress, fear and emotional

aspects of memory [187, 190, 191, 194, 216, 217]. Physiologically, the phenomena of long-term

potentiation (LTP) and long-term depression (LTD), along with NMDA signaling are thought to

currently represent our best cellular and molecular models of the processes involved in

acquisition and consolidation of learned behavior [183, 218, 219]. Incoming sensory

information arising from primary centers in the CNS are subsequently transferred to higher

cortical stations [220]. Such incoming information is subject to consolidation and processing

within the hippocampus, which allows contextual modification of sensory experiences [221,

222]. Therefore, alterations in the consolidation or processing of spatial and/or sensory-related

memories within the hippocampus can result in significant modification of learned behaviors.

Previous work by Siegel and colleagues [203-205, 223] have demonstrated that context

dependent cues and Pavlovian conditioning exert significant functional influences over the

strength of morphine-dependent analgesia and tolerance. Re-examination of Siegel's context

dependent paradigm of morphine tolerance in EphB2 null mutants and littermate controls

demonstrated findings similar to those previously published. Morphine tolerized wild-type

109

animals demonstrated an apparent enhancement in morphine analgesia following the transfer to

a novel environment. In contrast, morphine tolerized EphB2 null mice demonstrated a complete

inhibition of response in this context dependent paradigm of learning. This suggests that EphB2

null mice exhibit impairment in morphine-dependent contextual learning. This is consistent with

previously published results by our lab and others, that found EphB2 null mice exhibit both an

attenuation in the magnitude and stability of hippocampal LTP, compared to littermate controls

[150, 157]. These effects on LTP have been demonstrated to be due to EphB2 influences on the

relative synaptic localization and lifetime of NMDA receptors on the postsynaptic membrane of

hippocampal CA1 neurons [150, 157]. Thus, we investigated whether the defects in contextual

learning observed in EphB2 null mice were in fact dependent upon an inhibition of dorsal

hippocampal function, by performing bilateral electrolytic lesions to the rostral component of

the dorsal hippocampus in wild-type mice. We examined the behavioural response of EphB2

null mice and controls in several paradigms of context dependent learning. We compared these

behavioural responses to those of lesioned animals, to determine whether such hippocampal

inhibition could explain the modified morphine responses seen in EphB2 null mice.

One behavioural assessment employed was the passive avoidance, which is a well-

characterized paradigm of context-dependent learning that tests hippocampal learning [200]. It

examines an animal's ability to distinguish between two distinct contextual frameworks [224-

226]. Hippocampal dependence of this task (and others such as the Morris water maze) has been

confirmed using animals with bilateral lesions to the dorsal hippocampus, which demonstrated a

significant attenuated capacity to discriminate between the two contextual cues [224-226].

When examined on this task, EphB2 null mice performed poorly relative to controls, similar to

the performance seen in animals with bilateral lesions to the dorsal hippocampus. In both cases,

EphB2 null and lesioned animals demonstrated significantly lower transfer latency times into

the aversive environment compared to that observed in wild type and sham operated control

110

littermates. These findings suggest that loss of EphB2 impairs contextual/spatial memory in a

hippocampal-dependent manner.

The other behavioural assessment employed was the open field test to measure

exploratory activity upon placement into a novel environment. Studies have suggested the open

field test represents measures of contextual learning, novelty seeking and anxiety [201, 227].

Thus, we have utilized such activity measurements to assess the relative exploratory activity of

EphB2 null mice compared littermate controls. Although, no baseline differences in motor and

sensory performance were observed between EphB2 null and control littermates; EphB2 null

mice exhibit consistently higher levels of exploratory activity compared to wild type littermates.

The inability of EphB2 null mice to habituate to the novel environment in comparison to

controls suggests they may exhibit problems in contextual learning, and/or increased levels of

anxiety. Both such behavioral responses could be linked to problems in hippocampal dependent

learning, consistent with known deficiencies of EphB2 null mice in CA1 dependent LTP. Thus,

upon examination of the bilateral lesioned animals on the open field test, lesioned animals

similar to EphB2 null mice also exhibit higher levels of exploratory activity compared to sham

operated control animals. Taken together, the above findings suggest that EphB2 null mice

exhibit deficiencies in contextual learning which are similar to that seen in animals following

disruption of dorsal hippocampal function.

4.3 Impaired opiate-dependent responses seen in EphB2 null mice arise from

hippocampal-dependent deficiencies in contextual learning.

Several studies have recently examined the role of the EphB-ephrinB signalling in

modulating the perception of neuropathic pain [162, 164, 165, 167, 168], and the role of EphB

family receptors in influencing opiate dependence and withdrawal [167, 168]. In general, these

studies have postulated a model where interruption of the Eph-ephrin signaling by receptors

111

such as EphB1 induces responses similar to that seen following NMDA inhibition (i.e.

impediment in the development of morphine tolerance) [93, 168]. However, loss of EphB2

induces the opposite behavior; actually accelerating the development opiate tolerance.

Therefore, EphB2 plays a role in the regulation of morphine dependent response, distinct from

direct enhancement of NMDA function and signalling. In contrast to published results, EphB2

null mice fail to exhibit enhancement in morphine analgesia following the transfer to a novel

environment compared to littermate controls. As well, consistent with our previous description

of impairments in hippocampal LTP in EphB2 null mice [150, 157], our analysis of contextual

learning in EphB2 null mice also demonstrated impairments in hippocampal dependent learning.

Therefore, we investigated whether EphB2 null mice exhibit impairment in morphine-dependent

contextual learning.

In a morphine naive state, we demonstrated that animals with bilateral dorsal

hippocampal lesions exhibit similar learning behavioural impairments to the EphB2 null

animals. Thus, we were interested in investigating whether opiate-dependent learning was also

hippocampal dependent. Morphine has been shown to induce hyperactivity/ hyperlocomotion

following injection. This morphine induced motor hyperexcitability, termed “running fit,” is

species specific [206, 207]. Not all strains of mice exhibit this behavioral characteristic [206,

207]. EphB2 null mice display normal motor and sensory responses, and their initial response

upon morphine exposure is identical to that seen in wild-type mice. Upon examination of

morphine induced hyperactivity, both EphB2 null and wild-type mice exhibit morphine induced

hyperactivity following initial exposure to morphine. However, despite the similar baseline

responses between EphB2 null and wild-type mice, EphB2 null mice display distinct and

abnormal response to opiates which may be attributed to their hippocampal learning

impairments. For example, although EphB2 null animals displayed morphine induced

hyperactivity, this behaviour was observed for a short period of time before it begins to decay.

112

This in contrast to EphB2 wild-type controls which exhibited morphine induced hyperactivity

for a longer sustained duration. Similarly, in morphine induced state bilateral lesioned animals

display strikingly similar characteristics to EphB2 null mutants, while the sham operated

controls behaved as the wild-types. Interestingly, the decay of morphine induced hyperactivity

observed in EphB2 null mice and lesioned animals is reminiscent of the induction of LTP in

EphB2 null mice. EphB2 null mice induce LTP that is not stable and which decays rapidly

compared to control littermates [150]. As well, the pattern displayed by the EphB2 null animals

and wild-types is consistent with the analgesic pattern of morphine tolerance displayed by the

animals on day 1. On day 1 analysis of morphine tolerance initially both groups of animals

displayed similar analgesic responses, however, EphB2 null mutants quickly display decreased

analgesic response as observed with the morphine induced hyperactivity. The similar behavioral

findings between EphB2 null and lesioned animals suggest EphB2 null mice exhibit impairment

in morphine-dependent contextual learning due to inhibition of dorsal hippocampal processing.

To further investigate EphB2 null mice impairment in morphine-dependent contextual

learning, we performed Siegel's context-dependent paradigm of morphine tolerance in the

animals with bilateral lesions in the dorsal hippocampus and the sham operated controls.

Environmental cues have been shown to be important in predicting the analgesic actions of

morphine following the development of tolerance. That is because opiate tolerance has been

shown to involve a learned response of the association between the systemic actions of an opiate

and the environmental cues. Thus, contextual learning can modulate an animal’s response to a

drug depending on the environment. In contrast, to previously described results [203-205]

lesioned animals that developed morphine tolerance like the EphB2 null mice, failed to display

enhancement in morphine analgesia following the transfer to a novel environment compared to

sham operated controls. All together, the findings suggest that lesioned animals exhibit similar

behavioral responses in morphine naive state, and altered pattern of responses in morphine

113

induced state, similar to those observed of EphB2 null mice. Therefore, this suggests that EphB2

null mice exhibit impairment in hippocampal-dependent learning that fail to undergo contextual

learning of the environmental cues associated with morphine exposure. Hence, both EphB2 null

mice and lesioned animals display impairment in morphine-dependent contextual learning. This

suggests that loss of CA1 neurons in the dorsal hippocampus is important for learning of

contextual cues in opiate-dependent learning paradigms. This is consistent, with previous work

in the lab that demonstrated that loss of EphB2 results in reduced levels of active postsynaptic

NMDA receptor expression in CA1 neurons of the hippocampus, resulting in reduced LTP at

this site within the hippocampus [150, 157]. Thus, EphB2-dependent changes in the

hippocampus may be mediating the observed opiate dependent learning.

4.4. Concluding Remarks & Future Studies

The mechanisms underlying physiologic responses to opiates are complex and varied.

They not only depend on signaling interactions triggered by binding of opiate ligands to their

respective opiate receptors, but also the integration of information from higher-order cortical

centers. Consistent with previous studies, in this study we have shown that contextual cues

associated with exposure to opiates can modulate and exert an influence over physiologic

responses such as sensory analgesia and motor excitability. In the present study, I have tried to

understand the role by which EphB2, a receptor tyrosine kinase, plays in regulating opiate-

dependent function. Despite overlapping expression of EphB2 and MORs at several CNS loci

directly involved in mediating the sensory effects of opiates, EphB2 appears to exert a majority

of its effects by modifying the perception of contextual cues associated with morphine exposure.

The complexity of these contextual associations is highlighted by the fact that loss of EphB2

does not simply inhibit NMDA receptor function. That is because, based on previously

published studies, one might expect the effects of EphB2 ablation to mimic those seen following

114

the infusion of the non-competitive NMDA receptor antagonist, MK-801, which in contrast to

EphB2 actually inhibit the development of morphine tolerance rather than accelerate it [93].

Instead inhibition of EphB2 appears to alter morphine-dependent response at sites such as the

hippocampus, through modification of contextual learning.

The data presented in the current thesis demonstrates an important role for EphB2

signaling in the hippocampus in regulating opiate-dependent learning. Analyses of the EphB2

receptor mutants in which the kinase domain is inactivated further demonstrate that this effect is

dependent upon reverse signaling through EphB2 ligands (unpublished data). Our findings

suggest that that loss of EphB2 receptors does not alter MOR avidity or distribution in the CNS,

nor does it significantly alter morphine metabolism. However, this study is one of many more

studies to come in order to establish a thorough understanding of the physiologic interactions

regulating time-dependent morphine response. Further studies will be required to examine

whether loss of EphB2 alters other aspects of morphine-dependent signaling such as β-arrestin

translocation within primary neurons, or phosphorylation of MORs. With respect to the

hippocampal role of EphB2 in regulating morphine-dependent response, a strong test of this

model would be local inhibition of EphB2 within the hippocampus through means such as

protein-based EphB2 blocking receptor bodies, or via local lentiviral inhibition of EphB2

mRNA using siRNA approaches.

115

References

1. Le Merrer, J., et al., Reward processing by the opioid system in the brain. Physiological reviews, 2009. 89(4): p. 1379-412.

2. McQuay, H., Opioids in pain management. Lancet, 1999. 353(9171): p. 2229-32. 3. Kalant, H., D.M. Grant, and J. Mitchell, Principles of medical pharmacology. 7th

ed2007, Toronto: Saunders Elsevier. xvii, 1022 p. 4. Pert, C.B. and S.H. Snyder, Opiate receptor: demonstration in nervous tissue. Science,

1973. 179(4077): p. 1011-4. 5. Simon, E.J., J.M. Hiller, and I. Edelman, Stereospecific binding of the potent narcotic

analgesic (3H) Etorphine to rat-brain homogenate. Proceedings of the National Academy of Sciences of the United States of America, 1973. 70(7): p. 1947-9.

6. Terenius, L., Characteristics of the "receptor" for narcotic analgesics in synaptic plasma membrane fraction from rat brain. Acta pharmacologica et toxicologica, 1973. 33(5): p. 377-84.

7. Hughes, J., et al., Identification of two related pentapeptides from the brain with potent opiate agonist activity. Nature, 1975. 258(5536): p. 577-80.

8. Goldstein, A., Opioid peptides endorphins in pituitary and brain. Science, 1976. 193(4258): p. 1081-6.

9. Goldstein, A., et al., Dynorphin-(1-13), an extraordinarily potent opioid peptide. Proceedings of the National Academy of Sciences of the United States of America, 1979. 76(12): p. 6666-70.

10. Akil, H., et al., Endogenous opioids: biology and function. Annual Review of Neuroscience, 1984. 7: p. 223-55.

11. Martin, W.R., et al., The effects of morphine- and nalorphine- like drugs in the nondependent and morphine-dependent chronic spinal dog. The Journal of pharmacology and experimental therapeutics, 1976. 197(3): p. 517-32.

12. Knapp, R.J., et al., Molecular biology and pharmacology of cloned opioid receptors. FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 1995. 9(7): p. 516-25.

13. Quigley, D.I., et al., Orphanin FQ is the major OFQ1-17-containing peptide produced in the rodent and monkey hypothalamus. Peptides, 1998. 19(1): p. 133-9.

14. Bunzow, J.R., et al., Molecular cloning and tissue distribution of a putative member of the rat opioid receptor gene family that is not a mu, delta or kappa opioid receptor type. FEBS letters, 1994. 347(2-3): p. 284-8.

15. Meunier, J.C., et al., Isolation and structure of the endogenous agonist of opioid receptor-like ORL1 receptor. Nature, 1995. 377(6549): p. 532-5.

16. Reinscheid, R.K., et al., Orphanin FQ: a neuropeptide that activates an opioidlike G protein-coupled receptor. Science, 1995. 270(5237): p. 792-4.

17. Thomsen, C. and R. Hohlweg, (8-Naphthalen-1-ylmethyl-4-oxo-1-phenyl-1,3,8-triaza-spiro[4. 5]dec-3-yl)-acetic acid methyl ester (NNC 63-0532) is a novel potent nociceptin receptor agonist. British journal of pharmacology, 2000. 131(5): p. 903-8.

18. Mansour, A., et al., Opioid-receptor mRNA expression in the rat CNS: anatomical and functional implications. Trends in neurosciences, 1995. 18(1): p. 22-9.

19. Higgins, G.A., et al., Influence of the selective ORL1 receptor agonist, Ro64-6198, on rodent neurological function. Neuropharmacology, 2001. 41(1): p. 97-107.

20. Kieffer, B.L. and C. Gaveriaux-Ruff, Exploring the opioid system by gene knockout. Progress in neurobiology, 2002. 66(5): p. 285-306.

116

21. Alfaras-Melainis, K., et al., Modulation of opioid receptor function by protein-protein interactions. Frontiers in bioscience : a journal and virtual library, 2009. 14: p. 3594-607.

22. Lagerstrom, M.C. and H.B. Schioth, Structural diversity of G protein-coupled receptors and significance for drug discovery. Nature reviews. Drug discovery, 2008. 7(4): p. 339-57.

23. van Rijn, R.M., J.L. Whistler, and M. Waldhoer, Opioid-receptor-heteromer-specific trafficking and pharmacology. Current opinion in pharmacology, 2010. 10(1): p. 73-9.

24. Lord, J.A., et al., Endogenous opioid peptides: multiple agonists and receptors. Nature, 1977. 267(5611): p. 495-9.

25. Kane, B.E., B. Svensson, and D.M. Ferguson, Molecular recognition of opioid receptor ligands. The AAPS journal, 2006. 8(1): p. E126-37.

26. Mansour, A., et al., The cloned mu, delta and kappa receptors and their endogenous ligands: evidence for two opioid peptide recognition cores. Brain research, 1995. 700(1-2): p. 89-98.

27. Chen, Y., et al., Molecular cloning and functional expression of a mu-opioid receptor from rat brain. Molecular Pharmacology, 1993. 44(1): p. 8-12.

28. Wang, J.B., et al., mu opiate receptor: cDNA cloning and expression. Proceedings of the National Academy of Sciences of the United States of America, 1993. 90(21): p. 10230-4.

29. Thompson, R.C., et al., Cloning and pharmacological characterization of a rat mu opioid receptor. Neuron, 1993. 11(5): p. 903-13.

30. Doyle, G.A., et al., Identification of five mouse mu-opioid receptor (MOR) gene (Oprm1) splice variants containing a newly identified alternatively spliced exon. Gene, 2007. 395(1-2): p. 98-107.

31. Pan, Y.X., Diversity and complexity of the mu opioid receptor gene: alternative pre-mRNA splicing and promoters. DNA and cell biology, 2005. 24(11): p. 736-50.

32. Majumdar, S., et al., Truncated G protein-coupled mu opioid receptor MOR-1 splice variants are targets for highly potent opioid analgesics lacking side effects. Proceedings of the National Academy of Sciences of the United States of America, 2011. 108(49): p. 19778-83.

33. Zimprich, A., T. Simon, and V. Hollt, Cloning and expression of an isoform of the rat mu opioid receptor (rMOR1B) which differs in agonist induced desensitization from rMOR1. FEBS letters, 1995. 359(2-3): p. 142-6.

34. Schulz, S., et al., Immunolocalization of two mu-opioid receptor isoforms (MOR1 and MOR1B) in the rat central nervous system. Neuroscience, 1998. 82(2): p. 613-22.

35. Pan, Y.X., et al., Involvement of exon 11-associated variants of the mu opioid receptor MOR-1 in heroin, but not morphine, actions. Proceedings of the National Academy of Sciences of the United States of America, 2009. 106(12): p. 4917-22.

36. Pan, Y.X., et al., Generation of the mu opioid receptor (MOR-1) protein by three new splice variants of the Oprm gene. Proceedings of the National Academy of Sciences of the United States of America, 2001. 98(24): p. 14084-9.

37. von Zastrow, M., Mechanisms regulating membrane trafficking of G protein-coupled receptors in the endocytic pathway. Life sciences, 2003. 74(2-3): p. 217-24.

38. Manglik, A., et al., Crystal structure of the micro-opioid receptor bound to a morphinan antagonist. Nature, 2012. 485(7398): p. 321-6.

39. Borsodi, A. and G. Toth, Characterization of opioid receptor types and subtypes with new ligands. Annals of the New York Academy of Sciences, 1995. 757: p. 339-52.

117

40. Hollt, V., Opioid peptide processing and receptor selectivity. Annual review of pharmacology and toxicology, 1986. 26: p. 59-77.

41. Garzon, J., et al., Endogenous opioid peptides: comparative evaluation of their receptor affinities in the mouse brain. Life sciences, 1983. 33 Suppl 1: p. 291-4.

42. Lahti, R.A., et al., [3H]U-69593 a highly selective ligand for the opioid kappa receptor. European journal of pharmacology, 1985. 109(2): p. 281-4.

43. Handa, B.K., et al., Analogues of beta-LPH61-64 possessing selective agonist activity at mu-opiate receptors. European journal of pharmacology, 1981. 70(4): p. 531-40.

44. Mosberg, H.I., J.R. Omnaas, and A. Goldstein, Structural requirements for delta opioid receptor binding. Molecular Pharmacology, 1987. 31(6): p. 599-602.

45. Matthes, H.W., et al., Activity of the delta-opioid receptor is partially reduced, whereas activity of the kappa-receptor is maintained in mice lacking the mu-receptor. The Journal of neuroscience : the official journal of the Society for Neuroscience, 1998. 18(18): p. 7285-95.

46. Al-Hasani, R. and M.R. Bruchas, Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology, 2011. 115(6): p. 1363-81.

47. De Gregori, S., et al., Morphine metabolism, transport and brain disposition. Metabolic brain disease, 2012. 27(1): p. 1-5.

48. Ishikawa, K., et al., A sensitive procedure for determination of morphine in mouse whole blood by high performance liquid chromatography with electrochemical detection. Japanese journal of pharmacology, 1982. 32(5): p. 969-71.

49. King, C.D., et al., The glucuronidation of exogenous and endogenous compounds by stably expressed rat and human UDP-glucuronosyltransferase 1.1. Archives of biochemistry and biophysics, 1996. 332(1): p. 92-100.

50. Coffman, B.L., et al., The glucuronidation of opioids, other xenobiotics, and androgens by human UGT2B7Y(268) and UGT2B7H(268). Drug metabolism and disposition: the biological fate of chemicals, 1998. 26(1): p. 73-7.

51. Frolich, N., et al., Distinct pharmacological properties of morphine metabolites at G(i)-protein and beta-arrestin signaling pathways activated by the human mu-opioid receptor. Biochemical pharmacology, 2011. 81(10): p. 1248-54.

52. Xie, R., et al., The role of P-glycoprotein in blood-brain barrier transport of morphine: transcortical microdialysis studies in mdr1a (-/-) and mdr1a (+/+) mice. British journal of pharmacology, 1999. 128(3): p. 563-8.

53. Zelcer, N., et al., Mice lacking multidrug resistance protein 3 show altered morphine pharmacokinetics and morphine-6-glucuronide antinociception. Proceedings of the National Academy of Sciences of the United States of America, 2005. 102(20): p. 7274-9.

54. Handal, M., et al., Morphine-3-glucuronide inhibits morphine induced, but enhances morphine-6-glucuronide induced locomotor activity in mice. Pharmacology, biochemistry, and behavior, 2007. 86(3): p. 576-86.

55. Handal, M., et al., Pharmacokinetic differences of morphine and morphine-glucuronides are reflected in locomotor activity. Pharmacology, biochemistry, and behavior, 2002. 73(4): p. 883-92.

56. Frances, B., et al., Further evidence that morphine-6 beta-glucuronide is a more potent opioid agonist than morphine. The Journal of pharmacology and experimental therapeutics, 1992. 262(1): p. 25-31.

57. Lipkowski, A.W., et al., Morphine-3-glucuronide: silent regulator of morphine actions. Life sciences, 1994. 55(2): p. 149-54.

118

58. Hewett, K., A.H. Dickenson, and H.J. McQuay, Lack of effect of morphine-3-glucuronide on the spinal antinociceptive actions of morphine in the rat: an electrophysiological study. Pain, 1993. 53(1): p. 59-63.

59. Suzuki, Y., et al., Susceptibility to Chronic Infection with Toxoplasma-Gondii Does Not Correlate with Susceptibility to Acute Infection in Mice. Infection and Immunity, 1993. 61(6): p. 2284-2288.

60. Osborne, R., et al., The Analgesic Activity of Morphine-6-Glucuronide. British journal of clinical pharmacology, 1992. 34(2): p. 130-138.

61. Gong, Q.L., et al., Antinociceptive and Ventilatory Effects of the Morphine Metabolites - Morphine-6-Glucuronide and Morphine-3-Glucuronide. European Journal of Pharmacology, 1991. 193(1): p. 47-56.

62. Paul, D., et al., Pharmacological Characterization of Morphine-6-Beta-Glucuronide, a Very Potent Morphine Metabolite. Journal of Pharmacology and Experimental Therapeutics, 1989. 251(2): p. 477-483.

63. Bourasset, F., et al., Evidence for an active transport of morphine-6-beta-d-glucuronide but not P-glycoprotein-mediated at the blood-brain barrier. Journal of neurochemistry, 2003. 86(6): p. 1564-7.

64. Barchfeld, C.C. and F. Medzihradsky, Receptor-mediated stimulation of brain GTPase by opiates in normal and dependent rats. Biochemical and Biophysical Research Communications, 1984. 121(2): p. 641-8.

65. Childers, S.R. and S.H. Snyder, Guanine nucleotides differentiate agonist and antagonist interactions with opiate receptors. Life sciences, 1978. 23(7): p. 759-61.

66. Connor, M. and M.D. Christie, Opioid receptor signalling mechanisms. Clinical and experimental pharmacology & physiology, 1999. 26(7): p. 493-9.

67. Sharma, S.K., W.A. Klee, and M. Nirenberg, Opiate-dependent modulation of adenylate cyclase. Proceedings of the National Academy of Sciences of the United States of America, 1977. 74(8): p. 3365-9.

68. Fields, H., State-dependent opioid control of pain. Nature reviews. Neuroscience, 2004. 5(7): p. 565-75.

69. Law, P.Y., Y.H. Wong, and H.H. Loh, Molecular mechanisms and regulation of opioid receptor signaling. Annual review of pharmacology and toxicology, 2000. 40: p. 389-430.

70. Herz, A., Endogenous opioid systems and alcohol addiction. Psychopharmacology, 1997. 129(2): p. 99-111.

71. Jalabert, M., et al., Neuronal circuits underlying acute morphine action on dopamine neurons. Proceedings of the National Academy of Sciences of the United States of America, 2011. 108(39): p. 16446-50.

72. Johnson, S.W. and R.A. North, Opioids excite dopamine neurons by hyperpolarization of local interneurons. The Journal of neuroscience : the official journal of the Society for Neuroscience, 1992. 12(2): p. 483-8.

73. Connor, M., P.B. Osborne, and M.J. Christie, Mu-opioid receptor desensitization: is morphine different? British journal of pharmacology, 2004. 143(6): p. 685-96.

74. Yu, Y., et al., Mu opioid receptor phosphorylation, desensitization, and ligand efficacy. The Journal of biological chemistry, 1997. 272(46): p. 28869-74.

75. Keith, D.E., et al., mu-Opioid receptor internalization: opiate drugs have differential effects on a conserved endocytic mechanism in vitro and in the mammalian brain. Molecular Pharmacology, 1998. 53(3): p. 377-84.

119

76. Borgland, S.L., et al., Opioid agonists have different efficacy profiles for G protein activation, rapid desensitization, and endocytosis of mu-opioid receptors. The Journal of biological chemistry, 2003. 278(21): p. 18776-84.

77. Arden, J.R., et al., Phosphorylation and agonist-specific intracellular trafficking of an epitope-tagged mu-opioid receptor expressed in HEK 293 cells. Journal of neurochemistry, 1995. 65(4): p. 1636-45.

78. Finn, A.K. and J.L. Whistler, Endocytosis of the mu opioid receptor reduces tolerance and a cellular hallmark of opiate withdrawal. Neuron, 2001. 32(5): p. 829-39.

79. Keith, D.E., et al., Morphine activates opioid receptors without causing their rapid internalization. The Journal of biological chemistry, 1996. 271(32): p. 19021-4.

80. Celver, J., et al., Distinct domains of the mu-opioid receptor control uncoupling and internalization. Molecular Pharmacology, 2004. 65(3): p. 528-37.

81. Zhang, J., et al., Role for G protein-coupled receptor kinase in agonist-specific regulation of mu-opioid receptor responsiveness. Proceedings of the National Academy of Sciences of the United States of America, 1998. 95(12): p. 7157-62.

82. Lefkowitz, R.J., et al., Mechanisms of beta-adrenergic receptor desensitization and resensitization. Advances in pharmacology, 1998. 42: p. 416-20.

83. Chavkin, C., J.P. McLaughlin, and J.P. Celver, Regulation of opioid receptor function by chronic agonist exposure: constitutive activity and desensitization. Molecular Pharmacology, 2001. 60(1): p. 20-5.

84. Schulz, S., et al., Morphine induces terminal micro-opioid receptor desensitization by sustained phosphorylation of serine-375. The EMBO journal, 2004. 23(16): p. 3282-9.

85. Whistler, J.L. and M. von Zastrow, Morphine-activated opioid receptors elude desensitization by beta-arrestin. Proceedings of the National Academy of Sciences of the United States of America, 1998. 95(17): p. 9914-9.

86. Bohn, L.M., et al., Enhanced morphine analgesia in mice lacking beta-arrestin 2. Science, 1999. 286(5449): p. 2495-8.

87. Bohn, L.M., et al., Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature, 2000. 408(6813): p. 720-3.

88. Raehal, K.M. and L.M. Bohn, The role of beta-arrestin2 in the severity of antinociceptive tolerance and physical dependence induced by different opioid pain therapeutics. Neuropharmacology, 2011. 60(1): p. 58-65.

89. Salmanzadeh, F., et al., Dependence on morphine impairs the induction of long-term potentiation in the CA1 region of rat hippocampal slices. Brain research, 2003. 965(1-2): p. 108-13.

90. Bao, G., et al., Morphine and heroin differentially modulate in vivo hippocampal LTP in opiate-dependent rat. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, 2007. 32(8): p. 1738-49.

91. Zhu, H., et al., Region-specific changes in NMDA receptor mRNA induced by chronic morphine treatment are prevented by the co-administration of the competitive NMDA receptor antagonist LY274614. Brain research. Molecular brain research, 2003. 114(2): p. 154-62.

92. Mendez, I.A. and K.A. Trujillo, NMDA receptor antagonists inhibit opiate antinociceptive tolerance and locomotor sensitization in rats. Psychopharmacology, 2008. 196(3): p. 497-509.

93. Trujillo, K.A. and H. Akil, Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science, 1991. 251(4989): p. 85-7.

94. Noda, Y. and T. Nabeshima, Opiate physical dependence and N-methyl-D-aspartate receptors. European journal of pharmacology, 2004. 500(1-3): p. 121-8.

120

95. Ueda, H., M. Inoue, and T. Matsumoto, Protein kinase C-mediated inhibition of mu-opioid receptor internalization and its involvement in the development of acute tolerance to peripheral mu-agonist analgesia. The Journal of neuroscience : the official journal of the Society for Neuroscience, 2001. 21(9): p. 2967-73.

96. Bailey, C.P., E. Kelly, and G. Henderson, Protein kinase C activation enhances morphine-induced rapid desensitization of mu-opioid receptors in mature rat locus ceruleus neurons. Molecular Pharmacology, 2004. 66(6): p. 1592-8.

97. Chen, L. and L.Y. Huang, Protein kinase C reduces Mg2+ block of NMDA-receptor channels as a mechanism of modulation. Nature, 1992. 356(6369): p. 521-3.

98. Chen, L. and L.Y. Huang, Sustained potentiation of NMDA receptor-mediated glutamate responses through activation of protein kinase C by a mu opioid. Neuron, 1991. 7(2): p. 319-26.

99. Schuller, A.G.P., et al., Retention of heroin and morphine-6 beta-glucuronide analgesia in a new line of mice lacking exon 1 of MOR-1. Nature Neuroscience, 1999. 2(2): p. 151-156.

100. Sora, I., et al., Opiate receptor knockout mice define mu receptor roles in endogenous nociceptive responses and morphine-induced analgesia. Proceedings of the National Academy of Sciences of the United States of America, 1997. 94(4): p. 1544-1549.

101. Matthes, H.W.D., et al., Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene. Nature, 1996. 383(6603): p. 819-823.

102. Loh, H.H., et al., mu opioid receptor knockout in mice: effects on ligand-induced analgesia and morphine lethality. Molecular Brain Research, 1998. 54(2): p. 321-326.

103. Hirai, H., et al., A novel putative tyrosine kinase receptor encoded by the eph gene. Science, 1987. 238(4834): p. 1717-20.

104. Gale, N.W., et al., Eph receptors and ligands comprise two major specificity subclasses and are reciprocally compartmentalized during embryogenesis. Neuron, 1996. 17(1): p. 9-19.

105. Unified nomenclature for Eph family receptors and their ligands, the ephrins. Eph Nomenclature Committee. Cell, 1997. 90(3): p. 403-4.

106. Pasquale, E.B., Eph receptor signalling casts a wide net on cell behaviour. Nature reviews. Molecular cell biology, 2005. 6(6): p. 462-75.

107. Murai, K.K. and E.B. Pasquale, 'Eph'ective signaling: forward, reverse and crosstalk. Journal of cell science, 2003. 116(Pt 14): p. 2823-32.

108. Egea, J. and R. Klein, Bidirectional Eph-ephrin signaling during axon guidance. Trends in cell biology, 2007. 17(5): p. 230-8.

109. Davis, S., et al., Ligands for EPH-related receptor tyrosine kinases that require membrane attachment or clustering for activity. Science, 1994. 266(5186): p. 816-9.

110. Himanen, J.P., et al., Repelling class discrimination: ephrin-A5 binds to and activates EphB2 receptor signaling. Nature neuroscience, 2004. 7(5): p. 501-9.

111. Flanagan, J.G. and P. Vanderhaeghen, The ephrins and Eph receptors in neural development. Annual Review of Neuroscience, 1998. 21: p. 309-45.

112. Himanen, J.P., M. Henkemeyer, and D.B. Nikolov, Crystal structure of the ligand-binding domain of the receptor tyrosine kinase EphB2. Nature, 1998. 396(6710): p. 486-91.

113. Labrador, J.P., R. Brambilla, and R. Klein, The N-terminal globular domain of Eph receptors is sufficient for ligand binding and receptor signaling. The EMBO journal, 1997. 16(13): p. 3889-97.

121

114. Wybenga-Groot, L.E., et al., Structural basis for autoinhibition of the Ephb2 receptor tyrosine kinase by the unphosphorylated juxtamembrane region. Cell, 2001. 106(6): p. 745-57.

115. Himanen, J.P., N. Saha, and D.B. Nikolov, Cell-cell signaling via Eph receptors and ephrins. Current opinion in cell biology, 2007. 19(5): p. 534-42.

116. Zisch, A.H., et al., Replacing two conserved tyrosines of the EphB2 receptor with glutamic acid prevents binding of SH2 domains without abrogating kinase activity and biological responses. Oncogene, 2000. 19(2): p. 177-87.

117. Holland, S.J., et al., Bidirectional signalling through the EPH-family receptor Nuk and its transmembrane ligands. Nature, 1996. 383(6602): p. 722-5.

118. Bruckner, K., E.B. Pasquale, and R. Klein, Tyrosine phosphorylation of transmembrane ligands for Eph receptors. Science, 1997. 275(5306): p. 1640-3.

119. Himanen, J.P. and D.B. Nikolov, Eph signaling: a structural view. Trends in neurosciences, 2003. 26(1): p. 46-51.

120. Smalla, M., et al., Solution structure of the receptor tyrosine kinase EphB2 SAM domain and identification of two distinct homotypic interaction sites. Protein science : a publication of the Protein Society, 1999. 8(10): p. 1954-61.

121. Stapleton, D., et al., The crystal structure of an Eph receptor SAM domain reveals a mechanism for modular dimerization. Nature structural biology, 1999. 6(1): p. 44-9.

122. Zhuang, G., et al., Regulation of EphA2 receptor endocytosis by SHIP2 lipid phosphatase via phosphatidylinositol 3-Kinase-dependent Rac1 activation. The Journal of biological chemistry, 2007. 282(4): p. 2683-94.

123. Hock, B., et al., PDZ-domain-mediated interaction of the Eph-related receptor tyrosine kinase EphB3 and the ras-binding protein AF6 depends on the kinase activity of the receptor. Proceedings of the National Academy of Sciences of the United States of America, 1998. 95(17): p. 9779-84.

124. Pasquale, E.B., Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nature reviews. Cancer, 2010. 10(3): p. 165-80.

125. Nakamoto, M., Eph receptors and ephrins. The international journal of biochemistry & cell biology, 2000. 32(1): p. 7-12.

126. Himanen, J.P., et al., Crystal structure of an Eph receptor-ephrin complex. Nature, 2001. 414(6866): p. 933-8.

127. Lackmann, M., et al., Distinct subdomains of the EphA3 receptor mediate ligand binding and receptor dimerization. Journal of Biological Chemistry, 1998. 273(32): p. 20228-20237.

128. Noren, N.K. and E.B. Pasquale, Eph receptor-ephrin bidirectional signals that target Ras and Rho proteins. Cellular Signalling, 2004. 16(6): p. 655-66.

129. Sahin, M., et al., Eph-dependent tyrosine phosphorylation of ephexin1 modulates growth cone collapse. Neuron, 2005. 46(2): p. 191-204.

130. Shamah, S.M., et al., EphA receptors regulate growth cone dynamics through the novel guanine nucleotide exchange factor ephexin. Cell, 2001. 105(2): p. 233-244.

131. Murai, K.K. and E.B. Pasquale, New exchanges in Eph-dependent growth cone dynamics. Neuron, 2005. 46(2): p. 161-163.

132. Irie, F. and Y. Yamaguchi, EphB receptors regulate dendritic spine development via intersectin, Cdc42 and N-WASP. Nature Neuroscience, 2002. 5(11): p. 1117-1118.

133. Penzes, P., et al., Rapid induction of dendritic spine morphogenesis by trans-synaptic EphrinB-EphB receptor activation of the Rho-GEF kalirin. Neuron, 2003. 37(2): p. 263-274.

122

134. Pratt, R.L. and M.S. Kinch, Activation of the EphA2 tyrosine kinase stimulates the MAP/ERK kinase signaling cascade. Oncogene, 2002. 21(50): p. 7690-7699.

135. Tong, J.F., et al., Manipulation of EphB2 regulatory motifs and SH2 binding sites switches MAPK signaling and biological activity. Journal of Biological Chemistry, 2003. 278(8): p. 6111-6119.

136. Elowe, S., et al., Downregulation of the Ras-mitogen-activated protein kinase pathway by the EphB2 receptor tyrosine kinase is required for ephrin-induced neurite retraction. Molecular and Cellular Biology, 2001. 21(21): p. 7429-7441.

137. Palmer, A., et al., EphrinB phosphorylation and reverse signaling: regulation by Src kinases and PTP-BL phosphatase. Molecular Cell, 2002. 9(4): p. 725-737.

138. Tanaka, M., R. Kamata, and R. Sakai, Phosphorylation of ephrin-B1 via the interaction with claudin following cell-cell contact formation. The EMBO journal, 2005. 24(21): p. 3700-11.

139. Ran, X. and J. Song, Structural insight into the binding diversity between the Tyr-phosphorylated human ephrinBs and Nck2 SH2 domain. The Journal of biological chemistry, 2005. 280(19): p. 19205-12.

140. Armstrong, J.N., et al., B-ephrin reverse signaling is required for NMDA-independent long-term potentiation of mossy fibers in the hippocampus. Journal of Neuroscience, 2006. 26(13): p. 3474-3481.

141. Kayser, M.S., et al., Intracellular and trans-synaptic regulation of glutamatergic synaptogenesis by EphB receptors. Journal of Neuroscience, 2006. 26(47): p. 12152-12164.

142. Davy, A., et al., Compartmentalized signaling by GPI-anchored ephrin-A5 requires the Fyn tyrosine kinase to regulate cellular adhesion. Genes & development, 1999. 13(23): p. 3125-35.

143. Hattori, M., M. Osterfield, and J.G. Flanagan, Regulated cleavage of a contact-mediated axon repellent. Science, 2000. 289(5483): p. 1360-1365.

144. Janes, P.W., et al., Adam meets Eph: An ADAM substrate recognition module acts as a molecular switch for ephrin cleavage in trans. Cell, 2005. 123(2): p. 291-304.

145. Zimmer, M., et al., EphB-ephrinB bi-directional endocytosis terminates adhesion allowing contact mediated repulsion. Nature Cell Biology, 2003. 5(10): p. 869-878.

146. Marston, D.J., S. Dickinson, and C.D. Nobes, Rac-dependent trans-endocytosis of ephrinBs regulates Eph-ephrin contact repulsion. Nature Cell Biology, 2003. 5(10): p. 879-888.

147. Parker, M., et al., Reverse endocytosis of transmembrane ephrin-B ligands via a clathrin-mediated pathway. Biochemical and Biophysical Research Communications, 2004. 323(1): p. 17-23.

148. Goldshmit, Y., S. McLenachan, and A. Turnley, Roles of Eph receptors and ephrins in the normal and damaged adult CNS. Brain research reviews, 2006. 52(2): p. 327-45.

149. Henkemeyer, M., et al., Nuk controls pathfinding of commissural axons in the mammalian central nervous system. Cell, 1996. 86(1): p. 35-46.

150. Henderson, J.T., et al., The receptor tyrosine kinase EphB2 regulates NMDA-dependent synaptic function. Neuron, 2001. 32(6): p. 1041-56.

151. Birgbauer, E., et al., Kinase independent function of EphB receptors in retinal axon pathfinding to the optic disc from dorsal but not ventral retina. Development, 2000. 127(6): p. 1231-41.

152. Grunwald, I.C., et al., Hippocampal plasticity requires postsynaptic ephrinBs. Nature Neuroscience, 2004. 7(1): p. 33-40.

123

153. Klein, R., Bidirectional modulation of synaptic functions by Eph/ephrin signaling. Nature Neuroscience, 2009. 12(1): p. 15-20.

154. Savelieva, K.V., et al., Learning and memory impairment in Eph receptor A6 knockout mice. Neuroscience letters, 2008. 438(2): p. 205-9.

155. Dalva, M.B., et al., EphB receptors interact with NMDA receptors and regulate excitatory synapse formation. Cell, 2000. 103(6): p. 945-56.

156. Henkemeyer, M., et al., Multiple EphB receptor tyrosine kinases shape dendritic spines in the hippocampus. The Journal of cell biology, 2003. 163(6): p. 1313-26.

157. Grunwald, I.C., et al., Kinase-independent requirement of EphB2 receptors in hippocampal synaptic plasticity. Neuron, 2001. 32(6): p. 1027-40.

158. Takasu, M.A., et al., Modulation of NMDA receptor-dependent calcium influx and gene expression through EphB receptors. Science, 2002. 295(5554): p. 491-5.

159. Dalva, M.B., A.C. McClelland, and M.S. Kayser, Cell adhesion molecules: signalling functions at the synapse. Nature reviews. Neuroscience, 2007. 8(3): p. 206-20.

160. Simon, A.M., et al., Early changes in hippocampal Eph receptors precede the onset of memory decline in mouse models of Alzheimer's disease. Journal of Alzheimer's disease : JAD, 2009. 17(4): p. 773-86.

161. Cisse, M., et al., Reversing EphB2 depletion rescues cognitive functions in Alzheimer model. Nature, 2011. 469(7328): p. 47-52.

162. Battaglia, A.A., et al., EphB receptors and ephrin-B ligands regulate spinal sensory connectivity and modulate pain processing. Nature neuroscience, 2003. 6(4): p. 339-40.

163. Kobayashi, H., et al., Involvement of EphB1 receptor/EphrinB2 ligand in neuropathic pain. Spine, 2007. 32(15): p. 1592-8.

164. Song, X.J., et al., Upregulation and redistribution of ephrinB and EphB receptor in dorsal root ganglion and spinal dorsal horn neurons after peripheral nerve injury and dorsal rhizotomy. European journal of pain, 2008. 12(8): p. 1031-9.

165. Song, X.J., et al., EphrinB-EphB receptor signaling contributes to neuropathic pain by regulating neural excitability and spinal synaptic plasticity in rats. Pain, 2008. 139(1): p. 168-80.

166. Han, Y., et al., Targeted mutation of EphB1 receptor prevents development of neuropathic hyperalgesia and physical dependence on morphine in mice. Molecular Pain, 2008. 4: p. 60.

167. Liu, W.T., et al., EphB receptor signaling in mouse spinal cord contributes to physical dependence on morphine. FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2009. 23(1): p. 90-8.

168. Liu, S., et al., Blocking EphB1 receptor forward signaling in spinal cord relieves bone cancer pain and rescues analgesic effect of morphine treatment in rodents. Cancer research, 2011. 71(13): p. 4392-402.

169. Squire, L.R., The legacy of patient H.M. for neuroscience. Neuron, 2009. 61(1): p. 6-9. 170. Scoville, W.B. and B. Milner, Loss of recent memory after bilateral hippocampal

lesions. Journal of neurology, neurosurgery, and psychiatry, 1957. 20(1): p. 11-21. 171. Corkin, S., What's new with the amnesic patient H.M.? Nature reviews. Neuroscience,

2002. 3(2): p. 153-60. 172. Squire, L.R., Memory systems of the brain: a brief history and current perspective.

Neurobiology of learning and memory, 2004. 82(3): p. 171-7. 173. Baddeley, A., Working memory. Current biology : CB, 2010. 20(4): p. R136-40. 174. Squire, L.R. and J.T. Wixted, The cognitive neuroscience of human memory since H.M.

Annual Review of Neuroscience, 2011. 34: p. 259-88.

124

175. Okano, H., T. Hirano, and E. Balaban, Learning and memory. Proceedings of the National Academy of Sciences of the United States of America, 2000. 97(23): p. 12403-4.

176. Mayes, A.R., et al., The relationship between retrograde and anterograde amnesia in patients with typical global amnesia. Cortex; a journal devoted to the study of the nervous system and behavior, 1997. 33(2): p. 197-217.

177. Squire, L.R., Memory and brain systems: 1969-2009. The Journal of neuroscience : the official journal of the Society for Neuroscience, 2009. 29(41): p. 12711-6.

178. Packard, M.G. and B.J. Knowlton, Learning and memory functions of the Basal Ganglia. Annual Review of Neuroscience, 2002. 25: p. 563-93.

179. McDonald, R.J. and N.M. White, A triple dissociation of memory systems: hippocampus, amygdala, and dorsal striatum. Behavioral Neuroscience, 1993. 107(1): p. 3-22.

180. Ito, M., Mechanisms of motor learning in the cerebellum. Brain research, 2000. 886(1-2): p. 237-245.

181. Meyers, R.A., A.R. Zavala, and J.L. Neisewander, Dorsal, but not ventral, hippocampal lesions disrupt cocaine place conditioning. Neuroreport, 2003. 14(16): p. 2127-31.

182. Olmstead, M.C. and K.B. Franklin, The development of a conditioned place preference to morphine: effects of lesions of various CNS sites. Behavioral Neuroscience, 1997. 111(6): p. 1313-23.

183. Bliss, T.V. and G.L. Collingridge, A synaptic model of memory: long-term potentiation in the hippocampus. Nature, 1993. 361(6407): p. 31-9.

184. Lisman, J., A mechanism for the Hebb and the anti-Hebb processes underlying learning and memory. Proceedings of the National Academy of Sciences of the United States of America, 1989. 86(23): p. 9574-8.

185. Collingridge, G.L., et al., Long-term depression in the CNS. Nature reviews. Neuroscience, 2010. 11(7): p. 459-73.

186. Ito, M., Long-term depression. Annual Review of Neuroscience, 1989. 12: p. 85-102. 187. Moser, M.B. and E.I. Moser, Functional differentiation in the hippocampus.

Hippocampus, 1998. 8(6): p. 608-19. 188. Andersen, P., et al., Lamellar organization of hippocampal excitatory pathways. Acta

physiologica Scandinavica, 1969. 76(1): p. 4A-5A. 189. Pothuizen, H.H., et al., Dissociation of function between the dorsal and the ventral

hippocampus in spatial learning abilities of the rat: a within-subject, within-task comparison of reference and working spatial memory. The European journal of neuroscience, 2004. 19(3): p. 705-12.

190. Bannerman, D.M., et al., Ventral hippocampal lesions affect anxiety but not spatial learning. Behavioural brain research, 2003. 139(1-2): p. 197-213.

191. Bannerman, D.M., et al., Regional dissociations within the hippocampus--memory and anxiety. Neuroscience and biobehavioral reviews, 2004. 28(3): p. 273-83.

192. Jarrard, L.E., On the role of the hippocampus in learning and memory in the rat. Behavioral and neural biology, 1993. 60(1): p. 9-26.

193. Tsien, J.Z., P.T. Huerta, and S. Tonegawa, The essential role of hippocampal CA1 NMDA receptor-dependent synaptic plasticity in spatial memory. Cell, 1996. 87(7): p. 1327-38.

194. Moser, M.B., et al., Spatial learning with a minislab in the dorsal hippocampus. Proceedings of the National Academy of Sciences of the United States of America, 1995. 92(21): p. 9697-701.

195. Moser, E., M.B. Moser, and P. Andersen, Spatial learning impairment parallels the magnitude of dorsal hippocampal lesions, but is hardly present following ventral lesions.

125

The Journal of neuroscience : the official journal of the Society for Neuroscience, 1993. 13(9): p. 3916-25.

196. Best, P.J., A.M. White, and A. Minai, Spatial processing in the brain: the activity of hippocampal place cells. Annual Review of Neuroscience, 2001. 24: p. 459-86.

197. Moser, E.I., E. Kropff, and M.B. Moser, Place cells, grid cells, and the brain's spatial representation system. Annual Review of Neuroscience, 2008. 31: p. 69-89.

198. Maren, S., Neurobiology of Pavlovian fear conditioning. Annual Review of Neuroscience, 2001. 24: p. 897-931.

199. Anagnostaras, S.G., G.D. Gale, and M.S. Fanselow, Hippocampus and contextual fear conditioning: recent controversies and advances. Hippocampus, 2001. 11(1): p. 8-17.

200. Stubley-Weatherly, L., J.W. Harding, and J.W. Wright, Effects of discrete kainic acid-induced hippocampal lesions on spatial and contextual learning and memory in rats. Brain research, 1996. 716(1-2): p. 29-38.

201. Walsh, R.N. and R.A. Cummins, The Open-Field Test: a critical review. Psychological bulletin, 1976. 83(3): p. 482-504.

202. Maren, S. and M.S. Fanselow, Electrolytic lesions of the fimbria/fornix, dorsal hippocampus, or entorhinal cortex produce anterograde deficits in contextual fear conditioning in rats. Neurobiology of learning and memory, 1997. 67(2): p. 142-9.

203. Siegel, S., Morphine analgesic tolerance: its situation specificity supports a Pavlovian conditioning model. Science, 1976. 193(4250): p. 323-5.

204. Siegel, S., Morphine tolerance acquisition as an associative process. Journal of experimental psychology. Animal behavior processes, 1977. 3(1): p. 1-13.

205. Siegel, S., Evidence from rats that morphine tolerance is a learned response. Journal of comparative and physiological psychology, 1975. 89(5): p. 498-506.

206. Brase, D.A., H.H. Loh, and E.L. Way, Comparison of the effects of morphine on locomotor activity, analgesia and primary and protracted physical dependence in six mouse strains. The Journal of pharmacology and experimental therapeutics, 1977. 201(2): p. 368-74.

207. Stevens, K.E., G.A. Mickley, and L.J. McDermott, Brain areas involved in production of morphine-induced locomotor hyperactivity of the C57B1/6J mouse. Pharmacology, biochemistry, and behavior, 1986. 24(6): p. 1739-47.

208. Andersen, J.M., et al., Increased locomotor activity induced by heroin in mice: pharmacokinetic demonstration of heroin acting as a prodrug for the mediator 6-monoacetylmorphine in vivo. The Journal of pharmacology and experimental therapeutics, 2009. 331(1): p. 153-61.

209. Karinen, R., et al., Determination of heroin and its main metabolites in small sample volumes of whole blood and brain tissue by reversed-phase liquid chromatography-tandem mass spectrometry. Journal of analytical toxicology, 2009. 33(7): p. 345-50.

210. Mansour, A., et al., Autoradiographic differentiation of mu, delta, and kappa opioid receptors in the rat forebrain and midbrain. The Journal of neuroscience : the official journal of the Society for Neuroscience, 1987. 7(8): p. 2445-64.

211. Arvidsson, U., et al., Distribution and targeting of a mu-opioid receptor (MOR1) in brain and spinal cord. The Journal of neuroscience : the official journal of the Society for Neuroscience, 1995. 15(5 Pt 1): p. 3328-41.

212. Lee, I., M.R. Hunsaker, and R.P. Kesner, The role of hippocampal subregions in detecting spatial novelty. Behavioral Neuroscience, 2005. 119(1): p. 145-53.

213. Morris, R.G., et al., Place navigation impaired in rats with hippocampal lesions. Nature, 1982. 297(5868): p. 681-3.

126

214. Squire, L.R., Memory and the hippocampus: a synthesis from findings with rats, monkeys, and humans. Psychological review, 1992. 99(2): p. 195-231.

215. Nyberg, L., et al., Activation of medial temporal structures during episodic memory retrieval. Nature, 1996. 380(6576): p. 715-7.

216. Fanselow, M.S. and H.W. Dong, Are the dorsal and ventral hippocampus functionally distinct structures? Neuron, 2010. 65(1): p. 7-19.

217. White, N.M. and S. Gaskin, Dorsal hippocampus function in learning and expressing a spatial discrimination. Learning & memory, 2006. 13(2): p. 119-22.

218. Moser, E.I., et al., Impaired spatial learning after saturation of long-term potentiation. Science, 1998. 281(5385): p. 2038-42.

219. Bear, M.F. and W.C. Abraham, Long-term depression in hippocampus. Annual Review of Neuroscience, 1996. 19: p. 437-62.

220. Gilbert, C.D. and M. Sigman, Brain states: top-down influences in sensory processing. Neuron, 2007. 54(5): p. 677-96.

221. Kentros, C.G., et al., Increased attention to spatial context increases both place field stability and spatial memory. Neuron, 2004. 42(2): p. 283-95.

222. Burwell, R.D., The parahippocampal region: corticocortical connectivity. Annals of the New York Academy of Sciences, 2000. 911: p. 25-42.

223. Krank, M.D., R.E. Hinson, and S. Siegel, Conditional hyperalgesia is elicited by environmental signals of morphine. Behavioral and neural biology, 1981. 32(2): p. 148-57.

224. Winocur, G. and D. Bindra, Effects of additional cues on passive avoidance learning and extinction in rats with hippocampal lesions. Physiology & behavior, 1976. 17(6): p. 915-20.

225. Lorenzini, C.A., et al., Role of dorsal hippocampus in acquisition, consolidation and retrieval of rat's passive avoidance response: a tetrodotoxin functional inactivation study. Brain research, 1996. 730(1-2): p. 32-9.

226. Munoz, C. and S.P. Grossman, Spatial discrimination, reversal and active or passive avoidance learning in rats with KA-induced neuronal depletions in dorsal hippocampus. Brain research bulletin, 1981. 6(5): p. 399-406.

227. Light, K.R., et al., General learning ability regulates exploration through its influence on rate of habituation. Behavioural brain research, 2011. 223(2): p. 297-309.

127