an intranasal delivery method for novel peptide ... · figure 3-1 representative images of...

130
An Intranasal Delivery Method for Novel Peptide Therapeutics Designed to treat Major Depressive Disorder by Virginia Joan Margaret Brown A thesis submitted in conformity with the requirements for the degree of Masters of Science Department of Physiology University of Toronto © Copyright by Virginia Brown, 2013

Upload: others

Post on 23-Sep-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

An Intranasal Delivery Method for Novel Peptide Therapeutics

Designed to treat Major Depressive Disorder

by

Virginia Joan Margaret Brown

A thesis submitted in conformity with the requirements

for the degree of Masters of Science

Department of Physiology

University of Toronto

© Copyright by Virginia Brown, 2013

Page 2: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

ii

An Intranasal Delivery Method for Novel Therapeutics designed to treat

Major Depressive Disorder

Virginia Brown

Masters of Science

Department of Physiology

University of Toronto

2013

Abstract

A problem in designing drugs that act upon the central nervous system is developing

effective delivery methods. Major depressive disorder (MDD) affects 12% of men and 20% of

women in the United States, and treatment options are often inadequate. In patients, the

interaction between dopamine D1 and D2 receptors is correlated with major depressive disorder.

A small peptide that disrupts this interaction can be delivered to brain areas using intranasal

delivery. The D1-D2 interfering peptide has an antidepressant effect comparable to imipramine

in the forced swimming test (FST), a test for antidepressant efficacy. At doses greater than 5.75

mg/kg, the D1-D2 interfering peptide has antidepressant action in the FST for 2 hours after

intranasal administration. The D1-D2 interfering peptide disrupts the D1-D2 receptor interaction

in the PFC after intranasal administration. This study provides preclinical support for intranasal

administration of the D1-D2 interfering peptide as a new treatment option for MDD.

Page 3: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

iii

Acknowledgments

I would like to thank my supervisor, Dr. Fang Liu, for her guidance and support over the

last 18-20 months. Her encouragement and positive attitude have made working in the lab a

pleasure. Her scientific guidance over the course of this project as well as her personal guidance

has been invaluable.

I would also like to thank my Masters’ committee members, Dr. Paul Fletcher and Dr.

Paul Frankland. Their expertise, positive encouragement and guidance significantly contributed

to my learning throughout this project and to its overall success.

Impel NeuroPharma, the company that developed the POD used throughout my study,

provided training and important input into the development of the protocol we used to administer

substances intranasally. I am grateful to them, especially to John Hokeman, for their patience and

encouragement.

Finally, I would like to thank my wonderful family, friends and roommates for their

encouragement, understanding and support throughout the last two years.

Page 4: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

iv

Table of Contents

Acknowledgments .......................................................................................................................... iii

Table of Contents ........................................................................................................................... iv

List of Tables ................................................................................................................................. vi

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

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

1 Introduction .............................................................................................................................. 1

1.1 Dopamine neurotransmission in the mammalian brain ...................................................... 3

1.1.2 Dopaminergic pathways in the mammalian CNS ................................................. 10

1.1.3 Heterodimerization of Dopamine Receptors ........................................................ 16

1.2 Major Depressive Disorder ............................................................................................... 19

1.2.1 Epidemiology of MDD ......................................................................................... 20

1.2.2 Symptoms and clinical presentation of MDD ....................................................... 21

1.2.3 Treatments for MDD ............................................................................................. 22

1.2.4 Neurobiological changes and pathophysiology of MDD ...................................... 25

1.2.5 Preclinical models of MDD .................................................................................. 31

1.3 Intranasal delivery to the CNS .......................................................................................... 33

1.3.1 Mechanisms of intranasal delivery to the CNS ..................................................... 34

1.3.2 Experimental considerations for successful intranasal delivery to the CNS ........ 36

1.4 Rationale ........................................................................................................................... 36

1.5 Hypothesis ......................................................................................................................... 39

2 Materials and Methods .......................................................................................................... 41

2.1 Animals ............................................................................................................................. 41

2.2 Intranasal administration procedures ................................................................................ 41

2.2.1 Intranasal administration using the POD .............................................................. 41

2.2.2 Verification of POD delivery to the olfactory epithelium .................................... 42

2.2.3 Substances injected intranasally ........................................................................... 42

2.3 Intra-peritoneal injection procedures ................................................................................ 44

2.4 Immunofluorescence and confocal microscopy ................................................................ 44

2.4.1 Tissue fixation and storage ................................................................................... 44

2.4.2 Immunofluorescent staining procedures ............................................................... 45

2.5 The Forced Swimming Test .............................................................................................. 46

2.5.1 FST Procedure ...................................................................................................... 46

2.5.2 FST behavioral scoring method ............................................................................ 48

2.6 FST experiments: experimental design ............................................................................. 50

2.6.1 Effect of the D1-D2 interfering peptide in the FST .............................................. 50

2.6.2 Effect of the D1-D2-FLAG interfering peptide in the FST .................................. 50

2.6.3 Efficacy of the D1-D2 interfering peptide at various intranasal doses ................. 51

2.6.4 Duration of behavioral effect of D1-D2 interfering peptide in the FST ............... 53

2.7 Locomotor activity test ..................................................................................................... 53

2.9 Co-immunoprecipitation and western blots ...................................................................... 55

2.9.1 Tissue Collection .................................................................................................. 55

2.9.2 Co-Immunoprecipitation of D1 receptor by anti-D2DR ....................................... 56

2.9.3 Western Blots ........................................................................................................ 57

3 Results ..................................................................................................................................... 58

Page 5: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

v

3.1 Experiment 1: The POD preferentially deposits substances on the olfactory

epithelium within the rat nasal cavity ............................................................................... 58

3.2 Experiment 2: The D1-D2-FLAG interfering peptide can be detected in the prefrontal

cortex after intranasal administration ................................................................................ 58

3.3 Experiment 3: Intranasal administration of the D1-D2 interfering peptide has an

antidepressant effect in the forced swimming test ............................................................ 60

3.3.1 The D1-D2 Interfering Peptide has an Anti-Immobility Effect in the FST .......... 62

3.3.2 The D1-D2-FLAG interfering peptide has an anti-immobility effect in the FST

after intranasal administration ............................................................................... 64

3.4 Experiment 4: Efficacy of the D1-D2 interfering peptide at various intranasal doses ..... 67

3.4.1 D1-D2 interfering peptide dose: 4.0nmol/g (13.72 mg/kg) .................................. 67

3.4.2 D1-D2 interfering peptide dose: 2.0nmol/g (6.86 mg/kg) .................................... 68

3.4.3 D1-D2 interfering peptide dose: 1.67nmol/g (5.75 mg/kg) .................................. 68

3.4.4 D1-D2 interfering peptide dose: 1.0nmol/g (3.43 mg/kg) .................................... 71

3.5 Experiment 5: Duration of the behavioral effect of the D1-D2 interfering peptide ......... 75

3.5.1 Behavioral Effect in FST 2 hours after intranasal administration ........................ 75

3.5.2 Behavioral Effect in FST 3 hours after intranasal administration ........................ 75

3.5.3 Behavioral effect in the FST 4 hours after intranasal administration ................... 76

3.6 Experiment 6: The D1-D2 interfering peptide does not increase locomotor activity ....... 81

3.6.1 Overall locomotor activity .................................................................................... 81

3.6.2 Effect of time on locomotor activity during 30-minute test ................................. 82

3.7 Experiment 7: Intranasal administration of the D1-D2 interfering peptide disrupts the

interaction between dopamine D1 and D2 receptors in the PFC ...................................... 85

3.8 Experiment 8: The D1-D2 interfering peptide does not change the expression of

dopamine D1 or D2 receptors in the PFC ......................................................................... 85

3.8.1 Expression of Dopamine D1 receptors in the PFC after intranasal

administration of the D1-D2 interfering peptide .................................................. 87

3.8.2 Expression of Dopamine D2 receptors in the PFC after intranasal

administration of the D1-D2 interfering peptide .................................................. 87

4 Discussion ................................................................................................................................ 91

4.1 Overall Findings ................................................................................................................ 91

4.2 The POD delivers biologically active peptides to the CNS .............................................. 92

4.3 Mechanism of transport to the CNS after intranasal administration ................................. 94

4.4 The D1-D2 interfering peptide is effective at intranasal doses ≥ 5.75 mg/kg for up to

2 hours after intranasal administration. ............................................................................. 95

4.5 Possible neurobiological mechanisms of the D1-D2 interfering peptide’s

antidepressant effect .......................................................................................................... 97

4.6 Limitations of the FST as a preclinical test for antidepressant efficacy ........................... 99

4.7 The D1-D2 interfering peptide, TAT-peptide and imipramine significantly decrease

locomotor activity ........................................................................................................... 100

4.8 Future Directions ............................................................................................................ 104

References ................................................................................................................................... 107

Appendix 1: Sufficient intranasal D1-D2 interfering peptide dose to produce antidepressant

effect in the Forced Swimming Test (Calculation) ................................................................ 120

Page 6: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

vi

List of Tables

Table 2-1 Efficacy of the D1-D2 interfering peptide at various doses: overall experimental

design and Treatment Groups. ...................................................................................................... 52

Table 2-2 Duration of the anti-immobility effect of the D1-D2 interfering peptide: treatment

groups and overall experimental design ....................................................................................... 54

Page 7: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

vii

List of Figures

Figure 1-1 Dopamine receptors: structure and function ............................................................... 9

Figure 1-2 Schematic Representation of D1-D2R receptor interaction and activation of

intracellular signalling pathways. ................................................................................................. 18

Figure 1-3 Previous findings demonstrating the role of the D1-D2R interaction in MDD. ........ 29

Figure 2-1 Pressurized Olfactory Device (POD) for intranasal administration: apparatus ......... 43

Figure 2-2 Overall experimental procedure for FST. .................................................................. 47

Figure 2-3 Representative photographs of behaviors exhibited during the FST. ........................ 49

Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after

correct POD administration .......................................................................................................... 59

Figure 3-2 Immunofluorescent staining for anti-FLAG antibodies is visible in PFC slices of

animals who were administered TAT-D1-D2-FLAG-IPep (A) but not those who were

administered saline (B). ................................................................................................................ 61

Figure 3-3 The D1-D2 interfering peptide has an antidepressant effect in the FST when

administered intranasally. ............................................................................................................. 63

Figure 3-4 The D1-D2-FLAG interfering peptide has an anti-immobility effect in the FST. ..... 65

Figure 3-5 The D1-D2 interfering peptide and D1-D2-FLAG tagged interfering peptide have

similar behavioral effects in the FST. ........................................................................................... 66

Figure 3-6 The D1-D2 interfering peptide has an anti-immobility effect in the FST at an

intranasal dose of 4.0nmol/g ......................................................................................................... 69

Figure 3-7 The D1-D2 interfering peptide has an anti-immobility effect in the FST at an

intranasal dose of 2.0nmol/g ......................................................................................................... 70

Figure 3-9 The D1-D2 interfering peptide has an anti-immobility effect in the FST at an

intranasal dose of 1.67 nmol/g. ..................................................................................................... 72

Figure 3-8 The D1-D2 interfering peptide does not have an anti-immobility effect in the FST at

an intranasal dose of 1.0nmol/g .................................................................................................... 73

Figure 3-10 Efficacy of the D1-D2 interfering peptide at various doses in the FST: summary of

findings. ........................................................................................................................................ 74

Figure 3-12 The D1-D2 interfering peptide has an anti-immobility effect in the FST 2 hours

after intranasal administration. ...................................................................................................... 77

Page 8: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

viii

Figure 3-11 The D1-D2 Interfering Peptide does not have an anti-immobility effect in the FST 3

hours after intranasal administration. ............................................................................................ 78

Figure 3-13 The D1-D2 interfering peptide does not have an anti-immobility effect in the FST 4

hours after intranasal administration ............................................................................................. 79

Figure 3-14 The D1-D2 interfering peptide no longer has a behavioral effect in the FST 3 hours

after it is administered via intranasal injections. ........................................................................... 80

Figure 3-15 The D1-D2 interfering peptide does not increase locomotor activity during a 30-

minute open field test. ................................................................................................................... 83

Figure 3-16 The D1-D2 interfering peptide decreases overall locomotor activity but does not

change the activity pattern during a 30-minute open field test. .................................................... 84

Figure 3-17 Co-Immunoprecipitation of D1 by anti-D2R is reduced in the PFC of animals who

received intransal injections of TAT-D1-D2-IPep (Dose: 1.67nmol/g) ....................................... 86

Figure 3-18 Intranasal administration of the D1-D2 interfering peptide does not change the

expression of the dopamine D1 receptor in the PFC. ................................................................... 88

Figure 3-19 Intranasal administration of the D1-D2 interfering peptide does not change the

expression of the dopamine D2 Receptor in the PFC ................................................................... 89

Figure 3-20 Representative immunoblot of α-tubulin expression in rat PFC tissue. ................... 90

Page 9: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

ix

List of Abbreviations

5-HT serotonin

AC adenyl cyclase

ADHD attention deficit hyperactivity disorder

ANOVA Analysis of Variance

anti α-tubulin immunoglobulin against α-tubulin protein

anti-cy2 immunoglobulin conjugated to cyanine 2 fluorescent dye

anti-D1DR immunoglobulin against dopamine D1 receptor

anti-D2DR immunoglobulin against dopamine D2 receptor

ATP adenosine triphosphate

BDNF brain derived neurotrophic factor

CaMKII calmodulin kinase II

cAMP cyclic adenosine monophosphate

cDNA complementary DNA (deoxyribonucleic acid)

CNS central nervous system

CSF cerebrospinal fluid

C-terminal carboxy-terminal of protein

D1 dopamine D1 receptor

D1-D2 dopamine D1-D2 receptor interaction

D2 dopamine D2 receptor

D2L dopamine D2 receptor - long isoform

D2S dopamine D2 receptor - short isoform

D3 dopamine D3 receptor

D4 dopamine D4 receptor

D5 dopamine D5 receptor

DA dopamine

DAT dopamine transporter protein

DDC DOPA decarboxylase

DSM-IV-TR Diagnostic and Statistical Manual, 4th edition, text revision (2000)

FLAG FLAG octapeptide (protein tag)

FST forced swimming test

GABA gamma-Aminobutyric acid

GABAAR gamma-Aminobutyric acid receptor type A

Gi/o G protein, α subunit type i/o

GPCR G protein coupled receptor

Gq G protein, α subunit type q

Gs

GSK-3β

G protein, α subunit type s

Glycogen Synthase Kinase 3β

GTP guanine triphosphate

Gα G protein, α subunit

Page 10: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

x

HIV1 human immunodeficiency virus type 1

IGF-1 insulin-like growth factor 1

IP intraperitoneal injection

IP3 inositol triphosphate

kD kiloDalton

L-DOPA L-3,4-dihydroxyphenylalanine

LH learned helplessness

MAO monomaine oxidase

MAOI monoamine oxidase inhibitors

MCI mild cognitive impairment

MDD major depressive disorder

mg/kg milligrams per kilogram

mRNA messenger RNA (ribonucleic acid)

MSN medium spiny neuron

NAc nucleus accumbens

NE norepinephrine

NGF nerve growth factor

NMDARs n-methyl-D-aspartate receptors

nmol/g nanomoles per gram

OEC olfactory ensheathing cell

ORN olfactory receptor neuron

PBS phosphate buffered saline

PD Parkinson's Disease

PFA 4 % paraformaldehyde

PFC prefrontal cortex

PLC phospholipase C

POD pressurized olfactory device

RGP regulators of G proteins

SNpc substantia nigra pars compacta

SSRI selective serotonin reuptake inhibitor

STAR*D sequenced treatment alternatives to relieve depression clinical trial

TAT membrane permeable protein from HIV1

TAT-D1-D2-FLAG-Ipep TAT-linked membrane permeable D1-D2 interfering peptide with c-

terminal 8-amino acid FLAG tag

TAT-D1-D2-Ipep TAT-linked membrane permeable D1-D2 interfering peptide

TAT-Pep 9-amino acid membrane permeable peptide fragment from HIV1 TAT

protein

TH tyrosine hydroxylase

TrkB tyrosine receptor kinase type B

VMAT2 vesicular monoamine transporter 2

VTA ventral tegmental area

Page 11: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

1

1 Introduction

The neurotransmitter dopamine is involved in many processes within the brain, including

motor control, cognition, reward, emotion and pleasure. Dopamine exerts its effects through five

unique dopamine receptors, termed D1 through D5. These receptors are G-protein coupled

receptors (GPCRs) that contain seven trans-membrane domains and initiate intracellular

signaling cascades.4 In addition to existing as unique receptors, dopamine receptors can also

couple with other proteins and receptors to form functional heterodimers that activate signaling

cascades, independent from those activated by each component receptor.5,6

Recently, scientific

evidence has shown that these heterodimers can play a pathological role in the progression of

psychiatric conditions.7 Dopamine D1 and D2 receptors couple in this manner and are thought to

play a role in psychiatric conditions such as Major Depressive Disorder (MDD).3,8

Our laboratory has found a pathophysiological role for the dopamine D1-D2 heterodimer

in MDD.3 MDD is a common, serious psychiatric condition that accounts for 4.4% of total global

disease burden9 and is often left undiagnosed and untreated in patients.

10-12 Furthermore, many

patients do not respond to available pharmacological or psychological treatment for MDD with

over 50% of patients not responding to first-line pharmacological treatment.13,14

Pei et al3

demonstrated that the D1-D2 heterodimer is up-regulated in the striatum of patients with MDD.

Disrupting this interaction using a membrane permeable peptide (the D1-D2 interfering peptide)

had an antidepressant effect in the Forced Swimming Test (FST) and the Learned Helplessness

(LH) task, two strongly validated preclinical tests for antidepressant efficacy.3 These results are

promising, but lack clinical validity since invasive, direct administration methods were used to

deliver the peptide to the prefrontal cortex (PFC). In order for the D1-D2 interfering peptide to

Page 12: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

2

become a clinically relevant antidepressant treatment, a less invasive, clinically applicable

method of drug delivery must be developed.

The purpose of this project is to test whether we can effectively administer the D1-D2

interfering peptide to the brain using intranasal delivery. Intranasal delivery is clinically

applicable, offers a direct pathway to the brain and is a non-invasive method to target

therapeutics to the central nervous system (CNS). A number of proteins including insulin and

nerve growth factor have been delivered to the CNS intranasally, both in animals and in

humans.15

The goals of this project are to (1) confirm that the D1-D2 interfering peptide is able

to disrupt the interaction between D1 and D2 when administered intranasally, (2) test whether

the D1-D2 peptide has an antidepressant effect in the FST when administered intranasally, and

(3) further investigate the pharmacological properties of the D1-D2 interfering peptide.

In the introduction, I will briefly review the scientific literature relating the role of

dopamine in cognitive and behavioral processes within the mammalian brain as well as currently

held hypotheses about dopamine receptor heterodimerization and its role in psychiatric

conditions (Section 1.1). I will focus specifically on the Dopamine D1 and D2 receptor-receptor

interaction our laboratory has previously identified (Section 1.1.3). Next, the etiology,

symptoms, and neurobiology of MDD will be reviewed, along with currently available

antidepressant treatment options and their efficacy in treating this disorder (Section 1.2). I will

also discuss preclinical models of depression, and their strengths and weaknesses for identifying

new therapeutics for this complex psychiatric disorder (Section 1.2.5). Finally, I will discuss the

evidence supporting the use of intranasal administration methods to target therapeutic substances

to the CNS (Section 1.3).

Page 13: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

3

1.1 Dopamine neurotransmission in the mammalian brain

Dopamine is a catecholamine neurotransmitter synthesized from the amino acid

tyrosine.16

Discovered in the mid-20th

century,17,18

dopamine and its role in the central nervous

system have been the subject of extensive scientific investigation. It is involved in a wide variety

of cognitive and behavioral processes in the brain, such as reward seeking and motivation,

voluntary motor movement, emotional and cognitive processing, attention, and working memory.

It also plays a role in numerous neurological and psychiatric illnesses, including but not limited

to Parkinson’s Disease19

, Huntington’s Disease20

, schizophrenia21

, major depression22

, and

addiction23,24

. For example, Parkinson’s Disease (PD) is a neurodegenerative disorder that

occurs due to a loss of dopaminergic neurons in the substantia nigra (SN) and dopaminergic

innervations to the striatum, a brain area involved in voluntary motor movements.19

Dopamine itself is a small organic compound made up of a benzene ring, an amine group

attached to a 3-carbon chain, and two hydroxyl groups.25

In the central nervous system,

dopamine is synthesized from the amino acid tyrosine in neurons containing the enzymes

necessary for this conversion. These neurons originate in three distinct areas: the substantia nigra

pars compacta (SNpc), the ventral tegmental area (VTA) and the arcuate nucleus of the

hypothalamus.16

Briefly, the amino acid tyrosine is converted into L-DOPA by the enzyme

tyrosine hydroxylase (TH), the rate-limiting enzyme in the production of dopamine.16

L-DOPA

is converted into dopamine by DOPA decarboxylase (DDC).16

Once synthesized, dopamine is

transported from the cytosol into synaptic vesicles by the vesicular monoamine transporter 2

(VMAT2), from where it is released into the synaptic cleft when dopamine neurons fire.4 Once

released into the synaptic cleft, it binds to and activates dopamine receptors on the post-synaptic

(and pre-synaptic) membranes. Subsequently, it can be transported back into the presynaptic

dopaminergic neuron by the dopamine transporter protein (DAT) for re-use or degradation.16

Page 14: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

4

Dopamine is broken down into inactive metabolites by a sequence of reactions catalyzed by

monoamine oxidases (MAOs) resulting in the production of homovanillic acid, which is released

into the cerebrospinal fluid (CSF) as metabolic waste. Dopamine also serves as the precursor for

norepinephrine, as the two neurotransmitters differ by the addition of one β-hydroxyl group, a

reaction catalyzed by the enzyme dopamine β-hydroxylase.16

Presynaptic neurons that produce and release dopamine originate from distinct areas in

the basal ganglia and innervate cortical areas, the hippocampus and limbic cortex, and brain

areas related to movement and endocrine function (see Section 1.1.2).4 Like many

neurotransmitters, dopamine exerts its effects in the brain through specific receptors termed D1

through D5, G-protein coupled receptors that each have specific downstream effects in cells that

effect complex intracellular signaling pathways.26,27

Dopamine receptors can form functional

interactions with different types of dopamine receptors3,5,28

while also interacting with other

types of neurotransmitter receptors and a variety of other proteins to facilitate cross-talk between

neurotransmitter systems29-31

(Section 1.1.3). Unlike other neurotransmitters such as glutamate

and GABA, dopamine is not considered an excitatory or an inhibitory neurotransmitter, as the

ultimate effect of dopamine on a given neuronal population depends on the type of dopamine

receptor and the ultimate effect of the intracellular signaling cascade that is activated.

1.1.1 Dopamine receptors and their intracellular effects

As stated above, dopamine exerts its intracellular effects through five distinct receptors,

D1 through D5. These receptors belong to the guanine nucleotide-blinding (G-protein) coupled

receptors superfamily. G-proteins are signal transducers that mediate the transduction of

intracellular signals for a vast number of endocrine, neurotransmitter, autocrine and paracrine

compounds. There are four main types of G proteins, Gs, Gt, Gi and Go, each consisting of three

Page 15: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

5

subunits (Gα, Gβ and Gγ).32

There are 20 known Gα, 6 Gβ and 11 Gγ subunits, and G-proteins

are typically named by the identity of their α-subunit.32,33

As a result of their heterogeneity, a

large number of GPCRs can form with resultant activation of vastly different intracellular

signaling pathways.32

Dopamine receptors, like all GPCRs, exert their downstream signaling effects by

activating (or inhibiting) intracellular second messenger cascades.25,34

In the case of dopamine

receptors, the receptor itself contains 7 transmembrane domains, and is coupled to G protein

subunits on the intracellular side (See Figure 1-1).4,25

After dopamine binds to its binding site,

the G-protein becomes activated and causes downstream intracellular effects, which underlie the

cognitive and behavioral changes mediated by dopamine in the brain.35

The existence of dopamine receptors was first proposed in the 1970s when Kebabian and

Greengard published evidence for a dopamine-selective adenyl cyclase36

(AC, an enzyme that

converts ATP to cAMP, a potent second-messenger signaling molecule).4 After the initial

discovery of cAMP-coupled dopamine receptors, Spano et al37

demonstrated that dopamine

receptors exist in two groups, one that is positively coupled to cAMP production and one that is

not.4 Based on their opposing effects on cAMP signaling, these receptors types were named D1

(cAMP-activating) and D2 (cAMP-inhibiting).26

Quickly, the hypothesis that dopaminergic

signaling was mediated by two dopamine receptors with opposing effects was proven to be an

oversimplification,35

as the advent of molecular biology and genetic cloning techniques allowed

for the identification of D1 and D2 cDNA38

and three additional distinct dopamine receptors

activated by dopamine: D339

, D440

and D541

. Currently, dopaminergic pathways in the CNS are

thought to be mediated by these five dopamine receptors, that are separated into two families

based on their effects on cAMP: D1-like and D2-like.4

Page 16: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

6

1.1.1.1 D1-like dopamine receptors

The D1-like dopamine receptor family is comprised of dopamine D1 and D5 (formerly

D1B) receptors.41

When agonists bind to these receptors they activate the Gs/α family of GPCRs.

Activation of Gs/α results in activation of AC, and cAMP production.25

D1-like receptors can also

couple to Golf/α (also stimulating AC and cAMP production) in specific brain areas (caudate,

nucleus accumbens (NAc) and olfactory tubercle).42

The genes for both D1 and D5 receptors do

not contain introns in their coding sequences, and thus there are no splice variants of D1 or

D5.34,43

Some pharmacological differences exist between D1 and D5 receptors, as the D5

receptor is more pharmacologically sensitive to dopamine than D1 receptor.34

D1 receptors are the most common dopamine receptors in the CNS, and are highly

expressed in post-synaptic targets of the mesocortical, mesolimbic and nigrostriatal dopamine

pathways (see Section 1.1.2), including the striatum, NAc, amygdala and PFC.25,44,45

D5

receptors are expressed at lower levels in the PFC, the cingulate cortex, substantia nigra,

hypothalamus, hippocampus and dentate gyrus.45-47

D1 and D5 receptors are co-expressed in

pyramidal neurons of the prefrontal, premotor, and cingulate cortices and the dentate gyrus.48,49

Unlike the D2-like dopamine receptors, D1-like receptors are, for the most part,

expressed mostly in the post-synaptic membrane of neurons receiving dopaminergic input4,25

,

although recent evidence suggests that D5 is expressed presynaptically in the basolateral

amygdala and other brain structures.49

Thus, it is probable that dopamine D1-type receptors are

responsible for mediating the diverse effects of dopamine on its post-synaptic cellular targets.

For example, dopaminergic signaling through D1-type receptors in the PFC is critical to working

memory processes50

and to the occurrence of motor movements gated by basal ganglia circuits.51

Page 17: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

7

1.1.1.2 D2-like dopamine receptors

The D2-like family of dopamine receptors includes dopamine D2, D3 and D4 receptors.

Originally identified as those receptors that were not coupled to AC activation and cAMP

production26

, they have since been found to play complex roles in various intracellular signaling,

cognitive and behavioral processes. When the dopamine D2, D3 and D4 receptors were cloned in

the early 1990s, it emerged that inhibition of AC was a general property of the D2-like

receptors, although the degree to which AC is inhibited varies by receptor subtype.25

This

property of D2-like dopamine receptors is mediated by coupling to the Gαi/o GPCR subunit,

which inhibits AC function.32,52

Dopamine D2-type receptors also activate intracellular signaling cascades independently

of G-protein activation. For example, the D2 receptor complexes with the regulatory protein β-

arrestin, protein phosphotase 2A and Akt ( a serine/threonine kinase), and this pathway regulates

the function of Glycogen synthase kinase 3β (GSK-3β).53,54

This signaling pathway typically

takes longer to become active, and stays active for a much longer period of time than the G-

protein mediated pathways. GSK-3β is also involved in signaling pathways activated by other

neurotransmitters, such as serotonin. The D2 receptor involvement in GSK-3β function may

represent a point where signaling from numerous neurotransmitters is integrated.4,54

Furthermore,

β-arrestin plays a role in GPCR desensitization and regulation, and thus, dopaminergic signaling

through D2-like receptors may also be involved in dopamine receptor desensitization processes.4

Unlike the D1-type dopamine receptors, the D2-like receptor coding genes contain intron

sequences, allowing for the generation of receptor splice variants.43

The most widely studied of

these splice variants are the D2S (short) and D2L (long) dopamine D2 receptors, generated by

alternative splicing of an 87-base pair exon between introns 4 and 5 of the D2 receptor gene.55,56

As a result, the D2L receptor isoform contains a 29-amino acid sequence in the third intracellular

Page 18: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

8

loop that is missing in the D2S receptor isoform.55,56

Interestingly, these isoforms localize

differently within the CNS, with the D2L receptor isoform predominantly located in post-

synaptic targets of dopamine pathways and the D2S isoform located presynaptically, in

dopaminergic neurons.57

D2-like receptors are expressed presynaptically, indicating that they can act as

autoreceptors, providing an important negative feedback mechanism by modulating dopamine

synthesis, neuronal firing rate and dopamine release in response to extracellular dopamine

levels.4,25

The D2S receptor isoform, and not the D2L receptor isoform, of the dopamine D2

receptor is likely at least partially responsible for this autoregulation, as generation of a D2L -/-

transgenic model did not affect the ability of dopaminergic neurons to auto regulate.45,57,58

Dopamine D2 receptors are found in various brain areas, including the striatum, NAc, substantia

nigra, VTA, hypothalamus, cortical areas including the PFC and the hippocampus.39,59,60

D3

dopamine receptors are more limited in their expression than D2s, and are mostly expressed in

the limbic areas.60,61

D4 receptors have the lowest expression of all dopamine receptor subtypes,

but are expressed in the frontal cortex, amygdala, hippocampus, hypothalamus and other brain

areas.62,63

1.1.1.3 Regulation of dopamine receptors

After activation of dopamine receptors and activation of intracellular signaling cascades,

(e.g. cAMP in the case of D1 receptors), the downstream events initiated in the cell occur

regardless of whether dopamine remains bound to its receptor.4 To control these intracellular

signaling events, dopamine receptors are regulated through a number of mechanisms including

G-protein regulatory proteins (RGP family), phosphorylation of intracellular loops, receptor

sensitization and desensitization to agonist binding, and receptor internalization.5,25,32

The

Page 19: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

9

D1-type and D2-type Dopamine receptors act on adenyl cyclase (AC) in opposing ways. D1-type receptors activate

AC via coupling with Gs/olf, while D2-type dopamine receptors inhibit AC via coupling with Gi/o G-proteins.

All dopamine receptors contain 7 trans-membrane domains, an extracellular N-terminus and intracellular C-terminal

tail. D2-like receptors have shorter C-terminal cytosolic tails and a larger third intracellular loops. Receptor

function is modulated in part by phosphorylation sites on intracellular loops and C-terminal tails, which can mediate

receptor desensitization and endocytosis. Figure Prepared with help from S.Chen, Liu Lab (2011).

Figure 1-1 Dopamine receptors: structure and function

Page 20: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

10

regulators of G-protein family (RGP) typically increase the rate of G-protein GTP hydrolysis,

decreasing the amount of time the proteins spend active, modulating the efficacy of G-protein

mediated signaling.64

For example, activation of protein kinase A by cAMP elevations in

response to D1-type activation will phosphorylate residues on the C-terminal tail of D1

receptors, and initiate the recruitment of adaptor proteins (typically, β-arrestins) that prevent

further G-protein activation.65

Arrestins can also recruit proteins such as clatherin and β-adaptin

that mediate receptor endocytosis, which can reduce signaling in response to high extracellular

levels of dopamine.66

Dopamine receptors are also regulated by their interactions with other

proteins and other transmembrane receptors (see Section 1.1.3), which can change the receptor’s

affinity for agonists and the intracellular pathways activated by each component receptor.5

1.1.2 Dopaminergic pathways in the mammalian CNS

Although neurons producing dopamine are relatively few in number in the brain, they

project extensively to numerous cortical and subcortical structures. There are four main

dopaminergic pathways in the CNS: the nigrostriatal, mesocortical, mesolimbic and

tuberoinfundibular.25

Each pathway plays an important role in the functions of its target areas

and creates a complex system of dopamine-modulated circuits within the brain. The functions of

the mesolimibic (Section 1.1.2.2) and mesocortical (Section 1.1.2.3) dopaminergic pathways

will be the focus of this review, as these are the most relevant to this project and to the

pathogenesis of MDD. The role of the nigrostriatal pathway in motor behavior will be briefly

discussed (Section 1.1.2.1). The tuberoinfundibular pathway, in which dopamine functions as a

neuroendocrine hormone to inhibit prolactin secretion from the anterior pituitary16

, will not be

reviewed here.

Page 21: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

11

1.1.2.1 The nigrostriatal dopamine pathway

The nigrostriatal pathway originates in the SNpc and projects to the striatum.16,67

The

striatum plays a major role in the gating of motor movements, and the vast majority of the

neurons originating from the striatum are GABA-ergic (inhibitory) Medium Spiny Neurons

(MSNs).16

Striatal neuronal firing activity has opposing functions: depending on the area that the

neurons project to, it can control both the direct (favoring movement) and indirect (favoring no

movement) basal ganglia circuits.68

D1, D2 and D3 receptors are all involved in the effects of dopamine on motor

movements.4 When dopamine D1 receptors on MSNs are activated and converge with cortical

premotor inputs, the firing of MSN projections “disinhibits” the thalamus, favoring the

behavioral occurrence of that movement.25,51

The role of D2 and D3 receptors in the gating of

locomotor activity is more complex than that of D1 receptors, as they function both as

presynaptic auto-receptors (decrease dopamine release when activated) and post-synaptic

receptors.4,25

In PD, the dopaminergic neurons originating in the SNpc are gradually lost,

gradually reducing dopaminergic input to the striatum, resulting in the symptoms of stiffness and

reduced movement in PD.16,19

1.1.2.2 The mesolimbic dopamine pathway

The mesolimbic dopamine pathway originates in a second brain area containing

dopaminergic neurons, the VTA.69

This pathway projects predominantly the NAc (also known as

the ventral striatum).70

The NAc is highly interconnected with other limbic areas including the

amygdala, cingulate cortex, parahippocampal gyrus, hippocampal formation, anterior thalamic

nuclei and NAc.16

The amygdala, the area most involved with fear and emotional experience,

Page 22: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

12

sends neuronal projections to the hypothalamus, which, among other functions, is thought to

regulate the physiological and endocrine changes associated with emotional states.16

Animal models of disrupted mesolimbic dopaminergic input to the limbic areas and

amygdala have demonstrated that dopamine transmission in the amygdala is associated with the

acquisition of Pavlovian-conditioned fear responses.71

Both D1-type and D2-type dopamine

receptors seem to play a role in fear conditioning. Briefly, D1/D5 antagonists diminish

conditioned fear responses69

, and D1 agonists potentiate them72

, indicating that dopamine

signaling via D1 receptors potentiates fear responses in the amygdala. Paradoxically, both D2-

type agonists and antagonists, impair fear conditioning and recall of emotional memory.71

Dopaminergic input to the NAc is highly involved in behavioral reinforcement

mechanisms and reward-dependent learning.16

Most addictive recreational drugs such as

amphetamine, cocaine and nicotine, along with naturally rewarding experiences such as food and

social interactions increase the levels of dopamine in the NAc at the terminals of dopaminergic

projections originating from the VTA.73

Evidence from rodents, non-human primates and human

neuroimaging studies strongly suggest that the mesolimbic system is involved in cue association

to positive rewards, and the reinforcement of behaviors that result in acquisition of a reward.74

The increased levels of dopamine in the NAc are thought to contribute to the strong

reinforcing and addictive properties of recreational drugs and other substances.74-76

Di Chiara et

al75

found that drugs with aversive effects reduced dopamine release in the NAc, implying that

dopamine release is correlated with hedonistic, reinforcing events. In fact, many recreational

drugs enhance dopamine neurotransmission, either by blocking DAT (cocaine and

amphetamine), enhancing release of dopamine through pre-synaptic modulation (nicotine) or

inhibiting inhibitory, GABA-ergic neurons that suppress dopaminergic neurons in the VTA (mu-

opioid agonists).77

Due to the involvement of the dopaminergic system in the CNS response to

Page 23: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

13

recreational drugs and other hedonistic experiences, dopaminergic system dysfunction is thought

to play a major role in the pathophysiological changes that accompany addiction.23,24

The reward circuit modulated by dopamine neurotransmission has also been implicated in

the pathogenesis of MDD and other psychiatric conditions. This is not surprising, given the

symptoms of anhedonia, lack of motivation present in MDD and the role of the mesolimibic

pathway in reward-based learning, motivation and emotional processing.22,78

Dopamine signaling

in the VTA – NAc mesolimbic reward circuit modulates motivation for rewards and pleasure,

implying that these common symptoms of MDD could be due to pathological changes in this

circuit.79,80

Interestingly, dopamine does not, as was initially suggested, code for “pleasure” in the

mesolimibic reward pathway.77,81

Studies in mice missing tyrosine hydroxylase (the rate-limiting

enzyme in dopamine synthesis) show that these mice still have hedonic preferences, preferring

sweetened water over unsweetened.77,82

At the same time, dopamine-deficient mice did not seek

rewards during reward-directed tasks, that is, although they enjoyed the reward, they did not seek

it out.83

These studies support the hypothesis that dopamine in the NAc is required for “wanting”

a reward, but not for “liking” it, that is, dopamine signaling encodes incentive salience, leading

to the modification of behavior in order to obtain the reward.77,84,85

The large number of studies

investigating mesolimbic dopaminergic signaling strongly suggest that dopamine is involved in

motivation, reward-based learning and emotional processing in the limbic areas, and that

dysfunction within the mesolimbic system could lead to addiction, MDD and other psychiatric

illnesses.

Page 24: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

14

1.1.2.3 The mesocortical dopamine pathway

The mesocortical dopamine pathway consists of dopaminergic neurons originating in the

VTA and projecting to the PFC, insular cortex and cingulate cortex.86

The PFC is highly

involved in higher-order cognitive processing including motivation, planning, attention to salient

stimuli, decision making, behavioral flexibility, and working memory. Working memory is

conceptualized as the manipulation of a number of items in short-term memory storage in order

to effectively plan and organize future thought or actions.86

Early findings by Brozoski et al87

demonstrated that depletion of dopamine in the PFC of monkeys produced cognitive and

working memory deficits comparable to those observed when the frontal lobes were completely

removed. Subsequent research into the role of dopaminergic neurotransmission in working

memory indicated that an optimal range of dopaminergic signaling in the PFC existed, where

“too little” or “too much” signaling through D1 receptors increased errors in the radial arm maze

and other working memory tests mediated by the PFC.50,88,89

The PFC also modulates behavioral flexibility, or the ability to alter behavior in response

to changing environmental conditions. A common test for behavioral flexibility and set-shifting

is the Wisconsin Card Sort Task, which requires the human or animal to disregard a previously

beneficial strategy (e.g. sort cards by shape) and engage in a novel one (e.g. sort cards by color)

to obtain a reward.50,86

Patients with damage to the dorsolateral PFC are unable to alter their

sorting strategy when they are required to organize cards by another dimension, a finding

replicated in non-human primate90,91

and rodent92-94

versions of dimensional set-shifting tasks

and reversal learning (where the animal must discriminate between two or more stimuli, only one

of which is relevant to reinforcement) tasks.50,86

In microdialysis studies that measured PFC

dopamine levels in freely-behaving rats during a set-shifting task, dopamine levels in the PFC

Page 25: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

15

increased when the rat had to shift to a different rule in conflict with the first, indicating a role

for DA signaling in behavioral flexibility.50,95

Both attention deficit hyperactivity disorder (ADHD) and schizophrenia patients show

marked impairments in set-shifting, 96,97

and both disorders are associated with various changes

in the mesocortical dopaminergic pathway.98

Pharmacological treatments for ADHD such as

methylphenidate, which increases mesocortical dopamine transmission, are able to decrease

impairments in set shifting seen in patients.95

These clinical findings suggest that dopaminergic

systems are involved in modulating behavioral flexibility, but the exact mechanisms through

which this modulation occurs remain unknown.

The PFC is also very important in decision making processes, specifically when weighing

the advantages and disadvantages of a given choice. Bechara et al99

demonstrated that patients

with damage to the ventromedial PFC were impaired on behavioral tasks designed to simulate

real-life decisions, and the uncertainty and rewards involved.98

In rodents, one can model this

cost/benefit decision making by manipulating the cost (i.e. increasing the delay to reward

delivery, increasing the amount of physical activity required, or making reward delivery

probabilistic) of a reward (typically more, or better, food).98

These different forms of cost/benefit

decision making are regulated by anatomically-distinct regions of the PFC, and all are sensitive

to manipulations in dopamine PFC levels.100,101

Both D1-type and D2-type dopamine receptors

seems to be implicated in cost/benefit decision making paradigms, but they seem to play a

complex role, with their specific function dependent on PFC area and the type of cost/benefit

decision being made.50,102,103

Dopaminergic signaling in the PFC occurs through both D1-type and D2-type dopamine

receptors. In both rodent and monkey PFC, the distribution of D1 receptor messenger RNA

(mRNA) is significantly greater than the other dopamine receptor subtypes.104

Both D1 and D2

Page 26: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

16

receptors are found on excitatory, glutamatergic pyramidal neurons and non-pyramidal, GABA-

ergic interneurons in the PFC.86,105,106

In fact, a subset of layer V pyramidal neurons

(approximately 25 %)107

as well as non-pyramidal PFC neurons express both D1 and D2

receptors, indicating that these receptors may co-localize within these cells.8,105,106

Beyond

cellular and sub-cellular expression patterns of dopamine receptors in the CNS, little is currently

understood regarding how intracellular signaling pathways activated by dopamine receptors

eventually modulate the higher-order cognitive processes mediated by the PFC.

1.1.3 Heterodimerization of Dopamine Receptors

After the cloning of the five distinct dopamine receptor subtypes in the early 1990s,

structural, pharmacological and biochemical studies suggested that each receptor had unique

properties, although they fell into the two previously described families of dopamine receptors

(D1-like and D2-like).4 Over the last 20 years, it has become apparent that dopamine receptors

function both as independent entities and form heterodimers with members of the same family

and with structurally divergent families of receptors.5 The pharmacological and functional

profiles of dopamine receptor heterodimers are often very different from that of the component

receptors and these are thought to contribute to the numerous heterogeneous functions of

dopaminergic signaling in the CNS.5,6

Dopamine receptors have been shown to form

heterodimers through direct protein-protein interactions between D1 and D2 receptors,3,108-110

D1 and D3 receptors in the striatum111,112

, D2 and D5 receptors109,113

, D1 receptors and NMDA

receptors (NMDARs) 30,114

and D5 and GABA-A receptors115

, among other transmembrane and

cytoplasmic proteins.

All dopamine receptors subtypes form non-obligatory heterodimers, that is, dimerization

is not necessary in order for the receptor to function.4 However, a large degree of complexity in

Page 27: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

17

the signaling effects of dopamine receptors results from their ability to heterodimerize.5 For

example, dopamine receptor heteromers could create novel ligand binding sites, activation of one

or both component receptors could initiate different intracellular signaling pathways than those

initiated by the component receptors, or a synergistic increase in signaling could occur when

both agonists are present.5 The D1-D2 receptor interaction and the activation of independent

intracellular signaling pathways that occurs will be the focus of this section, as this interaction is

implicated in the pathogenesis of MDD3 and is the target of the D1-D2 interfering peptide used

in this project.

1.1.3.1 The Dopamine D1-D2 Receptor Interaction

An interaction between the dopamine D1 and D2 receptors was first investigated because

of the observation that a D1-like receptor could activate Inisitol Phosphate 3 (IP3) production

(leading to increases in intracellular calcium) in various brain regions including striatum,

hippocampus and cortex.116,117

An interaction between dopamine D1 and D2 receptor was

proposed because of the observations that the presence of calcium signaling activated by D1 was

absent in D1-transfected cells, and present in cells transfected with both D1 and D2 receptors.2

Research from our laboratory identified the specific regions through which D1 and D2 receptors

interact as a 15 amino acid sequence within the 30-amino acid insert in the third intracellular

loop of the D2L receptor isoform, and the D1 intracellular C-terminal tail.3 This finding provided

indirect evidence that the D1-D2 receptor interaction occurs in post-synaptic membranes, as both

D1 and D2L dopamine receptors are generally localized to post-synaptic areas.25,57

Page 28: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

18

Figure 1-2 Schematic representation of D1-D2R receptor interaction and activation of

intracellular signaling pathways.

(A) Activation of D1 or D2 when those receptors are in complex is thought to activate PLC, resulting

in release of calcium from the endoplasmic reticulum and subsequent activation of CamKII.1,2

(B) The

D1-D2 interfering peptide (TAT-D1-D2-IPep) disrupts the interaction between D1 and D2L receptors,

resulting in disruption in the Gq-mediated downstream signaling pathways.3 (A) and (B) prepared with

help from S.Chen, Liu Lab.

Page 29: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

19

Research by George and colleagues2,118

confirmed the interaction between D1 and D2

using co-immunoprecipitation and through fluorescence resonance energy transfer (FRET)

techniques. They also demonstrated that the D1-D2 receptor complex induces intracellular

calcium via a Gq GPCR-dependent pathway in the striatum.119

In the Gq-pathway, Phospholipase

C (PLC) becomes activated by Gq, resulting in an increase in inisitol triphosphate (IP3) which

then causes activation of downstream molecules resulting in increased calcium concentration in

the cytoplasm (Figure 1-2A).32,119

The colocalization of D1 and D2 receptors seems to occur in a

number of brain regions including the dorsal and ventral striatum, and the PFC.105-108

Although the intracellular pathway activated by the D1-D2 receptor heterodimer is

characterized, the physiological relevance of this interaction and its role in neurological and

psychiatric illnesses is not yet clear. Recently, our laboratory demonstrated that the D1-D2

interaction may have a role in the pathogenesis of MDD. Most importantly, the D1-D2 receptor

interaction was up-regulated in post-mortem samples from the striatum of patients with MDD,

implying that this interaction may be disrupted in this illness.3 Uncoupling the D1-D2 receptor

interaction using a small, membrane permeable peptide (TAT-D1-D2-IPep) results in an

antidepressant effect in animal model of depression (Figure 1-2B, Figure 1-3).3 This study

suggests that the D1-D2 receptor interaction may play a role in the pathogenesis of MDD, and

warrants further investigation into possible therapies based on disrupting this interaction.

1.2 Major Depressive Disorder

MDD is the most common psychiatric illness in the world, with the lifetime incidence in

the United States 12% in men and 20% in women.120

Despite its prevalence, many patients who

have MDD are not adequately treated with current antidepressant therapies. Since the discovery

of the first antidepressant compounds over 50 years ago, much progress has been made

Page 30: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

20

investigating the neurobiological changes underlying MDD.121

Although numerous hypotheses

attempt to explain the neurobiological and pathological changes underlying the clinical

presentation of MDD, no unitary hypothesis explaining all the pathological changes and the

complex symptoms observed in MDD exists. Here, the epidemiology (Section 1.2.1) and

symptoms of MDD (Section 1.2.2) will be reviewed, along with currently available treatments

and their efficacy in treating MDD (Section 1.2.3). Next, a number of different hypotheses

regarding the pathophysiology underlying MDD will be explored (Section 1.2.4). Since the

treatment in our current investigation targets a protein-protein interaction between D1 and D2

dopamine receptors, the evidence for the involvement of the dopaminergic system in MDD will

be reviewed (Section 1.2.4.2). To conclude, the use of preclinical models to model MDD and

test new antidepressant treatments will be discussed (Section 1.2.5).

1.2.1 Epidemiology of MDD

Depression is a common psychiatric illness characterized by 2 or more weeks of a distinct

change in mood, sadness and/or constant irritability, as well as feelings of hopelessness and loss

of interest in pleasurable activities.122

The lifetime incidence of depression in the United States is

12% in men and 20% in women120

. In fact, women are 70% more likely than men to experience

depression in their lifetimes.123,124

Although depression is closely related to the normal emotions

of sadness, it often does not regress when the external cause of these emotions dissipates, and

can be disproportionate to their cause.78

Often, episodes of depression will recur two or more

times and become classified as MDD.122

At its most severe, MDD can lead to suicide attempts,

which can result in loss of life or significant disability.

MDD is responsible for 4.4 % of the worldwide disease burden and, is the leading cause

of disability worldwide when considering total years lost to disability.125

Depression often goes

Page 31: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

21

undiagnosed and untreated because of the prevalent societal stigma associated with psychiatric

conditions and seeking treatment for these conditions. According to statistics from the National

Institute of Mental Health, only 57% of patients with MDD in the United States are receiving

any kind of treatment for the disorder, and only 19% of patients with MDD are receiving

adequate treatment.120,126

Additionally, MDD can often occur in conjunction with other serious illnesses such as

cancer, chronic pain, epilepsy and cardiovascular disease.9,120,127-129

When this is the case, both

MDD and the co-morbid illness are adversely affected, as treatment outcomes in patients who

have diabetes, epilepsy or ischemic heart disease along with MDD have poorer outcomes than

those without MDD.130

Overwhelmingly, epidemiological data regarding the prevalence of MDD

indicates that it is extremely common, often undiagnosed and, in the majority of cases, not

adequately treated.

1.2.2 Symptoms and clinical presentation of MDD

The Diagnostics and Statistical Manual of Mental Disorders IV (DSM IV-TR)122

criteria

for MDD are the most commonly used criteria for MDD diagnosis. A single depressive episode

is categorized by the presence of five or more of the following symptoms during a two-week

period where at least one of the symptoms is either depressed mood most of the day, nearly every

day, or loss of interest or pleasure in almost all activities.122,131

Recurrent MDD occurs when a

patient experiences two or more major depressive episodes, with a symptom-free period of two

or more months separating them. Other symptoms include a change of more than 5% in body

weight over the course of 1 month, insomnia or hypersomnia, psychomotor agitation or

retardation, persistent fatigue and loss of energy, feelings of worthlessness or guilt, diminished

ability to think or concentrate, indecisiveness, and/or recurrent thoughts of death or

Page 32: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

22

suicide.8,122,131

These symptoms can cause significant impairment in the patient’s social,

occupation and other functioning. MDD and its symptoms are often variable in both clinical

presentation and severity, which may contribute to the large number of patients left undiagnosed

and untreated.120

1.2.3 Treatments for MDD

Patients diagnosed with MDD are typically treated with pharmaceutical agents that

increase the amount of monoaminergic neurotransmitters at the synapse. Currently, the “first-

line” pharmaceutical therapy for MDD is a class of drugs termed selective serotonin re-uptake

inhibitors (SSRIs).78

When given to patients, these drugs produce an increase in the

neurotransmitter serotonin in the brain by inhibiting its re-uptake into the presynpatic neurons

from where it was released.9 In the CNS, serotonergic neurons project to numerous cortical and

subcortical areas from the brainstem raphe nuclei and are involved in regulation of mood,

appetite, sleeping behavior, learning and memory.78

Serotonin act through serotonin receptors, of

which there are seven families that have diverse effects in cells.16

SSRIs quickly increases the total amount of serotonin available at serotonergic synapses

and, as such, increase the amount of serotonin-mediated neurotransmission in the brain.132

The

side-effects of SSRIs are often apparent almost immediately after the initiation of treatment,

while any therapeutic antidepressant effect from these medications takes approximately three

weeks to become apparent in patients.78

The delay in onset of any therapeutic effect suggests that

the ability to increase serotonin at the synapse may not be the only mechanism through which

SSRIs have an antidepressant effect and that they may be mediating other, longer-term effects in

the CNS responsible for its therapeutic efficacy.8,133

Page 33: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

23

In a large clinical trial for antidepressant efficacy, the Sequenced Treatment Alternatives

to Relieve Depression (STAR*D) trial, it was shown that only 27- 33 % of patients with MDD

adequately responded to first line treatment with citalopram, an SSRI medication.134

If, after a

number of weeks of citalopram or other SSRI treatment, little or no improvement on a

standardized rating scale such as the Hamilton Depression Rating Scale135

, is observed, patients

can be treated by increasing the SSRI dose (if side effects are tolerable), switched to a new SSRI,

or started on another antidepressant along with the original SSRI.9 Studies have also indicated

that SSRI efficacy is correlated with the severity of depression when treatment is initiated,

implying that SSRIs are more effective for patients with severe depression and may not be

significantly more effective than placebo for patients with mild or moderate depression.136,137

For patients who do not respond to SSRIs, pharmaceutical treatment alternatives include

SNRIs (Selective norepinephrine reuptake inhibitors), Triple reuptake inhibitors (inhibit re-

uptake of serotonin, dopamine and norepinephrine)138

, tricyclic antidepressants such as

imipramine, and monoamine oxidase inhibitors (MAOIs). MAOIs exert their antidepressant

effect by blocking the breakdown of monoaminergic neurotransmitters (serotonin,

norepinephrine and dopamine) and are effective in the treatment of MDD, but also have strong

and often intolerable side effects.139,140

A final, invasive option in the case of severe, unremitting

MDD is electro-convulsive therapy, which remains the most effective treatment for severe,

unremitting depression.139,141

1.2.3.1 Efficacy of current antidepressant treatments: The STAR*D trial

The STAR*D trial134

is a large clinical trial designed to investigate remission rates after

antidepressant treatment in a large and generalizable sample of patients. In the first level of the

trail, all patients enrolled were treated with citalopram (an SSRI) as a first-line treatment, with

Page 34: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

24

their depressive symptoms evaluated every 2 weeks after initiation of treatment.134,142

Between

28 and 33 % of patients treated with citalopram achieved remission within 12 to 14 weeks of

treatment.134

In Level 2 of the STAR*D trial, those patients who did not respond to citalopram

treatment after 14 weeks were given the choice between pharmacotherapy augmentation,

psychotherapy or switching to a different SSRI medication for 12 weeks.14

Of the patients in

Level 2, approximately 30% achieved remission of symptoms within 12 weeks, with no

significant differences in remission rates with any of the treatment strategies employed.14,142

In

Levels 3 and 4 of the trial, patients who had not responded to antidepressant treatments or

psychotherapy in Level 1 or 2 were treated with alternative pharmacotherapies, including

tricyclic antidepressants. In Level 3 and 4 of the STAR*D trial, the remission rats dropped

substantially, ranging from 12 % to 25%, depending on the treatment used.142

In all, only 67% of

patients originally enrolled in the STAR*D achieved remission of their depressive symptoms.142

The STAR*D trial reveals that almost one third of patients with unipolar depression do

not respond to multiple trials of SSRI and other antidepressant treatments. After the first two

levels of the trial, patients were much less likely to respond to further pharmaceutical treatment

trials, indicating the importance of achieving a treatment response with the first few

antidepressants prescribed to patients.142

The STAR*D trial also provides a strong rationale for

further investigation into new antidepressant therapies that could help treat MDD in the subset of

patients who currently do not respond to available antidepressant treatments.

Page 35: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

25

1.2.4 Neurobiological changes and pathophysiology of MDD

Currently, there is no unitary hypothesis that explains the various pathological and

neurobiological changes that occur in MDD. What remains clear, however, is that MDD is a

heterogeneous disorder with complex pathological mechanisms that vary considerably between

individuals affected by the disease. In human neuroimaging studies, the brain regions that are

consistently found to be involved in MDD are the PFC, the cingulate cortex (area Cg25), the

hippocampus and the amygdala.79,143,144

These findings are consistent at both the structural level,

where magnetic resonance imaging (MRI) data and other neuroimaging data suggests decreased

hippocampal and PFC volume in depressed patients, and the functional level, where functional

MRI (fMRI) and positron emission tomography studies suggest abnormal connectivity and

decreased functionality of limbic, cingulate and prefrontal areas in depression.143

The monoamine and catecholamine neurotransmitters, serotonin, dopamine and

norepinephrine innervate these areas through extensive axonal projections from the dorsal raphe

nucleus (serotonin), VTA (dopamine) and locus coeruleus (norepinephrine).16,73

Disruptions in

these systems are thought to contribute to the pathogenesis of MDD (Section 1.2.4.1). More

recent investigations into the neurobiological mechanisms behind MDD have also demonstrated

that neurotrophic factors such as Brain Derived Neurotrophic Factor (BDNF) also play a large

role in MDD (Section 1.2.4.2). The evidence implicating dopamine in the pathogenesis of MDD

will be reviewed, as the D1-D2 interfering peptide used in this project specifically targets the

heterodimerization of two dopamine receptors (Section 1.2.4.3). Although these theories of

MDD all attempt to explain the complex symptoms of the disease, it is currently unclear how

neurobiological, genetic, societal and environmental factors result in the complex and variable

clinical presentation of MDD.

Page 36: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

26

1.2.4.1 The Monoaminergic deficiency hypothesis of depression

The most widely accepted hypothesis of the neurobiological basis of depression is the

monoamine deficiency hypothesis. This hypothesis states that depressive symptoms occur due to

a decrease in the amount of monoaminergic neurotransmitters in brain areas implicated in MDD

such as the prefrontal and limbic cortex.78,138

The hypothesis was first proposed in the 1960s

because of the antidepressant actions of two structurally unrelated compounds (tricyclic

compounds and MAOIs). Both tricyclic compounds and MAOIs were found to increase overall

levels of monoamines in the brain, thereby increasing mood in patients being treated with

them.145

The early MAOI – type antidepressants inhibited MAOs, enzymes that break down

serotonin, dopamine or norepinephrine in the presynaptic neuron, rendering these

neurotransmitters inactive.78

Due to their action on MAOs, MAOIs increase the available stores

of monoamines in the CNS, which is the proposed mechanism for their antidepressant effect and

the basis of the monoaminergic hypothesis of MDD.

There is extensive clinical and pre-clinical evidence that suggests that serotonin,

dopamine and norepinephrine are highly involved in the neurobiology of MDD, and that a

sustained deficiency in any one could result in depression.78,138

The clinical efficacy of SSRIs,

SNRIs, and triple reuptake inhibitors, which all increase the availability of monoaminergic

neurotransmitters in the synapse, supports the monoaminergic hypothesis of MDD

pathophysiology. Although these drugs all produce immediate, substantial increases in serotonin

and other monoamines in the brain any antidepressant effect takes a number of weeks to become

apparent.9,134

Thus. it seems probable that the longer-term, antidepressant effects of SSRIs and

other antidepressants are due to adaptive responses in the brain secondary to the effects of

increasing monoaminergic neurotransmission.133

Furthermore, most antidepressant therapies

Page 37: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

27

currently in use are based on increasing monoamine levels, and are only effective in

approximately 50% of patients with MDD.121

It is probable that a more complex pathological mechanism underlies MDD than the

monoaminergic deficiency hypothesis. Human dietary studies suggest that depleting tryptophan

stores (rate-limiting for synthesis of serotonin in the brain) or depleting TH (required for

catecholamine synthesis) does not cause depressive symptoms in healthy subjects, but can cause

relapse in patients previously treated for MDD.146

Furthermore, post-mortem studies on human

brain tissue from patients with MDD have not consistently shown decreases in brain monoamine

levels.79,147,148

These findings suggest that monoamines and catecholamine neurotransmitter

levels play an important role in MDD, but depleting these neurotransmitters alone may not be

sufficient to cause MDD.78

Thus, the monoaminergic deficiency hypothesis of MDD may be too

simplistic to explain the pathological changes and clinical presentation of MDD.

1.2.4.2 Role of dopamine in the pathophysiology of MDD

The majority of theories regarding the neurobiology of MDD focus on the role of

disruptions in serotonin and norepinephrine neurotransmitter systems. Disruptions in brain

dopamine levels, as well as changes in dopaminergic neurotransmission have also been identified

as factors contributing to in the neurobiology of MDD, and are the target of a number of

clinically effective antidepressants.8 For example, nomifensine and bupropion block the reuptake

of norepinephrine and dopamine and are both effective antidepressants when used alone or in

conjunction with other antidepressant treatments.149

The involvement of dopamine in MDD is

also supported by clinical studies, as the turnover rate of dopamine, as measured by CSF or

plasma levels of dopamine metabolite homovanillic acid, is decreased in patients with MDD

compared with controls.22,150

Page 38: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

28

Many of the common clinical symptoms of MDD, such as anhedonia, loss of ability to

concentrate, flattened affect and motor changes are present in disorders in which dopaminergic

signaling is disrupted, such as PD and schizophrenia.151

Cognitive and behavioral functions

modulated by the mesolimibic and mesocortical pathways such as emotional processing,

planning, motivation and other executive functions, can be impaired in patients with MDD,

implying that dopaminergic signaling in areas such as the cingulate, prefrontal and limbic

cortices may be disrupted in MDD.80,152

Preclinical models of depression have also been used to study the role of dopamine in

MDD. In the LH model of depression, dopamine levels are reduced in the caudate nucleus and

the NAc of animals with MDD, and depressive-like behavior can be prevented by treatment with

a dopamine agonist prior to the behavioral task.22,153,154

In the FST, a commonly used test for

antidepressant efficacy, dopamine agonists and DAT blockers tend to increase mobility,

indicating that these substances have antidepressant-like effects.22,155

Furthermore, dopamine D1

and D2 receptor antagonists can inhibit the effects of antidepressants in the chronic

unconditioned stress model of MDD.156

It is apparent that dopamine is implicated in the

pathology of MDD, but given the complexities of dopaminergic signaling and its role in

prefrontal and limbic processes, we do not have a complete understanding of these mechanisms.

Our laboratory has recently shown that heterodimerization of the dopamine D1 and D2 receptors

is up-regulated in patients with MDD, and disrupting this interaction has an antidepressant effect

in preclinical models of depression (See Figure 1-3 for a summary of these findings) 3,8

Despite

these promising findings, the mechanism by which the D1-D2 receptor interaction is involved in

the neurobiology of depression is unclear. Overall, it is clear that dopamine and dopaminergic

signaling is disrupted in MDD, and may be involved in the pathogenesis of the disorder,

Page 39: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

29

Figure 1-3 Previous findings demonstrating the role of the D1-D2R interaction in MDD.

(A) The D1-D2 interaction is significantly increased in post-mortem samples from patients with MDD as assessed

by Co-immunoprecipitation of D1 by anti-D2R. (B) A 15-maino acid, membrane permeable peptide capable of

disrupting the interaction between D1-D2LR (TAT-D2-Il3-29-2) has an anti-immobility effect in the FST when

infused directly into the PFC. (C) Co-imunoprecipitation of D1 by anti-D2R is significantly decreased in the PFC

after infusion with the D1-D2 Interfering peptide (TAT-D2-Il3-29-2). Figures prepared by Pei et al. (2010) and used by

F.Liu in conference presentations. This data was also published, in different figures, in Nature Medicine 16, 1393-

1395 (2010).3

Page 40: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

30

although we currently do not have a complete understanding, or a unitary hypothesis, to explain

dopamine’s role in this complex illness.

1.2.4.3 The Neurotrophic Hypothesis of Depression

Neurotrophic factors such as Brain Derived Neurotrophic Factor (BDNF) are expressed

in the brain and act through transmembrane receptors (tropomyosin receptor kinase B receptors

(TrkB)) to promote neuronal survival.157

The neurotrophic hypothesis of MDD postulates that in

MDD, expression of neurotrophic factors such as BDNF is altered in the brain. These alterations

in BDNF expression may be caused by chronic stress often associated with MDD78

, and result in

decreases in BDNF, which may account for its expression in brain areas involved in MDD

pathogenesis.157,158

This hypothesis is supported by the observed decrease in adult neurogenesis

markers in the hippocampus and the PFC of patients with MDD who had committed suicide.159-

161 Antidepressants may cause long-term changes in BDNF expression as post-mortem and

serum BDNF levels increased in the hippocampus and cortex after long-term antidepressant use

compared with patients not taking antidepressants.162

Rodent models of MDD have provided contradictory evidence about the role of BDNF

and other neurotrophic factors in MDD. Infusing BDNF into the hippocampus and surrounding

brain areas has an antidepressant effect in rodent models of depression.163,164

Diverse

antidepressant pharmacological agents increase signaling pathways activated by the BDNF

membrane receptor, TrkB, in a BDNF-mediated manner.165,166

However, male mice who were

knockouts for the BDNF membrane receptor, TrkB, did not exhibit endogenous depressive

behavior in a number of preclinical tests for depression and anxiety,167

and hippocampal BDNF

infusions in male rats did not prevent learned helplessness behaviors.168

Interestingly, BDNF

infusion directly into the VTA and NAc, two areas highly involved in the mesolimibic and

Page 41: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

31

mesocortical dopamine pathways, had a pro-depression effect and enhanced social aversion

behaviors in mice.169,170

Since BDNF works to promote neuronal survival (among many other

functions) in areas where it is expressed, it is likely that its role in the neurobiology of MDD

depends on the brain area where it is elevated, and its underlying functions. As such, considering

it as an “antidepressant” in the classical sense oversimplifies its role in the neurobiology of

MDD.

1.2.5 Preclinical models of MDD

Clinical studies investigating the pathological mechanisms in MDD can only provide

correlative, and not causative, clues into this complex disorder. New pharmacological treatment

options for MDD must be validated using preclinical models of MDD to ensure their efficacy

and safety before being tested in the clinical setting. However, accurately modeling a complex,

multi-faceted disease like MDD with unknown neurobiological mechanisms in a laboratory

preclinical animal model is a challenging task. As such, the development of reliable animal

models that model one symptom of MDD (versus attempting to model the spectrum of human

MDD symptoms) has been more useful in the laboratory setting.171

A number of criteria for the validity of animal models of MDD have been proposed.

These include strong predictive validity (i.e. all antidepressants that are clinically effective

produce a similar ‘antidepressant-like’ effect in the behavioral model), that the behavioral output

of the model is reliable within and between laboratories and that a similarity between the

behavioral output in animals and clinical symptoms of depression is present.171-173

In general,

animals must be exposed to some type of stress, either acutely or over a sustained period, in

order to produce depressive-like symptoms.171

In animals, chronic stress can result in

helplessness (inability and unwillingness to escape from a stressful situation), anhedonia (lack of

Page 42: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

32

interest in otherwise pleasurable activities), or social aversion (avoidance of other animals).79,171

These behaviors resemble specific human symptoms of depression, albeit in a simplified manner.

Although a number of well-validated models of depression exist including social aversion tests,

and chronic unconditioned stress paradigms,171,172

only the FST and the LH task will be

discussed here, as the D1-D2 interfering peptide used in this study was shown to have an

antidepressant effect using these models.

The FST is a two-day acute test of antidepressant efficacy developed by Porsolt et al.174-

176 The FST is not considered a chronic model of depression, as it is an acute test used to screen

substances that may act as antidepressants.171

Briefly, the animal is placed in an inescapable

plexiglass cylinder for 15 minutes before being given a treatment intervention. The next day, the

animal is replaced in the cylinder for a 5 minute period, and its behavior is scored. An animal

that didn’t receive an antidepressant treatment will display ‘behavioral despair’ or ‘helplessness’

and will assume a floating, immobility posture for the majority of the 5-minute test.176,177

On the

other hand, an animal that receives pharmacological agent with antidepressant properties will

remain active and try to escape for the majority of the 5-minute test (this is referred to as an anti-

immobility effect).175,176

The FST is a useful test because it has high predictive validity, as all

antidepressants currently used in the clinic have an anti-immobility effect in the FST.176,178

On

the other hand, it is not considered an animal model of MDD because it bears little resemblance

to the etiology and symptoms of MDD in patients.

Another widely used depression model is the LH task, a 5-day test in which the animal is

exposed to inescapable shock. Subsequently, its passive response to subsequent shocks (from

which the animal can escape) is measured.179,180

This model is thought to have clinical relevance

and etiological validity since evidence exists indicating that stressful life events perceived as

uncontrollable (such as death of a loved one and romantic breakups) are major predictors of

Page 43: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

33

MDD onset and severity.180-182

This clinical finding can be replicated in rodents by controlling

the onset and duration of uncontrollable, aversive events, as is done in the LH paradigm.183,184

The LH model also has pharmacological validity, as administration of substances that act as

antidepressants over the 5-day task results in an increase in the animal’s escape attempts

compared to untreated animals.3,180,185-187

Thus, the LH model is a useful model of both

helplessness in human MDD and has pharmacological validity as a test for novel antidepressant

therapies.

1.3 Intranasal delivery to the CNS

The nasal anatomy, both in humans and rodents, contains a number of features that make

it attractive as a delivery pathway for proteins and peptides targeted to the CNS.15,188

A number

of properties of the vasculature in the CNS limit the entry of molecules, ions, pathogens and

toxins into the CSF, and are together called the Blood Brain Barrier (BBB).189

Tight control of

the extracellular environment in the CNS provided by the BBB is required to maintain proper

neuronal function and prevent injury within the CNS. At the same time, the BBB makes it

difficult to effectively deliver therapeutic substances unable to cross it, such as foreign peptides

and proteins, to the CNS and the brain.189

A number of features of the nasal anatomy make

intranasal administration of peptides and proteins an effective way to bypass the BBB and

specifically deliver these substances to the CNS.15,188

A number of protein and peptide therapies have been effectively delivered to the CNS

using the intranasal pathway. For example, in recent years, intranasal administration of insulin

has been shown to slow memory impairments in rodent models of Alzheimer’s disease (AD).190-

192 A recent meta-analysis suggested an overall beneficial effect of intranasal insulin on cognitive

functions in human trials of intranasal insulin delivery in healthy patients, patients with mild

Page 44: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

34

cognitive impairment (MCI) and those with AD, with few detectable side effects and low

systemic levels of insulin.193

Other protein and peptide therapies that have been successfully

delivered to the CNS after intranasal delivery include nerve growth factor (NGF)194,195

other

neurotrophic factors such as BDNF196

, Insulin-like Growth factor 1 (IGF-1)197,198

and numerous

other proteins and drugs (reviewed in Dhuria et al15

).

Furthermore, a TAT-linked membrane permeable peptide similar in size to the D1-D2

interfering peptide was effectively delivered to the CNS using the intranasal approach.196

In fact,

the authors196

found that less than 10% of the intravenous dose administered intranasally resulted

in equivalent brain concentrations of their 22-amino acid TAT-linked peptide. The success of

intranasal insulin delivery to the CNS, along with other proteins and peptides that were delivered

successfully to the CNS after intranasal delivery, suggests that a direct pathway exists between

the nasal olfactory epithelium and the central nervous system.

1.3.1 Mechanisms of intranasal delivery to the CNS

Both the human and the rodent nasal anatomy have several features that make it

conducive to drug transport to the CNS while minimizing systemic exposure to the drug (see

Figure 1-5). The olfactory nerve pathways that connect the olfactory sensing region of the nasal

cavity to the olfactory bulbs and other CNS areas are important for intranasal drug delivery to the

CNS. For example, a fluorescently-labeled 3 kiloDalton (kD) Dextran allowed visualization of

the olfactory pathway after intranasal administration, and demonstrated that the Dextran was

transported to the olfactory bulbs along olfactory nerve pathways in approximately 15

minutes.199

Olfactory receptor neurons (ORNs) are responsible for conveying information about

odors to the CNS.16

ORNs are bipolar cells whose cell bodies are located within the olfactory

Page 45: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

35

epithelium, with chemoreceptor-containing dendrites extending into the nasal mucosal layer and

axons travelling via the cribiform plate and olfactory nerve bundle into the CNS and olfactory

bulb.200

The cribiform plate of the ethmoid bone contains many small perforations that allow

ORN axons to extend into the CNS, effectively bypassing the BBB.15,200

Extracellular channels between the olfactory ensheathing cells (OECs) (which protect

the axonal projections of the ORNs) and the ORN axons allow drugs and peptides administered

intranasally to access the CSF and brain directly.15,188

Intracellular transport mechanisms along

the ORN axons may be important for certain substances201,202

but are not currently thought to be

the predominant mode of transportation into the CNS. This is because most intranasal delivery

studies, particularly of proteins and peptides, have demonstrated rapid transport from the

olfactory epithelium and nasal cavity into the CNS, suggesting that these substances are

transported extracellularly via the channels between OECs and ORNs.203-205

In addition,

intracellular transport of intranasally administered substances requires uptake of the substance

into the ORNs, necessitating receptor-mediated transport mechanisms, or the ability of the

substance to cross the phospholipid bilayer, which cannot account for the large variety of drugs,

proteins and peptides that have successfully been delivered to the CNS using the intranasal

route.15

Other potential transport mechanisms from the nasal cavity to the CNS include transport

via the trigeminal nerve pathways and via vascular pathways. The trigeminal nerve innervates

the respiratory and olfactory epithelium of the nose and enters the CNS in the brainstem.200

It is

possible that substances administered to the nasal cavity are also transported to the CNS via this

pathway, as this has been demonstrated for radioactively-labeled IGF-1 and other proteins and

peptides.203,206,207

Secondly, the nasal passages are highly vascularized structures, and

intranasally administered substances can be absorbed into the bloodstream through the

Page 46: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

36

endothelial cells making up the capillary wall.15

In order for successful delivery to the CNS to

occur after absorption into the systemic circulation, substances must cross the BBB. This

approach is not thought to mediate the transport of proteins and peptides to the CNS after

intranasal delivery, because they lack the ability to cross the BBB in appreciable amounts.15

1.3.2 Experimental considerations for successful intranasal delivery to the

CNS

For successful delivery of proteins and peptides to the CNS using the olfactory pathway,

experimental considerations including intranasal administration technique, head position and

drug formulation must be taken into account. The vast majority of preclinical studies involving

intranasal administration to rodents have administered substances intranasally in anaesthetized

animals. To optimize delivery to the CNS, the substance being administered must reach the

olfactory epithelium and upper third of the nasal cavity, and different head positions can alter

absorption of substances into the bloodstream and CSF.208

In anaesthetized rodents, a number of

studies have demonstrated that intranasal administration targeted to the CNS can be achieved via

the insertion of flexible tubing into the nostrils, localizing delivery to the olfactory epithelium

and surrounding tissue.208-210

The Pressurized Olfactory Device (POD) used in our present study

combines delivery to the olfactory epithelium using flexible tubing and aerosolized delivery to

deliver substances preferentially to the olfactory epithelium and surrounding tissue, favoring

delivery to the CNS.210,211

1.4 Rationale

After the discovery of the antidepressant effects of MAOIs and tricyclic antidepressants

nearly 60 years ago9, there has been extensive investigation into the pathogenesis of MDD in the

Page 47: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

37

human brain and new pharmacological approaches to treating it. Most effective antidepressant

therapies target the monoaminergic (serotonin, norepinephrine and dopamine) neurotransmitter

systems. A disadvantage of these therapies is that they broadly increase monoaminergic

signaling, resulting in a number of unpleasant side effects in patients. These often appear

immediately, a couple of weeks before any appreciable benefit of the antidepressant becomes

apparent.9 As many as 50% of patients do not respond to current antidepressant treatments,

121

necessitating new and more effective therapeutic options for this disorder. New approaches to

treating MDD, and the development of new therapies targeting specific pathological changes

occurring in MDD, are necessary in order to advance the treatment as well as our scientific

understanding of this illness.

Peptides specifically designed to disrupt pathological interactions between

neurotransmitter receptors are promising therapeutics for psychiatric and neurological disorders

because they allow for specific targeting of these pathological interactions.8 They disrupt the

pathological interaction between the two proteins without having an effect on either receptor’s

independent function, minimizing the likelihood of side-effects. Our laboratory has

demonstrated that an interfering peptide specifically designed to disrupt the dopamine D1-D2

receptor heterodimer has a significant antidepressant effect in the FST and the LH task, a

preclinical model of depression, when given directly to the PFC of rats.3 Pei et al

3 also

demonstrated that the interaction between dopamine D1 and D2L receptors is significantly

increased in post-mortem striatal samples from patients with MDD compared with controls. This

finding suggests that the efficacy of the D1-D2 interfering peptide in preclinical models of

depression is relevant to the clinical pathogenesis of this disorder, warranting further

investigation into its pharmacological and biochemical functions.

Page 48: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

38

Although the previous findings from our laboratory are promising, they are not yet

clinically applicable, as the administration methods (direct microinjections to the brain and

intracerebral ventricular (ICV) injections) Pei et al3 used are extremely invasive and not feasible

in the clinical setting. For this interfering peptide, and any other peptide that has a therapeutic

effect in animal models of psychiatric or neurological conditions, to translate to the clinical

setting a non-invasive, clinically applicable method of administration must be tested and

developed.

A major challenge in the development of novel therapies for psychiatric or neurological

disorders is successfully and non-invasively delivering them to the central nervous system,

without substantial accumulation of these therapies in the systemic circulation.15

In the last 20

years, intranasal delivery of peptide and proteins targeted to the CNS has been extensively

studied (for review, see Dhuria et al.)15

This relatively non-invasive approach exploits the

weakened blood-brain barrier at the olfactory epithelium to deliver therapeutic substances such

as peptides and proteins to the central nervous system.192,195,196

Many substances, including TAT-

linked membrane permeable peptides212

, insulin190,192

, IGF-1203

and NGF195

, have been

successfully delivered to the CNS using the intranasal route. The POD used to administer the

D1-D2 interfering peptide intranasally is designed to preferentially deposit substances on the

olfactory epithelium within the nasal cavity, favoring absorption of substances into the CNS.

Studies on intranasal delivery to the CNS have also indicated that the intranasal pathway

preferentially delivers substances to anterior brain regions such as the olfactory bulbs, PFC and

adjacent areas.15

Since many patients suffering from MDD do not respond to current antidepressant

therapies, or cannot tolerate these therapies because of aversive side effects, newer and better

therapeutic options must be investigated. For this novel therapeutic approach targeting the D1-

Page 49: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

39

D2 interaction in MDD to become a relevant treatment, translational studies in rodents must be

carried out investigating non-invasive methods to deliver the D1-D2 interfering peptide to the

CNS.

This project is designed to test whether the D1-D2 interfering peptide can be successfully

delivered to the central nervous system, and the PFC in particular, using the intranasal approach

and whether it will have an antidepressant effect in the FST after intranasal delivery. We will

also investigate the pharmacological properties of the D1-D2 interfering peptide, including the

intranasal doses required to observe an antidepressant effect in the FST and the amount of time it

remains biologically active in the body. Overall, this project will indicate whether the intranasal

pathway is a viable method to deliver peptides like the D1-D2 interfering peptide to the CNS,

and better inform whether the D1-D2 interfering peptide is suitable for further development as a

novel treatment for depression.

1.5 Hypothesis

Based on our laboratory’s previous findings3 that the D1-D2 interfering peptide, when

infused directly into the PFC, has an antidepressant effect in the FST and the LH task,3 we

hypothesize that it will have this same effect after intranasal delivery. In order for this hypothesis

to be correct, a number of criteria must be met.

First, we hypothesize that the POD used in this study preferentially deposits substances

on the olfactory epithelium, favoring uptake into the CNS via olfactory nerve pathways, and that

after intranasal delivery, we will be able to visualize a FLAG-tagged D1-D2 interfering peptide

in the PFC.

Second, the efficiency of delivery of peptides and protein therapies to the CNS is largely

unknown, but some studies have suggested that between 1 and 5% of the dose delivered

Page 50: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

40

intranasally reaches the CNS and anterior brain areas.15

Based on these estimates, we

hypothesize that the D1-D2 interfering peptide will be effective in the FST at a dose 100-fold

larger than that delivered directly to the PFC.

Third, since we currently do not have any information about the efficacy of the D1-D2

interfering peptide after intranasal administration, we will also test whether it is effective at

doses higher or lower than our original 100-fold dose. We predict that at doses 100-fold or

greater than those given directly to the PFC in the previous study, the D1-D2 interfering peptide

will have an antidepressant effect in the FST. Along these same lines, we will investigate the

length of time the D1-D2 interfering peptide remains biologically active in the body, as we are

unsure about how long it remains stable in the CNS once it is administered intranasally.

Finally, the D1-D2 interfering peptide displayed an antidepressant effect when it was

delivered directly to the PFC, but not to other brain areas such as the NAc and hippocampus3.

We hypothesize that if the D1-D2 interfering peptide has an antidepressant effect after intranasal

administration, it will be due, at least in part, to the ability of the D1-D2 interfering peptide to

disrupt the D1-D2 receptor-receptor interaction in the PFC. Taken together, these experiments

will further our understanding of the intranasal delivery route for small interfering peptides as

well as reveal whether the D1-D2 interfering peptide is a promising novel therapeutic for the

treatment of MDD.

Page 51: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

41

2 Materials and Methods

2.1 Animals

Adult Male Sprague-Dawley rats (Charles River Laboratories, Wilmington, MA) were

used in all experiments. Rats were pair-housed at a constant temperature (20-23°C) on a 12-hour

light/dark cycle (light on at 8:00AM) with unrestricted food and water. After arriving at the

facility, rats were given 1 week to acclimatize before being subjected to behavioral testing and

injections. All rats weighed between 300 and 350g when they underwent behavioral testing. All

experimental procedures were approved by the Animal Care Committee at the Centre for

Addiction and Mental Health (Toronto, ON).

2.2 Intranasal administration procedures

2.2.1 Intranasal administration using the POD

All animals were anaesthetized using 5% isoflurane, an inhalant anesthetic (Benson

Medical Industries, Inc.) for 3-4 minutes. Rats were then placed in a supine position and dosed

with the POD developed by Impel NeuroPharma (Seattle, Washington). When dosing the

animals, the POD tip (with the relevant dose) was inserted approximately 8-10mm into the rat’s

nostril, angled towards the olfactory epithelium (towards the top of the head) and the propellant

can was fired for 1 second. 2 seconds later, the POD tip was slowly removed. The propellant can

and the POD tip were attached by a 30cm-long piece of plastic tubing (see Figure 2-1). This

allowed for maximum maneuverability of the POD tip in the nose. This protocol was adapted

from the POD administration procedure originally developed by Impel NeuroPharma.213

Page 52: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

42

After intranasal administration animals were replaced in the anesthetic chamber in the

supine position for 4 minutes at decreasing isoflurane concentrations. This step allows for

maximum absorption of the peptide dose, while decreasing sneezing and other behaviors that

would result in expulsion of the substance from the nose. Animals were then replaced in their

home cages and returned to their housing rooms once they had recovered from the anesthesia and

had regained complete locomotor control. Rats that bled after intranasal administration were

eliminated from the study. The time of injection was recorded as the time at which the animals

woke up from the anesthesia and regained locomotor control.

2.2.2 Verification of POD delivery to the olfactory epithelium

In order to verify that the POD deposits substances preferentially onto the olfactory

epithelium, we gave intranasal injections (IN) of Richard Allen Scientific Mark-It Blue Dye

(5000BL, Thermo Scientific). The Mark-It blue dye allowed us to visualize where within the rat

nose the POD was preferentially depositing substances. Using the POD, we gave 10uL per

nostril using the administration method outlined in Section 2.2a). After POD administration,

animals were immediately sacrificed using trans-cardiac perfusion of 1X Phosphate Buffered

saline (pH=7.4). The nasal anatomy was then dissected and examined for traces of Mark-IT Blue

Dye. After dissection, the nasal anatomy was photographed for qualitative evaluation of the

deposition of substances by the POD.

2.2.3 Substances injected intranasally

In each experiment, animals received the same total number of intranasal injections (IN)

(3-4) in alternating nostrils over a 24-hour period. Animals received either filtered saline (0.9%

9- NaCl, 8-12 L/injection), the D1-D2 interfering peptide (TAT-D1-D2-IPep) (8-12µL, 50mM);

Page 53: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

43

Figure 2-1 Pressurized Olfactory Device (POD) for intranasal administration: apparatus

(A) POD apparatus: the Propellant can is attached to the administration tip with a 30cm long piece of plastic

tubing. This allows for maneuverability when inserting the administration tip into the nose and administering the

intranasal dose. (B) Optimal position of the administration tip within the nose for preferential dose deposition

onto the olfactory epithelium.

Page 54: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

44

a amino acid membrane permeable TAT peptide (TAT-Pep) (8-12 µL, 50mM) from the Human

Immunodeficiency Virus 1(HIV1) TAT protein214

; or D1-D2-FLAG tagged interfering peptide

(TAT-D1-D2-FLAG-IPep) (8-12µL, 50mM). The 9 amino acid TAT-peptide sequence from the

HIV1 TAT protein (YGRKKQRRR)214

rendered all peptides used in these studies cell-

permeable. All peptides were custom synthesized by Gen Script (New Jersey, USA) and/or

Biomatik, Inc (Cambridge, Ontario) and had purity levels between 95 and 99%. All peptides

were dissolved in filtered saline at a concentration of 50mM and stored at -80°C.

2.3 Intra-peritoneal injection procedures

We administered imipramine hydrochloride (15mg/mL, Sigma-Aldrich) at a dose of

15mg/kg using intra-peritoneal (IP) injections into the abdominal cavity. To control for

anesthetic exposure, these animals were also anaesthetized before administration of IP injections.

Rats were anaesthetized in an induction chamber using 5% isoflurane. Rats were placed in a

supine position and given imipramine via IP injections. After the injection, animals were

replaced in the induction chamber at decreasing isoflurane concentrations for 4 minutes. Animals

were allowed to recover in their home cages and the time of injection was recorded as the time

when the rats regained locomotor control.

2.4 Immunofluorescence and confocal microscopy

2.4.1 Tissue fixation and storage

The purpose of this study was to test whether a biologically active peptide that has an

anti-immobility effect in the FST, can be visualized in the PFC and after intranasal

administration. To do this, we used a peptide with the same sequence as the D1-D2 interfering

Page 55: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

45

peptide3, but with an 8-amino acid FLAG-tag (Sequence: DYKDDDDK)

215 fused to the C-

terminal of the peptide. This modification allowed us to use immunofluorescent (see section

2.4.1) methods to detect it in the brain after intranasal administration and completion of the

FST.216

For this experiment, a small number of animals were sacrificed by transcardiac perfusion.

After being anaesthetized for 4 minutes in an induction chamber with 5% isoflurane, animals

were perfused transcardially with 60mL phosphate buffered saline (PBS, pH =7.4) followed by

60mL 4% Paraformaldehyde ( 4% PFA, in PBS). Subsequently, whole brains were dissected and

stored in 4% PFA overnight. The next day, brains were transferred to a 20% sucrose

cryoprotection solution for approximately 48 hours. The tissue was then stored at -80°C for

subsequent use.

2.4.2 Immunofluorescent staining procedures

Rats that had completed the FST and were assigned to the TAT-D1-D2-FLAG-IPep or

saline treatment groups were sacrificed, their tissue fixed and collected as described in Section

2.4.1. The olfactory bulbs and PFC of 2 brains from each condition (TAT-D1-D2-FLAG-IPep

and saline) were cut into 12 M sections using a cryostat. After slicing, we blocked non-specific

antibody interactions using 5% donkey serum for 1 hour, before staining overnight with an anti-

FLAG monoclonal antibody (mouse monoclonal, M2, Sigma-Aldrich). Sections were then

incubated with a secondary immunofluorescent antibody (donkey anti-mouse Cy2-conjugated

antibody, Jackson Immuno Research Laboratories, Inc.) before being counter-stained with

NeuroTrace 530/615 red fluorescent Nissl Stain (Molecular Probes, Invitrogen). Sections were

mounted on slices with PureGold anti-fade mounting reagent (Molecular Probes, Invitrogen.)

and stored at 4°C.

Page 56: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

46

Sections were visualized and imaged using a Zeiss LSM 510 confocal microscope.

Images of PFC coronal sections in both conditions were taken under 25X magnification. Cy2

immunofluorescence was imaged using an argon laser with maximum excitation at 488nm. To

detect the NeuroTrace 530/615 stain, we used a helium 1 laser with maximum excitation at 530

nm. Images were overlaid using Image J software.

2.5 The Forced Swimming Test

2.5.1 FST Procedure

The FST is an acute test for antidepressant efficacy originally developed by Porsolt et al.

in 1977.174

On the first day of the test, animals undergo a training session where they are placed

in an inescapable plexiglass cylinder for 15 minutes. The plexiglass cylinders were 60cm high

and 20cm in diameter. The plexiglass cylinders were filled to a height of 40 cm with water at a

temperature of 25 +/- 0.2°C, which was changed between each animal (See Figure 2-3 for a

picture of the FST cylinder).

In accordance with the dosing schedule established in the literature and used

previously3,174-176

subjects were given the treatment intervention three times after the training and

testing sessions: 30 minutes after the 15-minute training session, 5 hours after training and 1 hour

before undergoing the FST. 24 hours after the training session and 1 hour after the last

behavioral intervention, the rat was replaced in the same cylinder for 5 minutes. The session was

video recorded and scored blindly at a later date. After both the training and test FST sessions,

rats were towel-dried and placed in a heated cage for a minimum of 15 minutes. See Figure 2-2

for a detailed schematic of the FST experimental design.

Page 57: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

47

Figure 2-2 Overall experimental procedure for FST.

See section 2.5.1 for detailed description of the FST

experimental procedure

Page 58: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

48

2.5.2 FST behavioral scoring method

The 5-minute session of the FST was video recorded and animal behavior during the FST

was scored at a later date after the experimenter was blinded to the treatment groups. All FST

videos were scored by the same experimenter who had been blinded to the experimental

conditions of the animals. Behaviors were scored in five-second bins with the predominant

behavior (immobility, swimming, climbing, diving) in each 5-second period recorded for a total

of 60 behavioral counts.

The animal’s activity during the FST was segregated into four behaviors, in keeping with

the literature and previous studies3,174,176

: immobility, which consisted of the animal only making

those movements necessary to keep its nose above the water; swimming, consisting of active

movement of the forepaws and legs and movement around the cylinder; climbing, consisting of

vigorous movement of the forepaws along the sides of the cylinder, as if trying to climb out of it;

and diving, when the animal entered head-first into the water and spent a minimum of 2 seconds

completely submerged. Figure 2-3A-D displays representative photographs of each of these

behaviors.

Mean immobility counts across all groups were analyzed by 1-way independent groups

analysis of variance (ANOVA). Post-hoc Newman-Keuls multiple comparisons tests were used

to evaluate differences across individual groups, as necessary. In some cases, other behaviors

(swimming, diving and climbing) were also analyzed using 1-way independent groups

ANOVAs, followed by post-hoc Newman-Keuls multiple comparisons tests to evaluate

differences between treatment groups. Data were analyzed using Prism (GraphPad Software,

Inc.).

Page 59: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

49

Figure 2-3 Representative photographs of behaviors exhibited during the FST.

Top L to Bottom R: immobility, or passive floating with nose out of the water, swimming,

actively moving forepaws and legs to remain afloat, climbing, vigorous movement of

forepaws and legs in tandem, as if to climb up the walls of the FST cylinder, and diving,

head-first submersion of the entire body in the FST cylinder.

Page 60: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

50

2.6 FST experiments: experimental design

2.6.1 Effect of the D1-D2 interfering peptide in the FST

We tested whether the D1-D2 interfering peptide would have an anti-immobility effect in

the FST when administered at a dose 100-fold that which was administered directly to the PFC3

(for detailed calculations of the D1-D2 interfering peptide intranasal dose, see Appendix 1.)

Animals were randomly assigned into four treatment groups: TAT-D1-D2-IPep (IN, 1.67nmol/g,

n=7), TAT-Pep (IN, 1.67nmol/g, n=6), saline (IN, 1.67nmol/g, n=6), and imipramine (IP,

15mg/kg, n=6). This group was included in order to confirm that the 9-amino acid, membrane

permeable TAT peptide fragment did not have any behavioral effect in the FST. We followed the

2-day FST protocol and behavioral analysis procedure outlined in Section 2.5. These treatments

were given 30 minutes after the 15-minute FST training session, 5 hours later and 1 hour before

the 5-minute FST test. The 5-minute FST behavioral tests were video-recorded, scored and

analyzed as described in Section 2.5.2.

2.6.2 Effect of the D1-D2-FLAG interfering peptide in the FST

The purpose of this study was to test whether a biologically active peptide that can be

visualized in the PFC areas after intranasal administration also has an anti-immobility effect in

the FST. To do this, we synthesized a peptide with the same sequence as the D1-D2 interfering

peptide3, but with an 8-amino acid FLAG-tag (Sequence: DYKDDDDK)

215 fused to the C-

terminal of the peptide, tested whether it had an anti-immobility effect in the FST and whether

we could visualize it in the PFC.

To test whether the D1-D2-FLAG interfering peptide had an anti-immobility effect in

the FST (for detailed procedure, see Section 2.5), animals were randomly assigned into four

treatment groups: TAT-D1-D2-IPep-FLAG (IN, 1.67nmol/g, n=4), TAT-Pep (IN, 1.67nmol/g,

Page 61: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

51

n=4), saline (IN, 1.67nmol/g, n=5), and imipramine (IP, 15mg/kg, n=4). We followed the 2-day

FST protocol and behavioral analysis procedure outlined in Section 2.5. The 5-minute FST

behavioral tests were video-recorded, scored and analyzed as outlined in Section 2.5.2.

2.6.3 Efficacy of the D1-D2 interfering peptide at various intranasal doses

In order to better understand the pharmacological effects of the D1-D2 interfering peptide

upon intranasal administration, information regarding its efficacy at various doses is required. To

address this, we varied the dose of the D1-D2 interfering peptide and analyzed their anti-

immobility effects in the FST. We used saline and imipramine groups as a negative and positive

control, respectively. We followed the general FST design and behavioral analysis procedure

outlined in Section 2.5. Table 2-1 details the treatment groups, doses and the overall

experimental design employed in this study.

Immobility counts during the 5-minute FST in each treatment dose was compared with

saline and imipramine groups using 1-way independent groups ANOVAs, followed by post-hoc

Newman-Keuls multiple comparisons tests. From our results, we identified the minimum dose of

the D1-D2 interfering peptide with the maximal behavioral effect as 1.67nmol/g, which we used

in all subsequent experiments. In order to control for any behavioral effect of the TAT-peptide,

we included two treatment groups that received TAT-pep at a dose of 1.67 nmol/g and 2.0nmol/g

in order to test whether the TAT-peptide’s effect in the FST changes if the dose is increased.

Page 62: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

52

Table 2-1 Efficacy of the D1-D2 interfering peptide at various doses: overall experimental design

and Treatment Groups.

Treatment Doses, nmol/g (mg/kg), number of animals per group

saline

Volume-

Controlled

n=4

n=8 (same

group as

1.0nmol/g)

n=6

n=8 (same

group as

2.0nmol/g)

Imipramine

15mg/kg, IP

n=3

n=8 (same

group as

1.0nmol/g)

n=6 n=8 ( same

group as

2.0nmol/g)

TAT-Pep n=7

2.0

nmol/g

n=6

1.67nmol

/g

TAT-D1-D2-

IPep

4.0nmol/g

(13.72mg

/kg)

n=3

2.0nmol/g

(6.86mg/

kg)

n=7

1.67nmol/g

(5.75mg/

kg)

n=7

1 nmol/g

(3.45mg/k

g)

n=7

Page 63: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

53

2.6.4 Duration of behavioral effect of D1-D2 interfering peptide in the FST

It is unclear how long the D1-D2 interfering peptide remains active in the CNS after

intranasal administration. To test this, we compared the anti-immobility effect of the D1-D2

interfering peptide in the FST at 2, 3 and 4 hours after intranasal administration. We used the

general FST protocol and analysis procedure outlined in Section 2.5. The overall experimental

design and treatment groups are shown in Table 2-2.

Each animal in this study received intranasal or IP injections 30 min and 5 hrs after the

FST training session while the time point of the last intranasal injection was increased to 2, 3 and

4 hours before the 5-minute FST session. The amount of immobility behavior in treatment

groups was compared using 1-way independent groups ANOVA at each time point (2h, 3h and

4h). Saline and imipramine groups were compared to at all time points (2h, 3h, and 4h).

2.7 Locomotor activity test

We tested whether the increased mobility we observed in the FST after intranasal

administration was due to the antidepressant effect of the D1-D2 interfering peptide or due to an

overall increase in locomotor activity. To do this, we tested whether the D1-D2 interfering

peptide had an effect on activity during a 30-minute locomotor activity test after intranasal

administration. 25 rats that had already been exposed to the FST were used in this experiment.

Rats were given TAT-D1-D2-IPep (IN, 2.0nmol/g, n=5); TAT-D1-D2-IPep (IN, 1.67nmol/g,

n=5); saline (IN, n=5); TAT-Pep (IN 2.0nmol/g, n=5); or 15mg/kg imipramine (IP, n=5) three

times before the open field test: 24 hours before the test, 19 hours before the test, and one hour

before testing. This dosing schedule was identical to that used in the forced swimming

Page 64: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

54

Treatment Time point before FST, number of animals/group

Saline 1 hour before FST

Volume-Controlled (10ul/dose)

n=6

Imipramine 1 hour before FST

15mg/kg, IP

n=6

TAT-Pep

(1.67nmol/g)

1 hour before

FST

n=7

2 hours before

FST

n=6

3 hours before

FST

n=6

4 hours before FST

n=6

TAT-D1-D2-

IPep

(1.67nmol/g)

1 hour before

FST

n=6

2 hours before

FST

n=6

3 hours before

FST

n=6

4 hours before FST

n=6

Table 2-2 Duration of the anti-immobility effect of the D1-D2 interfering peptide: treatment groups and

overall experimental design

Page 65: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

55

experiments (see Section 2.5.1). Animals were kept in their original treatment groups, for

example if an animal had been assigned to the saline treatment group during the FST, it remained

in the saline group during the locomotor experiments.

To record locomotor activity, rats were placed in a custom-made locomotor activity

recording apparatus. Each animal was placed in a 20cm high, 20 cm wide and 30 cm long cage

(standard housing cage) in the locomotor apparatus for 30 minutes in a dark room. Rats had not

previously been exposed to the testing room or to the locomotor activity boxes. An array of 11

infrared photocells was placed along the long axis of the cages. Interruption of infrared beams

(Beam Breaks) was used as a measure of locomotor activity. The total amount of locomotor

activity was recorded in 5 minute intervals and for the entire 30 minute session. Total activity

data were analyzed via 1-way independent groups ANOVA according to the locomotor measure

followed by Newman-Keuls multiple comparisons tests, using Prism Software (GraphPad

Software, Inc.). The locomotor activity at various intervals during the test was analyzed by 2-

way independent factors ANOVA with treatment group (saline, imipramine, TAT-Pep and TAT-

D1-D2-IPep) and Time Point (5min, 10min, 15min, 20min, 25min, 30min) as main factors.

2.9 Co-immunoprecipitation and western blots

2.9.1 Tissue Collection

Animals were sacrificed the day they completed the FST or, if applicable, the open field

test. Animals were anaesthetized for 3 minutes with 5% isoflurane and were decapitated. Their

brains were quickly removed and relevant tissue areas were dissected on ice. These areas

included the olfactory bulbs, PFC, striatum, hippocampus and VTA. Brain tissue was stored at -

80°C for subsequent use in biochemical assays.

Page 66: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

56

2.9.2 Co-Immunoprecipitation of D1 receptor by anti-D2DR

Since the peptide we are testing in the FST is designed to disrupt the interaction between

the D1 and D2 receptors, we investigated the co-immunoprecipitation of the D1 receptor by an

antibody against the D2 receptor in the PFC of animals who had received intranasal injections of

the D1-D2 interfering peptide or saline. We compared animals who had been given intranasal

injections of TAT-D1-D2-IPep (IN, 1.67nmol/g, n=3), or saline (IN, n=3) and had been exposed

to the FST and the open field test.

For the co-immunoprecipitation, solubilized proteins from the PFC and striatum (500 µg)

from each animal were incubated with 1ug goat polyclonal anti-D2DR (N-19, Santa Cruz

Biotechnology) and protein A/G PLUS agarose beads (Santa Cruz Biotechnologies) overnight. A

control sample was incubated with polyclonal Goat IgG (Sigma-Aldrich) to confirm the absence

of non-specific immunoprecipitation.

After incubation, the immunoprecipitated proteins were washed and incubated with SDS

sample buffer (BioRad, Inc.) at 37°C for 40 minutes before being separated from the Protein A/G

PLUS-agarose beads using centrifugation. Immunoprecipitated proteins were then subjected to

separation using 10% SDS-Page gels, transferred onto nitrocellulose membranes and

immunoblotted overnight using anti-D1DR (D187, Sigma-Aldrich). Each immunoblot included

samples from saline and TAT-D1-D2-IPep treatment groups, along with a control sample

incubated with goat IgG (Sigma-Aldrich) and 75µg of tissue-extracted input protein from PFC

tissue. After overnight incubation, secondary antibodies conjugated with horseradish peroxidase

were applied to the blots for approximately two hours. After washing, immunoblots were

developed with ECL reagent (GE Healthcare, Inc.) and imaged using a BioRad ChemiDoc MP

system (BioRad Technologies, Inc.). To quantify the expression of protein, we conducted

Page 67: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

57

densitometry analyses using ImageLab software (BioRad Technologies, Inc.). Densitometry data

were analyzed using two-tailed, unpaired Student’s t-tests (Prism Software, GraphPad, Inc).

2.9.3 Western Blots

We compared the expression of the D1 and D2 receptors using western blots in animals

given intranasal injections of TAT-D1-D2-IPep (IN, 1.67nmol/g, n=3) or saline (IN, n=3). All

groups were given intranasal injections and exposed to the FST and open field tests. 80 µg of

solubilized protein extracts from the PFC, in SDS sample buffer (BioRad Technologies, Inc.)

were denatured by boiling for 5 minutes before being separated by 10% SDS page gel.

After transfer onto nitrocellulose membranes, we immunoblotted for D1 and D2 receptors

with monoclonal anti-D1DR (rat IgG, D187, Sigma-Aldrich) and monoclonal anti-D2DR

(Mouse IgG, B-10, Santa Cruz Biotechnologies). In order to confirm that the total amount of

protein in each sample were equal, we separated 20µg solubilized protein extracts with 10% SDS

page gel and immunoblotted using monoclonal anti-α-tubulin (Mouse IgG, DM1A clone, Sigma-

Aldrich).

After overnight incubation, immunoblots were incubated for 2 hours with species specific

Horseradish Peroxidase conjugated secondary antibodies. After washing, immunoblots were

developed with ECL reagent (GE HealthCare, Ltd.) and visualized using a BioRad ChemiDoc

MP system (BioRad Technologies, Inc). To quantify the expression of protein, we conducted

densitometry analyses using ImageLab software (BioRad Technologies, Inc). Densitometry data

were analyzed with two-tailed, unpaired Student’s T-tests using Prism Software (GraphPad

Software, Inc.).

Page 68: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

58

3 Results

3.1 Experiment 1: The POD preferentially deposits substances on the

olfactory epithelium within the rat nasal cavity

In order to verify that the POD deposits substances preferentially onto the olfactory

epithelium, we administered intranasal injections of Richard Allen Scientific Mark-It Blue Dye

using the POD. The Mark-It dye allowed us to visualize where the POD was preferentially

depositing substances within the rat nasal cavity. It also allowed us to optimize our POD

delivery protocol (see Section 2.2.1) in order to maximize deposition of substances on the

olfactory epithelium and surrounding tissue.

We assessed the deposition of the Mark-It dye after delivery using the POD by grossly

dissecting the nasal anatomy. Figure 3-1A and B show representative images of Mark-It dye

deposition after POD delivery on the olfactory epithelium within the rats’ nasal cavity (A) and

visible through the cribiform plate (B), the porous bone through which the olfactory axons travel

to the olfactory bulb and CNS.15

As the images show, the POD preferentially deposited the

Mark-It Dye on the olfactory epithelium, favoring transport mechanisms that deliver substances

to the CNS.

3.2 Experiment 2: The D1-D2-FLAG interfering peptide can be detected in

the prefrontal cortex after intranasal administration

After confirming that the POD preferentially deposits substances on the olfactory

epithelium and olfactory sensory areas of the rat’s nasal anatomy, we next assessed whether a

TAT-linked, membrane-permeable peptide would reach the CNS, and the PFC in particular, after

Page 69: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

59

Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after correct POD

administration

(A) Deposition of Blue Mark-It dye on olfactory epithelium after intranasal administration using the POD

following the protocol outlined in Section 2.2.1. Animal’s nasal anatomy was dissected through the midline in

order to visualize both the left and right nasal cavities. (B) Blue Mark-It Dye is visible through the cribiform plate

(bone separating the nasal olfactory epithelium from the CNS, CSF and olfactory bulbs. The cribiform plate

contains perforations through which the olfactory receptor neuron (ORN) axons travel to the olfactory bulb and

CNS. Representative images shown.

Page 70: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

60

administration with the POD. We focused on detecting the peptide in the PFC because Pei et al’s

study3 showed that the D1-D2 interfering peptide only had an antidepressant effect in the FST

when it was administered directly to this brain area.3 After staining selected sections of the PFC

from animals that received TAT-D1-D2-FLAG-IPep (1.67nmol/g, n=2) and saline (n=2) with

anti-FLAG antibodies, followed by a Cy2-conjugated fluorescent secondary antibody, we

visualized the resulting immunofluorescence using a confocal microscope. Figure 3-2A-B shows

representative images of prefrontal cortical slices from individual animals who received

intranasal injections of TAT-D1-D2-IPep (A) or saline (B).

We detected the presence of a TAT-D1-D2-IPep peptide in the PFC, as the Cy-2-

conjugated immunofluorescent signal was visible in slices from animals that had received TAT-

D1-D2-FLAG-IPep intranasal injections and not visible in the PFC of animals that received

saline injections. Immunofluorescence was visible through the anterior PFC coronal sections,

with no extreme variations in staining in dorsal, ventral medial or lateral areas. The results of this

experiment suggest that intranasal administration is a viable method to deliver membrane-

permeable peptides to the PFC.

3.3 Experiment 3: Intranasal administration of the D1-D2 interfering

peptide has an antidepressant effect in the forced swimming test

Although the experiment outlined in Section 3.2 allowed us confirm the presence of the

D1-D2-FLAG interfering peptide in the PFC after intranasal administration, it did not indicate

whether the accumulation of the peptide in the PFC was sufficient to produce a behavioral

antidepressant effect in the FST. Thus, it remained unclear whether intranasal administration

resulted in sufficient accumulation of the D1-D2 interfering peptide in the PFC to produce an

observable behavioral effect in the FST.

Page 71: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

61

Figure 3-2 Immunofluorescent staining for anti-FLAG antibodies is visible in PFC slices of animals who

were administered TAT-D1-D2-FLAG-IPep (A) but not those who were administered saline (B). A) 1,2: Representative images of PFC sections of two separate animals who received TAT-D1-D2-FLAG-

Ipep (1.67nmol/g) intranasally. B) 1,2 Representative images of PFC sections from two separate animals who

received intranasal injections of Saline. Representative fluorescent images taken with a Zeiss LM confocal

microscope (25X magnification ) of anti-FLAG (Cy2 secondary antibodies) and NeuroTrace 530/615 neuronal

cell body stain. Representative images shown.

Page 72: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

62

3.3.1 The D1-D2 Interfering Peptide has an Anti-Immobility Effect in the

FST

We compared immobility, swimming and diving behaviors during the 5-minute FST

across four treatment groups: animals that received saline (IN, n=6, Volume-controlled)

imipramine (IP, n=6, 15mg/kg), TAT-D1-D2-IPep (IN, n=7, 1.67nmol/g), or TAT-Pep (IN, n=6,

1.67nmol/g).

The D1-D2 interfering peptide had a significant anti-immobility effect in the FST that was

comparable to that of imipramine and significantly greater than that of saline or TAT-Pep. A 1-

way Independent Groups ANOVA revealed a significant difference in immobility behavior

between all groups: TAT-D1-D2-IPep, saline, imipramine and TAT-Pep (n=6-7 per group,

F(3,21)=12.25, p<0.0001). Post-Hoc Newman-Keuls multiple comparisons tests showed

significant differences when comparing TAT-D1-D2-IPep and saline (p<0.01), TAT-D1-D2-

IPep and TAT-Pep (p<0.01), imipramine and saline (p<0.001) and imipramine and TAT-Pep

(p<0.001). No significant differences were present between imipramine and TAT-D1-D2-IPep

groups (p>0.05) and saline and TAT-Pep groups (p>0.05).

For the swimming behavior, a 1-way Independent Groups ANOVA revealed a significant

difference between all treatment groups, (F(3,21)=12.57 p<0.0001). Post-hoc Newman Keuls

multiple comparisons tests revealed significant differences between TAT-D1-D2-IPep and saline

(p<0.01), TAT-D1-D2-IPep and TAT-Pep (p<0.01), imipramine and saline (p<0.001) and

imipramine and TAT-Pep (p<0.001). No significant differences were present between

imipramine and TAT-D1-D2-IPep (p>0.05) and saline and TAT-Pep (p>0.05) treatment groups.

There was no significant difference in diving behavior between saline, imipramine, TAT-D1-D2-

IPep and TAT-Pep groups (F(3,21)=0.99, p>0.05). Data is represented graphically in Figure 3-3.

Page 73: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

63

Figure 3-3 The D1-D2 interfering peptide has an antidepressant effect in the FST when administered

intranasally.

After intranasal administration using the POD, the D1-D2 interfering peptide (dose = 1.67nmol/g) significantly

decreases immobility and increases swimming behavior in the FST. Data for each standard behavior (immobility,

swimming, diving) analyzed via 1-way independent groups ANOVA followed by Newman Keuls post-hoc multiple

comparisons tests. ** p<0.01, *** p<0.01 compared to saline; ^^ p<0.01, ^^^ p<0.001 compared to TAT-Pep. No

significant difference was observed between behavior in animals who received TAT-D1-D2-Ipep or imipramine.

Error bars represent SEM.

Page 74: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

64

3.3.2 The D1-D2-FLAG interfering peptide has an anti-immobility effect

in the FST after intranasal administration

We tested whether the D1-D2 FLAG peptide produced an anti-immobility effect in the

FST, and whether we could detect it in the PFC of animals who had been exposed to the FST(see

Section 3.2). We hypothesized that, at a sufficient dose, the TAT-linked D1-D2 interfering

peptide with a FLAG tag on the C-terminal end (TAT-D1-D2-FLAG-IPep) would have an anti-

immobility effect in the FST comparable to that of imipramine and the D1-D2 interfering

peptide. The D1-D2-FLAG interfering peptide had a significant anti-immobility effect in the

FST that was comparable to imipramine and significantly greater than that of saline and TAT-

Pep.

A 1-way independent groups ANOVA of immobility behavior across treatment groups

demonstrated a significant difference between all groups (saline, imipramine, TAT-D1-D2-IPep

and TAT-Pep, n=4-5 per group, F(3,14)=7.746, p<0.01). Post-hoc Newman-Keuls multiple

comparisons tests revealed significant differences between imipramine and saline groups (n=4-5,

p<0.01), imipramine and TAT-Pep groups (n=4-5, p<0.05), TAT-D1-D2-FLAG-IPep and saline

groups (n=4-5, p<0.01) and TAT-D1-D2-FLAG-IPep and TAT-Pep groups (n=4-5, p<0.05).

There was no significant differences between TAT-Pep and saline groups (p>0.05) or

imipramine and TAT-D1-D2-IPep groups (p>0.05). Data is represented graphically in Figure 3-

4.

To investigate whether the behavior during the 5-minute FST was significantly different

in animals that received the D1-D2-FLAG peptide and those that received the D1-D2 peptide, we

compared immobility, swimming and diving behavior in these two groups. The D1-D2

interfering peptide and the D1-D2-FLAG tagged interfering peptide had indistinguishable

Page 75: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

65

Figure 3-4 The D1-D2-FLAG interfering peptide has an anti-immobility effect in the FST.

The D1-D2-FLAG interfering peptide significantly decreases immobility behaviors in the FST after

intranasal administration. TAT-D1-D2-FLAG-IPep and TAT-Pep administered intranasally at a dose of

1.67nmol/g . Data analyzed via 1-way independent groups ANOVA followed by Newman Keuls Post-Hoc

Multiple Comparisons Tests. ** p<0.01, *** p<0.01 compared to saline; ^^ p<0.01, ^^^ p<0.001 compared

to TAT-Pep. No significant difference was observed between behavior in animals who received saline or

imipramine. Error bars represent SEM. Numbers within bars represent number of animals per group.

Page 76: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

66

Figure 3-5 The D1-D2 interfering peptide and D1-D2-FLAG tagged interfering peptide have similar

behavioral effects in the FST. We compared immobility, swimming and diving behavior in treatment groups that that received TAT-D1-D2-

IPep (see Section 3.3.1) and TAT-D1-D2-FLAG-IP (see Section 3.3.2). Two-tailed, unpaired student’s t-tests

revealed no significant differences in immobility (p=0.83), swimming (p=0.73) or diving (p=0.18) behaviors

between these groups, indicating that the D1-D2 interfering peptide and the D1-D2-FLAG interfering peptide’s

effect in the FST are indistinguishable. Error bars represent SEM.

Page 77: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

67

behavioral effects during the 5-minute FST. Two-tailed, unpaired Student’s T-tests revealed no

significant differences between immobility, swimming or diving behavior in animals who

received TAT-D1-D2-IPep or TAT-D1-D2-FLAG-IPep (n=4-7 per group, Immobility, t(9)=0.21

, p=0.83; Swimming, t(9)=0.32 , p=0.74 ; Diving t(9)=1.447 , p=0.18).(Figure 3-5).

3.4 Experiment 4: Efficacy of the D1-D2 interfering peptide at various

intranasal doses

The D1-D2 interfering peptide has an antidepressant effect in the FST when given

intranasally at a dose of 1.67nmol/g (5.75mg/kg) (Section 3.3.1., Figure 3-3.). The 1.67nmol/g

dose administered intranasally represents an estimated 100-fold increase over the dose given

directly to the PFC (5nmol/injection).3 Although this particular intranasal dose can produce an

antidepressant effect in the FST, the behavioral effects of the D1-D2 interfering peptide at doses

higher or lower than 1.67nmol/g (5.75 mg/kg) remains unknown.

To investigate the antidepressant effects of the D1-D2 interfering peptide at various

intranasal doses, we varied its dose, exposed animals to the 2-day FST, and analyzed the

resulting immobility behavior in the treatment groups (see Section 2.6.3 and Table 2-1 for

detailed experimental design). We hypothesized that at intranasal doses larger than 1.67nmol/g,

the D1-D2 interfering peptide would have a significant anti-immobility effect in the FST.

3.4.1 D1-D2 interfering peptide dose: 4.0nmol/g (13.72 mg/kg)

At a dose of 4.0nmol/g, the D1-D2 interfering peptide had an anti-immobility effect in

the FST comparable to that of imipramine and significantly greater than that of saline. We

compared immobility behavior during the 5-minute FST in rats that received saline (IN, n=3),

imipramine (IP, 15mg/kg, n=3) and TAT-D1-D2-IPep (IN, n=3, 4.0 nmol/g). A 1-way

Page 78: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

68

independent groups ANOVA revealed a significant difference between all groups (F(2,6) =

9.207, p<0.01). Post-hoc Newman-Keuls multiple comparisons tests showed significant

differences between TAT-D1-D2-IPep and saline (p<0.05) and saline and imipramine (p<0.05).

No significant difference was present between TAT-D1-D2-IPep and imipramine groups (Figure

3-6, 3-10).

3.4.2 D1-D2 interfering peptide dose: 2.0nmol/g (6.86 mg/kg)

At an intranasal dose of 2.0nmol/g, the D1-D2 interfering peptide had an anti-immobility

effect in the FST comparable to that of imipramine and significantly greater than that of saline.

We compared immobility behavior during the 5-minute FST in rats that received saline (IN,

n=8), imipramine (IP, 15 mg/kg, n=8), TAT-D1-D2-IPep (IN, 2nmol/g, n=8), and TAT-Pep (IN,

2nmol/g, n=7). A 1-way independent groups ANOVA revealed a significant difference between

immobility behavior in all groups ( F(3,27)=6.836, p<0.01). Post-hoc Newman-Keuls Multiple

comparisons tests revealed significant differences between TAT-D1-D2-IPep and saline

(p<0.01), imipramine and saline (p<0.01) and imipramine and TAT-Pep (p <0.05). There were

no significant differences between saline and TAT-Pep (p>0.05), imipramine and TAT-D1-D2-

IPep (P>0.05) and TAT-Pep and TAT-D1-D2-IPep (p>0.05). Immobility behavioral counts are

represented graphically in Figure 3-7 and summarized in Figure 3-10.

3.4.3 D1-D2 interfering peptide dose: 1.67nmol/g (5.75 mg/kg)

For the purposes of comparison, data relating to immobility behavior in rats that received

our original intranasal dose of 1.67nmol/g (5.57mg/kg) is included here (for complete analysis,

see Section 3.3.1.). Briefly, a 1-way independent groups ANOVA revealed a significant

Page 79: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

69

Figure 3-6 The D1-D2 interfering peptide has an anti-immobility effect in the FST at an intranasal dose of

4.0nmol/g

Immobility behavioral of animals who received intranasal injections of TAT-D1-D2-IPep (dose = 4nmol/g) was

compared to those who received saline or IP injections of imipramine. Immobility behavior between groups

compared by 1-way independent groups ANOVA followed by post-hoc Newman Keuls multiple comparisons tests.

* p<0.05 compared to saline. Error bars represent SEM. Numbers within bars represent number of animals per

group.

Page 80: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

70

Figure 3-7 The D1-D2 interfering peptide has an anti-immobility effect in the FST at an intranasal dose of

2.0nmol/g

Immobility behavioral of animals who received intranasal injections of TAT-D1-D2-IPep (dose = 2nmol/g) was

compared to those who received saline, TAT-Pep or IP injections of imipramine. Immobility behavior between

groups compared by 1-way Independent Groups ANOVA followed by post-hoc Newman-Keuls multiple

comparisons tests. ** p<0.01 compared to saline, ^ p<0.05 compared to TAT-Pep. Error Bars Represent SEM.

Numbers within bars represent number of animals per group.

Page 81: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

71

difference between groups who received saline (IN, n=6), imipramine (IP, 15mg/kg, n=6), TAT-

D1-D2-IPep (IN, 1.67nmol/g, n=7) and TAT-Pep (IN, 1.67nmol/g, n=6), (n=6-7, F(3,21)=12.25,

p<0.001). Post-Hoc Newman-Keuls multiple comparisons tests revealed significant differences

when comparing TAT-D1-D2-IPep and saline (p<0.01), TAT-D1-D2-IPep and TAT-Pep

(p<0.01), imipramine and saline (p<0.001) and imipramine and TAT-Pep (p<0.001). No

significant differences were found between imipramine and TAT-D1-D2-IPep (p>0.05) and

saline and TAT-Pep (p>0.05). Data is represented graphically in Figure 3-3, 3-8 and

summarized in Figure 3-10.

3.4.4 D1-D2 interfering peptide dose: 1.0nmol/g (3.43 mg/kg)

At an intranasal dose of 1.0nmol/g, the D1-D2 interfering peptide did not have an anti-

immobility effect in the FST, as it was not significantly different from that of saline, and

significantly less than that of imipramine. We compared immobility behavior during the 5-

minute FST in rats that received saline (IN, n=8), imipramine (IP, 15 mg/kg, n=8) and TAT-D1-

D2-IPep (IN, n=6, 1.0 nmol/g). A 1-way independent groups ANOVA revealed a significant

difference between all groups (n=6-8 per group, F(2,19)=9.653, p<0.01). Post-hoc Newman-

Keuls multiple comparisons tests revealed significant differences between imipramine and saline

(P <0.001) and imipramine and TAT-D1-D2-IPep (P <0.05). No significant difference was found

between saline and TAT-D1-D2-IPep (p>0.05). Data is represented graphically in Figure 3.9

and summarized in Figure 3.10.

Page 82: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

72

Figure 3-8 The D1-D2 interfering peptide has an anti-immobility effect in the FST at an intranasal dose of 1.67

nmol/g.

Immobility behavioral of animals who received intranasal injections of TAT-D1-D2-IPep (dose = 1.67nmol/g) was

compared to those who received saline, TAT-Pep or IP injections of imipramine. Immobility behavior between groups

compared by 1-way independent groups ANOVA followed by post-hoc Newman-Keuls multiple comparisons tests. ***

p<0.001, ** p<0.01 compared to saline, ^^^ p<0.001, ^^ p<0.01 compared to TAT-Pep. Error bars represent SEM.

Numbers within bars represent number of animals per group.

Page 83: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

73

Figure 3-9 The D1-D2 interfering peptide does not have an anti-immobility effect in the FST at an

intranasal dose of 1.0nmol/g

Immobility behavioral of animals who received intranasal injections of TAT-D1-D2-IPep (dose = 1nmol/g) was

compared to those who received saline or IP injections of imipramine. Immobility behavior between groups

compared by 1-way independent groups ANOVA followed by post-hoc Newman-Keuls multiple comparisons

tests *** p<0.001 compared to saline, # p<0.05 compared to imipramine. Error bars represent SEM. Numbers

within bars represent number of animals per group.

Page 84: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

74

Figure 3-10 Efficacy of the D1-D2 interfering peptide at various doses in the FST: summary of findings.

Data Points in imipramine and saline group are combined from experiments outlined in Sections 3.4.1 – 3.4.4. For

each dose, complete results represented graphically in Figure 3.6 – 3.9. Data at each dose compared using 1-way

independent groups ANOVA. *** p <0.001, ** p<0.01, * p<0.01 compared to Saline, # p<0.05 compared to

imipramine. Numbers below data points are number of animals per group. Error bars represent SEM. Data from

treatment groups who received TAT-Pep excluded from figure for the purposes of clarity (Figure 3-7, 3-8).

Page 85: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

75

3.5 Experiment 5: Duration of the behavioral effect of the D1-D2

interfering peptide

We investigated the length of time that the D1-D2 interfering peptide remains

behaviorally active (has a detectable anti-immobility effect in the FST) after intranasal

administration. To do this, we increased the amount of time between the last intranasal injection

of either the D1-D2 interfering peptide or the 9-amino acid TAT peptide and the beginning of the

FST to 2, 3 or 4 hours. Overall results at all time points are summarized in Figure 3-14.

3.5.1 Behavioral Effect in FST 2 hours after intranasal administration

The D1-D2 interfering peptide had a significant anti-immobility effect in the FST two

hours after the final intranasal injection. We compared immobility behavior during the FST in

rats that had received saline (IN, 1 hr before FST, n=6), imipramine (15mg/kg, IP, 1 hr before

FST, n=6), TAT-D1-D2-IPep (1.67nmol/g, IN 2 hrs before FST, n=6) or TAT-Pep (1.67nmol/g,

IN, 2 hrs before FST, n=5). A 1-way independent groups ANOVA revealed a significant

difference between all groups (n=5-6 per group, F(3,19)=5.399, p<0.01). Post-Hoc Newman-

Keuls multiple comparisons tests revealed a significant difference between TAT-D1-D2-IPep

and saline (p<0.05), TAT-D1-D2-IPep and TAT-Pep (p<0.05), imipramine and saline (p<0.05)

and imipramine and TAT-Pep (p<0.05). No significant differences exist between imipramine and

TAT-D1-D2-IPep groups (p>0.05) or TAT-Pep and saline groups (p>0.05). Data is represented

graphically in Figure 3-11 and summarized in Figure 3-14.

3.5.2 Behavioral Effect in FST 3 hours after intranasal administration

Three hours after the last intranasal injection, the D1-D2 interfering peptide did not have a

significant anti-immobility effect in the FST. We compared immobility behavior during the 5-

Page 86: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

76

minute FST in rats that had received saline (IN, 1 hr before FST, n=6), imipramine (15mg/kg, IP,

1 hr before FST, n=6), TAT-D1-D2-IPep (1.67nmol/g, IN, 3 hrs before FST, n=7) or TAT- Pep

(1.67nmol/g, IN, 3 hrs before FST, n=5). A 1-way independent groups ANOVA revealed a

significant difference between immobility behaviors across all groups (n=5-6 per group,

F(3,20)=4.669, p<0.01). Post-Hoc Newman-Keuls Multiple Comparisons Tests indicated

significant differences between imipramine and saline groups (p<0.05), imipramine and TAT-

Pep groups (p<0.05) and imipramine and TAT-D1-D2-IPep groups (p<0.05) groups. No

significant differences were present between TAT-D1-D2-IPep and saline groups (p>0.05),

TAT-D1-D2-Ipep and TAT-Pep groups (p>0.05) or TAT-Pep and saline groups (p>0.05). Data is

represented graphically in Figure 3-12 and summarized in Figure 3-14.

3.5.3 Behavioral effect in the FST 4 hours after intranasal administration

Four hours after the last intranasal injection, the D1-D2 interfering peptide did not have a

significant anti-immobility effect in the FST. We compared immobility behavior during the FST

in rats that had received injections of saline (IN, 1 hr before FST, n=6), imipramine (15mg/kg,

IP, 1 hr before FST, n=6), TAT-D1-D2-IPep (1.67nmol/g, IN, 4 hrs before FST, n=6) or TAT-

Pep (1.67nmol/g, IN, 3 hrs before FST, n=6). A 1-way independent groups ANOVA revealed a

significant difference between immobility behaviors across all groups (n=5-6 per group,

F(3,19)=3.727, p<0.05). Post-Hoc Newman-Keuls multiple comparisons tests indicated

significant differences between imipramine and saline groups (p<0.05), imipramine and TAT-

Page 87: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

77

Figure 3-11 The D1-D2 interfering peptide has an anti-immobility effect in the FST 2 hours after intranasal

administration.

Saline and imipramine were administered via IN and IP injections, respectively, 1 hour before the FST. TAT-D1-

D2-IPep and TAT-Pep administered via IN injections 2 hours before the FST. Immobility behavioral counts

analyzed by 1 way independent groups ANOVA (p<0.01) followed by post hoc Newman-Keuls multiple

comparisons tests. * p<0.05 compared with saline, ^ p<0.05 compared with TAT-Pep. Error bars represent SEM.

Numbers within bars represent number of animals per group.

Page 88: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

78

Figure 3-12 The D1-D2 Interfering Peptide does not have an anti-immobility effect in the FST 3 hours after

intranasal administration.

Saline and imipramine were administered via IN and IP injections, respectively, 1 hour before the FST. TAT-D1-D2-

IPep and TAT-Pep administered via IN injections 3 hours before the FST. Immobility behavioral counts analyzed by 1

way independent groups ANOVA (p<0.01) followed by post hoc Newman-Keuls multiple comparisons tests. * p<0.05

compared with saline, ^ p<0.05 compared with TAT-Pep, # p<0.05 compared with imipramine. Error bars represent

SEM.

Page 89: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

79

Figure 3-13 The D1-D2 interfering peptide does not have an anti-immobility effect in the FST 4 hours after

intranasal administration

Saline and Imipramine were administered via IN and IP injections, respectively, 1 hour before the FST. TAT-D1-D2-

IPep and TAT-Pep administered via IN injections 4 hours before the FST. Immobility behavioral counts analyzed by 1

way independent groups ANOVA (p<0.05) followed by post hoc Newman-Keuls multiple comparisons tests. * p<0.05

compared with saline, ^ p<0.05 compared with TAT-Pep, # p<0.05 compared with imipramine. Error bars represent

SEM. Numbers within bars represent number of animals per group.

Page 90: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

80

Figure 3-14 The D1-D2 interfering peptide no longer has a behavioral effect in the FST 3 hours after it is

administered via intranasal injections.

Summarized data from Figures 3-11 to 3-13 and Figure 3-3. Immobility data from our original time point of 1 hour after

intranasal administration is included for the purposes of comparison. Imipramine and saline treatment groups received

IP and IN injections, respectively, 1 hour before the FST. Immobility behavioral data from each time point (1,2,3 and 4

hrs) analyzed via 1-way independent groups ANOVA followed by Newman Keuls post hoc tests. *** p<0.001, * p<0.05

compared to saline ^^ p<0.01, ^ p<0.05 compared to TAT-Peptide # p<0.05 compared to imipramine. Numbers below

data points represent number of animals per group. Error bars represent SEM.

Page 91: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

81

Pep groups (p<0.05) and imipramine and TAT-D1-D2-IPep groups (p<0.05). No significant

differences were present between TAT-D1-D2-IPep and saline (p>0.05), TAT-D1-D2-Ipep and

TAT-Pep (p>0.05) or TAT-Pep and saline (p>0.05). Data is represented graphically in Figure 3-

13 and summarized in Figure 3-14.

3.6 Experiment 6: The D1-D2 interfering peptide does not increase

locomotor activity

In order to investigate whether the anti-immobility effect of the D1-D2 interfering

peptide in the FST was due to its specific anti-depressant effects, we examined the effect of the

D1-D2 interfering peptide on locomotor activity. To test this, we compared the amount of

locomotor activity during a 30-minute test in animals given intranasal injections of saline (n=5),

TAT-Pep (IN, 2nmol/g, n=5), TAT-D1-D2-IPep (IN, 2nmol/g, n=5) or imipramine (IP, 15mg/kg,

n=5).

3.6.1 Overall locomotor activity

Animals in the D1-D2 interfering peptide, TAT-peptide or imipramine treatment groups

had significantly lower overall locomotor activity during the 30-minute test than animals in the

saline group. A 1-way independent groups ANOVA of locomotor activity (as measured by Beam

Breaks) revealed a significant difference between all four treatment groups (n=5 per group,

F(3,16)=9.775 p<0.001). Post-Hoc Newman-Keuls multiple comparisons tests revealed

significant differences between TAT-D1-D2-IPep and saline groups (p<0.001), TAT-Pep and

saline groups (p<0.01), and imipramine and saline groups (p<0.001). No significant difference

was present between imipramine, TAT-Pep or TAT-D1-D2-IPep groups (p>0.05 for all

comparisons). Data is represented graphically in Figure 3-15A.

Page 92: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

82

to test whether the D1-D2 interfering peptide’s effect on locomotor activity was consistent across

different doses, we compared the locomotor activity of animals in treatment groups that received

the D1-D2 interfering peptide at two doses: 2.0 nmol/g (n=5) and 1.67nmol/g (n=5). A two-

tailed, unpaired student’s t-test revealed no significant differences between overall locomotor

activity in these groups (n=5 per group, t(8)=1.203, p>0.05) (Figure 3-15B).

3.6.2 Effect of time on locomotor activity during 30-minute test

We examined locomotor activity across the treatment groups during the open-field test in

5-minute intervals. There was a significant difference in the amount of locomotor activity across

different treatment groups, as well as the amount at different time points in the test. However, all

treatment groups displayed the same pattern of locomotor activity (highest at the beginning of

the test) (Figure 3-16). Locomotor activity at various time course data was analyzed by 2-way

Independent Groups ANOVA, with treatment groups (saline, imipramine, TAT-D1-D2-IPep or

TAT-Pep) and time point (5min, 10min, 15min, 20min, 25min, 30min) as independent factors.

The analysis revealed a significant main effect of treatment groups (F(3,96)= 20.24, p<0.001)

and time point (F(5,96)= 30.66, p<0.001). The interaction between treatment group and time

point was not significant (F(15,96)=1.06, p>0.05).

Page 93: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

83

Figure 3-15 The D1-D2 interfering peptide does not increase locomotor activity during a 30-minute open field

test.

(A) Total locomotor activity during a 30-minute open field test. Locomotor activity was assessed during a 30-

minute open field test in a novel environment, and measured by the amount of movement around the testing

chamber (Beam Breaks). Animals were given intranasal injections of saline, TAT-D1-D2-IPep or TAT Pep

(2.0nmol/g) or IP injections of imipramine 24 hours, 19hours and 1 hour before the open field test. Locomotor

activity data was analyzed by 1-way independent groups ANOVA (p<0.001) followed by post-hoc Newman-Keuls

multiple comparisons tests. *** p<0.001, ** p<0.01, compared to saline. (B) The TAT-D1-D2-IPep has a similar

effect in the open field test at 2.0nmol/g and 1.67nmol/g. Differences in locomotor activity produced by TAT-D1-

D2-IPep dose evaluated by unpaired, two-tailed Student’s t-test (p>0.05).

Page 94: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

84

Figure 3-16 The D1-D2 interfering peptide decreases overall locomotor activity but does not change the

activity pattern during a 30-minute open field test.

Each Point on the test represents total number of beam breaks during the previous 5-minutes of the open field

test. Data across treatment group and time points analyzed using 2-way independent groups ANOVA with

time point and treatment group as independent factors. There was a significant main effect for time course

(p<0.001) and treatment group (p<0.001) but no significant interaction between the two factors. n=5 animals

per group. TAT-D1-D2-Ipep and TAT-Pep were given at an intranasal dose of 2.0nmol/g. Error bars represent

SEM.

Page 95: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

85

3.7 Experiment 7: Intranasal administration of the D1-D2 interfering

peptide disrupts the interaction between dopamine D1 and D2 receptors

in the PFC

The D1-D2 interfering peptide is designed to disrupt the interaction between the

Dopamine D1 receptor and the Dopamine D2-Long Receptor isoform by interacting with a 15-

amino acid segment in the third intracellular loop of the D2L receptor.3 In previous studies from

our laboratory, administering this peptide directly to the PFC decreased the interaction between

dopamine D1 and D2 receptors in the PFC (as assessed by co-immunoprecipitation) and had an

anti-immobility effect in the FST (see Figure 1-4).3 The D1-D2 interfering peptide also had a

significant antidepressant effect in the FST after intranasal administration (Figure 3-3, 3-10),

thus we examined whether there was a concurrent decrease in the D1-D2 dopamine receptor

interaction in the PFC after intranasal administration of the D1-D2 interfering peptide.

A representative immunoblot of D1R (anti-D1DR) immunoprecipitated by anti-D2DR is

shown in Figure 3-17A. Densitometry analysis of immunoblots revealed that the detectable

interaction between the Dopamine D1 and D2 receptors was significantly reduced in prefrontal

tissue from animals that received TAT-D1-D2-IPep compared with animals who received saline

(Student’s t-test, n=3 per group, t(4)=3.872, p=0.018) (Figure 3-17B).

3.8 Experiment 8: The D1-D2 interfering peptide does not change the

expression of dopamine D1 or D2 receptors in the PFC

After intranasal administration, the D1-D2 interfering peptide can disrupt the interaction

between Dopamine D1 and D2 receptors (Figure 3-17). We tested if this disruption is due to the

D1-D2 interfering peptide’s pharmacological effect or due to a change in the expression of either

Page 96: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

86

Figure 3-17 Co-Immunoprecipitation of D1 by anti-D2R is reduced in the PFC of animals who received

intranasal injections of TAT-D1-D2-IPep (Dose: 1.67nmol/g)

(A) Representative immunoblot of anti-D2DR immunoprecipitated tissue from the PFC of rats that received

intranasal injections of saline (n=3) or TAT-D1-D2-Ipep (IN, 1.67nmol/g, n=3). Input lane: 75 µg solubilized PFC

tissue, IgG: tissue incubated with non-specific immunoglobulin antibody. (B) The interaction between D1 and D2R

is significantly reduced in the PFC of animals who received TAT-D1-D2-IPep. The interaction between D1 and

D2R quantified by densitometry analysis of immunoblots. All samples were standardized to control (saline) samples

and analyzed by two-tailed, unpaired student’s T-test (n=3 per group=0.018).

Page 97: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

87

the dopamine D1 or D2 receptor proteins. To do this, we evaluated the expression of D1 and D2

receptors in the PFC of animals that were given intranasal injections of TAT-D1-D2-IPep (n=3)

or saline (n=3) using Western Blot analysis.

3.8.1 Expression of Dopamine D1 receptors in the PFC after intranasal

administration of the D1-D2 interfering peptide

We compared prefrontal expression of D1 in the same subsample of animals used in the

co-immunoprecipitation experiment (Section 3.8). The Western Blot analysis showed no change

in the overall expression of the D1 dopamine receptor in the PFC after intranasal administration

of the D1-D2 interfering peptide (IN, 1.67nmol/g, n=3) compared with saline (IN, n=3)

(Student’s t-test, t(4)=0.167, p>0.05), Figure 3-18A-B.

3.8.2 Expression of Dopamine D2 receptors in the PFC after intranasal

administration of the D1-D2 interfering peptide

We compared prefrontal expression of D2 in the same subsample of animals used in the

co-immunoprecipitation analysis described in Section 3.7. We found no change in the overall

expression of D2 dopamine receptors in the PFC after intranasal administration of the D1-D2

interfering peptide (IN, 1.67nmol/g, n=3) compared with saline (IN, n=3) (Student’s t-test,

t(4)=0.009, p>0.05), Figure 3-19A-B. We also performed an immunoblot for α-tubulin (Figure

3-20) to verify that the amount of protein in each sample was equivalent.

Page 98: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

88

Figure 3-18 Intranasal administration of the D1-D2 interfering peptide does not change the expression of the

dopamine D1 receptor in the PFC.

(A) Representative immunoblot of D2R in PFC tissue from animals who received intranasal injections of TAT-D1-

D2-IPep (IN, 1.67nmol/g, n=3) or saline (IN, n=3). (B) Expression of D2 is unchanged in animals who received

intranasal injections of TAT-D1-D2-IPep compared with those who received Saline (Control). Data quantified using

densitometry and analyzed using unpaired, two-tailed student’s t-test (n=3 per group, p>0.05). Error bars represent

SEM.

Page 99: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

89

Figure 3-19 Intranasal administration of the D1-D2 interfering peptide does not change the expression of the

dopamine D2 Receptor in the PFC

(A) Representative Immunoblot of D1 in PFC tissue from animals who received intranasal injections of TAT-D1-

D2-IPep (IN, 1.67nmol/g, n=3) or saline (IN, 1.67nmol/g, n=3). (B) Expression of D2 is unchanged in animals who

received intranasal injections of TAT-D1-D2-IPep compared with those who received saline (control). Data

quantified using densitometry and analyzed using unpaired, two-tailed student’s t-test (n=3 per group, p>0.05). Error

bars represent SEM.

Page 100: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

90

Figure 3-20 Representative immunoblot of α-tubulin expression in rat PFC tissue.

Rats received intranasal injections of saline (IN, n=3) or TAT-D1-D2-IPep (IN, 1.67nmol/g, n=3). 20 µg protein

from PFC of each animal resolved using SDS page and immunoblotted with anti- α-tubulin. α-tubulin levels were

used as a loading control for experiments assessing expression of D1 or D2R in the PFC.

Page 101: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

91

4 Discussion

4.1 Overall Findings

This project addressed whether we can effectively deliver peptide therapies designed to

disrupt pathological interactions between two cell membrane receptors to the CNS in a non-

invasive manner. We show that the D1-D2 interfering peptide, designed to disrupt the interaction

between the D1 and D2 dopamine receptors3, has an antidepressant effect in the FST after

intranasal delivery (Figure 3-3 and 3-10). These findings, along with experiments investigating

the pharmacological and biological properties of the D1-D2 interfering peptide, provide a solid

basis for further development of this therapy as a novel treatment for MDD.

We demonstrated that the POD used in this study deposits substances preferentially on

the olfactory epithelium and that a FLAG-tagged biologically active peptide can be detected in

the PFC after intranasal administration. Administration of the D1-D2 interfering peptide at doses

greater than 5.75 mg/kg (1.67nmol/g) had a significant anti-immobility effect that was

comparable to that of imipramine in the FST, for two, but not three, hours after intranasal

administration. The D1-D2 interfering peptide, the TAT-peptide and imipramine all significantly

decreased locomotor activity during a 30-minute locomotor activity test, suggesting that these

substances do not increase the overall occurrence of motor movements and that the anti-

immobility effect observed in the FST is specific to its antidepressant effects. Together, these

finding demonstrated the efficacy of the intranasal pathway for peptide delivery to the CNS,

expanded upon our laboratory’s previous findings that the D1-D2 interfering peptide had an

antidepressant effect in animal models of depression, and developed a non-invasive, clinically

applicable method to deliver this peptide to the CNS.3

The D1-D2 interfering peptide significantly disrupted the interaction between D1 and D2

in the PFC without changing the expression of either receptor, indicating that the antidepressant

Page 102: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

92

effect of the peptide may be due to its ability to disrupt this protein-protein interaction. I will

discuss the implications and the shortcomings of these findings and propose future experiments

to further understand the pharmacological effects of the D1-D2 interfering peptide.

4.2 The POD delivers biologically active peptides to the CNS

The POD preferentially deposits substances on the olfactory epithelium and surrounding

tissue when using the administration protocol we developed (outlined in Section 2.2.1). After

intranasal delivery with the POD, Mark-It tissue dye was preferentially deposited onto the

olfactory epithelium and surrounding tissue (Figure 3.1). Numerous previous studies regarding

protein and peptide delivery to the CNS have indicated that proteins such as NGF194,195

, BDNF196

and IGF-1197,198

preferentially enter the CNS through the extracellular channels between the

ORN axons and the OECs protecting them from the cribiform plate.16

It is probable that

preferentially depositing substances onto the olfactory epithelium, as the POD does, where ORN

axons originate would increase the amount of the D1-D2 interfering peptide delivered to the

CNS while minimizing loss to the periphery.

Next, we investigated whether we could detect the D1-D2 interfering peptide in the PFC

after intranasal administration. To do this, we added an 8-amino acid, immunoreactive FLAG215

tag to the C-terminal end of the D1-D2 interfering peptide. We detected fluorescent Cy-2-

conjugated antibodies for FLAG both intracellularly and in extracellular areas of PFC coronal

sections (Figure 3-2). As well, the immunofluorescence was visible in most areas of the PFC

coronal section, with no extreme variations in fluorescent signals between dorsal and ventral,

lateral or medial PFC areas. We focused on investigating whether the D1-D2-FLAG peptide

could be detected in the PFC because in our laboratory’s previous study, the D1-D2 interfering

Page 103: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

93

peptide only had an antidepressant effect when infused directly to the PFC and not in other brain

areas.3

These experiments were designed to qualitatively assess where substances are deposited

by the POD, and whether they can be transported to the CNS. As a result, we are unable to make

direct conclusions about the quantitative aspects of this process, such as the efficiency of

delivery to the CNS, and the mechanisms through which this occurs. One of the shortcomings of

this method of detecting the D1-D2-FLAG peptide in the PFC is that we were not able to

quantify the amount of D1-D2 interfering peptide effectively delivered to the PFC. Thus, the

proportion of the original intranasal dose of the D1-D2-FLAG peptide present after intranasal

administration is not known. In addition, these experiments did not provide any information

about the mechanism through which the D1-D2 interfering peptide gains access to the central

nervous system.

These results do not provide a quantitative measure of the efficiency of delivery to the

CNS after intranasal administration. However, we observed an anti-immobility effect in the FST

at an intranasal D1-D2 interfering peptide dose equal to or greater than 1.67nmol/g. This dose

was approximately 100-fold larger than that given directly to the PFC (5nmol per injection) in

Pei et al’s previous study.3 We chose this dose based on previous studies of intranasal

administration of small proteins such as BDNF, NGF and insulin to the CNS that indicated that

the efficiency of delivery to the CNS is between 1 and 5%.192,194,196,212,217

Using these studies, we

estimated that approximately 1-5% of the dose originally given intranasally will be delivered to

the CNS in a biologically-active form (see Appendix 1 for detailed calculations) and our

subsequent results (see Figure 3-10) are consistent with this estimate.

Page 104: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

94

4.3 Mechanism of transport to the CNS after intranasal administration

The results outlined above show that the POD preferentially deposits substances on the

olfactory epithelium, and, hypothetically, once there, they are transported to the CNS via

extracellular pathways around the olfactory receptor neuron axons (see section 1.3 for review of

the intranasal pathways to the CNS). However, the mechanism by which the D1-D2 interfering

peptide is transported to the CNS and PFC is not known. As stated above, it is likely that the

peptide is being transported to the CNS via extracellular transport pathways. This hypothesis is

supported by our experimental observations that the POD preferentially deposits substances on

the olfactory epithelium, the relatively short (1 hour) amount of time required for transport to the

PFC, and previous studies demonstrating that peptides similar to the one used in this study are

also transported to the CNS via extracellular mechanisms.203,212

Alternately, the peptide could be

transported by the ORNs via intracellular mechanisms, a process that would require diffusion of

the peptide into the ORNs and subsequent transport via axonal transport mechanisms to the axon

terminals in the olfactory bulbs.15

This is unlikely, as previous studies have shown that

intracellular transport of this type takes significantly longer than 1 hour.15,218

An alternate possibility is that the D1-D2 interfering peptide is absorbed into the systemic

circulation through the nasal capillary bed, as the nasal cavity contains extensive capillaries and

vascularization.15

After uptake into the bloodstream, the peptide would be transported throughout

the body via the systemic circulation, resulting in diffuse administration of the peptide and

proportionally less of the intranasal dose transported to the CNS.15

Second, the peptide would

need to cross the BBB, and, although peptides containing a membrane-permeable TAT sequence

are permeable to the BBB214

, they require an extremely high systemic dose to enter the CNS in

appreciable amounts. This is supported by a number of studies from our laboratory suggesting

that TAT-linked peptides need to be administered at a systemic dose of ≥ 3nmol/g in order to be

Page 105: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

95

transported to the CNS, and have any detectable CNS-mediated pharmacological effects in vivo

(unpublished data, Liu laboratory, 2008-2013). In the present study, the minimum effective dose

we tested was 1.67nmol/g, 55% that of the typical systemic dose. This suggests that our

intranasal delivery method has some specificity to the CNS, although it is not conclusive as we

have not yet tested the antidepressant effects of the D1-D2 interfering peptide after systemic

administration. In conclusion, although it is likely that after intranasal administration some of the

D1-D2 peptide is taken up into the circulation, it is unlikely that this is the principle mechanism

of transport to the CNS.

A recent study by Yang et al212

showed that for a 22-amino acid, TAT-linked membrane

permeable peptide similar in size to the D1-D2 interfering peptide, an intranasal dose only 7%

that of the IV doses previously administered219

was able to alleviate hypoxia-induced ischemic

brain injury in a rat preclinical model. Yang et al212

also showed that their TAT-linked peptide

was detectable in the olfactory bulbs and anterior brain areas such as the PFC 10-30 minutes

after delivery, a result that suggests that their peptide was being transported to the CNS via the

extracellular pathways between the olfactory epithelium and the CNS. Overall, when the results

from this project are evaluated within the context of the scientific literature regarding intranasal

delivery of peptides of this type, it seems likely that the D1-D2 interfering peptide is being

delivered to the CNS via extracellular transport pathways along olfactory receptor axons.

4.4 The D1-D2 interfering peptide is effective at intranasal doses ≥ 5.75

mg/kg for up to 2 hours after intranasal administration.

The D1-D2 interfering peptide has a significant anti-immobility effect in the FST at doses

greater than or equal to 1.67nmol/g (5.75 mg/kg) (Figure 3-10). At these doses, the

antidepressant effect in the FST was comparable to that of imipramine, a clinically effective

Page 106: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

96

tricyclic antidepressant. We tested if the behavioral effect of the D1-D2 interfering peptide in the

FST would differ at doses higher or lower than the original dose (1.67nmol/g) we tested. We did

not observe any appreciable increase or decrease in immobility behavior at D1-D2 interfering

peptide doses greater than 1.67nmol/g. Thus, there may be some threshold dose of the D1-D2

interfering peptide that is required to produce an anti-immobility effect in the FST, beyond

which no additional antidepressant efficacy is conferred. This experiment also allowed us to

identify the minimum intranasal dose of the D1-D2 interfering peptide with maximal behavioral

effect in the FST as 1.67nmol/g.

The lowest intranasal dose of the D1-D2 interfering peptide we tested was 1.0nmol/g. At

this dose, we did not observe a significant difference between the behavior of animals who

received the D1-D2 interfering peptide and those who received saline (Figure 3-9). However,

the mean amount of immobility in the saline group was greater than in the D1-D2 interfering

peptide group. (Figure 3-9, 3-10) It is possible that the D1-D2 interfering peptide has some sub-

threshold antidepressant effect in the FST when administered at this dose. In order to test this

hypothesis and to gain a more complete understanding of the antidepressant efficacy of the D1-

D2 interfering peptide at various intranasal doses, doses of the D1-D2 interfering peptide lower

than 1.0nmol/g should be tested in the FST.

We evaluated the duration of the effect of the D1-D2 interfering peptide in the FST. We

show that at the minimally effective intranasal dose, 1.67nmol/g, the D1-D2 interfering peptide

has an anti-immobility effect in the FST for two, but not three, hours after intranasal

administration. (Figure 3-14) At the 3 and 4 hour time points, we were no longer able to detect a

significant anti-immobility effect in animals that were given intranasal administrations of the D1-

D2 interfering peptide. The absence of a behavioral effect in the FST 3 or 4 hours after

intranasal administration is likely due to degradation of the D1-D2 interfering peptide as it is

Page 107: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

97

transported from the olfactory epithelium to the central nervous system. The mucous membranes

in the nasal cavity and covering the olfactory epithelium contain proteases capable of degrading

peptide bonds.15

Furthermore, small peptides like the D1-D2 interfering peptide disrupt the

interaction between two proteins by competitively binding to the interacting regions, and, as

such, must remain properly folded in order to effectively bind to these regions. Likely, the longer

the D1-D2 interfering peptide in the body, the more degradation occurs, resulting in loss of its

pharmacological effects in the CNS. Thus, strategies to improve the stability of the D1-D2

interfering peptide in the CNS will be useful in increasing the duration of its antidepressant

effect.

4.5 Possible neurobiological mechanisms of the D1-D2 interfering peptide’s

antidepressant effect

In Pei et al.’s study,3 they document an increase in D1-D2 interaction in the striatum of

patients with MDD. This observation is correlative in nature and could be due to neurobiological

changes from antidepressant treatments or a result of other confounding factors inherent in

human studies. As such, no causal role for the D1-D2 in the pathogenesis of MDD in the

striatum has been shown. Beyond the D1-D2 interfering peptide’s ability to disrupt the

interaction between D1 and D2 in the PFC, the cellular and neurobiological mechanisms leading

to its antidepressant efficacy remain unknown. It is unclear how disrupting the D1-D2 interaction

translates into a behavioral antidepressant effect in the FST and, in Pei et al’s previous study, the

LH task.3

However, a number of studies have indicated that the D1-D2 heterodimer results in

increases in intracellular calcium levels through activation of the Gq – PLC pathway.2 Increases

in cytoplasmic calcium levels have a myriad of effects within neurons, including changing

Page 108: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

98

neuronal excitability, activating intracellular processes such as transcription and translation of

proteins and trophic factors.220

Any of these sub-cellular changes could have effects on

mesocortical dopaminergic circuits and the higher-order cognitive processes, thereby having an

effect on the neurobiology of MDD.70,152

One possible mechanistic explanation for the antidepressant effects of the D1-D2

interfering peptide is that it causes an increase in expression of BDNF in the PFC and other brain

areas. Unpublished data from our laboratory indicates that after ICV delivery of the D1-D2

interfering peptide to animals undergoing the LH task, BDNF protein levels were elevated in

striatal tissue compared to animals who had received saline infusions. (T.Lai, Liu lab,

Unpublished Data, 2011). Presumably, the D1-D2 interfering peptide has this effect because it

blocks intracellular pathways that inhibit BDNF expression. Furthermore, evidence from post-

mortem human studies indicate that BDNF serum levels are decreased in the PFC of patients

who had MDD and committed suicide, and increased in patients on long term antidepressant

treatment.159-161

This hypothesis remains unproven, as a study by Hasbi et al. 118

contradicts these findings

and demonstrates a link between activation of the D1-D2 heterodimer and subsequent increases

in BDNF expression in the NAc. The authors hypothesize that an increase in intracellular

calcium caused by activation of the D1-D2 heteromer results in phosphorylation of calmodulin

Kinase II (CamKII), which then acts as a transcription factor and activates transcription of the

BDNF gene.118

One limitation of Hasbi et al’s118

study is that it was conducted in cultured

neurons from the ventral striatum, and, as such, may not be generalizable to the in vivo

pathogenesis of complex psychiatric conditions such as MDD.

A number of studies have demonstrated that directly infusing BDNF into the NAc and the

VTA results in a ‘pro-depression’ phenotype in mice, increasing social aversion

Page 109: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

99

behaviors.121,169,170

Overall, these contradictory results suggest that the effect of BDNF on the

pathogenesis of MDD is complex and may depend on the brain region where it is expressed.

Thus, the specific pathway linking D1-D2 heteromerization and upregulation or down-regulation

of BDNF expression with the pathogenesis of MDD remains unknown.

The intranasal administration method used to deliver the D1-D2 interfering peptide to the

brain is much less anatomically specific than the direct microinjections to the PFC used in Pei et

al’s study.3 Thus, it is likely that, after intranasal administration, the D1-D2 interfering peptide

was transported diffusely to numerous brain areas after intranasal administration. As a result, it is

possible that the anti-immobility effect we observed in the FST may not be solely due to the

effect of the D1-D2 interfering peptide in the PFC, but also to its concurrent effects in other brain

areas.

There are numerous lines of evidence that implicate both the NAc and the mesolimbic

dopaminergic reward system in the pathogenesis of MDD.3,80,152

In the NAc, there is a

population of neurons that express both D1 and D2 receptors where these receptors co-

localize.116,118,119

It is possible that, after intranasal administration, the D1-D2 interfering peptide

interferes with the interaction between D1 and D2 in the PFC and these other areas. To better

understand the regions and mechanisms involved in the antidepressant effect of the D1-D2

interfering peptide, biochemical studies in brain areas other than the PFC along with studies

investigating the mechanisms behind the antidepressant effect of the D1-D2 interfering peptide

should be conducted.

4.6 Limitations of the FST as a preclinical test for antidepressant efficacy

It is important to note that the behavioral test we used throughout this study, the FST, is

not a model of MDD. Instead, it is a pharmacologically valid test for antidepressant efficacy, as

Page 110: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

100

all clinically effective therapeutics have the same behavioral effect in the FST, that of decreasing

the time spent immobile during the test.171

One disadvantage of the FST is that it lacks

etiological validity, as it does not accurately model the clinical symptoms and manifestation of

MDD.79,171

Notwithstanding, the FST was an appropriate test for this study, as we were

examining whether intranasal administration of the D1-D2 interfering peptide would result in a

behavioral antidepressant effect in the FST, as well as evaluating pharmacological parameters of

this administration method.

A disadvantage of using the FST is that we were unable to examine whether the D1-D2

interfering peptide, after intranasal administration, would continue to have an antidepressant

effect in a chronic animal model of depression such as chronic mild stress or the LH task. We

hypothesize that this is likely the case as intra-cerebral administration of the D1-D2 interfering

peptide decreased escape failures in the 5-day LH Task.3 Additionally, the acute nature of the

FST also presented limitations in investigating whether intranasal administration of the D1-D2

interfering peptide has any appreciable neurobiological changes other than that of disrupting the

interaction between D1 and D2 receptors in vivo. Future studies investigating the antidepressant

efficacy of the D1-D2 interfering peptide should examine its effects in a chronic model of MDD

such as chronic mild stress paradigms.

4.7 The D1-D2 interfering peptide, TAT-peptide and imipramine

significantly decrease locomotor activity

We tested whether the anti-immobility effect of the D1-D2 interfering peptide and of

imipramine during the FST was due to the specific antidepressant effects of these treatments or

due to an overall increase in locomotor activity. To determine this, we tested the effect of these

treatments in a 30-minute locomotor activity test (Figure 3-15). Surprisingly, we found that the

Page 111: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

101

D1-D2 interfering peptide, TAT-peptide and imipramine all significantly decreased locomotor

activity during a 30-minute open field test (Figure 3-15). When we examined the animals’

locomotor activity over time, we found no significant interaction between treatment and time

(p>0.05, 2-way Independent Groups ANOVA), indicating that while the amount of locomotor

activity was decreased in animals that received imipramine, TAT peptide or D1-D2 interfering

peptide compared with saline, the behavioral patterns over time did not change dramatically

(Figure 3-16).

In the previous study done in our laboratory, direct microinjections of the D1-D2

interfering peptide or the TAT-peptide to the PFC, NAc or hippocampus did not significantly

alter locomotor activity during a 30-minute open field test.3 Animals in the imipramine treatment

group displayed decreased locomotor activity in the open field test, a result consistent with other

studies of imipramine’s sedative effects.3,221

Thus, the difference in our results may be due to

differences in testing methodology between this study and our laboratory’s previous study.3

These include the method of administration (Intranasal administration vs. direct microinjections),

the intranasal doses of the D1-D2 interfering peptide and TAT peptide were much larger than

those administered directly to the PFC, and the repeated use of isoflurane anesthetic in all

groups.

Repeated use of isoflurane anesthetic can result in changes in animals’ behavior, and

studies have demonstrated changes in animals’ performance on learning and memory tasks but

not on locomotor activity tests.222,223

What may be more likely is that repeated exposure to

isoflurane anesthesia is an aversive, stressful experience for the animal that, coupled with

exposure to the FST, could result in changes in the animal similar to those that occurring in

animals exposed to a chronic mild stress (CMS) paradigm.224

Some CMS protocols induce

decreases in spontaneous locomotor activity and exploratory behavior in the open field test.225-227

Page 112: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

102

Although this hypothesis may support our observation of decreased activity in the open field test,

not all chronic mid stress paradigms cause a decrease in locomotor activity and this effect has not

be replicated.224,228

Additionally, if combined exposure to anesthetic, intranasal administration

and the FST resulted in a CMS-like response in the animals, we would expect the locomotor

activity of animals to be equivalent in all treatment groups, which was not the case in our

experiment.

Although it is likely that aversive experiences from repeated intranasal administrations

under anesthetic and exposure to the FST cause stress in the experimental animals and

contributes to these results, it may not be the only factor at play. To determine if this is the case,

we could test this hypothesis by repeating the locomotor activity experiment using

experimentally naive animals. If all groups have the same locomotor activity profiles, then it can

be concluded that the stress caused by the previous experiments resulted in the observed decrease

in locomotor activity.

A second possibility is that the D1-D2 interfering peptide has an effect on the function of

dopamine in the striatum and reduces the occurrence of motor movements by inhibiting the

functions of the nigrostriatal dopamine pathway. As discussed above, it is likely that after

intranasal administration and transport to the CNS, the D1-D2 interfering peptide and other

substances are delivered to numerous brain areas in a diffuse manner, thus it is probable that the

D1-D2 peptide was present in some amount in the striatum after intranasal administration.

Furthermore, D1 and D2 receptors are known to complex in a specific subset of medium spiny

neurons in the basal ganglia and striatum.108

It is possible that this subset of neurons, as Perrault

et al.108

suggest, are responsible for mediating some aspects of dopaminergic signaling within the

nigrostriatal pathway. Thus, signaling through the D1-D2 receptor complex may mediate

Page 113: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

103

locomotor activity, and disruption of this signaling may reduce the incidence of locomotor

activity, although no studies have been done directly address this hypothesis.

This hypothesis could account for the decreased locomotor activity observed in the D1-

D2 interfering peptide treatment group but it does not address the decrease in locomotor activity

observed in animals treated with the TAT peptide. Secondly, this explanation may be unlikely

because the D1-D2 interfering peptide had a robust antidepressant effect (i.e. increased mobility)

in the FST. If it had caused a reduction in locomotor activity through the nigrostriatal pathway,

then it is unlikely we would have observed an anti-immobility effect in the FST, as the

occurrence of motor movements would have been reduced in a global manner.

Another possible explanation for our results is that they are an artifact of the experimental

design we used. In order to maintain consistency between the studies, we followed the protocol

used in our laboratory’s previous study of the D1-D2 interfering peptide’s antidepressant effect

(see Section 2.7 for detailed description).3 Briefly, animals were administered D1-D2 interfering

peptide, TAT-Peptide, saline or imipramine 24 hours, 19 hours and 1 hour before the 30-minute

locomotor activity test. This may not have been the best approach, as the locomotor activity

changes observed in the treatment groups could have been due to the animal’s exploration of a

novel environment and not due to any real changes in locomotor behavior across the treatment

groups.

The above is supported by our experimental data, as after 15 minutes in the novel

locomotor activity box, there was no difference in locomotor activity between any of the

treatments groups (Figure 3-16). To test this theory, we could repeat the locomotor activity

experiment and include a sensitization period where the animal is exposed to the locomotor

activity testing apparatus, before the three treatment administrations and subsequent locomotor

activity test. This method may also be a better control for activity in the FST paradigm because

Page 114: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

104

animals are replaced in the same plexiglass cylinder on Day 2 of the test as on Day 1, and thus

not exposed to a novel environment during the 5-minute test for antidepressant efficacy.

4.8 Future Directions

The results of this project raise a number of interesting questions for future investigation.

Although this project demonstrated that the intranasal pathway can be used to deliver peptide

therapies to the central nervous system, we did not directly investigate the mechanisms through

which these peptides gain access to the CNS after intranasal delivery. A radioactively-labeled

peptide with the same sequence as the D1-D2 interfering peptide could be used to address this

question, as its pathway to the central nervous system and its presence in various brain areas

could be easily traced and quantified by measuring the resulting levels of radioactivity in these

tissues. This strategy has been successfully used to trace the intranasal transport route of insulin

to the CNS and identify the areas with the most accumulation within the CNS.190,192

A second area warranting further investigation is strategies for improving the efficiency

of D1-D2 interfering peptide transport to the central nervous system after intranasal delivery.

This would allow us to reduce the minimum effective dose of the D1-D2 interfering peptide,

thereby decreasing the potential toxicity of the peptide and accumulation in non-CNS tissues.

One strategy to improve the efficiency of delivery to the CNS is using mucoadhesive solutions to

increase the residence time of the D1-D2 interfering peptide at the olfactory epithelium.229

Mucoadhesive solutions of this type have been shown to increase the proportion of the total dose

of proteins or peptides delivered to the CNS after intranasal administration.229-231

Finally, this project and previous studies done in our laboratory have demonstrated that

the D1-D2 interfering peptide selectively uncouples the D1-D2 receptor heterodimer and has an

antidepressant effect in the FST and other models of depression.3 Other laboratories have

Page 115: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

105

investigated the downstream effects of the D1-D2 receptor heterodimerization, and shown that it

causes an increase in cytoplasmic calcium levels via a Gq-dependent mechanism.1,2

Beyond

these observations, there is currently little known about the pathological role of the D1-D2

interaction in MDD, or the exact mechanisms behind the antidepressant effect of the D1-D2

interfering peptide. More studies both in preclinical models and in vitro at the cellular level will

be necessary to fully understand why the D1-D2 interfering peptide has an antidepressant effect.

Although intranasal delivery of the D1-D2 interfering peptide has an antidepressant effect in the

FST, it is unclear whether it will be effective in other preclinical models of MDD when delivered

intranasally. The antidepressant effect of the D1-D2 interfering peptide after intranasal

administration should be tested in other animal models of MDD, such as the LH task and social

aversion paradigms.

Overall, this project has raised a number of interesting questions about the antidepressant

mechanism of the D1-D2 interfering peptide and the intranasal pathway used to deliver these

peptides to the central nervous system. At the same time, the results presented here demonstrate

that the D1-D2 interfering peptide can have an antidepressant effect after intranasal delivery, in a

pharmacologically valid preclinical model. Furthermore, these results have provided important,

clinically relevant results vital to the development of the D1-D2 interfering peptide as a novel

therapeutic for MDD, warranting further preclinical studies should be conducted to conclusively

determine whether the D1-D2 interfering peptide can be tested as a novel antidepressant in the

clinical setting.

Page 116: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

106

Page 117: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

107

References

1 Hasbi, A., O'Dowd, B. F. & George, S. R. Heteromerization of dopamine D2 receptors

with dopamine D1 or D5 receptors generates intracellular calcium signaling by different

mechanisms. Current opinion in pharmacology 10, 93-99 (2010).

2 Lee, S. P. et al. Dopamine D1 and D2 receptor Co-activation generates a novel

phospholipase C-mediated calcium signal. The Journal of biological chemistry 279,

35671-35678 (2004).

3 Pei, L. et al. Uncoupling the dopamine D1-D2 receptor complex exerts antidepressant-

like effects. Nature medicine 16, 1393-1395 (2010).

4 Beaulieu, J. M. & Gainetdinov, R. R. The physiology, signaling, and pharmacology of

dopamine receptors. Pharmacological reviews 63, 182-217 (2011).

5 Missale, C., Fiorentini, C., Collo, G. & Spano, P. The neurobiology of dopamine

receptors: evolution from the dual concept to heterodimer complexes. Journal of receptor

and signal transduction research 30, 347-354 (2010).

6 Maggio, R., Aloisi, G., Silvano, E., Rossi, M. & Millan, M. J. Heterodimerization of

dopamine receptors: new insights into functional and therapeutic significance.

Parkinsonism & related disorders 15 Suppl 4, S2-7 (2009).

7 Niciu, M. J., Ionescu, D. F., Mathews, D. C., Richards, E. M. & Zarate, C. A. Second

messenger/signal transduction pathways in major mood disorders: moving from

membrane to mechanism of action, part I: major depressive disorder. CNS spectrums, 1-

10 (2013).

8 Wong, A. H. & Liu, F. Uncoupling the dopamine D1-D2 receptor complex: a novel target

for antidepressant treatment. Clinical pharmacology and therapeutics 91, 298-302

(2012).

9 Mann, J. J. The medical management of depression. The New England journal of

medicine 353, 1819-1834 (2005).

10 Cuijpers, P. The patient perspective in research on major depression. BMC psychiatry 11,

89 (2011).

11 Mojtabai, R. Unmet need for treatment of major depression in the United States.

Psychiatric Services 60, 297-305 (2009).

12 van Beljouw, I. et al. Reasons and determinants for not receiving treatment for common

mental disorders. Psychiatric Services 61, 250-257 (2010).

13 Papakostas, G. I. Managing partial response or nonresponse: switching, augmentation,

and combination strategies for major depressive disorder. The Journal of clinical

psychiatry 70 Suppl 6, 16-25 (2009).

14 Thase, M. E. et al. Cognitive therapy versus medication in augmentation and switch

strategies as second-step treatments: a STAR*D report. The American journal of

psychiatry 164, 739-752 (2007).

15 Dhuria, S. V., Hanson, L. R. & Frey, W. H., 2nd. Intranasal delivery to the central

nervous system: mechanisms and experimental considerations. Journal of

pharmaceutical sciences 99, 1654-1673 (2010).

16 Kandel, E. R., Schwartz, J. H. & Jessell, T. M. Principles of Neural Science, 4th Edition.

4th edn, (McGraw-Hill, 2000).

17 Carlsson, A., Lindqvist, M. & Magnusson, T. 3,4-Dihydroxyphenylalanine and 5-

hydroxytryptophan as reserpine antagonists. Nature 180, 1200 (1957).

Page 118: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

108

18 Carlsson, A. Perspectives on the discovery of central monoaminergic neurotransmission.

Annual review of neuroscience 10, 19-40 (1987).

19 Samii, A., Nutt, J. G. & Ransom, B. R. Parkinson's disease. Lancet 363, 1783-1793

(2004).

20 Walker, F. O. Huntington's disease. Lancet 369, 218-228 (2007).

21 Kuepper, R., Skinbjerg, M. & Abi-Dargham, A. The dopamine dysfunction in

schizophrenia revisited: new insights into topography and course. Handbook of

experimental pharmacology, 1-26 (2012).

22 Kapur, S. & Mann, J. J. Role of the dopaminergic system in depression. Biological

psychiatry 32, 1-17 (1992).

23 Volkow, N. D., Wang, G. J., Fowler, J. S., Tomasi, D. & Telang, F. Addiction: beyond

dopamine reward circuitry. Proceedings of the National Academy of Sciences of the

United States of America 108, 15037-15042 (2011).

24 Volkow, N. D., Wang, G. J., Fowler, J. S. & Tomasi, D. Addiction circuitry in the human

brain. Annual review of pharmacology and toxicology 52, 321-336 (2012).

25 Missale, C., Nash, S. R., Robinson, S. W., Jaber, M. & Caron, M. G. Dopamine

receptors: from structure to function. Physiological reviews 78, 189-225 (1998).

26 Kebabian, J. W. & Calne, D. B. Multiple receptors for dopamine. Nature 277, 93-96

(1979).

27 Snyder, S. H. What dopamine does in the brain. Proceedings of the National Academy of

Sciences of the United States of America 108, 18869-18871 (2011).

28 Aizman, O. et al. Anatomical and physiological evidence for D1 and D2 dopamine

receptor colocalization in neostriatal neurons. Nature neuroscience 3, 226-230 (2000).

29 Lee, F. J. et al. Dual regulation of NMDA receptor functions by direct protein-protein

interactions with the dopamine D1 receptor. Cell 111, 219-230 (2002).

30 Pei, L., Lee, F. J., Moszczynska, A., Vukusic, B. & Liu, F. Regulation of dopamine D1

receptor function by physical interaction with the NMDA receptors. The Journal of

neuroscience 24, 1149-1158 (2004).

31 Zou, S. et al. Protein-protein coupling/uncoupling enables dopamine D2 receptor

regulation of AMPA receptor-mediated excitotoxicity. The Journal of neuroscience 25,

4385-4395 (2005).

32 Neves, S. R., Ram, P. T. & Iyengar, R. G protein pathways. Science 296, 1636-1639

(2002).

33 Pierce, K. L., Premont, R. T. & Lefkowitz, R. J. Seven-transmembrane receptors. Nature

Reviews Molecular Cell Biology 3, 639-650 (2002).

34 Vallone, D., Picetti, R. & Borrelli, E. Structure and function of dopamine receptors.

Neuroscience and biobehavioral reviews 24, 125-132 (2000).

35 Niznik, H. B. & Van Tol, H. H. Dopamine receptor genes: new tools for molecular

psychiatry. Journal of psychiatry & neuroscience : JPN 17, 158-180 (1992).

36 Kebabian, J. W. & Greengard, P. Dopamine-sensitive adenyl cyclase: possible role in

synaptic transmission. Science 174, 1346-1349 (1971).

37 Spano, P. F., Memo, M., Stefanini, E., Fresia, P. & Trabucchi, M. Detection of multiple

receptors for dopamine. Advances in biochemical psychopharmacology 21, 243-251

(1980).

38 Bunzow, J. R. et al. Cloning and expression of a rat D2 dopamine receptor cDNA. Nature

336, 783-787 (1988).

Page 119: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

109

39 Sokoloff, P., Giros, B., Martres, M. P., Bouthenet, M. L. & Schwartz, J. C. Molecular

cloning and characterization of a novel dopamine receptor (D3) as a target for

neuroleptics. Nature 347, 146-151 (1990).

40 Van Tol, H. H. et al. Cloning of the gene for a human dopamine D4 receptor with high

affinity for the antipsychotic clozapine. Nature 350, 610-614 (1991).

41 Tiberi, M. et al. Cloning, molecular characterization, and chromosomal assignment of a

gene encoding a second D1 dopamine receptor subtype: differential expression pattern in

rat brain compared with the D1A receptor. Proceedings of the National Academy of

Sciences of the United States of America 88, 7491-7495 (1991).

42 Herve, D. et al. G(olf) and Gs in rat basal ganglia: possible involvement of G(olf) in the

coupling of dopamine D1 receptor with adenylyl cyclase. The Journal of neuroscience

13, 2237-2248 (1993).

43 Gingrich, J. A. & Caron, M. G. Recent advances in the molecular biology of dopamine

receptors. Annual review of neuroscience 16, 299-321 (1993).

44 Weiner, D. M. et al. D1 and D2 dopamine receptor mRNA in rat brain. Proceedings of

the National Academy of Sciences of the United States of America 88, 1859-1863 (1991).

45 Choi, W. S., Machida, C. A. & Ronnekleiv, O. K. Distribution of dopamine D1, D2, and

D5 receptor mRNAs in the monkey brain: ribonuclease protection assay analysis. Brain

research. Molecular brain research 31, 86-94 (1995).

46 Huntley, G. W., Morrison, J. H., Prikhozhan, A. & Sealfon, S. C. Localization of multiple

dopamine receptor subtype mRNAs in human and monkey motor cortex and striatum.

Brain Research: Molecular Brain Research 15, 181-188 (1992).

47 Rappaport, M. S., Sealfon, S. C., Prikhozhan, A., Huntley, G. W. & Morrison, J. H.

Heterogeneous distribution of D1, D2 and D5 receptor mRNAs in monkey striatum.

Brain research 616, 242-250 (1993).

48 Bergson, C. et al. Regional, cellular, and subcellular variations in the distribution of D1

and D5 dopamine receptors in primate brain. The Journal of Neuroscience 15, 7821-7836

(1995).

49 Muly, E. C. et al. Distribution of D1 and D5 dopamine receptors in the primate and rat

basolateral amygdala. Brain structure & function 213, 375-393 (2009).

50 Floresco, S. B. Prefrontal dopamine and behavioral flexibility: shifting from an "inverted-

U" toward a family of functions. Frontiers in neuroscience 7, 62 (2013).

51 Mink, J. W. The Basal Ganglia and involuntary movements: impaired inhibition of

competing motor patterns. Archives of neurology 60, 1365-1368 (2003).

52 Onali, P., Olianas, M. C. & Gessa, G. L. Characterization of dopamine receptors

mediating inhibition of adenylate cyclase activity in rat striatum. Molecular

pharmacology 28, 138-145 (1985).

53 Beaulieu, J. M. et al. An Akt/beta-arrestin 2/PP2A signaling complex mediates

dopaminergic neurotransmission and behavior. Cell 122, 261-273 (2005).

54 Beaulieu, J. M., Gainetdinov, R. R. & Caron, M. G. The Akt-GSK-3 signaling cascade in

the actions of dopamine. Trends in pharmacological sciences 28, 166-172 (2007).

55 Monsma, F. J., Jr., McVittie, L. D., Gerfen, C. R., Mahan, L. C. & Sibley, D. R. Multiple

D2 dopamine receptors produced by alternative RNA splicing. Nature 342, 926-929

(1989).

56 Giros, B. et al. Alternative splicing directs the expression of two D2 dopamine receptor

isoforms. Nature 342, 923-926 (1989).

Page 120: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

110

57 De Mei, C., Ramos, M., Iitaka, C. & Borrelli, E. Getting specialized: presynaptic and

postsynaptic dopamine D2 receptors. Current opinion in pharmacology 9, 53-58 (2009).

58 Usiello, A. et al. Distinct functions of the two isoforms of dopamine D2 receptors. Nature

408, 199-203 (2000).

59 Meador-Woodruff, J. H. et al. Distribution of D2 dopamine receptor mRNA in rat brain.

Proceedings of the National Academy of Sciences of the United States of America 86,

7625-7628 (1989).

60 Bouthenet, M. L. et al. Localization of dopamine D3 receptor mRNA in the rat brain

using in situ hybridization histochemistry: comparison with dopamine D2 receptor

mRNA. Brain research 564, 203-219 (1991).

61 Landwehrmeyer, B., Mengod, G. & Palacios, J. M. Differential visualization of dopamine

D2 and D3 receptor sites in rat brain. A comparative study using in situ hybridization

histochemistry and ligand binding autoradiography. The European journal of

neuroscience 5, 145-153 (1993).

62 Rondou, P., Haegeman, G. & Van Craenenbroeck, K. The dopamine D4 receptor:

biochemical and signalling properties. Cellular and molecular life sciences : CMLS 67,

1971-1986 (2010).

63 Noain, D. et al. Identification of brain neurons expressing the dopamine D4 receptor gene

using BAC transgenic mice. The European journal of neuroscience 24, 2429-2438

(2006).

64 Dohlman, H. G. & Thorner, J. RGS proteins and signaling by heterotrimeric G proteins.

The Journal of biological chemistry 272, 3871-3874 (1997).

65 Premont, R. T. & Gainetdinov, R. R. Physiological roles of G protein-coupled receptor

kinases and arrestins. Annual review of physiology 69, 511-534 (2007).

66 Laporte, S. A., Miller, W. E., Kim, K. M. & Caron, M. G. beta-Arrestin/AP-2 interaction

in G protein-coupled receptor internalization: identification of a beta-arrestin binging site

in beta 2-adaptin. The Journal of biological chemistry 277, 9247-9254 (2002).

67 Anden, N. E. et al. Demonstration and Mapping out of Nigro-Neostriatal Dopamine

Neurons. Life sciences 3, 523-530 (1964).

68 Mink, J. W. The basal ganglia: focused selection and inhibition of competing motor

programs. Progress in neurobiology 50, 381-425 (1996).

69 Le Moal, M. & Simon, H. Mesocorticolimbic dopaminergic network: functional and

regulatory roles. Physiological reviews 71, 155-234 (1991).

70 Ko, J. H. & Strafella, A. P. Dopaminergic neurotransmission in the human brain: new

lessons from perturbation and imaging. The Neuroscientist : a review journal bringing

neurobiology, neurology and psychiatry 18, 149-168 (2012).

71 Nader, K. & LeDoux, J. The dopaminergic modulation of fear: quinpirole impairs the

recall of emotional memories in rats. Behavioral neuroscience 113, 152-165 (1999).

72 Borowski, T. B. & Kokkinidis, L. The effects of cocaine, amphetamine, and the

dopamine D1 receptor agonist SKF 38393 on fear extinction as measured with

potentiated startle: implications for psychomotor stimulant psychosis. Behavioral

neuroscience 112, 952-965 (1998).

73 LeDoux, J. E. Emotion circuits in the brain. Annual review of neuroscience 23, 155-184

(2000).

74 Wise, R. A. Drug-activation of brain reward pathways. Drug and alcohol dependence 51,

13-22 (1998).

Page 121: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

111

75 Di Chiara, G. & Imperato, A. Drugs abused by humans preferentially increase synaptic

dopamine concentrations in the mesolimbic system of freely moving rats. Proceedings of

the National Academy of Sciences of the United States of America 85, 5274-5278 (1988).

76 Corrigall, W. A., Franklin, K. B., Coen, K. M. & Clarke, P. B. The mesolimbic

dopaminergic system is implicated in the reinforcing effects of nicotine.

Psychopharmacology 107, 285-289 (1992).

77 Hyman, S. E., Malenka, R. C. & Nestler, E. J. Neural mechanisms of addiction: the role

of reward-related learning and memory. Annual review of neuroscience 29, 565-598

(2006).

78 Belmaker, R. H. & Agam, G. Major depressive disorder. The New England journal of

medicine 358, 55-68 (2008).

79 Krishnan, V. & Nestler, E. J. The molecular neurobiology of depression. Nature 455,

894-902 (2008).

80 Nestler, E. J. & Carlezon, W. A., Jr. The mesolimbic dopamine reward circuit in

depression. Biological psychiatry 59, 1151-1159 (2006).

81 Palmiter, R. D. Dopamine signaling in the dorsal striatum is essential for motivated

behaviors: lessons from dopamine-deficient mice. Annals of the New York Academy of

Sciences 1129, 35-46 (2008).

82 Cannon, C. M. & Palmiter, R. D. Reward without dopamine. The Journal of neuroscience

3, 10827-10831 (2003).

83 Robinson, S., Sandstrom, S. M., Denenberg, V. H. & Palmiter, R. D. Distinguishing

whether dopamine regulates liking, wanting, and/or learning about rewards. Behavioral

neuroscience 119, 5-15 (2005).

84 Berridge, K. C. The debate over dopamine's role in reward: the case for incentive

salience. Psychopharmacology 191, 391-431 (2007).

85 Pecina, S., Cagniard, B., Berridge, K. C., Aldridge, J. W. & Zhuang, X.

Hyperdopaminergic mutant mice have higher "wanting" but not "liking" for sweet

rewards. The Journal of neuroscience 23, 9395-9402 (2003).

86 Seamans, J. K. & Yang, C. R. The principal features and mechanisms of dopamine

modulation in the prefrontal cortex. Progress in neurobiology 74, 1-58 (2004).

87 Brozoski, T. J., Brown, R. M., Rosvold, H. E. & Goldman, P. S. Cognitive deficit caused

by regional depletion of dopamine in prefrontal cortex of rhesus monkey. Science 205,

929-932 (1979).

88 Zahrt, J., Taylor, J. R., Mathew, R. G. & Arnsten, A. F. Supranormal stimulation of D1

dopamine receptors in the rodent prefrontal cortex impairs spatial working memory

performance. The Journal of Neuroscience 17, 8528-8535 (1997).

89 Sawaguchi, T. & Goldman-Rakic, P. S. The role of D1-dopamine receptor in working

memory: local injections of dopamine antagonists into the prefrontal cortex of rhesus

monkeys performing an oculomotor delayed-response task. Journal of neurophysiology

71, 515-528 (1994).

90 Dias, R., Robbins, T. W. & Roberts, A. C. Dissociable forms of inhibitory control within

prefrontal cortex with an analog of the Wisconsin Card Sort Test: restriction to novel

situations and independence from "on-line" processing. The Journal of Neuroscience 17,

9285-9297 (1997).

91 Dias, R., Robbins, T. W. & Roberts, A. C. Primate analogue of the Wisconsin Card

Sorting Test: effects of excitotoxic lesions of the prefrontal cortex in the marmoset.

Behavioral neuroscience 110, 872-886 (1996).

Page 122: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

112

92 Birrell, J. M. & Brown, V. J. Medial frontal cortex mediates perceptual attentional set

shifting in the rat. The Journal of neuroscience 20, 4320-4324 (2000).

93 McAlonan, K. & Brown, V. J. Orbital prefrontal cortex mediates reversal learning and

not attentional set shifting in the rat. Behavioural brain research 146, 97-103 (2003).

94 Floresco, S. B., Block, A. E. & Tse, M. T. Inactivation of the medial prefrontal cortex of

the rat impairs strategy set-shifting, but not reversal learning, using a novel, automated

procedure. Behavioural brain research 190, 85-96 (2008).

95 Pietrzak, R. H., Mollica, C. M., Maruff, P. & Snyder, P. J. Cognitive effects of

immediate-release methylphenidate in children with attention-deficit/hyperactivity

disorder. Neuroscience and biobehavioral reviews 30, 1225-1245 (2006).

96 Pantelis, C. et al. Comparison of set-shifting ability in patients with chronic

schizophrenia and frontal lobe damage. Schizophrenia research 37, 251-270 (1999).

97 Reeve, W. V. & Schandler, S. L. Frontal lobe functioning in adolescents with attention

deficit hyperactivity disorder. Adolescence 36, 749-765 (2001).

98 Floresco, S. B. & Magyar, O. Mesocortical dopamine modulation of executive functions:

beyond working memory. Psychopharmacology 188, 567-585 (2006).

99 Bechara, A., Damasio, H., Damasio, A. R. & Lee, G. P. Different contributions of the

human amygdala and ventromedial prefrontal cortex to decision-making. The Journal of

Neuroscience 19, 5473-5481 (1999).

100 Rudebeck, P. H., Walton, M. E., Smyth, A. N., Bannerman, D. M. & Rushworth, M. F.

Separate neural pathways process different decision costs. Nature neuroscience 9, 1161-

1168 (2006).

101 St Onge, J. R., Stopper, C. M., Zahm, D. S. & Floresco, S. B. Separate prefrontal-

subcortical circuits mediate different components of risk-based decision making. The

Journal of Neuroscience 32, 2886-2899 (2012).

102 Floresco, S. B., St Onge, J. R., Ghods-Sharifi, S. & Winstanley, C. A. Cortico-limbic-

striatal circuits subserving different forms of cost-benefit decision making. Cognitive,

affective & behavioral neuroscience 8, 375-389 (2008).

103 St Onge, J. R., Abhari, H. & Floresco, S. B. Dissociable contributions by prefrontal D1

and D2 receptors to risk-based decision making. The Journal of Neuroscience 31, 8625-

8633 (2011).

104 Lidow, M. S., Goldman-Rakic, P. S., Gallager, D. W. & Rakic, P. Distribution of

dopaminergic receptors in the primate cerebral cortex: quantitative autoradiographic

analysis using [3H]raclopride, [3H]spiperone and [3H]SCH23390. Neuroscience 40, 657-

671 (1991).

105 Vincent, S. L., Khan, Y. & Benes, F. M. Cellular colocalization of dopamine D1 and D2

receptors in rat medial prefrontal cortex. Synapse 19, 112-120 (1995).

106 Vincent, S. L., Khan, Y. & Benes, F. M. Cellular distribution of dopamine D1 and D2

receptors in rat medial prefrontal cortex. The Journal of Neuroscience 13, 2551-2564

(1993).

107 Zhang, Z. W., Burke, M. W., Calakos, N., Beaulieu, J. M. & Vaucher, E. Confocal

Analysis of Cholinergic and Dopaminergic Inputs onto Pyramidal Cells in the Prefrontal

Cortex of Rodents. Frontiers in neuroanatomy 4, 21 (2010).

108 Perreault, M. L. et al. The dopamine D1-D2 receptor heteromer localizes in

dynorphin/enkephalin neurons: increased high affinity state following amphetamine and

in schizophrenia. The Journal of biological chemistry 285, 36625-36634 (2010).

Page 123: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

113

109 So, C. H. et al. Calcium signaling by dopamine D5 receptor and D5-D2 receptor hetero-

oligomers occurs by a mechanism distinct from that for dopamine D1-D2 receptor hetero-

oligomers. Molecular pharmacology 75, 843-854 (2009).

110 Free, R. B. et al. D1 and D2 dopamine receptor expression is regulated by direct

interaction with the chaperone protein calnexin. The Journal of biological chemistry 282,

21285-21300 (2007).

111 Schwartz, J. C. et al. Functional implications of multiple dopamine receptor subtypes: the

D1/D3 receptor coexistence. Brain research. Brain research reviews 26, 236-242 (1998).

112 Ridray, S. et al. Coexpression of dopamine D1 and D3 receptors in islands of Calleja and

shell of nucleus accumbens of the rat: opposite and synergistic functional interactions.

The European journal of neuroscience 10, 1676-1686 (1998).

113 O'Dowd, B. F., Nguyen, T., Ji, X. & George, S. R. D5 dopamine receptor carboxyl tail

involved in D5-D2 heteromer formation. Biochemical and biophysical research

communications 431, 586-589 (2013).

114 Lee, F. J. & Liu, F. Direct interactions between NMDA and D1 receptors: a tale of tails.

Biochemical Society transactions 32, 1032-1036 (2004).

115 Liu, F. et al. Direct protein-protein coupling enables cross-talk between dopamine D5

and gamma-aminobutyric acid A receptors. Nature 403, 274-280 (2000).

116 Hasbi, A., O'Dowd, B. F. & George, S. R. Dopamine D1-D2 receptor heteromer

signaling pathway in the brain: emerging physiological relevance. Molecular brain 4, 26

(2011).

117 Jin, L. Q., Goswami, S., Cai, G., Zhen, X. & Friedman, E. SKF83959 selectively

regulates phosphatidylinositol-linked D1 dopamine receptors in rat brain. Journal of

neurochemistry 85, 378-386 (2003).

118 Hasbi, A. et al. Calcium signaling cascade links dopamine D1-D2 receptor heteromer to

striatal BDNF production and neuronal growth. Proceedings of the National Academy of

Sciences of the United States of America 106, 21377-21382 (2009).

119 Rashid, A. J. et al. D1-D2 dopamine receptor heterooligomers with unique pharmacology

are coupled to rapid activation of Gq/11 in the striatum. Proceedings of the National

Academy of Sciences of the United States of America 104, 654-659 (2007).

120 Kessler, R. C. et al. The epidemiology of major depressive disorder: results from the

National Comorbidity Survey Replication (NCS-R). JAMA : the journal of the American

Medical Association 289, 3095-3105 (2003).

121 Nestler, E. J. et al. Neurobiology of depression. Neuron 34, 13-25 (2002).

122 American Psychiatric Association. Diagnostic and statistical manual of mental disorders:

DSM-IV-TR. (American Psychiatric Publishing, Inc., 2000).

123 Centers for Disease Control and Prevention. Mental Illness Surveillance Among Adults

in the United States. (US Department of Health and Human Services, 2011).

124 Deecher, D., Andree, T. H., Sloan, D. & Schechter, L. E. From menarche to menopause:

exploring the underlying biology of depression in women experiencing hormonal

changes. Psychoneuroendocrinology 33, 3-17 (2008).

125 Mathers, C., Fat, D. M. & Boerma, J. The global burden of disease: 2004 update. (World

Health Organization, 2008).

126 Substance Abuse and Mental Health Services Administration. Results from the 2008

national survey on drug use and health: national findings. 305 (SAMHSA, Office of

Applied Studies, 2009).

Page 124: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

114

127 Tobet, S. A., Handa, R. J. & Goldstein, J. M. Sex-dependent pathophysiology as

predictors of comorbidity of major depressive disorder and cardiovascular disease.

European journal of physiology 465, 585-594 (2013).

128 Novy, J. et al. Psychiatric co-morbidities and cardiovascular risk factors in people with

lifetime history of epilepsy of an urban community. Clinical neurology and neurosurgery

114, 26-30 (2012).

129 Dear, B. F. et al. The Pain Course: A randomised controlled trial of a clinician-guided

Internet-delivered cognitive behaviour therapy program for managing chronic pain and

emotional well-being. Pain 154, 942-950 (2013).

130 Angst, F., Stassen, H. H., Clayton, P. J. & Angst, J. Mortality of patients with mood

disorders: follow-up over 34-38 years. Journal of affective disorders 68, 167-181 (2002).

131 National Institute of Mental Health. Depression. (National Institute of Health, 2011).

132 Blier, P. & El Mansari, M. Serotonin and beyond: therapeutics for major depression.

Philosophical transactions of the Royal Society of London. Series B, Biological sciences

368, 20120536 (2013).

133 Lee, S., Jeong, J., Kwak, Y. & Park, S. K. Depression research: where are we now?

Molecular brain 3, 8 (2010).

134 Trivedi, M. H. et al. Evaluation of outcomes with citalopram for depression using

measurement-based care in STAR*D: implications for clinical practice. The American

journal of psychiatry 163, 28-40 (2006).

135 Hamilton, M. A rating scale for depression. Journal of neurology, neurosurgery, and

psychiatry 23, 56-62 (1960).

136 Kirsch, I. Review: benefits of antidepressants over placebo limited except in very severe

depression. Evidence-based mental health 13, 49 (2010).

137 Kirsch, I. et al. Initial severity and antidepressant benefits: a meta-analysis of data

submitted to the Food and Drug Administration. PLoS medicine 5, e45 (2008).

138 Prins, J., Olivier, B. & Korte, S. M. Triple reuptake inhibitors for treating subtypes of

major depressive disorder: the monoamine hypothesis revisited. Expert opinion on

investigational drugs 20, 1107-1130 (2011).

139 Lisanby, S. H. Electroconvulsive therapy for depression. The New England journal of

medicine 357, 1939-1945 (2007).

140 Cohen, L. J. & Sclar, D. A. Issues in adherence to treatment with monoamine oxidase

inhibitors and the rate of treatment failure. The Journal of clinical psychiatry 73 Suppl 1,

31-36 (2012).

141 Shelton, R. C., Osuntokun, O., Heinloth, A. N. & Corya, S. A. Therapeutic options for

treatment-resistant depression. CNS drugs 24, 131-161 (2010).

142 Sinyor, M., Schaffer, A. & Levitt, A. The sequenced treatment alternatives to relieve

depression (STAR*D) trial: a review. Canadian journal of psychiatry. Revue canadienne

de psychiatrie 55, 126-135 (2010).

143 Ressler, K. J. & Mayberg, H. S. Targeting abnormal neural circuits in mood and anxiety

disorders: from the laboratory to the clinic. Nature neuroscience 10, 1116-1124 (2007).

144 Mayberg, H. S. et al. Reciprocal limbic-cortical function and negative mood: converging

PET findings in depression and normal sadness. The American journal of psychiatry 156,

675-682 (1999).

145 Coppen, A. The biochemistry of affective disorders. The British journal of psychiatry :

the journal of mental science 113, 1237-1264 (1967).

Page 125: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

115

146 Ruhe, H. G., Mason, N. S. & Schene, A. H. Mood is indirectly related to serotonin,

norepinephrine and dopamine levels in humans: a meta-analysis of monoamine depletion

studies. Molecular psychiatry 12, 331-359 (2007).

147 Leake, A., Fairbairn, A. F., McKeith, I. G. & Ferrier, I. N. Studies on the serotonin

uptake binding site in major depressive disorder and control post-mortem brain:

neurochemical and clinical correlates. Psychiatry research 39, 155-165 (1991).

148 Meyer, J. H. et al. Brain monoamine oxidase A binding in major depressive disorder:

relationship to selective serotonin reuptake inhibitor treatment, recovery, and recurrence.

Archives of general psychiatry 66, 1304-1312 (2009).

149 Kinney, J. L. Nomifensine maleate: a new second-generation antidepressant. Clinical

pharmacy 4, 625-636 (1985).

150 Lambert, G., Johansson, M., Agren, H. & Friberg, P. Reduced brain norepinephrine and

dopamine release in treatment-refractory depressive illness: evidence in support of the

catecholamine hypothesis of mood disorders. Archives of general psychiatry 57, 787-793

(2000).

151 Dailly, E., Chenu, F., Renard, C. E. & Bourin, M. Dopamine, depression and

antidepressants. Fundamental & clinical pharmacology 18, 601-607 (2004).

152 Yadid, G. & Friedman, A. Dynamics of the dopaminergic system as a key component to

the understanding of depression. Progress in brain research 172, 265-286 (2008).

153 Anisman, H., Remington, G. & Sklar, L. S. Effect of inescapable shock on subsequent

escape performance: catecholaminergic and cholinergic mediation of response initiation

and maintenance. Psychopharmacology 61, 107-124 (1979).

154 Anisman, H., Irwin, J. & Sklar, L. S. Deficits of escape performance following

catecholamine depletion: implications for behavioral deficits induced by uncontrollable

stress. Psychopharmacology 64, 163-170 (1979).

155 Yamada, J., Sugimoto, Y. & Yamada, S. Involvement of dopamine receptors in the anti-

immobility effects of dopamine re-uptake inhibitors in the forced swimming test.

European journal of pharmacology 504, 207-211 (2004).

156 Sampson, D., Willner, P. & Muscat, R. Reversal of antidepressant action by dopamine

antagonists in an animal model of depression. Psychopharmacology 104, 491-495 (1991).

157 Autry, A. E. & Monteggia, L. M. Brain-derived neurotrophic factor and neuropsychiatric

disorders. Pharmacological reviews 64, 238-258 (2012).

158 Duman, R. S. & Monteggia, L. M. A neurotrophic model for stress-related mood

disorders. Biological psychiatry 59, 1116-1127 (2006).

159 Pandey, G. N. et al. Brain-derived neurotrophic factor and tyrosine kinase B receptor

signalling in post-mortem brain of teenage suicide victims. International Journal of

Neuropsychopharmacology 11, 1047-1061 (2008).

160 Karege, F., Vaudan, G., Schwald, M., Perroud, N. & La Harpe, R. Neurotrophin levels in

postmortem brains of suicide victims and the effects of antemortem diagnosis and

psychotropic drugs. Brain research. Molecular brain research 136, 29-37 (2005).

161 Dwivedi, Y. et al. Altered gene expression of brain-derived neurotrophic factor and

receptor tyrosine kinase B in postmortem brain of suicide subjects. Archives of general

psychiatry 60, 804-815 (2003).

162 Sen, S., Duman, R. & Sanacora, G. Serum brain-derived neurotrophic factor, depression,

and antidepressant medications: meta-analyses and implications. Biological psychiatry

64, 527-532 (2008).

Page 126: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

116

163 Monteggia, L. M. et al. Brain-derived neurotrophic factor conditional knockouts show

gender differences in depression-related behaviors. Biological psychiatry 61, 187-197

(2007).

164 Shirayama, Y., Chen, A. C., Nakagawa, S., Russell, D. S. & Duman, R. S. Brain-derived

neurotrophic factor produces antidepressant effects in behavioral models of depression.

The Journal of Neuroscience 22, 3251-3261 (2002).

165 Saarelainen, T. et al. Activation of the TrkB neurotrophin receptor is induced by

antidepressant drugs and is required for antidepressant-induced behavioral effects. The

Journal of Neuroscience 23, 349-357 (2003).

166 Rantamaki, T. et al. Pharmacologically diverse antidepressants rapidly activate brain-

derived neurotrophic factor receptor TrkB and induce phospholipase-Cgamma signaling

pathways in mouse brain. Neuropsychopharmacology : official publication of the

American College of Neuropsychopharmacology 32, 2152-2162 (2007).

167 Zorner, B. et al. Forebrain-specific trkB-receptor knockout mice: behaviorally more

hyperactive than "depressive". Biological psychiatry 54, 972-982 (2003).

168 Greenwood, B. N., Strong, P. V., Foley, T. E., Thompson, R. S. & Fleshner, M. Learned

helplessness is independent of levels of brain-derived neurotrophic factor in the

hippocampus. Neuroscience 144, 1193-1208 (2007).

169 Berton, O. et al. Essential role of BDNF in the mesolimbic dopamine pathway in social

defeat stress. Science 311, 864-868 (2006).

170 Eisch, A. J. et al. Brain-derived neurotrophic factor in the ventral midbrain-nucleus

accumbens pathway: a role in depression. Biological psychiatry 54, 994-1005 (2003).

171 Cryan, J. F., Markou, A. & Lucki, I. Assessing antidepressant activity in rodents: recent

developments and future needs. Trends in pharmacological sciences 23, 238-245 (2002).

172 Geyer, M. A. & Markou, A. Animal Models of Psychiatric Disorders. (Raven Press,

1995).

173 McKinney, W. T., Jr. & Bunney, W. E., Jr. Animal model of depression. I. Review of

evidence: implications for research. Archives of general psychiatry 21, 240-248 (1969).

174 Porsolt, R. D., Le Pichon, M. & Jalfre, M. Depression: a new animal model sensitive to

antidepressant treatments. Nature 266, 730-732 (1977).

175 Porsolt, R. D., Anton, G., Blavet, N. & Jalfre, M. Behavioural despair in rats: a new

model sensitive to antidepressant treatments. European journal of pharmacology 47, 379-

391 (1978).

176 Lucki, I. The forced swimming test as a model for core and component behavioral effects

of antidepressant drugs. Behavioural pharmacology 8, 523-532 (1997).

177 Lino-de-Oliveira, C., De Lima, T. C. & de Padua Carobrez, A. Structure of the rat

behaviour in the forced swimming test. Behavioural brain research 158, 243-250 (2005).

178 Cryan, J. F., Valentino, R. J. & Lucki, I. Assessing substrates underlying the behavioral

effects of antidepressants using the modified rat forced swimming test. Neuroscience and

biobehavioral reviews 29, 547-569 (2005).

179 Overmier, J. B. & Seligman, M. E. Effects of inescapable shock upon subsequent escape

and avoidance responding. Journal of comparative and physiological psychology 63, 28-

33 (1967).

180 Sherman, A. D., Sacquitne, J. L. & Petty, F. Specificity of the learned helplessness model

of depression. Pharmacology, biochemistry, and behavior 16, 449-454 (1982).

181 Kendler, K. S., Hettema, J. M., Butera, F., Gardner, C. O. & Prescott, C. A. Life event

dimensions of loss, humiliation, entrapment, and danger in the prediction of onsets of

Page 127: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

117

major depression and generalized anxiety. Archives of general psychiatry 60, 789-796

(2003).

182 Green, J. G. et al. Childhood adversities and adult psychiatric disorders in the national

comorbidity survey replication I: associations with first onset of DSM-IV disorders.

Archives of general psychiatry 67, 113-123 (2010).

183 Pryce, C. R. et al. Helplessness: a systematic translational review of theory and evidence

for its relevance to understanding and treating depression. Pharmacology & therapeutics

132, 242-267 (2011).

184 Vollmayr, B. & Henn, F. A. Learned helplessness in the rat: improvements in validity

and reliability. Brain research. Brain research protocols 8, 1-7 (2001).

185 Martin, P., Soubrie, P. & Simon, P. The effect of monoamine oxidase inhibitors

compared with classical tricyclic antidepressants on learned helplessness paradigm.

Progress in neuro-psychopharmacology & biological psychiatry 11, 1-7 (1987).

186 Grappi, S. et al. The efficacy of reboxetine in preventing and reverting a condition of

escape deficit in rats. Biological psychiatry 53, 890-898 (2003).

187 Geoffroy, M., Tvede, K., Christensen, A. V. & Schou, J. S. The effect of imipramine and

lithium on "learned helplessness" and acetylcholinesterase in rat brain. Pharmacology,

biochemistry, and behavior 38, 93-97 (1991).

188 Li, Y., Field, P. M. & Raisman, G. Olfactory ensheathing cells and olfactory nerve

fibroblasts maintain continuous open channels for regrowth of olfactory nerve fibres.

Glia 52, 245-251 (2005).

189 Daneman, R. The blood-brain barrier in health and disease. Annals of neurology 72, 648-

672 (2012).

190 Renner, D. B. et al. Intranasal delivery of insulin via the olfactory nerve pathway. The

Journal of pharmacy and pharmacology 64, 1709-1714 (2012).

191 Marks, D. R., Tucker, K., Cavallin, M. A., Mast, T. G. & Fadool, D. A. Awake intranasal

insulin delivery modifies protein complexes and alters memory, anxiety, and olfactory

behaviors. The Journal of Neuroscience

29, 6734-6751 (2009).

192 Francis, G. J. et al. Intranasal insulin prevents cognitive decline, cerebral atrophy and

white matter changes in murine type I diabetic encephalopathy. Brain : a journal of

neurology 131, 3311-3334 (2008).

193 Shemesh, E., Rudich, A., Harman-Boehm, I. & Cukierman-Yaffe, T. Effect of intranasal

insulin on cognitive function: a systematic review. The Journal of clinical endocrinology

and metabolism 97, 366-376 (2012).

194 Chen, X. Q., Fawcett, J. R., Rahman, Y. E., Ala, T. A. & Frey, I. W. Delivery of Nerve

Growth Factor to the Brain via the Olfactory Pathway. Journal of Alzheimer's disease :

JAD 1, 35-44 (1998).

195 De Rosa, R. et al. Intranasal administration of nerve growth factor (NGF) rescues

recognition memory deficits in AD11 anti-NGF transgenic mice. Proceedings of the

National Academy of Sciences of the United States of America 102, 3811-3816 (2005).

196 Alcala-Barraza, S. R. et al. Intranasal delivery of neurotrophic factors BDNF, CNTF,

EPO, and NT-4 to the CNS. Journal of drug targeting 18, 179-190 (2010).

197 Liu, X. F., Fawcett, J. R., Thorne, R. G. & Frey, W. H., 2nd. Non-invasive intranasal

insulin-like growth factor-I reduces infarct volume and improves neurologic function in

rats following middle cerebral artery occlusion. Neuroscience letters 308, 91-94 (2001).

Page 128: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

118

198 Fletcher, L. et al. Intranasal delivery of erythropoietin plus insulin-like growth factor-I

for acute neuroprotection in stroke. Laboratory investigation. Journal of neurosurgery

111, 164-170 (2009).

199 Jansson, B. & Bjork, E. Visualization of in vivo olfactory uptake and transfer using

fluorescein dextran. Journal of drug targeting 10, 379-386 (2002).

200 Gray, H. Gray's Anatomy. 40th

Edition (Churchill Livingstone, 2008).

201 Perl, D. P. & Good, P. F. Uptake of aluminium into central nervous system along nasal-

olfactory pathways. Lancet 8540, 1028 (1987).

202 Baker, H. & Spencer, R. F. Transneuronal transport of peroxidase-conjugated wheat germ

agglutinin (WGA-HRP) from the olfactory epithelium to the brain of the adult rat.

Experimental brain research. Experimentelle Hirnforschung. Experimentation cerebrale

63, 461-473 (1986).

203 Thorne, R. G., Pronk, G. J., Padmanabhan, V. & Frey, W. H., 2nd. Delivery of insulin-

like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal

pathways following intranasal administration. Neuroscience 127, 481-496 (2004).

204 Nonaka, N., Farr, S. A., Kageyama, H., Shioda, S. & Banks, W. A. Delivery of galanin-

like peptide to the brain: targeting with intranasal delivery and cyclodextrins. The Journal

of pharmacology and experimental therapeutics 325, 513-519 (2008).

205 Yang, J. P. et al. Direct transport of VEGF from the nasal cavity to brain. Neuroscience

letters 449, 108-111 (2009).

206 Ross, T. M., Zuckermann, R. N., Reinhard, C. & Frey, W. H., 2nd. Intranasal

administration delivers peptoids to the rat central nervous system. Neuroscience letters

439, 30-33 (2008).

207 Dufes, C. et al. Brain delivery of vasoactive intestinal peptide (VIP) following nasal

administration to rats. International journal of pharmaceutics 255, 87-97 (2003).

208 van den Berg, M. P., Romeijn, S. G., Verhoef, J. C. & Merkus, F. W. Serial cerebrospinal

fluid sampling in a rat model to study drug uptake from the nasal cavity. Journal of

neuroscience methods 116, 99-107 (2002).

209 Chow, H. S., Chen, Z. & Matsuura, G. T. Direct transport of cocaine from the nasal

cavity to the brain following intranasal cocaine administration in rats. Journal of

pharmaceutical sciences 88, 754-758 (1999).

210 Hoekman, J. D. & Ho, R. J. Enhanced analgesic responses after preferential delivery of

morphine and fentanyl to the olfactory epithelium in rats. Anesthesia and analgesia 113,

641-651 (2011).

211 Hoekman, J. D. & Ho, R. J. Effects of localized hydrophilic mannitol and hydrophobic

nelfinavir administration targeted to olfactory epithelium on brain distribution. AAPS

PharmSciTech 12, 534-543 (2011).

212 Yang, D. et al. Intranasal delivery of cell-penetrating anti-NF-kappaB peptides (Tat-

NBD) alleviates infection-sensitized hypoxic-ischemic brain injury. Experimental

neurology (2013).

213 NeuroPharma, I. Rat POD Device Training Guide (Impel NeuroPharma, Seattle,

Washington, 2012).

214 Schwarze, S. R., Ho, A., Vocero-Akbani, A. & Dowdy, S. F. In vivo protein transduction:

delivery of a biologically active protein into the mouse. Science 285, 1569-1572 (1999).

215 Hopp, T. P. et al. A Short Polypeptide Marker Sequence Useful for Recombinant Protein

Identification and Purification. Nature Biotechnology 6, 1204-1210 (1988).

Page 129: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

119

216 Malhotra, A. Tagging for protein expression. Methods in enzymology 463, 239-258

(2009).

217 Wu, H., Hu, K. & Jiang, X. From nose to brain: understanding transport capacity and

transport rate of drugs. Expert opinion on drug delivery 5, 1159-1168 (2008).

218 Thorne, R. G., Emory, C. R., Ala, T. A. & Frey, W. H., 2nd. Quantitative analysis of the

olfactory pathway for drug delivery to the brain. Brain research 692, 278-282 (1995).

219 Nijboer, C. H. et al. Strong neuroprotection by inhibition of NF-kappaB after neonatal

hypoxia-ischemia involves apoptotic mechanisms but is independent of cytokines.

Stroke; a journal of cerebral circulation 39, 2129-2137 (2008).

220 Berridge, M. J. Neuronal calcium signaling. Neuron 21, 13-26 (1998).

221 Tucker, J. C. & File, S. E. The effects of tricyclic and 'atypical' antidepressants on

spontaneous locomotor activity in rodents. Neuroscience and biobehavioral reviews 10,

115-121 (1986).

222 Callaway, J. K., Jones, N. C. & Royse, C. F. Isoflurane induces cognitive deficits in the

Morris water maze task in rats. European journal of anaesthesiology 29, 239-245 (2012).

223 Qu, X. et al. Hippocampal glutamate level and glutamate aspartate transporter (GLAST)

are up-regulated in senior rat associated with isoflurane-induced spatial learning/memory

impairment. Neurochemical research 38, 59-73 (2013).

224 Willner, P. Validity, reliability and utility of the chronic mild stress model of depression:

a 10-year review and evaluation. Psychopharmacology 134, 319-329 (1997).

225 Grippo, A. J., Beltz, T. G. & Johnson, A. K. Behavioral and cardiovascular changes in

the chronic mild stress model of depression. Physiology & behavior 78, 703-710 (2003).

226 D'Aquila, P. S., Peana, A. T., Carboni, V. & Serra, G. Exploratory behaviour and

grooming after repeated restraint and chronic mild stress: effect of desipramine.

European journal of pharmacology 399, 43-47 (2000).

227 D'Aquila, P. S., Peana, A. T., Carboni, V. & Serra, G. Different effect of desipramine on

locomotor activity in quinpirole-treated rats after repeated restraint and chronic mild

stress. Journal of psychopharmacology 14, 347-352 (2000).

228 Schweizer, M. C., Henniger, M. S. & Sillaber, I. Chronic mild stress (CMS) in mice: of

anhedonia, 'anomalous anxiolysis' and activity. PloS one 4, e4326 (2009).

229 Jose, S. et al. Thermo-sensitive gels containing lorazepam microspheres for intranasal

brain targeting. International journal of pharmaceutics 441, 516-526 (2013).

230 El-Dakrouri, W. A., Ibrahim, H. K., Ghorab, M. K. & Ghorab, M. M. Enhancement of

the intranasal delivery of insulin via a novel mucoadhesive Carbopol gel. The Journal of

pharmacy and pharmacology 62, 866-872 (2010).

231 Jiang, L., Gao, L., Wang, X., Tang, L. & Ma, J. The application of mucoadhesive

polymers in nasal drug delivery. Drug development and industrial pharmacy 36, 323-336

(2010).

Page 130: An Intranasal Delivery Method for Novel Peptide ... · Figure 3-1 Representative images of deposition of Mark-It Blue tissue marker deposition after ... D2S dopamine D2 receptor -

120

Appendix 1: A Sufficient intranasal D1-D2 interfering peptide

dose to produce antidepressant effect in the Forced Swimming

Test (Calculation)

Efficiency of Intranasal Delivery to CNS: 1-5%15

of total intranasal dose

Previous D1-D2 Interfering peptide administered directly to the PFC: 5 nmol3

Estimate of Sufficient Intranasal dose: 100 × 5nmol = 500 nmol

Estimated Weight of Rats at time of behavioral testing : 300 – 325 g

Dose as nmol/g body weight: 500 nmol ÷ 300 g = 1.67 nmol/g

Dose as mg/kg body weight : 5.75 mg / kg