Вы находитесь на странице: 1из 103

THE ROLE OF THE VASOPRESSIN 1B RECEPTOR IN THE REGULATION OF

SENSORIMOTOR GATING

A thesis submitted to Kent State University


in partial fulfillment of the requirements
for the degree of Master of Science

By

Monica Dhakar

May 2011
Thesis written by
Monica Dhakar
M.B.B.S., Byramjee Jeejeebhoy Medical College, 2008
M.S., Kent State University, 2011

Approved by

________________________ Dr. H. Caldwell, Advisor

________________________Dr. R. Dorman, Director, School of Biomedical Sciences

________________________Dr. John Stalvey, Dean, College of Arts and Sciences

ii
TABLE OF CONTENTS

PAGE

LIST OF FIGURES………………………………………………………………vi

LIST OF TABLES……………………………………………………………...viii

ACKNOWLEDGEMENTS………………………………………………………ix

Introduction………………………………………………………………………1

Vasopressin………………………………………………………………..2

Sexual dimorphism in the central vasopressin system…………………….3

Vasopressin and social behavior…………………………………………..3

Vasopressin receptors……………………………………………………..5

The vasopressin 1b receptor………………………………………………6

Vasopressin 1b receptor knockout mice…………………………………..7

Vasopressin, the vasopressin 1b receptor, and neuropsychiatric disorders.8

Sensorimotor gating……………………………………………………...10

Prepulse inhibition of the startle reflex…………………………..11

Cortico-striato-pallido-pontine circuitry…………………………13

Neurotransmitter systems regulating prepulse inhibition of the

startle reflex………………………………………….…………..15

Interactions of vasopressin and the cortico-striato-pallido-pontine

circuitry…………………………………………………………..17

iii
Objective…………………………………………………………………………17

Methods…………………………………………………………………………..20

General Methods…………………………………………………………………20

Animals…………………………………………………………………..20

Prepulse inhibition the startle reflex……………………………………..20

Drugs……………………………………………………………..20

PPI………………………………………………………………..21

Procedure………………………………………………………...21

Study design……………………………………………………...24

Quantitative real time polymerase chain reaction ……………………….26

Procedure………………………………………………………...26

Individual Experiments…………………………………………………………..26

Experiment 1A and 1B…………………………………………………...26

Experiment 2A and 2B…………………………………………………...27

Experiment 3……………………………………………………………..27

Statistics………………………………………………………………….28

Experiment 4A and 4B…………………………………………………..28

Statistics………………………………………………………………….29

Results……………………………………………………………………………30

Experiment 1A…………………………………………………………...31

Experiment 1B…………………………………………………………...31

Experiment 2A…………………………………………………………...37

iv
Experiment 2B…………………………………………………………...43

Experiment 3……………………………………………………………..49

Experiment 4A…………………………………………………………...55

Experiment 4B…………………………………………………………...55

Discussion………………………………………………………………………..64

References………………………………………………………………………..74

Appendix I……………………………………………………………………….88

Appendix II………………………………………………………………………93

v
LIST OF FIGURES

PAGE

Introduction

Fig 1: Role of vasopressin in regulation of social behaviors……………...4

Fig 2: Schematic diagram of PPI………………………………………...12

Fig 3: CSPP circuit………………………………………………………14

Methods

Fig 4: Schematic diagram representing the components of SR-LAB……22

Fig 5: Acoustic startle chamber …………………………………………23

Fig 6: Study design for Experiments 1, 2 and 3………………………….25

Results

Fig7: Experiment 1A Habituation of startle in females………………….32

Fig 8: Experiment 1A Baseline PPI percentage in females ……………..34

Fig 9: Experiment 1A PPI percentage following drug treatment in

females…………………………………………………………….35

Fig 10: Experiment 1B Habituation of startle in males …………………38

Fig 11: Experiment 1B Baseline PPI percentage in males………………40

Fig 12: Experiment 1B PPI percentage following drug treatment in

male………………………………………………….…………..41

Fig 13: Experiment 2A Habituation of startle in females…..…………..44

Fig 14: Experiment 2A Baseline PPI percentage in females…………….46

vi
Fig 15: Experiment 2A PPI percentage following drug treatment in

females…………………………………………………………...47

Fig 16: Experiment 2B Habituation of startle in males………………….50

Fig 17: Experiment 2B Baseline PPI percentage in males………………52

Fig 18: Experiment 2B PPI percentage following drug treatment in

males……………………………………………………………..53

Fig 19: Experiment 3 Habituation of startle in males…………………....56

Fig 20: Experiment 3 Baseline PPI percentage in males………………...58

Fig 21: Experiment 3 PPI percentage following drug treatment in

males…………………………………………………………….59

Fig 22: Experiment 4A Expression of NMDAR1 in males………………62

Fig 23: Experiment 4B Expression of NMDAR2A in males……………..63

Appendix I

Fig 24: PPI percentage in Avpr1b +/− mice following drug treatment….90

Fig 25: PPI percentage following AMP and saline in Avpr1b +/− mice...91

vii
LIST OF TABLES

PAGE

Results

Table 1: Experiment 1A Startle amplitude in females………………...…33

Table 2: Experiment 1A Mean and SEM for PPI in females…………….36

Table 3: Experiment 1B Startle amplitude in males………………..........39

Table 4: Experiment 1B Mean and SEM for PPI in males………………42

Table 5: Experiment 2A Startle amplitude in females…………………...45

Table 6: Experiment 2A Mean and SEM for PPI in females…………….48

Table 7: Experiment 2B Startle amplitude in males……………………..51

Table 8: Experiment 2B Mean and SEM for PPI in males………………54

Table 9: Experiment 3 Startle amplitude in males……………………….57

Table 10: Experiment 3 Mean and SEM for PPI in males……………….60

Appendix II

Table 11: PPI program…………………………………………………...92

viii
ACKNOWLEDGEMENTS

I would like to thank my advisor and mentor Dr. Heather Caldwell for giving me an

opportunity to work in her lab and her invaluable guidance over the course of three years.

Dr. Caldwell has always provided me constant encouragement and endless support in all

my endeavors including research and future career goals. I would also like to thank Dr.

Eric Mintz and Dr. John Johnson for serving on my thesis committee and providing

valuable insights and expert advice on my research project.

I am grateful to my colleagues; Erica Stevenson, Megan Rich and Uju Dike for their

patience in teaching me all the lab techniques that I know today. Without their support

and friendship, this project could not have been completed. I would also like to thank

Shannah Witchey, and all the undergraduate students for their help in conducting some of

the experiments.

I would like to extend my gratitude to my friends; Vivek, Priyanka, Sagar and

Harsh for being the family to me for the last three years. A special thanks to my friend,

Charu; for always being there with me during the ups and downs of the graduate school.

Lastly, a heartfelt thanks to the people, whom I owe everything that I have ever

achieved; Mom, Dad, Neha, and Dhaval! I cannot thank them all enough for everything

that they are to me. I thank each one of them for believing in me and in my dreams.

ix
Without Dhaval’s love and support, I would not be where I am today. I am grateful to

you for being the pillar of strength and encouragement at every step of my life!

x
Introduction:

The expression of normal social behavior is crucial for the development and

survival of many species (Donaldson and Young, 2008). Broadly, social behavior can be

defined as interactions between individuals of the same species and includes affiliation,

aggression, reproduction, and communication (Sokolowski, 2010; Insel and Young,

2000). Successful social interactions require animals to observe the actions of others,

make predictions about their behavior, and respond appropriately (Skuse and Gallagher,

2009). In humans, aberrant social behavior is characteristic of a variety of

neuropsychiatric disorders, such as autism, schizophrenia, obsessive compulsive disorder

(OCD), anxiety, and depression. Across species, the neural substrates and the

mechanisms regulating social behavior are highly conserved (Donaldson and Young,

2008).

A number of hormones and neuropeptides are important to the neural regulation

of social behavior. In particular, two neurohormones, oxytocin (Oxt) and vasopressin

(Avp), have consistently been implicated in the neural regulation of social behavior (as

reviewed in Caldwell et al., 2008; Lee et al., 2009; Ross and Young, 2009).

Evolutionarily, Oxt and Avp and their gene families are largely conserved across species

(Acher et al., 1995). Both peptides are composed of nine amino acids and differ from one

another in only two amino acids, i.e., those in the third and eighth positions. Oxt and Avp

are hydrophilic in nature and are not thought to readily cross the blood brain barrier.

1
2

Further, both exert their actions through membrane bound G protein coupled receptors.

The effectiveness of Oxt and Avp at regulating behavior is attributed to three

characteristics: 1) their localized distribution in the brain 2) their slow and enduring

effects, and 3) their plasticity (Insel and Young, 2000).

Vasopressin

Avp is synthesized as a part of a preprohormone, which is then enzymatically

cleaved to produce the biologically active nonapeptide, a neurophysin, and a

glycopeptide. Once cleaved, Avp is transported along the axons and released (Brownstein

et al., 1980; Acher et al., 2002). Avp is synthesized primarily in the magnocellular

neurons of the supraoptic (SON) and paraventricular nucleus (PVN) of the hypothalamus

that project to the posterior pituitary. Upon appropriate stimulation, Avp is released into

the blood stream from the posterior pituitary to produce systemic effects which include

the regulation of water balance and maintenance of blood pressure by acting on the blood

vessels and the kidneys (Nishimura and Fan, 2003; Bankir, 2001). Centrally, small

quantities of Avp are released from the dendrites of the magnocellular neurons to produce

local effects. In many species, Avp is also made in small populations of parvocellular

neurons within the PVN, the bed nucleus of stria terminalis (BNST), the medial

amygdala (MeA), and the suprachiasmatic nucleus (SCN) (Sofroniew, 1983). These

neurons provide robust projections to a number of brain areas, including, hippocampus,

subiculum, diagonal band of Broca, locus coeruleus, solitary tract nucleus, dorsal motor

nucleus of vagus, olfactory tubercle, lateral septum (LS), lateral habenula (LH), ventral
3

tegmental area (VTA), and spinal cord (Buijs, 1978; De Vries and Buijs, 1983; Millan et

al., 1984).

Sexual dimorphisms in the central vasopressin system

The distribution of Avp immunoreactive (Avp-ir) neurons and fibers shows a

remarkable degree of sexual dimorphism. In most mammals especially rodents, males

have a higher density of Avp-ir neurons and fibers in the MeA, BNST, LH and LS, as

compared to females (De Vries et al., 1983). However, there are exceptions to these

sexual dimorphisms and the distribution varies across species. For example, in primates

such as marmosets, macaques, and humans, there is a similar distribution of Avp-ir

neurons between males and females (Fliers et al., 1986; Caffe et al., 1989; Wang et al.,

1997). In guinea pigs, there is reverse sexual dimorphism with females having higher

Avp-ir fibers in the inferior colliculus, ventral trapezoid body, and dorsal cochlear

nucleus than males (Dubois-Dauphin et al., 1987; Dubois-Dauphin et al., 1989). Since

Avp is involved in the modulation of sex specific behaviors, the distribution of Avp-ir

fibers are also sensitive to circulating gonadal hormones (for full review on the

interactions of gonadal steroids and vasopressin, please see Dhakar et al., in press). For

instance, castration reduces Avp-ergic innervations from BNST and MeA in both males

and female rats and administration of testosterone increases the density of Avp fibers in

these areas (de Vries et al., 1984).

Vasopressin and social behavior


4

VASOPRESSIN

Avpr1 Avpr1
a b

Aggression Social Behaviors


Stress

Social
Affiliation
Memory
Anxiety

Figure 1: Schematic diagram showing the role of vasopressin in the regulation of social
behaviors
5

Centrally released Avp acts as a neuromodulator for a variety of social behaviors

such as, social recognition, social motivation, social memory, aggression, affiliation, and

parental behavior (as reviewed in Caldwell et al., 2008; Donaldson et al., 2008; Lee et al.,

2009) (Figure 1). Some evidence which indicates the importance of Avp in the regulation

of social behaviors comes from studies in Brattleboro rat (BB-Ho), a naturally occurring

Avp mutant, that lacks the ability to synthesize Avp (Bohus and de, 1998), and thus has

diabetes insipidus. BB-Ho rats have deficits in memory, social recognition, social

motivation, and attention (Laycock et al., 1983; Williams et al., 1985; Williams et al.,

1983). Exogenous administration of Avp in these rats improves certain social behaviors

such as social recognition (Engelmann and Landgraf, 1994). Further, in rodents,

intracerebral injection of an Avp analogue facilitates aggression (Ferris et al., 1997;

Caldwell and Albers, 2004), partner preference (Winslow et al., 1993; Cho et al., 1999),

and paternal behavior (Wang et al., 1994). In humans, intranasal administration of Avp

enhances verbal memory and reaction time in males (Born et al., 1998; Beckwith et al.,

1983).

Vasopressin receptors

Avp exerts it effects via three different receptors: 1) the Avp 1a receptor (Avpr1a)

2) the Avp 1b receptor (Avpr1b) and 3) the Avp 2 receptor (Avpr2). While the Avpr1a

and the Avpr1b are found centrally (as well as peripherally), the Avpr2 is only expressed

peripherally (Tribollet et al., 1988; Lolait et al., 1992). Across species, the Avpr1a is

widely distributed in the central nervous system (CNS) in areas known to be important
6

for neural regulation of social behavior (Johnson et al., 1993; Young et al., 2000;

Tribollet et al., 1997). This thesis will focus specifically on the Avpr1b (for a review of

the role of the Avpr in the neural regulation of behavior please see Caldwell et al., 2008;

Bielsky et al., 2004).

The vasopressin 1b receptor

The Avpr1b is a seven transmembrane receptor and belongs to the family of G

protein coupled receptors. It is coupled to Gαq/11 GTP binding proteins which along with

Gβλ activates a downstream 1,4,5-inositol triphosphate (IP3) second messenger pathway

(Michell et al., 1979; Jard et al., 1987). In the rat brain, in situ hybridization and RT-PCR

have localized Avpr1b transcripts to a number of areas that are known to regulate social

behaviors; olfactory bulb, LS, cerebral cortex, hippocampus, PVN, SCN, cerebellum, red

nucleus, and piriform cortical layer II (Lolait et al., 1995; Vaccari et al., 1998; Saito et

al., 1995). However, it should be noted that the probes used in the above mentioned in

situ hybridization studies had some identity to the oxytocin receptor and the Avpr1a. A

subsequent study which used more specific riboprobes, found a more restricted

localization of Avpr1b in the anterior pituitary, and in the CA2 region of the

hippocampus in rat, mouse, and human as detected by in situ hybridization and RT-PCR

(Young et al., 2006). So far, the distribution of Avpr1b has not been studied using

receptor autoradiography, due to the lack of an available pharmacologically specific

radiolabeled ligand, or with immunocytochemistry, due to lack of a specific antibody. Of


7

the species in which the Avpr1b has been sequenced and mapped, there is no evidence of

sexual dimorphism in its distribution.

Evidence for the role of the Avpr1b in the regulation of social behaviors such as

aggression, social recognition and social motivation comes from studies using an oral

Avpr1b antagonist (SSR149415), Avpr1b agonists and genetically engineered Avpr1b

knockout (Avpr1b −/−) mice (Wersinger et al., 2002; Tanoue et al., 2004).The

development of Avpr1b −/− mice, in particular, has been helpful in elucidating the role of

the Avrp1b in the neural regulation of social behaviors. The work described in this thesis

utilized Avpr1b −/− mice to test our central hypothesis.

Vasopressin 1b receptor knockout mice

Avpr1b −/− mice were first generated in 2002 through a targeted disruption of the

Avpr1b gene (Wersinger et al., 2002). Initial characterization of Avpr1b −/− mice found

that the males have deficits in social aggression and social motivation. However,

predatory aggression in these mice seems to be normal, indicating that they do not have a

global deficit in processing and attacking the stimuli but rather have a deficit in

processing the social cues associated with social aggression (Wersinger et al., 2007).

They also have impaired social recognition, despite normal olfactory function (Wersinger

et al., 2004; Wersinger et al., 2002). Avpr1b −/− females have deficits in maternal

aggression and an abnormal Bruce Effect (Wersinger et al., 2008; Wersinger et al., 2007).

(The Bruce Effect is a pheromonally mediated response wherein the female terminates a

pregnancy in response to chemosensory cues from an unfamiliar male). Avpr1b −/−


8

females fail to terminate their pregnancy in presence of unfamiliar male. Given the

discreet localization of the Avpr1b within the CA2 region of the hippocampus in mice,

these data suggest that Avpr1b might be important for the formation and retrieval of

memories about social context (Young et al., 2006). This hypothesis fits well with the

phenotypic data in Avpr1b −/− mice demonstrating that they have deficits in social

recognition tasks. Avpr1b −/− mice also have an attenuated stress response with

suppressed activity of the hypothalamic-pituitary-adrenal (HPA) axis under resting as

well as stress condition (Tanoue et al., 2004). Specifically, Avpr1b −/− mice have a

blunted adrenocorticotrophic hormone (ACTH) and cortisol response to hypoglycemia

and chronic restraint stress (Lolait et al., 2007). Avpr1b −/− mice also have an impaired

stress response following acute immune stress (administration of bacterial

lipopolysaccharide) and alcohol intoxication (Lolait et al., 2007). In summary, these

studies implicate a role for the Avpr1b in the regulation of social behaviors and allow for

the possibility that abnormalities in the Avp system in humans could contribute to

abnormal social behavior.

Vasopressin, the vasopressin 1b receptor, and neuropsychiatric disorders

Alterations in the Avp system have been found in a number of neuropsychiatric

disorders in humans. Compared to controls, patients diagnosed with major depressive

disorder (van et al., 1997), post-traumatic stress disorder (PTSD) (de Kloet et al., 2008),

and OCD (Altemus et al., 1992) all have elevated plasma levels of Avp. In

schizophrenics, baseline levels of plasma Avp are low compared to controls (Elman et al.,
9

2003); however, acute psychosis is associated with increased serum levels of Avp

(Raskind et al., 1987). There is evidence that elevations in plasma Avp correlate with

central Avp, this is consistent with the effects of Avp on the coordination of physiology

and behavior (Dogterom et al., 1978). However, it should be noted that even elevations in

Avp within the CSF may not be reflective of changes in local release.

In depressed patients, Avp mRNA is upregulated within the SON which is

thought to be associated with elevated glucocorticoid levels (Meynen et al., 2006; Inder

et al., 1997; Chakrabarty et al., 2005). Recently, a study found that a single nucleotide

polymorphism (SNP) in the Avpr1b gene seems to be protective against recurrent major

depression in adults (van et al., 2004). In children, variations in the Avpr1b gene have

been associated with the onset of childhood mood disorders, particularly in females

(Dempster et al., 2007). In adolescents and adults with depression and OCD, treatment

with antidepressants such as fluoxetine, a selective serotonin reuptake inhibitor, and

clomipramine, a tricyclic antidepressant, decrease CSF levels of Avp (Altemus et al.,

1994; De et al., 1993). In addition, intranasal administration of Avp or its analogue

ameliorates some of the negative symptoms of schizophrenia and improves memory

(Brambilla et al., 1986; Brambilla et al., 1989; Iager et al., 1986; Korsgaard et al., 1981).

Studies in animal models also confirm a possible role for Avp and the Avpr1b in

psychiatric disorders. SSR149415, which is a specific orally active Avpr1b antagonist,

has been used to study the role of Avpr1b. In rodents, the administration of this agent

reduces aggression and anxiety-like behaviors, suggesting that the Avpr1b is important
10

for regulation of the circuitry that mediates aggression and anxiety (Griebel et al., 2002;

Blanchard et al., 2005; Stemmelin et al., 2005). Administration of SSR149415 in rodents

also produces antidepressant effects in various behavioral tests, such as forced swim in

rats (Griebel et al., 2002; Griebel et al., 2003).

In sum, all the above studies suggest that Avp could potentially contribute to the

pathogenesis of these neuropsychiatric disorders. However, as Avp is a neuromodulator,

it is not likely that it is the cause of neuropsychiatric disorders. Rather, Avp likely

interacts with the neurocircuitry and neurotransmitter systems that underlie

neuropsychiatric disorders, and in turn affects the susceptibility an individual to a certain

psychiatric disorder. Common to many neuropsychiatric disorders, including

schizophrenia, are abnormal thought processing, thought fragmentation, and difficulty

with attention and cognition. This cognitive fragmentation and disordered thinking is

thought to originate in part from deficits in sensorimotor gating.

Sensorimotor gating

Sensorimotor gating is the ability to process and filter incoming sensory and

cognitive information and selectively allocate attention to the specific information. In

normal awake individuals, sensorimotor gating is constantly active to ‘gate’ any excess or

trivial information and selectively process the informational stimuli. Any individual’s

gating abilities are viewed as plastic and are known to be shaped by a combination of

genetic and environmental factors. Sensorimotor gating is also influenced by changes in

the environmental, neurochemical, and hormonal milieu in the body (Swerdlow, 1996).
11

When this ‘gating window’ is pathologically broken, it can lead to abnormalities in

orderly processing of information; resulting in sensory flooding, and ultimately thought

fragmentation and disruption in cognitive processes (Mcghie and Chapman, 1961;

Venables, 1960). This breakdown of the gating window is thought to be responsible for

the cognitive abnormalities which are features of many neuropsychiatric disorders. An

operational measure of sensorimotor gating is called prepulse inhibition of the startle

reflex (PPI).

Prepulse Inhibition of the startle reflex

The startle reflex is a constellation of responses to sudden, relatively intense

stimuli (auditory, visual or tactile), which is usually defensive in nature (Swerdlow et al.,

2000). The primary mammalian acoustic startle circuit is composed of three synapses

from the auditory nerve to the spinal motor neuron (Davis et al., 1982; Koch and

Schnitzler, 1997). Startle demonstrates several forms of plasticity such as habituation,

and fear potentiation, which are regulated by forebrain neural circuitry that is conserved

across species (Brown et al., 1951; Grillon et al., 1994; Geyer and Braff, 1982).

PPI is the profound decrease in the startle magnitude when the startling pulse is

preceded by a prepulse of weaker intensity by a very brief time window (Graham, 1975;

Hoffman and Searle, 1968; Ison et al., 1973) (Figure 2). The ‘gating’ period is

determined to be approximately 30-500 msec and serves to protect the sensory

information within the weak stimulus so that it is processed without being disrupted by

subsequent stronger stimuli (Hoffman and Searle, 1968; Ison et al., 1973; Swerdlow,
12

Figure 2: Schematic diagram showing the phenomenon of prepulse inhibition (PPI). PPI
is the decrease in startle magnitude when a weaker prepulse precedes the startling pulse.
(Modified and adapted from Swerdlow et al., 2000)
13

1996). PPI is a normal phenomenon that occurs virtually in all mammals, including

primates, and does not exhibit habituation or extinction with multiple trials.

PPI is one form of startle plasticity that shows striking similarities across species

from rodents to humans, and is easily quantified (Swerdlow et al., 1999). It is a robust

experimental phenomenon that occurs irrespective of the sensory modality of the startling

stimulus and the prepulse. In humans, PPI is measured by using electromyography of the

orbicularis oculi muscle which is a part of the blink reflex of the startle component (Braff

et al., 1992). In rats and mice, the whole body reflexive flinch to startle stimulus can be

measured by using a stabilimeter chamber (Swerdlow et al., 2000).

Deficits in PPI can be seen in a number of disorders with thought fragmentation

such as schizophrenia (Bolino et al., 1994; Braff et al., 1992; Braff et al., 1978; Braff et

al., 1999; Grillon et al., 1992; Kumari et al., 1999; Weike et al., 2000), OCD (Swerdlow

et al., 1993; Schall et al., 1996), Huntington’s disease (Swerdlow et al., 1995), Tourette

syndrome (Castellanos et al., 1996), nocturnal enuresis, and attention deficit hyperactive

disorder (ADHD) (Ornitz et al., 1992). Deficits in PPI are not unique to a particular

disease but are characteristic of various disorders with pathological disruption of circuitry

regulating it called the cortico-striato-pallido-pontine (CSPP) circuit (Swerdlow et al.,

1992).

Cortico-striato-pallido-pontine circuitry

Studies in rats have identified the neural substrates that regulate PPI. This serial

and parallel neural circuitry is called the CSPP circuit (Figure 3). It connects the limbic
14

Ventral
tegmental area

Hippocampus
mPFC

Amygdala
Nucleus accumbens

Ventral pallidum CSPP Circuit

Pedunculopontine
tegmental nucleus

S R

Cochlear Caudal pontine Motor


nucleus reticular nucleus neuron

Acoustic stimulus
Response
Primary acoustic startle circuit

Figure 3: Schematic diagram showing the main components of the CSPP circuit and
its regulatory control over startle reflex. The acoustic stimulus S produces a motor
response R through the simple pontine circuit. Prepulse effects on R are mediated
via the pedunculopontine tegmental nucleus, which is under the control of serial
and parallel descending projections from the forebrain. (Modified and adapted from
Koch and Schnitzler et al., 1997; Swerdlow et al., 2001) (Abbreviations: mPFC,
medial prefrontal cortex)
15

cortex with the ventral striatum, ventral pallidum, pontine tegmentum and converges with

the primary startle circuit at the level of caudal pons (Koch and Schnitzler, 1997;

Swerdlow and Geyer, 1998; Swerdlow et al., 2001; Koch et al., 1993). Though the main

inhibitory effect of prepulse on startle is exerted at the level of pons, the CSPP circuit

regulates the quality of the inhibitory activity and thus the degree of inhibition of the

response to stimuli when preceded by a prepulse (Swerdlow et al., 2001). The CSPP

circuit is known to be regulated by a variety of neurotransmitter systems in humans and

rodents, including the dopaminergic, serotonergic, and glutamatergic systems.

Neurotransmitter systems regulating prepulse inhibition of the startle reflex

Abnormalities in the dopamine, the glutamate, and the serotonin system have

been consistently implicated in pathogenesis of a number of neuropsychiatric disorders

such as schizophrenia, major depression, panic disorders and OCD (Carlsson et al.,

1997). Since these neurotransmitters are also reported to act on various neural substrates

in the CSPP circuitry, pharmacological manipulation of these neurotransmitter systems

using various psychotomimetics, can be used to produce deficits in PPI in rodents.

The dopamine system has been studied extensively both in humans and rodents.

In healthy human subjects, direct dopamine agonists like bromocriptine (a D2 agonist)

and indirect dopamine agonists like amphetamine (AMP) have been shown to disrupt PPI

(Abduljawad et al., 1998; Abduljawad et al., 1999; Kumari et al., 1998; Hutchison and

Swift, 1999). Similarly, in many preclinical studies in rats and mice, apomorphine

(APO), which is a direct dopamine agonist, and AMP produce robust decreases in PPI
16

(Mansbach et al., 1988; Swerdlow et al., 2000; Dulawa and Geyer, 1996; Ralph et al.,

1999; Ralph et al., 2001). These PPI disrupting effects of dopamine agonists can be

effectively blocked by pretreatment with typical antipsychotics such as haloperidol

(which primarily acts on D2 receptors) suggesting that dopamine and its receptors are

important regulators of PPI (Abduljawad et al., 1998; Abduljawad et al., 1999). In

humans, dopamine hyperactivity has been implicated in the pathogenesis of

schizophrenia and deficits in sensorimotor gating. Few studies report that the PPI deficits

seen in schizophrenic patients can be successfully reversed by administration of typical

and atypical antipsychotics (which have primary actions on dopamine receptors),

suggesting the role of dopamine in the regulation of PPI (Kumari et al., 1999; Kumari et

al., 2000; Weike et al., 2000).

Another important neurotransmitter implicated in the regulation of PPI is

glutamate. Dysregulation of the glutamatergic system is thought to be involved in the

etiology of schizophrenia, OCD, and mania; all disorders with abnormal thought

processing (Chakrabarty et al., 2005; Javitt, 1987). In humans, schizophrenia-like

symptoms, including PPI deficits, can be produced by administering NMDA receptor

antagonists, like ketamine. In rodents and non-human primates, deficits in PPI can be

modeled using other NMDA receptor antagonists, like phencyclidine (PCP) or

dizocilpine (MK-801) (Curzon and Decker, 1998; Mansbach and Geyer, 1989; Furuya et

al., 1999; Linn et al., 1999). Further, the effects of NMDA receptor antagonists on PPI

can be reversed using atypical antipsychotics like clozapine, olanzapine, and quetiapine,

which have actions on multiple receptors (Swerdlow et al., 1996; Swerdlow et al., 1998;
17

Yamada et al., 1999). The reversal of PPI with atypical antipsychotics suggests that the

regulation of PPI by glutamate might not be due to a single receptor interaction, but

might involve many downstream receptors, synapses, and other neurotransmitter systems

such as serotonin (Olney et al., 1999).

Interactions of vasopressin and the cortico-striato-pallido-pontine circuitry

Avp is a neuromodulator that is known to interact with the dopamine, glutamate

and serotonin systems in various brain areas (Skuse and Gallagher, 2009; Syed et al.,

2007). Abnormalities of the Avp system have been implicated in a variety of

neuropsychiatric disorders which are all characterized by a dysfunction of these

neurotransmitters. The Avp mutant, BB-Ho rats have also been found to have

abnormalities in several neurotransmitter systems including dopamine and serotonin

(Feenstra et al., 1990). They also exhibit profound deficits in PPI at baseline, which can

be successfully reversed by using both typical and atypical antipsychotics (Feifel and

Priebe, 2001; Feifel et al., 2007), which suggests that Avp does play some role in

regulating PPI and might interact with more than one neurotransmitter system. Exactly

how Avp interacts with these neurotransmitter systems still needs to be explored further.

Moreover, whether this deficit in PPI is mediated via Avpr1a or Avpr1b has yet to be

investigated. The work described in this thesis set out to determine if the Avpr1b interacts

with the neural circuitry that regulates PPI.

Objective
18

Our central hypothesis is that Avp acting via the Avpr1b contributes to the

regulation of PPI through its interaction with either the glutamatergic or dopaminergic

systems. This hypothesis is based on several pieces of evidence suggesting a role of Avp

in regulation of PPI. First, BB-Ho rats, which are genetically deficient in the production

of Avp, have been shown to have deficits in baseline PPI. Second, these deficits in PPI

can be reversed by infusion of atypical antipsychotics such as clozapine and risperidone

(Feifel et al., 2007). We tested our central hypothesis by treating Avpr1b +/+ and Avpr1b

−/− mice with psychotomimetics and measuring their effects on PPI. The

psychotomimetics used in this study were dopamine agonists (APO and AMP) and

NMDA receptor antagonist (MK-801 and PCP), since these are the drugs most

commonly used to disrupt PPI.

Experiment 1, was performed to examine the effects of APO, AMP, and MK-801

on PPI in Avpr1b −/− and Avpr1b +/+ mice. It was hypothesized that the Avpr1b −/−

mice would be more susceptible to the PPI disruptive effects of psychotics. Experiment 2,

was performed to confirm the results of Experiments 1 by comparing the effects of a

single drug MK-801 to saline. It was hypothesized that Avpr1b −/− mice treated with

MK-801 would have more disrupted PPI as compared to Avpr1b +/+ mice. Experiment 3,

was performed to compare the effects of two different NMDA receptor antagonists MK-

801 and PCP on PPI in Avpr1b −/− and Avpr1b +/+ mice. It was hypothesized that a

stronger disruption of PPI will be seen following treatment with PCP in Avpr1b −/− mice

as compared to MK-801, since PCP augments the effects of glutamate through sigma
19

receptors. Experiment 4 was designed to find out where in the brain the Avp system

interacts with glutamatergic system to affect PPI. Therefore, in Experiment 4,

quantitative real time PCR was performed to quantify the expression of NMDA receptor

mRNA (NMDAR1 and NMDAR2A) in cortex, striatum and hippocampus in male Avpr1b

−/− and Avpr1b +/+ mice. It was hypothesized that developmental lack of Avpr1b in

Avpr1b −/− mice would cause significant changes in expression of NMDA receptor

mRNA as compared to Avpr1b +/+ mice.


Methods:

General Methods

Animals

This line was originally generated at the National Institute of Mental Health

(NIMH) by W. Scott Young, III (Wersinger et al., 2002). These mice were fully

backcrossed into a C57BL/6J background and shipped to Kent State University in 2007

where they have been bred in the vivarium in Cunningham Hall ever since. All the

animals used in the study (Avpr1b −/− and Avpr1b +/+ male and females) were offspring

from heterozygous breeding pairs. After weaning, animals were reared in the same-sex

sibling groups. At the time of weaning (21 days), tails were clipped in order to extract the

DNA, and PCR was performed using the specific primers for Avpr1b for genotyping the

animals (Wersinger et al., 2002). For each Experiment, the animals were adults between

three and six months of age. Food and water were available ad libitum throughout the

studies. All behavior testing was done during the light phase of the 12:12 light dark cycle.

Experiments were conducted in accordance with the protocol approved by the Kent State

University Institutional Animal Care and Use Committee.

Prepulse Inhibition of the Startle Reflex

Drugs

20
21

The following doses of APO, AMP, MK-801 and PCP were used and were

chosen based on the literature (Caldwell et al., 2009; Yee et al., 2004) and pilot

experiments (Appendix I); APO (10mg/kg), AMP (12mg/kg), MK-801 (0.7mg/kg) and

PCP (8mg/kg). All the drugs were dissolved in 0.9% saline. APO and AMP were diluted

to 1.5mg/ml, MK-801 to 10.5mg/ml and PCP to 1.2mg/ml. Saline was given in the same

volume as APO and AMP and served as control.

PPI

Two startle chambers were used for all the experiments (SR-LAB; San Diego

instruments, San Diego, CA, USA) (Figure 4 and 5). The startle chamber consisted of a

small plexiglass cylinder (8 cm diameter, 16cm long) which is mounted on a platform

inside a chamber. The loudspeaker inside the chamber generated the background noise as

well as all the prepulse and pulse alone tones. The startle response was measured from

the whole body reflexive flinch of the animals to acoustic startle stimuli. Vibrations of

the animal were sensed and transduced by the piezoelectric unit (motor sensor) attached

to the bottom of the platform which is then converted into arbitrary units by the

potentiometer and stored in the computer.

Procedure

Each testing session consisted of a 5 min acclimation period followed by 60 trials

for 20 min. During each session, background noise of 74dB was generated by the

loudspeaker. Sessions consisted of NO PULSE, PULSE ALONE and PREPULSE +

PULSE trials. During NO PULSE trails, no tone was presented. The PULSE ALONE
22

Figure 4: Schematic diagrams representing the components of the SR-LAB system.


23

Figure 5: Acoustic startle chamber (SR LAB, San Diego, CA)


24

trial consisted of 120dB pulse of 40ms duration. During the PREPULSE + PULSE trial,

three prepulse tones each 20ms long, of 77dB (3dB above the background), 80dB (6dB

above the background) and 86dB (12dB above the background) were presented 100 msec

prior to the PULSE tone. Each session began and ended with 5 trials of pulse alone tones.

In between the pulse alone trials, each session consisted of 10 blocks of 5 trials, during

which different stimuli were presented in a random order.

Study Design

Prior to drug testing, each animal was tested for baseline PPI without any drug

treatment. Following baseline testing, repeated measures testing with drugs was done

with each animal. APO, AMP, MK-801, PCP and saline (0.9%) were administered

intraperitoneally (i.p.) to animals in a counterbalanced manner, though which drug was

administered depended on the Experiment. A minimum of 1 week between testing was

observed in order to avoid any residual drug effects. PPI percentage was calculated using

the following equation: (1-(startle amplitude of the PREPULSE + PULSE/startle

amplitude of the PULSE ALONE)) × 100. Habituation of startle was calculated using the

average responses from the first 5 PULSE ALONE trials (Pulse 1) and last 5 PULSE

ALONE (Pulse 2) trials. Magnitude of the startle was calculated from the average of all

the PULSE ALONE trials during the entire session excluding the first and last blocks of

PULSE ALONE trials (Figure 6).


25

10 Blocks of 5 Trials
5 Pulse 5 Pulse
Alone Trials NO PULSE Alone Trials
PREPULSE + PULSE (3X)
PULSE 1 PULSE 2
PULSE ALONE

Startle amplitude was Habituation of startle PPI percentage was


calculated from the was measured from calculated from the
average responses to the average responses average response
PULSE ALONE of the first 5 (PULSE from the 10 blocks of
trials. Comparisons 1) and last 5 pulse 5 trials each using
were made between alone (PULSE 2) the equation: (1-
drug treatments as trials and (startle amplitude of
compared to saline. comparisons were the prepulse + pulse
made within drug trial / startle
treatment. amplitude of pulse
alone trial)) × 100.
Comparisons were
made between
genotypes and drug
treatment with
respect to saline.

Figure 6: Study design used to analyze startle amplitude, habituation of startle, and PPI
percentage.
26

Quantitative Real Time Polymerase Chain Reaction

Procedure

Brains were collected from male Avpr1b −/− and Avpr1b +/+ mice and frozen on

dry ice. Tissue punches of 1.00 mm diameter were collected from a 400 µm slab cut on

cryostat, from the striatum, hippocampus and frontal cortex. Samples were homogenized

using homogenizer and RNA was extracted using the RiboPure kit (Ambion, The RNA

Company, Austin, TX). The purity of the sample was established by using the Nanodrop

Spectrophotometer ND-100 (Thermo-Scientific) and 1 µl of extract. Samples having a

ratio of 260/280 between 1.8-2.0 were included in the assay. Purified RNA was reverse

transcribed into cDNA using a ThermoScript RT- PCR system (Invitrogen, Carlsbad, CA,

USA). Specific Taqman primers for NMDAR1 and NMDAR2A were purchased from

Applied Biosystems. Quantitative real time PCR was performed using Stratagene QPCR

System (Agilent Technologies, Foster City, CA, USA). The genes quantified were

NMDAR1 and NMDAR2A within the brain areas mentioned using specific primers.

Triplicates of 20 µl reaction with Taqman Universal master mix and 2 µl of cDNA in

each well were used to perform the assay. GAPDH was used as the control for

normalization.

Individual Experiments

Experiment 1: Assessment of prepulse inhibition of the startle reflex in A) female

Avpr1b +/+ and Avpr1b −/− mice and B) male Avpr1b +/+ and Avpr1b −/− mice

following treatment with APO, AMP, MK-801, and saline


27

In Experiment 1A, 13 Avpr1b +/+ and 10 Avpr1b −/− female mice were used as

subjects. In Experiment 1B, 11 Avpr1b +/+ and 12 Avpr1b −/− male mice were used. The

animals were run in two different cohorts at two different times. Each cohort consisted of

a mix of males and females. However, for analysis purposes, males and females were

considered separately. Each animal was initially tested for baseline PPI measurement

without any drugs. For the drug studies, APO, AMP, MK-801 and saline (0.9%) were

administered i.p. 10 min prior to testing in a counterbalanced manner. Habituation, startle

and PPI were measured as described under statistics section.

Experiment 2: Assessment of the effects of MK-801 and saline on prepulse inhibition

of the startle reflex in A) female Avpr1b +/+ and Avpr1b −/− mice and B) male

Avpr1b +/+ and Avpr1b −/− mice

In Experiment 2A, subjects were 9 Avpr1b +/+ and 7 Avpr1b −/− female mice. In

Experiment 2B, 9Avpr1b +/+ and 10 Avpr1b −/− male mice were used. Males and

females were run at the same time; however analyzed separately. MK-801 and saline

(0.9%) were given i.p. 10 min prior to testing. Data were analyzed as mentioned under

statistics section.

Experiment 3: Assessment of prepulse inhibition of startle reflex in response to MK-

801, PCP, and saline in Avpr1b +/+ and Avpr1b −/− male mice

Only 10 Avpr1b −/− and 7 Avpr1b +/+ male mice were used as study subjects

since a robust effect of MK-801 on PPI was seen only in males in the Experiment 1. MK-
28

801, PCP and saline (0.9%) were administered i.p. in a counterbalanced manner and data

were analyzed as described under statistics section.

Statistics

For Experiments 1, 2, and 3, baseline PPI percentage was analyzed across

genotypes using repeated measure of analysis of variance (ANOVA) comparing the main

effects of genotype, sex and prepulse intensity as well as their interactions. For the drug

studies, PPI percentage was determined using repeated ANOVA and main effects of

genotype, drug treatment, prepulse intensity and their interactions were compared.

Effects in both the sexes were analyzed independently. Animals with PPI percentage

within two standard deviations of the mean were included in statistical analysis. Since we

were interested in habituation across the test session; effect of drugs on habituation was

measured using repeated ANOVA with weight as a covariate. To correct for multiple

comparisons, the α was adjusted to p<0.013. Main effects of the first five PULSE

ALONE trials were compared to last five PULSE ALONE trials as well as those of

genotype and any interactions. Startle was compared using a repeated measures ANOVA.

Main effects of drug treatment, genotype, and their interactions were examined.

Experiment 4: Assessment of gene expression of A) NMDAR1 and B) NMDAR2A in

male Avr1b +/+ and Avpr1b −/− mice

Subjects were 5 Avpr1b −/− and 5 Avpr1b +/+ male mice. Animals were killed by

cervical dislocation and brains removed. Brains were immediately frozen on dry ice and
29

stored at -80C until tissue collection and quantitative real time PCR procedure were

performed as described under quantitative real time PCR methods section.

Statistics

For Experiment 4, the Ct values from the samples were normalized against

GAPDH. The expression values were then calculated using the equation: 2-∆Ct .

Comparisons were then made between genotypes by using t-test.


Results:

Experiment 1A: Assessment of PPI in Avpr1b +/+ and Avprb1 −/− female mice

following treatment with saline, APO, AMP and MK-801.

Habituation of startle

For all the drug treatments, there were no main effects of the drugs, genotype or any

interactions on habituation of startle. There was no habituation to startle between Pulse 1

and Pulse 2 trial sessions within any of the drugs tested (Figure 7).

Startle amplitude

There was a main effect of the drugs on startle amplitude (F3,54 = 16.042, p<0.05), with

APO and AMP decreasing the startle amplitude, whereas MK-801 increased the startle

amplitude. There was no main effect of genotype, or any interactions on the startle (Table

1).

PPI Percentage

Baseline: There were no genotypic differences in the baseline PPI percentage. There were

the expected main effects of prepulse intensity (F2,36 = 25.073, p<0.05), resulting in

increased PPI percentage with increasing prepulse tone (Figure 8).

30
31

Drug treatment: There was a main effect of the drug treatment (F3,54 = 11.636, p<0.05)

with APO, AMP and MK-801 all resulting in disruption of PPI as compared to saline.

There was also a main effect of prepulse intensity (F2,36 = 60.785, p<0.05). However,

there were no genotypic differences in the females on PPI percentage or any other

interactions (Figure 9) and (Table 2).

Experiment 1B: Assessment of PPI in male Avpr1b +/+ and Avprb1 −/− mice

following treatment with saline, APO, AMP and MK-801

Habituation of startle

There were no main effects of drug treatments, genotype or any interactions on the

habituation of startle across Pulse 1 and Pulse 2 sessions in male Avpr1b +/+ and Avpr1b

−/− mice (Figure 10).

Startle amplitude

There was a main effect of the drugs on startle amplitude (F3,60 = 34.775, p<0.05), with

APO decreasing the startle amplitude whereas AMP and MK-801 increased the startle

amplitude. There was no main effect of genotype, or any interactions on the startle (Table

3).

PPI Percentage
32

Saline APO

300 300
*
Pulse 1 Pulse 1
250 Pulse 2 250 Pulse 2

200
Startle Amplitude

200

Startle Amplitude
150 150

100 100

50 50

0 0
Avpr1b +/+ Avpr1b −/− Avpr1b +/+ Avpr1b −/−

AMP MK-801

300 300
*
Pulse 1 Pulse 1
250 Pulse 2 250 Pulse 2

200
Startle Amplitude

200
Startle Amplitude

150 150

100 100

50 50

0 0
Avpr1b +/+ Avpr1b −/− Avpr1b +/+ Avpr1b −/−

Figure 7: Habituation of startle in female Avpr1b +/+ and Avpr1b −/− mice. There was
no habituation of startle to drugs; Saline (0.9%), APO (10mg/kg), AMP (12mg/kg) and
MK-801 (0.7mg/kg), in Avpr1b +/+ and Avpr1b −/− female mice. Data are expressed as
means + standard error of the mean (Abbreviations: APO, apomorphine; AMP,
amphetamine; MK-801, dizocilpine).
33

Mean ± Standard Error of the Mean (Startle amplitude)

Drugs Avpr1b +/+ Avpr1b −/−

Saline 126.36 ± 18.25 111.83 ± 12.75

APO* 56.91 ± 9.70 47.88 ± 7.95

AMP* 82.50 ± 8.72 93.33 ± 10.01

MK-801* 127.97 ± 27.23 159.25 ± 9.21

Table 1: Startle amplitude in female Avpr1b +/+ and Avpr1b −/− mice. There was a main
effect of the drug treatment (p<0.05), with APO and AMP decreasing the startle
amplitude whereas MK-801 increased the startle amplitude in both the genotypes. There
was no genotypic difference between the Avpr1b +/+ and Avpr1b −/− mice. Startle was
calculated from the averages responses from the first and last pulse alone (120dB) trials.
(*p < 0.05 indicating a main effect of drug compared to saline) (Abbreviations: APO,
apomorphine; AMP, amphetamine; MK-801, dizocilpine).
34

Avpr1b +/+
100
Avpr1b −/−
*
*
80
% Prepulse Inhibition

60

40

20

0
3dB 6dB 12dB

Figure 8: Baseline PPI percentage in female Avpr1b +/+ and Avprb1 −/− mice. There was
no genotypic difference in baseline PPI percentage but there was a main effect of
prepulse intensity with the higher decibel prepulse tone resulting in greater PPI
percentage (* indicates p<0.05). Data are expressed as means + standard error of the
mean.
35

Avpr1b +/+

100 3dB
6dB
12dB

% Prepulse Inhibtion 80

60

40

20

0
SALINE APO AMP MK801

Avpr1b −/−

100 3dB
6dB
12dB

80
% Prepulse Inhibtion

60

40

20

0
SALINE APO AMP MK801

Figure 9: PPI percentage in female Avpr1b +/+ and Avpr1b −/− mice following drug
treatment. Treatment with all the three drugs, APO, AMP, and MK-801 resulted in
disruption of PPI as compared to saline (p<0.05). There were however no genotypic
differences between Avpr1b +/+ and Avpr1b −/− female mice. Data are expressed as
means + standard error of the mean. (Abbreviations: APO, apomorphine; AMP,
amphetamine; MK-801, dizocilpine).
36

Mean ± Standard Error of the Mean (PPI percentage)


Drugs and Prepulse
Avpr1b +/+ Avpr1b −/−
Levels
Saline 77 63.40 ± 4.56 66.61 ± 3.53

Saline 80 73.42 ± 3.19 74.14 ± 3.52

Saline 86 78.68 ± 2.21 82.47 ± 1.74

APO 77 48.91 ± 4.02 38.60 ± 9.87

APO 80 57.35 ± 3.62 52.87 ± 6.67

APO 86 58.46 ± 3.02 55.39 ± 10.41

AMP 77 46.51 ± 4.76 38.92 ± 7.58

AMP 80 55.60 ± 5.01 55.75 ± 2.71

AMP 86 63.22 ± 4.14 62.53 ± 3.51

MK-801 77 31.49 ± 9.29 40.22 ± 4.28

MK-801 80 47.78 ± 5.99 42.98 ± 6.62

MK-801 86 63.31 ± 5.62 68.43 ± 4.37

Table 2: Mean and standard error of the mean for drug treatment in female Avpr1b +/+
and Avpr1b −/− mice. (Abbreviations: APO, apomorphine; AMP, amphetamine; MK-
801, dizocilpine)
37

Baseline: There was a main effect of the prepulse intensity (F2,40 = 38.869, p<0.05), with

greater PPI percentage with higher intensity tone. However there were no genotypic

differences in baseline PPI percentage in Avpr1b +/+ and Avpr1b −/− males (Figure 11).

Drug treatment: There was a main effect of drug treatment (F3,60 = 28.954, p<0.05) and

prepulse intensity (F2,40 = 121.057, p<0.05). APO, AMP and MK-801 disrupted the PPI

as compared to saline in both Avpr1b +/+ and Avpr1b −/− male mice but there was no

main effect of genotype. However, there was a genotype × drug interaction (F3,60 = 6.018,

p<0.05). Post hoc tests with repeated ANOVA revealed that treatment with MK-801

resulted in more disruption of PPI in Avpr1b −/− mice as compared to Avpr1b +/+ mice

(p<0.05). There were no other interactions of genotype with any other drug treatment

(Figure 12) and (Table 4).

Experiment 2A: Assessment of prepulse inhibition of startle reflex in female

Avpr1b +/+ and Avpr1b −/− mice following administration of MK-801

Habituation of Startle

There was no habituation to startle reflex in female Avpr1b +/+ and Avpr1b −/− mice

with saline or MK-801 (Figure 13). There was no main effect of genotype, drug or any

other interaction.

Startle amplitude
38

Saline APO

300 300
* *
Pulse 1 Pulse 1
250 Pulse 2 250 Pulse 2

200
Startle Amplitude

200

Startle Amplitude
150 150

100 100

50 50

0 0
Avpr1b +/+ Avpr1b −/− Avpr1b +/+ Avpr1b −/−

AMP MK-801

300 300
* *
Pulse 1 Pulse 1
250 Pulse 2 250 Pulse 2

200
Startle Amplitude

200
Startle Amplitude

150 150

100 100

50 50

0 0
Avpr1b +/+ Avpr1b −/− Avpr1b +/+ Avpr1b −/−

Figure 10: Habituation of startle in male Avpr1b +/+ and Avpr1b −/− mice. There were
no main effects of the drugs; Saline (0.9%), APO (10mg/kg), AMP (12mg/kg) and MK-
801 (0.7mg/kg) on habituation of startle on Avpr1b +/+ and Avpr1b −/− mice. Data are
expressed as mean + standard error of the mean. (Abbreviations: APO, apomorphine;
AMP, amphetamine; MK-801, dizocilpine)
39

Mean ± Standard Error of the Mean (Startle amplitude)

Drugs Avpr1b +/+ Avpr1b −/−

Saline 137.12 ± 18.95 116.91 ± 13.95

APO* 76.19 ± 11.65 96.99 ± 11.38

AMP* 142.35 ± 13.80 117.23 ± 14.56

MK-801* 243.21 ± 29.89 232.95 ± 32.07

Table 3: Startle amplitude in male Avpr1b +/+ and Avpr1b −/− mice. There was a main
effect of the drug treatment on startle amplitude (p<0.05), with APO decreasing the
startle amplitude whereas AMP and MK-801 increased the startle amplitude in both
genotypes. There was no genotypic difference between the Avpr1b +/+ and Avpr1b −/−
mice. Startle was calculated from the averages responses from the first and last pulse
alone (120dB) trials. (*p < 0.05 indicating a main effect of drug compared to saline)
(Abbreviations: APO, apomorphine; AMP, amphetamine; MK-801, dizocilpine).
40

Avpr1b +/+
100
Avpr1b −/−
*
*
80
% Prepulse Inhibition

60

40

20

0
3dB 6dB 12dB

Figure 11: Baseline PPI percentage in male Avpr1b +/+ and Avprb1 −/− mice. There was
no genotypic difference in baseline PPI percentage but there was a main effect of
prepulse intensity with the higher decibel prepulse tone resulting in greater PPI
percentage (* indicates p<0.05). Data are expressed as mean + standard error of the
mean.
41

Avpr1b +/+

100 3dB
6dB
12dB

80

% Prepulse Inhibtion
60

40

20

0
SALINE APO AMP MK801

Avpr1b −/−

100 3dB
6dB
12dB

80

*
% Prepulse Inhibtion

60
*

40

20

0
SALINE APO AMP MK801

Figure 12: PPI percentage following drug treatment in males. Treatment with all the three
drugs, APO, AMP, and MK-801 resulted in disruption of PPI as compared to saline
(p<0.05). There was no main effect of the genotype; however there was a genotype ×
drug interaction (p<0.05). Specifically, treatment with MK-801 resulted in greater
disruption of PPI in male Avpr1b −/− mice as compared to Avpr1b +/+ mice (* indicates
p<0.05). Data are expressed as mean + standard error of the mean. (Abbreviations: APO,
apomorphine; AMP, amphetamine; MK-801, dizocilpine)
42

Mean ± Standard Error of the Mean (PPI percentage)


Drugs and Prepulse
Avpr1b +/+ Avpr1b −/−
Levels
Saline 77 67.33 ± 3.62 57.16 ± 3.89

Saline 80 73.25 ± 3.63 71.90 ± 2.08

Saline 86 78.06 ± 2.47 80.03 ± 1.79

APO 77 51.56 ± 6.30 50.17 ± 5.53

APO 80 66.23 ± 4.03 62.22 ± 4.41

APO 86 65.91 ± 4.11 65.54 ± 4.66

AMP 77 30.11 ± 2.26 39.46 ± 3.98

AMP 80 46.88 ± 5.10 54.62 ± 4.83

AMP 86 54.28 ± 3.62 62.29 ± 3.29

MK-801 77 45.37 ± 3.86 34.62 ± 5.09

MK-801 80 57.80 ± 6.35 42.88 ± 6.20

MK-801 86 73.44 ± 3.55 54.57 ± 7.72

Table 4: Mean and standard error of the mean for drug treatments on male Avpr1b +/+
and Avpr1b −/− mice. (Abbreviations: APO, apomorphine; AMP, amphetamine; MK-
801, dizocilpine)
43

There was a main effect of the drug on startle amplitude (F1,14 = 13.842, p<0.05), with

MK-801 increasing the startle amplitude. There was no main effect of genotype, or any

interactions on the startle amplitude (Table 5).

PPI Percentage

Baseline: There was no genotypic difference between Avpr1b +/+ and Avpr1b −/− female

mice in baseline PPI percentage. However, there was a main effect of prepulse intensity

(F2,26 = 14.393, p< 0.05), with higher prepulse tone resulting in greater PPI percentage

(Figure 14).

Drug treatment: There was a main effect of the drug treatment (F1,14 = 13.095, p<0.05)

and prepulse intensity (F2,28 = 34.016, p<0.05). There was no genotypic effect or any

interactions in Avpr1b +/+ and Avpr1b −/− female mice (Figure 15) and (Table 6).

Experiment 2B: Assessment of prepulse inhibition of startle reflex in Avpr1b

+/+ and Avpr1b −/− male mice following administration of MK-801

Habituation of startle reflex

Male Avpr1b +/+ and Avpr1b −/− mice did not show habituation to startle reflex across

the test sessions with either saline or MK-801 (Figure 16).

Startle amplitude

There was a main effect of the drug (MK-801) on startle amplitude, (F1,15 = 5.587,

p<0.05), with MK-801 increasing the amplitude of the startle reflex. There was no
44

Saline MK-801

300 300
* *
Pulse 1 Pulse 1
250 Pulse 2 250 Pulse 2

200 200
Startle Amplitude

Startle Amplitude
150 150

100 100

50 50

0 0
Avpr1b +/+ Avpr1b −/− Avpr1b +/+ Avpr1b −/−

Figure 13: Habituation of startle in Avpr1b +/+ and Avpr1b −/− female mice. There was
no habituation of startle between PULSE 1 and PULSE 2 trials with saline (0.9%) and
MK-801 (0.7mg/kg) in both genotypes. Data are expressed as mean + standard error of
the mean. (Abbreviations: MK-801, dizocilpine)
45

Mean ± Standard Error of the Mean (Startle amplitude)

Drugs Avpr1b +/+ Avpr1b −/−

Saline 49.94 ± 6.60 56.84 ± 11.46

MK-801* 95.06 ± 22.44 93.43 ± 18.18

Table 5: Startle amplitude in female mice following drug treatment. There was a main
effect of the drug on startle amplitude (p<0.05), with MK-801 increasing the startle
amplitude in both the genotypes. There was no genotypic difference between the Avpr1b
+/+ and Avpr1b −/− mice. Startle was calculated from the averages responses from the
first and last pulse alone (120dB) trials. (*p < 0.05 indicating a main effect of drug
compared to saline) (Abbreviations: MK-801, dizocilpine)
46

Avpr1b +/+
100
Avpr1b −/−
*
*
80
% Prepulse Inhibition

60

40

20

0
3dB 6dB 12dB

Prepulse Intensities
dBA above background

Figure 14: Baseline PPI percentage in females. There was no genotypic difference
between Avpr1b +/+ and Avpr1b −/− females, but an effect of prepulse intensity was
present (p<0.05), with higher prepulse tone resulting in greater PPI percentage. (*
indicates p<0.05). Data are expressed as mean + standard error of the mean.
47

Avpr1b +/+
100
3dB
6dB
12dB
% Prepulse Inhibtion 80

60

40

20

0
SALINE MK801

Avpr1b −/−

100 3dB
6dB
12dB

80
% Prepulse Inhibtion

60

40

20

0
SALINE MK801

Figure 15: PPI percentage following drug treatment in females. There was a main effect
of the drug (p<0.05) with MK-801 disrupting the PPI in both Avpr1b +/+ and Avpr1b
−/− female mice. However, there was no genotypic difference in PPI between Avpr1b +/+
and Avpr1b −/− females. Data are expressed as mean + standard error of the mean.
(Abbreviations: MK-801, dizocilpine)
48

Mean ± Standard Error of the Mean (PPI Percentage)


Drugs and Prepulse
Avpr1b +/+ Avpr1b −/−
Levels
Saline 77 64.24 ± 6.57 67.29 ± 4.05

Saline 80 71.46 ± 4.33 72.62 ± 3.25

Saline 86 78.86 ± 2.49 78.43 ± 2.98

MK-801 77 34.72 ± 7.63 46.68 ± 6.31

MK-801 80 51.19 ± 7.54 58.20 ± 6.63

MK-801 86 60.58 ± 4.53 65.38 ± 6.63

Table 6: Mean and standard error of the mean for drug treatment in female Avpr1b +/+
and Avpr1b −/− mice. (Abbreviations: MK-801, dizocilpine)
49

genotypic difference between Avpr1b +/+ and Avpr1b −/− male mice in startle amplitude

(Table 7).

PPI Percentage

Baseline: As expected there was a main effect of the prepulse intensity (F2,32 = 17.968,

p<0.05), but there was no genotypic difference (Figure 17).

Drug Treatment: There was a main effect of the drug treatment (F1,17 = 20.883, p<0.05)

with MK-801 disrupting the PPI as compared to saline in both Avpr1b +/+ and Avpr1b

−/− male mice. There was also a main effect of the prepulse intensity (F2, 34 = 71.671,

p<0.05), but there was no main effect of the genotype or any other interactions (Figure

18) and (Table 8).

Experiment 3: Assessment of prepulse inhibition of startle reflex in male Avpr1b +/+

and Avpr1b −/− mice following administration of MK-801, PCP and saline

Habituation of startle

There was no habituation to startle reflex across the test trials PULSE 1 and PULSE 2 in

both Avpr1b +/+ and Avpr1b −/− males (Figure 19).

Startle amplitude

There was a main effect of the drug on startle amplitude (F2,28 = 10.803, p<0.05), with

MK-801 increasing the startle amplitude and PCP decreasing the startle amplitude.
50

Saline MK-801

300 300
* *
Pulse 1 Pulse 1
250 Pulse 2 250 Pulse 2

200 200
Startle Amplitude

Startle Amplitude
150 150

100 100

50 50

0 0
Avpr1b +/+ Avpr1b −/− Avpr1b +/+ Avpr1b −/−

Figure 16: Habituation of startle reflex in males. There was no main effect of saline
(0.9%) and MK-801 (0.7mg/kg) on habituation of startle across test trials PULSE 1 and
PULSE 2 on male Avpr1b +/+ and Avpr1b −/− mice. Data are expressed as mean +
standard error of the mean. (Abbreviations: MK-801, dizocilpine)
51

Mean ± Standard Error of the Mean (Startle Amplitude)

Drugs Avpr1b +/+ Avpr1b −/−

Saline 58.00 ± 4.02 73.01 ± 8.09

MK-801* 110.33 ± 21.32 109.30 ± 20.55

Table 7: Effects of drugs on startle amplitude on males. Treatment with MK-801 resulted
in an increase in the startle amplitude as compared to saline in both Avpr1b +/+ and
Avpr1b −/− male mice (p<0.05). There was no genotypic difference between the Avpr1b
+/+ and Avpr1b −/− male mice. Startle was calculated from the averages responses from
the first and last pulse alone (120dB) trials. (*p < 0.05 indicating a main effect of drug
compared to saline) (Abbreviations: MK-801, dizocilpine)
52

Avpr1b +/+
100 *
Avpr1b −/−
*

80
% Prepulse Inhibition

60

40

20

0
3dB 6dB 12dB

Figure 17: Baseline PPI percentage in males. There was a main effect of the prepulse
intensity (p<0.05); with increased PPI percentage with higher decibel prepulse tone.
However, there was no effect of the genotype on baseline PPI percentage in male Avpr1b
+/+ and Avpr1b −/− mice. (* indicates p<0.05). Data are expressed as mean + standard
error of the mean.
53

Avpr1b +/+

100 3dB
6dB
12dB

80
% Prepulse Inhibtion

60

40

20

0
SALINE MK801

Avpr1b −/−

100 3dB
6dB
12dB

80
% Prepulse Inhibtion

60

40

20

0
SALINE MK801

Figure 18: PPI percentage following drug treatment in males. There was a main effect of
the drug (p<0.05), with MK-801 disrupting the PPI in both Avpr1b +/+ and Avpr1b
−/− male mice. However, there was no genotypic difference in PPI percentage between
Avpr1b +/+ and Avpr1b −/− males. Data are expressed as mean + standard error of the
mean. (Abbreviations: MK-801, dizocilpine)
54

Mean ± Standard Error of the Mean (PPI Percentage)

Drugs Avpr1b +/+ Avpr1b −/−

Saline 77 72.11 ± 5.88 58.29 ± 7.90

Saline 80 77.75 ± 3.78 64.66 ± 8.37

Saline 86 86.59 ± 1.58 77.40 ± 4.11

MK-801 77 48.53 ± 4.47 30.29 ± 6.78

MK-801 80 63.12 ± 5.46 51.21 ± 6.07

MK-801 86 72.03 ± 4.19 56.27 ± 6.40

Table 8: Mean and standard error of the mean of PPI percentage following drug treatment
in Avpr1b +/+ and Avpr1b −/− males. (Abbreviations: MK-801, dizocilpine)
55

However there was no main effect of genotype or any other interactions on Avpr1b+/+

and Avpr1b −/− mice (Table 9).

PPI Percentage

Baseline: There was a main effect of prepulse intensity (F2,28 = 24.079, p<0.05), with

greater PPI percentage with higher decibel prepulse tone. There was no main effect of the

genotype on baseline PPI percentage (Figure 20).

Drug Treatment: There was a main effect of drug treatment (F2,28 = 4.659, p<0.05) with

both MK-801 and PCP disrupting the PPI in Avpr1b +/+ and Avpr1b −/− mice as

compared to saline. There was also a main effect of the prepulse intensity (F2,28 =

105.553, p<0.05). However there was no genotypic effect or any other interactions

(Figure 21) (Table 10).

Experiment 4A: Gene expression of NMDAR1 in Avpr1b +/+ and Avpr1b −/− male

mice

In this experiment, expression of NMDAR1 was examined in cortex, striatum and

hippocampus in Avpr1b +/+ and Avpr1b −/− male mice. There were no genotypic

differences in mRNA levels of NMDAR1 receptors in cortex (p=0.365), striatum

(p=0.083) or hippocampus (p=0.137) (Figure 22).

Experiment 4B: Gene expression of NMDAR2A in Avpr1b +/+ and Avpr1b −/− male

mice
56

Saline

300
*
Pulse 1
250 Pulse 2

200

Startle Amplitude 150

100

50

0
Avpr1b +/+ Avpr1b −/−

MK-801 PCP

300 300
* *
Pulse 1 Pulse 1
250 Pulse 2 250 Pulse 2

200 200
Startle Amplitude

Startle Amplitude

150 150

100 100

50 50

0 0
Avpr1b +/+ Avpr1b −/− Avpr1b +/+ Avpr1b −/−

Figure 19: Habituation of startle in males. There was no habituation of startle between
PULSE 1 and PULSE 2 in both Avpr1b +/+ and Avpr1b −/− males on treatment with
saline (0.9%), MK-801 (0.7mg/kg) and PCP (1.2mg/kg). Data are expressed as mean +
standard error of the mean. (Abbreviations: MK-801, dizocilpine; PCP, phencyclidine)
57

Mean ± Standard Error of the Mean (Startle amplitude)

Drugs Avpr1b +/+ Avpr1b −/−

Saline 90.83 ± 7.11 68.51 ± 8.31

MK-801* 103.83 ± 16.36 143.71 ± 17.83

PCP* 85.91 ± 13.20 96.14 ± 11.53

Table 9: Effects of drugs on startle amplitude on males. There was a main effect of the
drugs on startle amplitude (p<0.05) with MK-801 increasing the startle amplitude and
PCP decreasing the startle amplitude. There was no genotypic difference between the
Avpr1b +/+ and Avpr1b −/− mice. Startle was calculated from the averages responses
from the first and last pulse alone (120dB) trials. (*p < 0.05 indicating a main effect of
drug compared to saline) (Abbreviations: MK-801, dizocilpine; PCP, phencyclidine)
58

Avpr1b +/+
100
Avpr1b −/−

*
80 *
% Prepulse Inhibition

60

40

20

0
3dB 6dB 12dB

Figure 20: Baseline PPI percentage in males. There was no genotypic difference between
male Avpr1b +/+ and Avpr1b −/− mice in baseline PPI percentage. There was the
expected main effect of prepulse intensity with greater PPI percentage with higher
decibel prepulse tone (* indicates p<0.05). Data are expressed as mean and standard error
of the mean.
59

Avpr1b +/+

100 3dB
6dB
12dB

80
% Prepulse Inhibtion

60

40

20

0
SALINE MK-801 PCP

Avpr1b −/−

100 3dB
6dB
12dB

80
% Prepulse Inhibtion

60

40

20

0
SALINE MK-801 PCP

Figure 21: PPI percentage following drug treatment in males. Treatment with MK-80 and
PCP resulted in disruption of PPI as compared to saline in both Avpr1b +/+ and Avpr1b
−/− mice (p<0.05). However there was no genotypic difference in PPI percentage. Data
are expressed as mean + standard error of the mean. (Abbreviations: MK-801,
dizocilpine; PCP, phencyclidine)
60

Mean ± Standard Error of the Mean


Drugs and Prepulse
Avpr1b +/+ Avpr1b −/−
Levels
Saline 77 57.89 ± 6.61 51.75 ± 6.45

Saline 80 70.53 ± 4.36 63.39 ± 4.59

Saline 86 78.39 ± 2.50 73.78 ± 5.38

MK-801 77 51.97 ± 6.27 35.92 ± 9.39

MK-801 80 63.81 ± 6.03 47.16 ± 7.83

MK-801 86 69.41 ± 7.37 60.94 ± 7.48

PCP 77 41.95 ± 6.00 39.72 ± 8.23

PCP 80 50.63 ± 6.00 53.48 ± 6.96

PCP 86 67.80 ± 4.93 62.27 ± 7.62

Table 10: Mean and stand error of the mean for drug treatment on PPI percentage in male
Avpr1b +/+ and Avpr1b −/− mice. (Abbreviations: MK-801, dizocilpine; PCP,
phencyclidine)
61

In this experiment, expression of NMDAR2A was evaluated in cortex, striatum and

hippocampus of Avpr1b +/+ and Avpr1b −/− male mice. Avpr1b −/− mice had

significantly higher mRNA levels of NMDAR2A receptors as compared to Avpr1b +/+

mice in the hippocampus (p= 0.003). No differences in NMDAR2A levels were found in

cortex (p=0.897) and striatum (p=0.527) (Figure 23).


62

200 p=0.14

Avpr1b +/+
Avpr1b −/−

150
Relative Expression

p=0.08

100

50

0
Cortex Striatum Hippocampus

Figure 22: Expression of NMDAR1 in male Avpr1b +/+ and Avpr1b −/− mice. There
were no genotypic differences in mRNA levels of NMDAR1 in cortex, striatum and
hippocampus. Data are expressed as mean + standard error of the mean.
63

**
200

Avpr1b +/+
Avpr1b −/−
150
Relative Expression

100

50

0
Cortex Striatum Hippocampus

Figure 23: Expression of NMDAR2A in male Avpr1b +/+ and Avpr1b −/− mice. Avpr1b
−/− males had significantly higher mRNA levels of NMDAR2A expression in the
hippocampus as compared to Avpr1b +/+ mice (** indicates p<0.01). There were no
genotypic differences in NMDAR2A expression in cortex and striatum. Data are
expressed as mean + standard error the mean.
Discussion

The work described in this thesis has helped elucidate the role of the Avpr1b in

the neural circuitry that underlies sensorimotor gating. In Experiments 1A and 2A, we

examined the effect of various PPI-disrupting drugs on PPI and habituation in female

Avpr1b +/+ and Avpr1b −/− mice. It was found that treatment with APO, AMP, MK-801,

all resulted in disruption of PPI in both Avpr1b +/+ and Avpr1b −/− females. However,

no genotype or genotype × drug differences were observed between Avpr1b +/+ and

Avpr1b −/− females. Our results are consistent with many other studies where no

genotypic differences have been found in PPI in females despite its presence in males.

Further, given the lack of effect in females, we did not include them in Experiment 3.

In Experiment 1B, we found that treatment with MK-801, but not APO and AMP,

resulted in greater disruption of PPI in male Avpr1b −/− mice as compared to male

Avpr1b +/+ mice, indicating that male Avpr1b −/− mice are more susceptible to the PPI-

disrupting effects of MK-801. As MK-801 is a selective NMDA receptor antagonist

(Wong et al., 1986), these data suggest that male Avpr1b −/− mice have an impairment in

their glutamatergic system.

Experiment 2B was performed to verify the results of Experiment 1B, though,

only two drugs were used, MK-801 and saline. We found that in male Avpr1b +/+ and

Avpr1b −/−mice, there were no genotypic differences in PPI percentage following

64
65

treatment with MK-801. Although the differences in PPI percentage between the

genotypes were not statistically significant, they showed a similar trend as in Experiment

1B; with male Avpr1b −/− mice having an inclination for greater PPI disruption by MK-

801 as compared to male Avpr1b +/+ mice (p=0.19).

In Experiment 3, we wanted to repeat aspects of Experiment 1 and 2 and add

another NMDA antagonist, PCP, which has a slightly different pharmacology than MK-

801. PCP is the most commonly used NMDA receptor antagonist to study PPI (Geyer et

al., 2001 and the references within). However, PCP binds to receptors other than NMDA

receptors, such as sigma opioid receptors (Contreras et al., 1988; Largent et al., 1986;

Sonders et al., 1988),which are known to further facilitate glutamate transmission (Su and

Hayashi, 2003). It was found that PCP produced a stronger disruption of PPI than MK-

801 in both Avpr1b +/+ and Avpr1b −/− mice. However, contrary to our hypothesis, there

were no drug specific genotypic differences.

Based on the finding from Experiment 1B, we decided to explore possible

changes in the glutamatergic system in male Avpr1b −/− mice in Experiment 4, using

quantitative real time PCR to quantify mRNA levels of the NMDA receptors subunits

NMDAR1 and NMDAR2A. Interestingly, we found that the NMDAR2A subunit is

increased in the hippocampus of Avpr1b −/− mice as compared to Avpr1b +/+ mice.

The lack of differences in PPI between Avpr1b +/+ and Avpr1b −/− females in

Experiment 1A and 2A was not surprising since there are other studies which have found

a differential response of females to these drugs as compared to males. The lack of a


66

genotypic effect in females may be due to the variability in the hormonal milieu

throughout the estrous cycle. This idea is supported by one study in humans, which found

that there are sex differences in PPI, with women having lower PPI than males

(Swerdlow et al., 1993). Further, there is evidence that cyclic changes in gonadal steroids,

in particular estrogens, can affect PPI across the menstrual cycle (Swerdlow et al., 1997).

These data from humans are consistent with data in female rats, that show that PPI varies

across the estrous cycle (Aurebach et al., 1997, (Koch, 1998). In a number of other

studies, estrogens have been reported to influence monoamines in brain regions which are

important for regulation of PPI (Becker, 1990; Joyce and van, 1984; Thompson and

Moss, 1997; Van and Joyce, 1986). These hormonal effects of estrogens on PPI in

females make it difficult to interpret the results seen in our study. It would be interesting

to investigate PPI in ovarectomized female mice treated with estradiol.

The data in Experiment 2B and 3 did not support our findings from Experiment

1B, where a genotypic difference was found in Avpr1b males following treatment with

MK-801. The lack of genotypic difference in PPI percentage with MK-801 and PCP seen

in Experiment 2B and 3, could be due to a number of reasons. First, in Experiment 2B, by

using just two drugs instead of four as in Experiment 1B, we might have eliminated the

possible drug interactions which could have resulted in a significant difference. Second, a

careful analysis of the PPI data in Experiment 2B revealed that saline treatment resulted

in lower PPI in Avpr1b −/− mice as compared to Avpr1b +/+ mice. Since the control

itself resulted in differences between Avpr1b +/+ and Avpr1b −/− males, it is possible

that the disruption of PPI that was due to MK-801 did not appear as robust. This
67

genotypic difference in PPI following saline treatment between Avpr1b +/+ and Avpr1b

−/− mice has not been observed in any previous experiments and we are unclear as to

why this is happening. We are hopeful that a repeat of this study will shed some light

onto these aspects. In Experiment 3, we are fairly confident that our lack of effect in

Avpr1b −/− mice is due to a decrease in the potency and efficacy of drugs in disrupting

PPI. The drugs used in this Experiment were at least a year old and it is likely that there

was a decrease in their effects due to their breakdown over time. This hypothesis is

supported by the fact that the disruptions of PPI by MK-801 and PCP were not as

profound as that observed in previous Experiments. Another reason that there were not

drug-specific genotypic differences in PPI may be due to the smaller numbers of animals

that were tested. It is likely that a repeat of this study with a larger sample size and using

freshly prepared drugs would reproduce the results of Experiment 1B.

With regards to startle magnitude, in Experiment 1B, the genotypic difference in

PPI percentage does not seem to be due to changes in the startle magnitude. So far, the

relationship between startle and PPI is not well understood. One study compared the

prepulse and startle magnitude in 12 different inbreed mouse strains and found no

association between the two; while another study with 20 different strains found a

positive correlation between the two (Paylor and Crawley, 1997; Logue et al., 1997). Our

present findings suggest that the Avpr1b plays a role in regulation of PPI by interacting

with the glutamatergic system.


68

Data suggesting a link between glutamate and PPI has been established by a

number of studies. Across species, treatment with PCP and MK-801 has been shown to

disrupt PPI through antagonism of NMDA receptors (Caldwell et al., 2009; Jerram et al.,

1996; Yee et al., 2004). The lack of genotypic difference in PPI following treatment with

AMP and APO in Experiment 1B suggests that the dopaminergic system is not involved

in the regulation of PPI in Avpr1b −/− male mice. However, it is naïve to think that an

altered glutamatergic system is solely responsible for the deficient sensorimotor gating

observed in Avpr1b −/− mice following treatment with MK-801. In a number of studies,

it has been shown that there is a great degree of crosstalk between the various

neurotransmitters- glutamate, dopamine and serotonin (Carlsson et al., 2000; Carlsson et

al., 1999). It has been reported that drugs that block NMDA receptors not only increase

the release of glutamate, but also dopamine and serotonin (margos-Bosch et al., 2006;

Martin et al., 1998; Mathe et al., 1999; Moghaddam et al., 1997; Schmidt and Fadayel,

1996). Further in rats and mice, PPI deficits induced by PCP and MK-801, which are

specific NMDA antagonists, can be effectively reversed by pretreatment with atypical

antipsychotics such as clozapine and olanzapine, which act on many different receptors

(Bubenikova et al., 2005). Additionally, there are few reports which show that the release

of serotonin and dopamine, due to administration of MK-801 or PCP, can be effectively

suppressed by atypical neuroleptics (Lopez-Gil et al., 2007). Hence it is possible that

altered functioning of glutamate receptors in Avpr1b −/− mice may lead to deficits in PPI

through some indirect mechanism involving other neurotransmitters, such as by


69

increasing the levels of dopamine. Further studies will be required to explore the

relationship of different neurotransmitters and their effects on PPI.

Our findings from Experiment 4 support our hypothesis that the glutamatergic

system is altered in male Avpr1b −/− mice. The increases in the NMDA receptor subunit

NMDAR2A within the hippocampus are consistent with Avpr1b localization in the brain,

with the highest density of Avpr1b in the CA2 region of hippocampus (Young et al.,

2006). Thus, the developmental lack of Avpr1b could alter the PPI neurocircuitry by

changing the levels of expression of NMDA receptor subunits in this area. Although the

levels of NMDAR1 did not differ significantly between Avpr1b +/+ and Avpr1b −/−

mice, the level of NMDAR1 was decreased in the striatum in Avpr1b −/− mice as

compared to Avpr1b +/+ (P=0.08). We are currently quantifying these same subunits

(NMDAR1 and NMDAR2A) in Avpr1b +/+ and Avpr1b −/− mice by western blot

analysis to determine whether these observed changes in mRNA result in changes in

protein levels as well.

Currently, hypo-functioning of the NMDA receptor is believed to partly

contribute to the PPI deficits seen in patients with schizophrenia or mood disorders.

However, in individuals with psychiatric disorders characterized by PPI deficits, the data

regarding NMDA transcripts within the brain are highly inconsistent. In one study by

Gao and colleagues (2000), they reported an increase in NMDAR2A and decrease in

NMDAR1 transcripts in the hippocampus of schizophrenic patients as compared to

healthy controls, with no changes in protein levels. Conversely, other studies of NMDA
70

receptors in individuals diagnosed with either bipolar disorder or schizophrenia have

shown decreases in NMDAR1 and NMDAR2A transcripts in hippocampus

(McCullumsmith et al., 2007). As NMDA receptors are tetrameric and composed of two

obligatory NMDAR1 subunits and either two facultative NMDAR2 (A-D) or NMDAR3

(A-B) subunits (Moriyoshi et al., 1991; Sugihara et al., 1992; Paoletti and Neyton, 2007),

it is likely that absolute numbers may not be particularly telling. Rather, the ratios of

subunits within the receptors are more likely to be altered in these diseased states which

in turn would result in altered functioning. Therefore, further studies are required to

confirm these results and explore their functional significance.

While the role of glutamate in PPI is well known, the interaction of Avp and

glutamate is still under investigation. In a couple of studies, it has been found that MK-

801 abolishes Avp-induced increases in memory, suggesting that Avp does interact with

NMDA receptors in the process of memory formation (Artemowicz and Wisniewski,

1998; Wisniewski et al., 1996). A recent study also reported that Avp increases

glutamate release in the astrocytes of the hippocampus and cortex (Syed et al., 2007).

This effect on hippocampal astrocytes was shown to be specifically mediated via Avpr1b

receptors. Given all these interactions between Avp and glutamate, it would be interesting

to examine the levels of glutamate within the hippocampus of Avpr1b −/− mice by

microdialysis. It can be speculated that the similar to the mRNA levels of NMDA

receptors, the amount of glutamate would also be different between Avpr1b +/+ and

Avpr1b −/− mice.


71

There is another line of Avpr1b −/− mice that were engineered by another

research group. Contrary to the work from this thesis, in this line there are reported

baseline differences in PPI between Avpr1b +/+ and Avpr1b −/− mice (Egashira et al.,

2005). There are several reasons why these inconsistencies may exist. First, the mice used

in the above study were hybrids between 129Sv and C57BL/6J from F3 to F5

generations, whereas the mice used our study were fully backcrossed into the C57BL/6J

background. This is significant because there are reports which indicate that PPI differs

between the different inbred mouse strains (Logue et al., 1997). Thus, it is possible that

the differences in the background could have resulted in differences in the results seen

with PPI. A second difference between the two studies is the technique of creating the

Avpr1b −/− mice (Tanoue et al., 2004; Wersinger et al., 2002). It can be speculated that

the differences in generation of knockout could have differently altered other systems as

well which resulted in discrepancy in the results.

Nevertheless, the study with two different Avpr1b −/− mice as well as BB-Ho rats

suggests that Avp and Avpr1b do play a role in the regulation of sensorimotor gating.

Interestingly, PPI deficits in Avpr1b −/− mice and BB-Ho rats are effectively reversed by

atypical antipsychotics (Feifel et al., 2004; Egashira et al., 2005). These deficits might be

sensitive to some particular property of these atypical neuroleptics. It would be

interesting to see if we can reverse the deficits with atypical antipsychotics.

One of caveat of our study is that Avpr1b −/− mice completely lack the Avpr1b

since the time of conception. This developmental lack of Avpr1b could have resulted in
72

some compensatory changes in these animals. It is also possible that all the results seen

are due to some compensatory changes due to developmental lack of Avpr1b and not

primarily due to its absence. The same reason also makes it difficult to extrapolate the

results for humans in the pathogenesis of neuropsychiatric disorders. Hence, while our

studies strongly suggest the role of Avpr1b in regulating sensorimotor gating; its

translational implications for humans need to be explored further.

Overall conclusions

In summary, our studies have provided evidence for the role of Avpr1b in the

regulation of sensorimotor gating. However, lack of Avpr1b by itself is not sufficient to

cause deficits in sensorimotor gating. We have demonstrated that the glutamatergic

system is altered in Avpr1b −/− mice, which might be contributing to the PPI deficits

found in Experiment 1B. Thus, we hypothesize that under altered glutamatergic signaling

following treatment with psychotomimetics, the lack of the Avpr1b increases the

susceptibility to develop deficits in sensorimotor gating. However, since glutamate and

Avp are both known to interact with other neurotransmitters systems, it is likely these

may also be altered in these mice. Therefore, further research will be required to explore

how and where Avpr1b is interacting with these neurotransmitter systems to regulate PPI.

In order to further explore the relationship of glutamate and Avp, it would be

interesting to examine the changes in NMDA receptors after acute and chronic

administration of MK-801 and PCP in Avpr1b +/+ and Avpr1b −/− mice. We could also

examine other aspects of glutamatergic system such as measuring the glutamate levels in
73

specific brain regions at baseline and following administration of psychotics to look for

changes.

In conclusion, our study further strengthened the evidence for the role of central

Avp system in pathogenesis of neuropsychiatric disorders. With more evidence emerging

implicating Avp in regulation of social behavior and its alteration in many diseases states,

further research could open new avenues for development of new treatment options.
Reference List

Abduljawad,K.A., Langley,R.W., Bradshaw,C.M., and Szabadi,E. (1998). Effects of


bromocriptine and haloperidol on prepulse inhibition of the acoustic startle response in
man. J. Psychopharmacol. 12, 239-245.

Abduljawad,K.A., Langley,R.W., Bradshaw,C.M., and Szabadi,E. (1999). Effects of


bromocriptine and haloperidol on prepulse inhibition: comparison of the acoustic startle
eyeblink response and the N1/P2 auditory-evoked response in man. J. Psychopharmacol.
13, 3-9.

Acher,R., Chauvet,J., and Chauvet,M.T. (1995). Man and the chimaera. Selective versus
neutral oxytocin evolution. Adv. Exp. Med. Biol. 395, 615-627.

Altemus,M., Pigott,T., Kalogeras,K.T., Demitrack,M., Dubbert,B., Murphy,D.L., and


Gold,P.W. (1992). Abnormalities in the regulation of vasopressin and corticotropin
releasing factor secretion in obsessive-compulsive disorder. Arch. Gen. Psychiatry 49, 9-
20.

Altemus,M., Swedo,S.E., Leonard,H.L., Richter,D., Rubinow,D.R., Potter,W.Z., and


Rapoport,J.L. (1994). Changes in cerebrospinal fluid neurochemistry during treatment of
obsessive-compulsive disorder with clomipramine. Arch. Gen. Psychiatry 51, 794-803.

Artemowicz,B. and Wisniewski,K. (1998). Role of NMDA receptor in the effects of


arginine-vasopressin on memory processes. Pol. J. Pharmacol. 50, 5-14.

Bankir,L. (2001). Antidiuretic action of vasopressin: quantitative aspects and interaction


between V1a and V2 receptor-mediated effects. Cardiovasc. Res. 51, 372-390.

Becker,J.B. (1990). Estrogen rapidly potentiates amphetamine-induced striatal dopamine


release and rotational behavior during microdialysis. Neurosci. Lett. 118, 169-171.

Beckwith,B.E., Couk,D.I., and Till,T.S. (1983). Vasopressin analog influences the


performance of males on a reaction time task. Peptides 4, 707-709.

Bielsky,I.F., Hu,S.B., Szegda,K.L., Westphal,H., and Young,L.J. (2004). Profound


impairment in social recognition and reduction in anxiety-like behavior in vasopressin
V1a receptor knockout mice. Neuropsychopharmacology 29, 483-493.

74
75

Blanchard,R.J., Griebel,G., Farrokhi,C., Markham,C., Yang,M., and Blanchard,D.C.


(2005). AVP V1b selective antagonist SSR149415 blocks aggressive behaviors in
hamsters. Pharmacol. Biochem. Behav. 80, 189-194.

Bohus,B. and de,W.D. (1998). The vasopressin deficient Brattleboro rats: a natural
knockout model used in the search for CNS effects of vasopressin. Prog. Brain Res. 119,
555-573.

Bolino,F., Di,M., V, Di,C.L., Manna,V., Daneluzzo,E., and Casacchia,M. (1994).


Sensorimotor gating and habituation evoked by electro-cutaneous stimulation in
schizophrenia. Biol. Psychiatry 36, 670-679.

Born,J., Pietrowsky,R., and Fehm,H.L. (1998). Neuropsychological effects of vasopressin


in healthy humans. Prog. Brain Res. 119, 619-643.

Braff,D., Stone,C., Callaway,E., Geyer,M., Glick,I., and Bali,L. (1978). Prestimulus


effects on human startle reflex in normals and schizophrenics. Psychophysiology 15, 339-
343.

Braff,D.L., Grillon,C., and Geyer,M.A. (1992). Gating and habituation of the startle
reflex in schizophrenic patients. Arch. Gen. Psychiatry 49, 206-215.

Braff,D.L., Swerdlow,N.R., and Geyer,M.A. (1999). Symptom correlates of prepulse


inhibition deficits in male schizophrenic patients. Am. J. Psychiatry 156, 596-602.

Brambilla,F., Aguglia,E., Massironi,R., Maggioni,M., Grillo,W., Castiglioni,R.,


Catalano,M., and Drago,F. (1986). Neuropeptide therapies in chronic schizophrenia:
TRH and vasopressin administration. Neuropsychobiology 15, 114-121.

Brambilla,F., Bondiolotti,G.P., Maggioni,M., Sciascia,A., Grillo,W., Sanna,F., Latina,A.,


and Picotti,G.B. (1989). Vasopressin (DDAVP) therapy in chronic schizophrenia: effects
on negative symptoms and memory. Neuropsychobiology 20, 113-119.

Brown,J.S., Kalish,H.I., and Farber,I.E. (1951). Conditioned fear as revealed by


magnitude of startle response to an auditory stimulus. J. Exp. Psychol. 41, 317-328.

Bubenikova,V., Votava,M., Horacek,J., Palenicek,T., and Dockery,C. (2005). The effect


of zotepine, risperidone, clozapine and olanzapine on MK-801-disrupted sensorimotor
gating. Pharmacol. Biochem. Behav. 80, 591-596.

Buijs,R.M. (1978). Intra- and extrahypothalamic vasopressin and oxytocin pathways in


the rat. Pathways to the limbic system, medulla oblongata and spinal cord. Cell Tissue
Res. 192, 423-435.
76

Caffe,A.R., Van Ryen,P.C., Van der Woude,T.P., and Van Leeuwen,F.W. (1989).
Vasopressin and oxytocin systems in the brain and upper spinal cord of Macaca
fascicularis. J. Comp Neurol. 287, 302-325.

Caldwell,H.K. and Albers,H.E. (2004). Effect of photoperiod on vasopressin-induced


aggression in Syrian hamsters. Horm. Behav. 46, 444-449.

Caldwell,H.K., Lee,H.J., Macbeth,A.H., and Young,W.S., III (2008). Vasopressin:


behavioral roles of an "original" neuropeptide. Prog. Neurobiol. 84, 1-24.

Caldwell,H.K., Stephens,S.L., and Young,W.S., III (2009). Oxytocin as a natural


antipsychotic: a study using oxytocin knockout mice. Mol. Psychiatry 14, 190-196.

Carlsson,A., Hansson,L.O., Waters,N., and Carlsson,M.L. (1997). Neurotransmitter


aberrations in schizophrenia: new perspectives and therapeutic implications. Life Sci. 61,
75-94.

Carlsson,A., Waters,N., and Carlsson,M.L. (1999). Neurotransmitter interactions in


schizophrenia-therapeutic implications. Eur. Arch. Psychiatry Clin. Neurosci. 249 Suppl
4, 37-43.

Carlsson,A., Waters,N., Waters,S., and Carlsson,M.L. (2000). Network interactions in


schizophrenia - therapeutic implications. Brain Res. Brain Res. Rev. 31, 342-349.

Castellanos,F.X., Fine,E.J., Kaysen,D., Marsh,W.L., Rapoport,J.L., and Hallett,M.


(1996). Sensorimotor gating in boys with Tourette's syndrome and ADHD: preliminary
results. Biol. Psychiatry 39, 33-41.

Chakrabarty,K., Bhattacharyya,S., Christopher,R., and Khanna,S. (2005). Glutamatergic


dysfunction in OCD. Neuropsychopharmacology 30, 1735-1740.

Cho,M.M., DeVries,A.C., Williams,J.R., and Carter,C.S. (1999). The effects of oxytocin


and vasopressin on partner preferences in male and female prairie voles (Microtus
ochrogaster). Behav. Neurosci. 113, 1071-1079.

Contreras,P.C., Contreras,M.L., O'Donohue,T.L., and Lair,C.C. (1988). Biochemical and


behavioral effects of sigma and PCP ligands. Synapse 2, 240-243.

Curzon,P. and Decker,M.W. (1998). Effects of phencyclidine (PCP) and (+)MK-801 on


sensorimotor gating in CD-1 mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 22,
129-146.

Davis,M., Gendelman,D.S., Tischler,M.D., and Gendelman,P.M. (1982). A primary


acoustic startle circuit: lesion and stimulation studies. J. Neurosci. 2, 791-805.
77

de Kloet,C.S., Vermetten,E., Geuze,E., Wiegant,V.M., and Westenberg,H.G. (2008).


Elevated plasma arginine vasopressin levels in veterans with posttraumatic stress
disorder. J. Psychiatr. Res. 42, 192-198.

De Vries,G.J., Best,W., and Sluiter,A.A. (1983). The influence of androgens on the


development of a sex difference in the vasopressinergic innervation of the rat lateral
septum. Brain Res. 284, 377-380.

De Vries,G.J. and Buijs,R.M. (1983). The origin of the vasopressinergic and


oxytocinergic innervation of the rat brain with special reference to the lateral septum.
Brain Res. 273, 307-317.

de Vries,G.J., Buijs,R.M., and Sluiter,A.A. (1984). Gonadal hormone actions on the


morphology of the vasopressinergic innervation of the adult rat brain. Brain Res. 298,
141-145.

De,B., Gold,P.W., Geracioti,T.D., Jr., Listwak,S.J., and Kling,M.A. (1993). Association


of fluoxetine treatment with reductions in CSF concentrations of corticotropin-releasing
hormone and arginine vasopressin in patients with major depression. Am. J. Psychiatry
150, 656-657.

Dempster,E.L., Burcescu,I., Wigg,K., Kiss,E., Baji,I., Gadoros,J., Tamas,Z.,


Kennedy,J.L., Vetro,A., Kovacs,M., and Barr,C.L. (2007). Evidence of an association
between the vasopressin V1b receptor gene (AVPR1B) and childhood-onset mood
disorders. Arch. Gen. Psychiatry 64, 1189-1195.

Dhakar M.B., Stevenson E.L., and Caldwell H.K. (in press). The interplay between
oxytocin, vasopressin and gonadal steroids. In Oxytocin, Vasopressin and Related
Peptides in the Regulation of Behavior, Elena Choleris, Donald Pfaff, and Martin
Cavaliers, eds. Cambridge University Press).

Dogterom,J., van Wimersma Greidanus,T.B., and De,W.D. (1978). Vasopressin in


cerebrospinal fluid and plasma of man, dog, and rat. Am. J. Physiol 234, E463-E467.

Donaldson,Z.R. and Young,L.J. (2008). Oxytocin, vasopressin, and the neurogenetics of


sociality. Science 322, 900-904.

Dubois-Dauphin,M., Tribollet,E., and Dreifuss,J.J. (1987). A sexually dimorphic


vasopressin innervation of auditory pathways in the guinea pig brain. Brain Res. 437,
151-156.

Dubois-Dauphin,M., Tribollet,E., and Dreifuss,J.J. (1989). Distribution of


neurohypophysial peptides in the guinea pig brain. I. An immunocytochemical study of
the vasopressin-related glycopeptide. Brain Res. 496, 45-65.
78

Dulawa,S.C. and Geyer,M.A. (1996). Psychopharmacology of prepulse inhibition in


mice. Chin J. Physiol 39, 139-146.

Egashira,N., Tanoue,A., Higashihara,F., Fuchigami,H., Sano,K., Mishima,K., Fukue,Y.,


Nagai,H., Takano,Y., Tsujimoto,G., Stemmelin,J., Griebel,G., Iwasaki,K., Ikeda,T.,
Nishimura,R., and Fujiwara,M. (2005). Disruption of the prepulse inhibition of the startle
reflex in vasopressin V1b receptor knockout mice: reversal by antipsychotic drugs.
Neuropsychopharmacology 30, 1996-2005.

Elman,I., Lukas,S., Shoaf,S.E., Rott,D., Adler,C., and Breier,A. (2003). Effects of acute
metabolic stress on the peripheral vasopressinergic system in schizophrenia. J.
Psychopharmacol. 17, 317-323.

Engelmann,M. and Landgraf,R. (1994). Microdialysis administration of vasopressin into


the septum improves social recognition in Brattleboro rats. Physiol Behav. 55, 145-149.

Feenstra,M.G., Snijdewint,F.G., Van,G.H., and Boer,G.J. (1990). Widespread alterations


in central noradrenaline, dopamine, and serotonin systems in the Brattleboro rat not
related to the local absence of vasopressin. Neurochem. Res. 15, 283-288.

Feifel,D., Melendez,G., Priebe,K., and Shilling,P.D. (2007). The effects of chronic


administration of established and putative antipsychotics on natural prepulse inhibition
deficits in Brattleboro rats. Behav. Brain Res. 181, 278-286.

Feifel,D., Melendez,G., and Shilling,P.D. (2004). Reversal of sensorimotor gating


deficits in Brattleboro rats by acute administration of clozapine and a neurotensin agonist,
but not haloperidol: a potential predictive model for novel antipsychotic effects.
Neuropsychopharmacology 29, 731-738.

Feifel,D. and Priebe,K. (2001). Vasopressin-deficient rats exhibit sensorimotor gating


deficits that are reversed by subchronic haloperidol. Biol. Psychiatry 50, 425-433.

Ferris,C.F., Melloni,R.H., Jr., Koppel,G., Perry,K.W., Fuller,R.W., and Delville,Y.


(1997). Vasopressin/serotonin interactions in the anterior hypothalamus control
aggressive behavior in golden hamsters. J. Neurosci. 17, 4331-4340.

Fliers,E., Guldenaar,S.E., van de,W.N., and Swaab,D.F. (1986). Extrahypothalamic


vasopressin and oxytocin in the human brain; presence of vasopressin cells in the bed
nucleus of the stria terminalis. Brain Res. 375, 363-367.

Furuya,Y., Kagaya,T., Ogura,H., and Nishizawa,Y. (1999). Competitive NMDA receptor


antagonists disrupt prepulse inhibition without reduction of startle amplitude in a
dopamine receptor-independent manner in mice. Eur. J. Pharmacol. 364, 133-140.
79

Gao,X.M., Sakai,K., Roberts,R.C., Conley,R.R., Dean,B., and Tamminga,C.A. (2000).


Ionotropic glutamate receptors and expression of N-methyl-D-aspartate receptor subunits
in subregions of human hippocampus: effects of schizophrenia. Am. J. Psychiatry 157,
1141-1149.

Geyer,M.A. and Braff,D.L. (1982). Habituation of the Blink reflex in normals and
schizophrenic patients. Psychophysiology 19, 1-6.

Geyer,M.A., Krebs-Thomson,K., Braff,D.L., and Swerdlow,N.R. (2001).


Pharmacological studies of prepulse inhibition models of sensorimotor gating deficits in
schizophrenia: a decade in review. Psychopharmacology (Berl) 156, 117-154.

Graham,F.K. (1975). Presidential Address, 1974. The more or less startling effects of
weak prestimulation. Psychophysiology 12, 238-248.

Griebel,G., Simiand,J., Serradeil-Le,G.C., Wagnon,J., Pascal,M., Scatton,B.,


Maffrand,J.P., and Soubrie,P. (2002). Anxiolytic- and antidepressant-like effects of the
non-peptide vasopressin V1b receptor antagonist, SSR149415, suggest an innovative
approach for the treatment of stress-related disorders. Proc. Natl. Acad. Sci. U. S. A 99,
6370-6375.

Griebel,G., Simiand,J., Stemmelin,J., Gal,C.S., and Steinberg,R. (2003). The vasopressin


V1b receptor as a therapeutic target in stress-related disorders. Curr. Drug Targets. CNS.
Neurol. Disord. 2, 191-200.

Grillon,C., Ameli,R., Charney,D.S., Krystal,J., and Braff,D. (1992). Startle gating deficits
occur across prepulse intensities in schizophrenic patients. Biol. Psychiatry 32, 939-943.

Grillon,C., Falls,W.A., Ameli,R., and Davis,M. (1994). Safety signals and human
anxiety: a fear-potentiated startle study. Anxiety. 1, 13-21.

Hoffman,H.S. and Searle,J.L. (1968). Acoustic and temporal factors in the evocation of
startle. J. Acoust. Soc. Am. 43, 269-282.

Hutchison,K.E. and Swift,R. (1999). Effect of d-amphetamine on prepulse inhibition of


the startle reflex in humans. Psychopharmacology (Berl) 143, 394-400.

Iager,A.C., Kirch,D.G., Bigelow,L.B., and Karson,C.N. (1986). Treatment of


schizophrenia with a vasopressin analogue. Am. J. Psychiatry 143, 375-377.

Inder,W.J., Donald,R.A., Prickett,T.C., Frampton,C.M., Sullivan,P.F., Mulder,R.T., and


Joyce,P.R. (1997). Arginine vasopressin is associated with hypercortisolemia and suicide
attempts in depression. Biol. Psychiatry 42, 744-747.
80

Insel,T.R. and Young,L.J. (2000). Neuropeptides and the evolution of social behavior.
Curr. Opin. Neurobiol. 10, 784-789.

Ison,J.R., McAdam,D.W., and Hammond,G.R. (1973). Latency and amplitude changes in


the acoustic startle reflex of the rat produced by variation in auditory prestimulation.
Physiol Behav. 10, 1035-1039.

Jard,S., Barberis,C., Audigier,S., and Tribollet,E. (1987). Neurohypophyseal hormone


receptor systems in brain and periphery. Prog. Brain Res. 72, 173-187.

Javitt,D.C. (1987). Negative schizophrenic symptomatology and the PCP (phencyclidine)


model of schizophrenia. Hillside. J. Clin. Psychiatry 9, 12-35.

Jerram,A.H., Smith,P.F., and Darlington,C.L. (1996). A dose-response analysis of the


behavioral effects of (+)MK-801 in guinea pig: comparison with CPP. Pharmacol.
Biochem. Behav. 53, 799-807.

Johnson,A.E., Audigier,S., Rossi,F., Jard,S., Tribollet,E., and Barberis,C. (1993).


Localization and characterization of vasopressin binding sites in the rat brain using an
iodinated linear AVP antagonist. Brain Res. 622, 9-16.

Joyce,J.N. and van,H.C. (1984). Behaviors induced by intrastriatal dopamine vary


independently across the estrous cycle. Pharmacol. Biochem. Behav. 20, 551-557.

Koch,M. (1998). Sensorimotor gating changes across the estrous cycle in female rats.
Physiol Behav. 64, 625-628.

Koch,M., Kungel,M., and Herbert,H. (1993). Cholinergic neurons in the


pedunculopontine tegmental nucleus are involved in the mediation of prepulse inhibition
of the acoustic startle response in the rat. Exp. Brain Res. 97, 71-82.

Koch,M. and Schnitzler,H.U. (1997). The acoustic startle response in rats--circuits


mediating evocation, inhibition and potentiation. Behav. Brain Res. 89, 35-49.

Korsgaard,S., Casey,D.E., mgaard Pedersen,N.E., Jorgensen,A., and Gerlach,J. (1981).


Vasopressin in anergic schizophrenia. A cross-over study with lysine-8-vasopressin and
placebo. Psychopharmacology (Berl) 74, 379-382.

Kumari,V., Mulligan,O.F., Cotter,P.A., Poon,L., Toone,B.K., Checkley,S.A., and


Gray,J.A. (1998). Effects of single oral administrations of haloperidol and d-
amphetamine on prepulse inhibition of the acoustic startle reflex in healthy male
volunteers. Behav. Pharmacol. 9, 567-576.
81

Kumari,V., Soni,W., Mathew,V.M., and Sharma,T. (2000). Prepulse inhibition of the


startle response in men with schizophrenia: effects of age of onset of illness, symptoms,
and medication. Arch. Gen. Psychiatry 57, 609-614.

Kumari,V., Soni,W., and Sharma,T. (1999). Normalization of information processing


deficits in schizophrenia with clozapine. Am. J. Psychiatry 156, 1046-1051.

Largent,B.L., Gundlach,A.L., and Snyder,S.H. (1986). Pharmacological and


autoradiographic discrimination of sigma and phencyclidine receptor binding sites in
brain with (+)-[3H]SKF 10,047, (+)-[3H]-3-[3-hydroxyphenyl]-N-(1-propyl)piperidine
and [3H]-1-[1-(2-thienyl)cyclohexyl]piperidine. J. Pharmacol. Exp. Ther. 238, 739-748.

Laycock,J.F., Gartside,I.B., and Chapman,J.T. (1983). A comparison of the learning


abilities of Brattleboro rats with hereditary diabetes insipidus and Long-Evans rats using
positively reinforced operant conditioning. Prog. Brain Res. 60, 183-187.

Lee,H.J., Macbeth,A.H., Pagani,J.H., and Young,W.S., III (2009). Oxytocin: the great
facilitator of life. Prog. Neurobiol. 88, 127-151.

Linn,G.S., O'Keeffe,R.T., Schroeder,C.E., Lifshitz,K., and Javitt,D.C. (1999). Behavioral


effects of chronic phencyclidine in monkeys. Neuroreport 10, 2789-2793.

Logue,S.F., Owen,E.H., Rasmussen,D.L., and Wehner,J.M. (1997). Assessment of


locomotor activity, acoustic and tactile startle, and prepulse inhibition of startle in inbred
mouse strains and F1 hybrids: implications of genetic background for single gene and
quantitative trait loci analyses. Neuroscience 80, 1075-1086.

Lolait,S.J., O'Carroll,A.M., Mahan,L.C., Felder,C.C., Button,D.C., Young,W.S., III,


Mezey,E., and Brownstein,M.J. (1995). Extrapituitary expression of the rat V1b
vasopressin receptor gene. Proc. Natl. Acad. Sci. U. S. A 92, 6783-6787.

Lolait,S.J., O'Carroll,A.M., McBride,O.W., Konig,M., Morel,A., and Brownstein,M.J.


(1992). Cloning and characterization of a vasopressin V2 receptor and possible link to
nephrogenic diabetes insipidus. Nature 357, 336-339.

Lolait,S.J., Stewart,L.Q., Roper,J.A., Harrison,G., Jessop,D.S., Young,W.S., III, and


O'Carroll,A.M. (2007). Attenuated stress response to acute lipopolysaccharide challenge
and ethanol administration in vasopressin V1b receptor knockout mice. J.
Neuroendocrinol. 19, 543-551.

Lopez-Gil,X., Babot,Z., margos-Bosch,M., Sunol,C., Artigas,F., and Adell,A. (2007).


Clozapine and haloperidol differently suppress the MK-801-increased glutamatergic and
serotonergic transmission in the medial prefrontal cortex of the rat.
Neuropsychopharmacology 32, 2087-2097.
82

Mansbach,R.S. and Geyer,M.A. (1989). Effects of phencyclidine and phencyclidine


biologs on sensorimotor gating in the rat. Neuropsychopharmacology 2, 299-308.

Mansbach,R.S., Geyer,M.A., and Braff,D.L. (1988). Dopaminergic stimulation disrupts


sensorimotor gating in the rat. Psychopharmacology (Berl) 94, 507-514.

margos-Bosch,M., Lopez-Gil,X., Artigas,F., and Adell,A. (2006). Clozapine and


olanzapine, but not haloperidol, suppress serotonin efflux in the medial prefrontal cortex
elicited by phencyclidine and ketamine. Int. J. Neuropsychopharmacol. 9, 565-573.

Martin,P., Carlsson,M.L., and Hjorth,S. (1998). Systemic PCP treatment elevates brain
extracellular 5-HT: a microdialysis study in awake rats. Neuroreport 9, 2985-2988.

Mathe,J.M., Nomikos,G.G., Blakeman,K.H., and Svensson,T.H. (1999). Differential


actions of dizocilpine (MK-801) on the mesolimbic and mesocortical dopamine systems:
role of neuronal activity. Neuropharmacology 38, 121-128.

McCullumsmith,R.E., Kristiansen,L.V., Beneyto,M., Scarr,E., Dean,B., and Meador-


Woodruff,J.H. (2007). Decreased NR1, NR2A, and SAP102 transcript expression in the
hippocampus in bipolar disorder. Brain Res. 1127, 108-118.

Mcghie,A. and Chapman,J. (1961). Disorders of attention and perception in early


schizophrenia. Br. J. Med. Psychol. 34, 103-116.

Meynen,G., Unmehopa,U.A., van Heerikhuize,J.J., Hofman,M.A., Swaab,D.F., and


Hoogendijk,W.J. (2006). Increased arginine vasopressin mRNA expression in the human
hypothalamus in depression: A preliminary report. Biol. Psychiatry 60, 892-895.

Michell,R.H., Kirk,C.J., and Billah,M.M. (1979). Hormonal stimulation of


phosphatidylinositol breakdown with particular reference to the hepatic effects of
vasopressin. Biochem. Soc. Trans. 7, 861-865.

Millan,M.J., Millan,M.H., Czlonkowski,A., and Herz,A. (1984). Vasopressin and


oxytocin in the rat spinal cord: distribution and origins in comparison to [Met]enkephalin,
dynorphin and related opioids and their irresponsiveness to stimuli modulating
neurohypophyseal secretion. Neuroscience 13, 179-187.

Moghaddam,B., Adams,B., Verma,A., and Daly,D. (1997). Activation of glutamatergic


neurotransmission by ketamine: a novel step in the pathway from NMDA receptor
blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex.
J. Neurosci. 17, 2921-2927.

Moriyoshi,K., Masu,M., Ishii,T., Shigemoto,R., Mizuno,N., and Nakanishi,S. (1991).


Molecular cloning and characterization of the rat NMDA receptor. Nature 354, 31-37.
83

Nishimura,H. and Fan,Z. (2003). Regulation of water movement across vertebrate renal
tubules. Comp Biochem. Physiol A Mol. Integr. Physiol 136, 479-498.

Olney,J.W., Newcomer,J.W., and Farber,N.B. (1999). NMDA receptor hypofunction


model of schizophrenia. J. Psychiatr. Res. 33, 523-533.

Ornitz,E.M., Hanna,G.L., and de,T.J. (1992). Prestimulation-induced startle modulation


in attention-deficit hyperactivity disorder and nocturnal enuresis. Psychophysiology 29,
437-451.

Paoletti,P. and Neyton,J. (2007). NMDA receptor subunits: function and pharmacology.
Curr. Opin. Pharmacol. 7, 39-47.

Paylor,R. and Crawley,J.N. (1997). Inbred strain differences in prepulse inhibition of the
mouse startle response. Psychopharmacology (Berl) 132, 169-180.

Ralph,R.J., Paulus,M.P., and Geyer,M.A. (2001). Strain-specific effects of amphetamine


on prepulse inhibition and patterns of locomotor behavior in mice. J. Pharmacol. Exp.
Ther. 298, 148-155.

Ralph,R.J., Varty,G.B., Kelly,M.A., Wang,Y.M., Caron,M.G., Rubinstein,M.,


Grandy,D.K., Low,M.J., and Geyer,M.A. (1999). The dopamine D2, but not D3 or D4,
receptor subtype is essential for the disruption of prepulse inhibition produced by
amphetamine in mice. J. Neurosci. 19, 4627-4633.

Raskind,M.A., Courtney,N., Murburg,M.M., Backus,F.I., Bokan,J.A., Ries,R.K.,


Dorsa,D.M., and Weitzman,R.E. (1987). Antipsychotic drugs and plasma vasopressin in
normals and acute schizophrenic patients. Biol. Psychiatry 22, 453-462.

Ross,H.E. and Young,L.J. (2009). Oxytocin and the neural mechanisms regulating social
cognition and affiliative behavior. Front Neuroendocrinol. 30, 534-547.

Saito,M., Sugimoto,T., Tahara,A., and Kawashima,H. (1995). Molecular cloning and


characterization of rat V1b vasopressin receptor: evidence for its expression in extra-
pituitary tissues. Biochem. Biophys. Res. Commun. 212, 751-757.

Schall,U., Schon,A., Zerbin,D., Eggers,C., and Oades,R.D. (1996). Event-related


potentials during an auditory discrimination with prepulse inhibition in patients with
schizophrenia, obsessive-compulsive disorder and healthy subjects. Int. J. Neurosci. 84,
15-33.

Schmidt,C.J. and Fadayel,G.M. (1996). Regional effects of MK-801 on dopamine


release: effects of competitive NMDA or 5-HT2A receptor blockade. J. Pharmacol. Exp.
Ther. 277, 1541-1549.
84

Skuse,D.H. and Gallagher,L. (2009). Dopaminergic-neuropeptide interactions in the


social brain. Trends Cogn Sci. 13, 27-35.

Sofroniew,M.V. (1983). Morphology of vasopressin and oxytocin neurones and their


central and vascular projections. Prog. Brain Res. 60, 101-114.

Sokolowski,M.B. (2010). Social interactions in "simple" model systems. Neuron 65, 780-
794.

Sonders,M.S., Keana,J.F., and Weber,E. (1988). Phencyclidine and psychotomimetic


sigma opiates: recent insights into their biochemical and physiological sites of action.
Trends Neurosci. 11, 37-40.

Stemmelin,J., Lukovic,L., Salome,N., and Griebel,G. (2005). Evidence that the lateral
septum is involved in the antidepressant-like effects of the vasopressin V1b receptor
antagonist, SSR149415. Neuropsychopharmacology 30, 35-42.

Su,T.P. and Hayashi,T. (2003). Understanding the molecular mechanism of sigma-1


receptors: towards a hypothesis that sigma-1 receptors are intracellular amplifiers for
signal transduction. Curr. Med. Chem. 10, 2073-2080.

Sugihara,H., Moriyoshi,K., Ishii,T., Masu,M., and Nakanishi,S. (1992). Structures and


properties of seven isoforms of the NMDA receptor generated by alternative splicing.
Biochem. Biophys. Res. Commun. 185, 826-832.

Swerdlow,N.R., Bakshi,V., and Geyer,M.A. (1996). Seroquel restores sensorimotor


gating in phencyclidine-treated rats. J. Pharmacol. Exp. Ther. 279, 1290-1299.

Swerdlow,N.R., Bakshi,V., Waikar,M., Taaid,N., and Geyer,M.A. (1998). Seroquel,


clozapine and chlorpromazine restore sensorimotor gating in ketamine-treated rats.
Psychopharmacology (Berl) 140, 75-80.

Swerdlow,N.R., Benbow,C.H., Zisook,S., Geyer,M.A., and Braff,D.L. (1993). A


preliminary assessment of sensorimotor gating in patients with obsessive compulsive
disorder. Biol. Psychiatry 33, 298-301.

Swerdlow,N.R., Braff,D.L., and Geyer,M.A. (1999). Cross-species studies of


sensorimotor gating of the startle reflex. Ann. N. Y. Acad. Sci. 877, 202-216.

Swerdlow,N.R. and Geyer,M.A. (1998). Using an animal model of deficient sensorimotor


gating to study the pathophysiology and new treatments of schizophrenia. Schizophr.
Bull. 24, 285-301.
85

Swerdlow,N.R., Geyer,M.A., and Braff,D.L. (2001). Neural circuit regulation of prepulse


inhibition of startle in the rat: current knowledge and future challenges.
Psychopharmacology (Berl) 156, 194-215.

Swerdlow,N.R., Hartman,P.L., and Auerbach,P.P. (1997). Changes in sensorimotor


inhibition across the menstrual cycle: implications for neuropsychiatric disorders. Biol.
Psychiatry 41, 452-460.

Swerdlow,N.R., Martinez,Z.A., Hanlon,F.M., Platten,A., Farid,M., Auerbach,P.,


Braff,D.L., and Geyer,M.A. (2000). Toward understanding the biology of a complex
phenotype: rat strain and substrain differences in the sensorimotor gating-disruptive
effects of dopamine agonists. J. Neurosci. 20, 4325-4336.

Swerdlow,N.R., Paulsen,J., Braff,D.L., Butters,N., Geyer,M.A., and Swenson,M.R.


(1995). Impaired prepulse inhibition of acoustic and tactile startle response in patients
with Huntington's disease. J. Neurol. Neurosurg. Psychiatry 58, 192-200.

Syed,N., Martens,C.A., and Hsu,W.H. (2007). Arginine vasopressin increases glutamate


release and intracellular Ca2+ concentration in hippocampal and cortical astrocytes
through two distinct receptors. J. Neurochem. 103, 229-237.

Tanoue,A., Ito,S., Honda,K., Oshikawa,S., Kitagawa,Y., Koshimizu,T.A., Mori,T., and


Tsujimoto,G. (2004). The vasopressin V1b receptor critically regulates hypothalamic-
pituitary-adrenal axis activity under both stress and resting conditions. J. Clin. Invest 113,
302-309.

Thompson,T.L. and Moss,R.L. (1997). Modulation of mesolimbic dopaminergic activity


over the rat estrous cycle. Neurosci. Lett. 229, 145-148.

Tribollet,E., Barberis,C., and Arsenijevic,Y. (1997). Distribution of vasopressin and


oxytocin receptors in the rat spinal cord: sex-related differences and effect of castration in
pudendal motor nuclei. Neuroscience 78, 499-509.

Tribollet,E., Barberis,C., Dreifuss,J.J., and Jard,S. (1988). Autoradiographic localization


of vasopressin and oxytocin binding sites in rat kidney. Kidney Int. 33, 959-965.

Vaccari,C., Lolait,S.J., and Ostrowski,N.L. (1998). Comparative distribution of


vasopressin V1b and oxytocin receptor messenger ribonucleic acids in brain.
Endocrinology 139, 5015-5033.

Van,H.C. and Joyce,J.N. (1986). Effects of estrogen on the basal ganglia. Neurosci.
Biobehav. Rev. 10, 1-14.
86

van,L.L., Goekoop,J.G., van Kempen,G.M., Frankhuijzen-Sierevogel,A.C.,


Wiegant,V.M., van,d., V, and De,W.D. (1997). Plasma levels of arginine vasopressin
elevated in patients with major depression. Neuropsychopharmacology 17, 284-292.

van,W.D., Del-Favero,J., Aulchenko,Y., Oswald,P., Souery,D., Forsgren,T., Sluijs,S.,


Bel-Kacem,S., Adolfsson,R., Mendlewicz,J., Van,D.C., Deboutte,D., Van,B.C., and
Claes,S. (2004). A major SNP haplotype of the arginine vasopressin 1B receptor protects
against recurrent major depression. Mol. Psychiatry 9, 287-292.

Venables,P.H. (1960). The effect of auditory and visual stimulation on the skin potential
response of schizophrenics. Brain 83, 77-92.

Wang,Z., Ferris,C.F., and De Vries,G.J. (1994). Role of septal vasopressin innervation in


paternal behavior in prairie voles (Microtus ochrogaster). Proc. Natl. Acad. Sci. U. S. A
91, 400-404.

Wang,Z., Toloczko,D., Young,L.J., Moody,K., Newman,J.D., and Insel,T.R. (1997).


Vasopressin in the forebrain of common marmosets (Callithrix jacchus): studies with in
situ hybridization, immunocytochemistry and receptor autoradiography. Brain Res. 768,
147-156.

Weike,A.I., Bauer,U., and Hamm,A.O. (2000). Effective neuroleptic medication removes


prepulse inhibition deficits in schizophrenia patients. Biol. Psychiatry 47, 61-70.

Wersinger,S.R., Caldwell,H.K., Christiansen,M., and Young,W.S., III (2007). Disruption


of the vasopressin 1b receptor gene impairs the attack component of aggressive behavior
in mice. Genes Brain Behav. 6, 653-660.

Wersinger,S.R., Ginns,E.I., O'Carroll,A.M., Lolait,S.J., and Young,W.S., III (2002).


Vasopressin V1b receptor knockout reduces aggressive behavior in male mice. Mol.
Psychiatry 7, 975-984.

Wersinger,S.R., Kelliher,K.R., Zufall,F., Lolait,S.J., O'Carroll,A.M., and Young,W.S., III


(2004). Social motivation is reduced in vasopressin 1b receptor null mice despite normal
performance in an olfactory discrimination task. Horm. Behav. 46, 638-645.

Wersinger,S.R., Temple,J.L., Caldwell,H.K., and Young,W.S., III (2008). Inactivation of


the oxytocin and the vasopressin (Avp) 1b receptor genes, but not the Avp 1a receptor
gene, differentially impairs the Bruce effect in laboratory mice (Mus musculus).
Endocrinology 149, 116-121.

Williams,A.R., Carey,R.J., and Miller,M. (1985). Altered emotionality of the


vasopressin-deficient Brattleboro rat. Peptides 6 Suppl 1, 69-76.
87

Williams,A.R., Carey,R.J., and Miller,M. (1983). Behavioral differences between


vasopressin-deficient (Brattleboro) and normal Long-Evans rats. Peptides 4, 711-716.

Winslow,J.T., Hastings,N., Carter,C.S., Harbaugh,C.R., and Insel,T.R. (1993). A role for


central vasopressin in pair bonding in monogamous prairie voles. Nature 365, 545-548.

Wisniewski,K., Artemowicz,B., and Lutostanska,A. (1996). The estimation of


interactions between arginine-vasopressin (AVP) and NMDA receptors in memory and
learning processes. Acta Physiol Hung. 84, 477-479.

Wong,E.H., Kemp,J.A., Priestley,T., Knight,A.R., Woodruff,G.N., and Iversen,L.L.


(1986). The anticonvulsant MK-801 is a potent N-methyl-D-aspartate antagonist. Proc.
Natl. Acad. Sci. U. S. A 83, 7104-7108.

Yamada,S., Harano,M., Annoh,N., Nakamura,K., and Tanaka,M. (1999). Involvement of


serotonin 2A receptors in phencyclidine-induced disruption of prepulse inhibition of the
acoustic startle in rats. Biol. Psychiatry 46, 832-838.

Yee,B.K., Chang,D.L., and Feldon,J. (2004). The Effects of dizocilpine and


phencyclidine on prepulse inhibition of the acoustic startle reflex and on prepulse-elicited
reactivity in C57BL6 mice. Neuropsychopharmacology 29, 1865-1877.

Young,L.J., Wang,Z., Cooper,T.T., and Albers,H.E. (2000). Vasopressin (V1a) receptor


binding, mRNA expression and transcriptional regulation by androgen in the Syrian
hamster brain. J. Neuroendocrinol. 12, 1179-1185.

Young,W.S., Li,J., Wersinger,S.R., and Palkovits,M. (2006). The vasopressin 1b receptor


is prominent in the hippocampal area CA2 where it is unaffected by restraint stress or
adrenalectomy. Neuroscience 143, 1031-1039.
APPENDIX I

PILOT STUDY

Objective: To determine the optimum dose of drug to disrupt the PPI in both male and

female mice

Methods: 6 female and 6 male heterozygous (Avpr1b +/−) animals of ages 3-6 months

were used for the study. All the testing was conducted in the light phase of the 12:12 light

dark cycle. APO, AMP and MK-801 were in the following doses; APO (10mg/kg), AMP

(10mg/kg) and (12mg/kg), MK-801 (0.7mg/kg). All the drugs were dissolved in 0.9%

saline. Saline was given in the same volume as APO and AMP and served as control.

Animals were tested as per the protocol mentioned in the methods section. PPI

percentage was calculated using the following equation: (1−(startle amplitude of the

PREPULSE + PULSE/startle amplitude of the PULSE ALONE)) × 100. PPI percentage

was determined using repeated ANOVA and main effects of drug treatment, prepulse

intensity and their interactions were compared. Data from both males and females were

collapsed and analyzed together.

Results:

PPI percentage: There was a main effect of the drug treatment with APO (10mg/kg) (F1,9

= 6.005, P<0.05) and MK-801(0.7mg/kg) (F1,9= 16.384, P<0.05) all resulting in

88
89

disruption of PPI as compared to saline. There was also a main effect of prepulse

intensity (F2,18 = 58.048, P<0.001) with higher decibel tone resulting in increased PPI

percentage. However, treatment with AMP (10mg/kg) did not result in significant

disruption of PPI. Hence the experiment was repeated with an increased dose of AMP-

12mg/kg. Treatment with AMP (12mg/kg) resulted in significant disruption of PPI (F1,9=

11.220, P<0.05) as compared to saline (P<0.05).

Conclusions:

APO (10mg/kg), AMP (12mg/kg) and MK-801 (0.7mg/kg) resulted in significant

disruption of PPI as compared to saline in Avpr1b +/− mice. The above mentioned doses

were selected for conducting experiments 1, 2, and 3.


90

100 3dB
6dB
12dB
a b
80
% Prepulse Inhibtion

60 3DB
6DB
12 DB
Col 8
40

20

0
SALINE APO AMP MK801
0.9% 10mg/kg 10mg/kg 0.7mg/kg

Figure 24: PPI percentage in Avpr1b +/− mice following drug treatment: Treatment with
APO (a), and MK-801 (b) resulted in disruption of PPI as compared to saline (P<0.05).
There was also a main effect of prepulse intensity, with higher decibel tone resulting in
increased PPI percentage. Treatment with AMP at 10mg/kg did not result in significant
disruption of PPI as compared to saline. Data are expressed as means and standard error
of means. (Abbreviations: APO, apomorphine; AMP, amphetamine; MK-801,
dizocilpine).
91

100 * 3dB
6dB
12dB

80
% Prepulse Inhibtion

60 3DB
6DB
12 DB
Col 8
40

20

0
SALINE AMP
0.9% 12mg/kg

Figure 25: PPI percentage in Avpr1b +/− mice following treatment with AMP: Treatment
with AMP (12mg/kg) resulted in disruption of PPI as compared to saline (P<0.05). Data
are expressed as means and standard error of means. (Abbreviations: AMP,
amphetamine)
APPENDIX II

PPI Program used in Experiment 1, 2, and 3

Trial Decibel
1 pulse120 Parameters
2 pulse120
PULSE 1 3 pulse120 5 min acclimation
4 pulse120 Background 74dB
5 pulse120
6 pulse120 Pulse 120- 40 msec duration
7 PPI77
Block 1 8 PPI80 After variable ITI- 12-30 sec
9 PPI86
10 nopulse Prepulse 77- 20 msec duration,
11 PPI80 100 msec wait and pulse 120
12 pulse120
Block 2 Prepulse 80- 20 msec duration,
13 nopulse
100 msec wait and pulse 120
14 PPI77
15 PPI86
Prepulse 86- 20 msec duration,
16 PPI77 100 msec wait and pulse 120
17 Nopulse
Block 3 18 PPI86
19 pulse120
20 PPI80
21 Nopulse
22 PPI86
Block 4 23 PPI80
24 PPI77
25 pulse120
26 PPI86
27 PPI80
Block 5 28 pulse120
29 nopulse
30 PPI77
31 pulse120
32 PPI86
Block 6 33 PPI77
92
93

34 nopulse
35 PPI80
36 PPI77
37 pulse120
Block 7 38 nopulse
39 PPI86
40 PPI80
41 pulse120
42 PPI80
Block 8 43 PPI86
44 nopulse
45 PPI77
46 PPI80
47 nopulse
Block 9 48 PPI86
49 PPI77
50 pulse120
51 nopulse
52 PPI86
Block 10 53 PPI77
54 PPI80
55 pulse120
56 pulse120
57 pulse120
PULSE 2 58 pulse120
59 pulse120
60 pulse120

Вам также может понравиться