13.07.2013 Views

APPROACHES TO MODELING SCHIZOPHRENIA FN THE RAT by ...

APPROACHES TO MODELING SCHIZOPHRENIA FN THE RAT by ...

APPROACHES TO MODELING SCHIZOPHRENIA FN THE RAT by ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>APPROACHES</strong> <strong>TO</strong> <strong>MODELING</strong> <strong>SCHIZOPHRENIA</strong> <strong>FN</strong> <strong>THE</strong> <strong>RAT</strong><br />

<strong>by</strong><br />

JOHN GEORGE HOWLAND<br />

B A. (Hons.), University of Saskatchewan, 1999<br />

M.A., The University of British Columbia, 2001<br />

A <strong>THE</strong>SIS SUBMITTED IN PARTIAL FULFILMENT OF<br />

<strong>THE</strong> REQUIREMENTS FOR <strong>THE</strong> DEGREE OF<br />

DOC<strong>TO</strong>R OF PHILOSOPHY<br />

in<br />

<strong>THE</strong> FACULTY OF GRADUATE STUDIES<br />

(Psychology)<br />

<strong>THE</strong> UNIVERSITY OF BRITISH COLUMBIA<br />

October 2005<br />

© John George Howland, 2005


ABSTRACT<br />

Schizophrenia is a complicated and variable disorder that is notoriously difficult<br />

to study. Converging lines of evidence support the hypothesis that schizophrenia is<br />

characterized <strong>by</strong> a diverse array of distributed changes in limbic and cortical areas of the<br />

brain involving abnormalities in dopamine and glutamate transmission. Furthermore,<br />

genetic and behavioral studies indicate that abnormalities in normal development<br />

contribute to the etiology of the disorder. The present dissertation used two general<br />

strategies in an attempt to model some of the basic characteristics of the disorder. In<br />

Chapter Two, experiments were conducted that demonstrate short periods of higher<br />

frequency stimulation applied to the ventral, but not dorsal, hippocampus in adult rats<br />

reversibly reduce prepulse inhibition, a pre-attentive processing mechanism that is<br />

disrupted in schizophrenic patients. In Chapters Three and Four, the behavioral effects of<br />

reversible pharmacological manipulation of glutamate receptors early in development on<br />

prepulse inhibition and locomotor activity were assessed both before and after puberty.<br />

Additional experiments tested the putative role of dopaminergic abnormalities following<br />

these manipulations. Results from Chapter Three demonstrate that administration of a<br />

convulsive dose of the glutamate receptor agonist kainic acid to neonatal rats on postnatal<br />

day seven results in the delayed emergence of PPI deficits in early adulthood. Levels of<br />

locomotor activity were not reliably altered in a novel environment or following<br />

amphetamine administration. The experiments conducted in Chapter Four were designed<br />

to assess alterations in prepulse inhibition and locomotor activity following<br />

administration of the NR2B-subunit selective NMDA antagonist Ro25-6981.<br />

Unexpectedly, Ro25-6981 administration resulted in behavioral convulsions when


administered during the first postnatal week; however, no consistent behavioral<br />

abnormalities were revealed in rats treated with the drug. Although the present results<br />

are somewhat mixed, the experiments were successful at providing novel insights into the<br />

symptoms and etiology of schizophrenia. In general, they support the assertion that short<br />

periods of altered activity in the limbic system, and hippocampus in particular, at<br />

different points during development may underlie the expression of some of the most<br />

basic symptoms of schizophrenia. These data also suggest that the nature and anatomical<br />

location of these alterations critically determines their long-term functional effects.<br />

iii


TABLE OF CONTENTS<br />

ABSTRACT ii<br />

TABLE OF CONTENTS iv<br />

LIST OF TABLES viii<br />

LIST OF FIGURES ix<br />

ACKNOWLEDGMENTS xi<br />

CO-AUTHORSHD? STATEMENT xii<br />

CHAPTER ONE: GENERAL INTRODUCTION 1<br />

Schizophrenia 2<br />

Brief Overview of the Disorder 2<br />

Etiological Theories 7<br />

Animal Models 8<br />

Genetic Models 11<br />

Adult Pharmacological Models 12<br />

Adult Lesion Models 14<br />

Developmental Animal Models 16<br />

Maternal Separation and Social Isolation Models 16<br />

Neonatal Lesion Models 18<br />

Neonatal Pharmacological Models 22<br />

References 24<br />

CHAPTER TWO: ELECTRICAL STIMULATION OF <strong>THE</strong> HIPPOCAMPUS<br />

DISRUPTS PREPULSE INHIBITION IN <strong>RAT</strong>S: FREQUENCY AND SITE<br />

DEPENDENT<br />

EFFECTS 37<br />

Introduction 37<br />

Methods 41<br />

Subjects 41<br />

Surgery 41<br />

Prepulse Inhibition Testing 42<br />

Neurochemical Experiments 44<br />

Histology 46<br />

Data Analysis 46<br />

Results 47<br />

Immediate Behavioral Effects of Stimulation 47<br />

Differential Effects of 20 Hz Stimulation of the vHip or dHip on PPI and Startle<br />

Amplitude : 48<br />

iv


Effects of 2 Hz Stimulation of the vHip on PPI and Startle Amplitude 50<br />

Effects of Stimulation of the vHip and dHip on NAc DA Efflux 52<br />

Histology 52<br />

Discussion 55<br />

Potent Disruption of Sensorimotor Gating Induced <strong>by</strong> 20 Hz Stimulation of the vHip,<br />

but not dHip 55<br />

Frequency Dependence of the vHip Stimulation-induced Disruption of PPI 57<br />

Increases in NAc DA Efflux following Hip Stimulation are Site and Hemisphere<br />

Specific 59<br />

Potential Mechanisms Underlying vHip Stimulation-induced Disruption in PPI 61<br />

Conclusion 62<br />

References 63<br />

CHAPTER THREE: DELAYED ONSET OF PREPULSE INHIBITION DEFICITS<br />

FOLLOWING KAINIC ACID TREATMENT ON POSTNATAL DAY SEVEN IN<br />

<strong>RAT</strong>S 70<br />

Introduction 70<br />

Methods 72<br />

Subjects 72<br />

Kainic Acid Administration 73<br />

Prepulse Inhibition 74<br />

Locomotor Activity 75<br />

Water Maze Testing 76<br />

Histology ". 77<br />

Data Analysis 77<br />

Results ., 78<br />

Immediate Behavioral Effects ofPND7 KA Administration 78<br />

Effects of PND7 KA Administration on Body Weight 79<br />

Prepulse Inhibition is Reduced in Post-Pubescent, but not Pre-pubescent Rats,<br />

Following PND7 KA Administration 81<br />

Locomotor Activity in Response to Novelty and Amphetamine in Rats Following PND7<br />

KA Administration 84<br />

Spatial Learning and Memory in the Morris Water Maze is not Altered in Rats<br />

Following PND7 KA Administration 87<br />

Histology 89<br />

Discussion 91<br />

Effects of Neonatal KA Administration on PPI 91<br />

Effects of Neonatal KA on Locomotor Activity 93<br />

Potential Mechanisms Underlying the Observed PPI Changes 94<br />

Conclusion 97<br />

References 99<br />

CHAPTER FOUR: BEHAVIORAL CONVULSIONS INDUCED BY EARLY<br />

POSTNATAL ADMINIST<strong>RAT</strong>ION OF <strong>THE</strong> NR2B ANTAGONIST R025-6981<br />

FAIL <strong>TO</strong> AFFECT SENSORIMO<strong>TO</strong>R GATING OR LOCOMO<strong>TO</strong>R


BEHAVIOR IN PRE- AND POST-PUBESCENT<br />

<strong>RAT</strong>S..... : 105<br />

Introduction 105<br />

Methods 110<br />

Experiment 1 - Incidence and Characteristics of Behavioral Convulsions in Postnatal<br />

Rats Following Antagonism of NMDA Receptors Containing the NR2B Subunit.... 110<br />

Subjects 110<br />

Experimental Procedures 110<br />

Experiment 2 - Long-term Behavioral Effects of Convulsions Resulting from Antagonism<br />

of NMDA Receptors Containing the NR2B Subunit 1111<br />

Subjects Ill<br />

Ro25-6981 Administration Ill<br />

Prepulse Inhibition 112<br />

Spontaneous Locomotor Activity 113<br />

Dopaminergic Challenges of PPI and Locomotor Activity 113<br />

Data Analysis 114<br />

Results 115<br />

Experiment 1 - Behavioral Effects ofRo25-6981 Administration in the Early Postnatal<br />

Period 115<br />

Experiment 2 - Effects of Neonatal Ro25-6981 Administration on Body Weight 116<br />

Neonatal Ro25-6981 Administration Does not Alter PPI Responding Either before<br />

Puberty or in Early Adulthood 118<br />

Apomorphine Challenge in Adulthood has Similar Effects on PPI in Neonatally<br />

Saline- or Ro25-6981 -treated Rats 121<br />

Neonatal Treatment with Ro25-6981 and Amphetamine-Induced Locomotor Activity in<br />

Adult Rats 121<br />

Discussion 125<br />

Antagonism of NR2B-containing NMDA Receptors Induces Behavioral Convulsions<br />

126<br />

The Long-Term Behavioral Effects of Ro25-6981 on PPI and Locomotor Activity.. 129<br />

Conclusion 131<br />

References 132<br />

CHAPTER FIVE: GENERAL DISCUSSION 138<br />

The Role of the Hippocampus in the Regulation of Prepulse Inhibition and Locomotor<br />

Activity 139<br />

The Utility of Adult Animal Models of Schizophrenia 142<br />

Developmental Models of Schizophrenia - Effects of Early Postnatal Glutamate<br />

Manipulations 143<br />

Developmental Models of Schizophrenia - The Time Course of Symptom Emergence<br />

148<br />

Criteria for Establishing Validity in Behavioral Models of Schizophrenia 149<br />

Conclusion 152<br />

References 153<br />

vi


LIST OF TABLES<br />

Table 4-1. The incidence of behavioral convulsions in rats administered<br />

NR2B-selective NMDA antagonists at various postnatal ages 117<br />

vii


LIST OF FIGURES<br />

Figure 2-1. Effects of 20 Hz stimulation of the hippocampus on PPI and<br />

acoustic startle amplitude 49<br />

Figure 2-2. Effects of 2 Hz stimulation of the vHip on PPI and acoustic<br />

startle amplitude. 51<br />

Figure 2-3. Unilateral stimulation (20 Hz, 10 s) of either the vHip or dHip,<br />

and its effect on NAc DA efflux in the ipsilateral or contralateral hemisphere 53<br />

Figure 2-4. Schematic diagram of the placements of stimulating electrodes and<br />

microdialysis probes in all experiments 54<br />

Figure 3-1. Prepulse inhibition scores from rats in Group 1 treated on<br />

postnatal day 7 with saline or kainic acid 80<br />

Figure 3-2. Prepulse inhibition scores from rats in Group 2 treated on<br />

postnatal day 7 with saline or kainic acid 83<br />

Figure 3-3. Effects of pretreatment with vehicle or apomorphine on<br />

average percent PPI scores in adulthood 85<br />

Figure 3-4. Locomotor activity levels in response to novelty or amphetamine<br />

of all rats tested on either postnatal day 36 or 57 86<br />

Figure 3-5. Average latencies to locate the hidden platform in the water<br />

maze of rats treated with saline or kainic acid on postnatal day 7 88<br />

Figure 3-6. A representative cresyl violet stained section of the dorsal<br />

hippocampus of a KA-treated and saline-treated rat 90<br />

Figure 4-1. Cartoon illustrating an NMDA receptor within the cell membrane 107<br />

Figure 4-2. Prepulse inhibition scores of rats in Group 1 treated on<br />

postnatal day 6 and 7 with saline or Ro25-6981 119<br />

Figure 4-3. Prepulse inhibition scores from rats in Group 2 treated on<br />

postnatal day 6 and 7 with saline or Ro25-6981 120<br />

Figure 4-4. Effects of pretreatment with vehicle or apomorphine on<br />

percent PPI scores in adulthood 122<br />

Figure 4-5. Locomotor activity levels in response to novelty or<br />

amphetamine of rats tested on either postnatal day 36 or 57 in Group 1 123<br />

viii


Figure 4-6. Locomotor activity levels in response to novelty or amphetamine of rats<br />

tested on either postnatal day 36 or 57 in Group 2. 124


ACKNOWLEDGMENTS<br />

The work contained within this dissertation would not have been completed without the<br />

help of many people. Those who directly assisted me with these projects are co-authors<br />

on my publications. Many of those who indirectiy assisted me in the lab are listed<br />

alphabetically below. I would like to especially acknowledge the wisdom and expert<br />

advise of Drs. Tony Phillips, Stanley B. Floresco, and Darren 'Twinkle-Toes' Hannesson<br />

throughout my graduate career and Dr. Karen Brebner for providing many helpful<br />

comments on the General Introduction and Discussion Sections of my dissertation.<br />

Finally, I would like to acknowledge NSERC, CIHR, and the Michael Smith Foundation<br />

for Health Research for funding.<br />

Thank you:<br />

Soyon Ahn, Steven Barnes, Alasdair Barr, Karen Brebner, Deanna Chavez, Christina<br />

Cheng, Michael Corcoran, Carine Dias, Brennan Eadie, Rachel Genn, Rosie Gilham,<br />

Natalia Gorelova, Lucy Greggorios-Pippas, Rebecca Harrison, Fred Lepiane, Sarah C.<br />

Lidstone, Brandi Ormerod, Julie Pongrac, Kitty So, Aline Stephan, Christina Thorpe,<br />

Giada Vacca, and liana Winrob.<br />

To my friends and family who may not have helped directly with my academic life, but<br />

in many other ways, don't worry, your thanks is coming (and you're likely not reading<br />

this anyway). USB UBC.


CO-AUTHORSHIP STATEMENT<br />

I am the first author on all manuscripts presented in this thesis. I was directly<br />

involved in all stages of the research including, but not limited to, conception of the<br />

ideas, designing the experiments, conducting the studies, analyzing the data, and writing<br />

the manuscripts. Research assistants and other students helped in gathering some of the<br />

data presented in this dissertation and they are included as authors on the manuscripts<br />

with which they were involved. Dr. Phillips was also involved in all aspects of the<br />

research included in this dissertation. He also acted as my PhD supervisor and is<br />

included as an author on all manuscripts.


CHAPTER ONE: GENERAL INTRODUCTION<br />

The present dissertation summarizes several novel approaches to the development<br />

of rodent models of schizophrenia that are reliable and valid. As will become clear in the<br />

introduction, schizophrenia is an incredibly complicated and variable disorder (or group<br />

of disorders). Therefore, it is difficult to put forth a unitary set of criteria <strong>by</strong> which to<br />

verify the success of putative models of schizophrenia. The following pages detail a<br />

number of strategies aimed at reproducing some characteristics of the disorder. As the<br />

experiments of Chapter Two demonstrate, acute changes in hippocampal activity in<br />

adulthood reversibly alter behavior in a manner consistent with schizophrenia.<br />

Additionally, Chapters Three and Four show that under some circumstances, transient<br />

disruption of normal activity patterns in the developing brain is sufficient to subtiy<br />

change adult behavior in a manner consistent with schizophrenia. Finally, the<br />

implications of these studies and a number of directions for future research are discussed<br />

in Chapter Five.<br />

This dissertation is presented in a manuscript-based format, rather than the<br />

traditional thesis format. Chapters Two through Four are stand-alone manuscripts,<br />

containing their own Introduction and Reference sections, while Chapter One and Five<br />

are the General Introduction and Discussion sections of the entire dissertation. While this<br />

format has numerous practical advantages, it has two clear limitations. First, there is<br />

inevitably a certain amount of repetition between chapters, and second, individual<br />

chapters do not necessarily flow into the next. In an effort to address the second problem,<br />

I will include a brief statement of the objectives of each chapter in the General<br />

Introduction, and summarize the findings from all chapters in the General Discussion.<br />

1


Schizophrenia<br />

Brief Overview of the Disorder<br />

Schizophrenia is a debilitating mental illness with a lifetime prevalence of 1.4-4.6<br />

per 1000 people worldwide (Jablensky, 2000). Symptoms usually appear in late<br />

adolescence or early adulthood, although cases with earlier and later onset certainly exist<br />

(Wong and Van Tol, 2003). For many patients, schizophrenia has a chronic course with<br />

unpredictable relapses that require repeated hospitalizations. Although antipsychotic<br />

drugs effectively reduce symptom severity in many patients, their side effects are severe,<br />

and the drugs often fail to improve many of the negative and cognitive symptoms of the<br />

disorder (see below). It is certainly worth noting that the complexities and<br />

inconsistencies of the disorder are far from understood. As a result, the behavioral<br />

symptoms and neuropathological characteristics of the disorder described below have<br />

been simplified. Within the following chapters, pertinent details related to relevant<br />

aspects of schizophrenia are described where necessary.<br />

As the pharmacology underlying antipsychotics drug action has been used to<br />

support hypotheses related to the neural bases of the symptoms of schizophrenia, a brief<br />

review of their main classes is required. Typical antipsychotic drugs, which antagonize<br />

dopamine receptors, were discovered in the 1950's and revolutionized treatment of<br />

schizophrenia (Wong and Van Tol, 2003). During the 1980's and 1990's, psychiatrists<br />

began prescribing numerous atypical antipsychotic drugs to patients who did not respond<br />

to typical antipsychotics (either due to symptom profiles, side effects, or unknown<br />

reasons). Atypical antipsychotics have unique pharmacological effects, including<br />

2


affinities for serotonin, adrenergic, dopamine, histamine, and muscarinic receptors, which<br />

may underlie their effectiveness in some patients (Wong and Van Tol, 2003).<br />

The behavioral symptoms of schizophrenia are varied and involve numerous<br />

advanced functions of the human brain. Generally, symptoms can be divided into three<br />

main categories: the classic psychotic or 'positive' symptoms, deficit or 'negative'<br />

symptoms, and cognitive impairment (Wong and Van Tol, 2003). Positive symptoms are<br />

those typically associated with the illness - disturbances such as hallucinations,<br />

delusions, and thought disorder. Importantly, the appearance of positive symptoms in<br />

late adolescence or early adulthood generally leads to the diagnosis of schizophrenia. A<br />

number of researchers have argued that altered activity of the dopamine system is the<br />

likely cause of positive symptoms, a hypothesis largely based on the effectiveness of<br />

antipsychotic drugs, which are dopamine D2 receptor antagonists, in reducing positive<br />

symptoms and dopamine agonists at inducing or aggravating positive symptoms (Snyder,<br />

1973;Kapur and Mamo, 2003).<br />

Whereas positive symptoms may be thought of as the presence of abnormal<br />

behaviors, negative symptoms may be regarded as a reduction or loss in a number of<br />

normally occurring behaviors. Symptoms such as social withdrawal, apathy, reduced<br />

expression of affect, and the ability to experience pleasure are common examples of<br />

negative symptoms. The causes and treatment of negative symptoms are more poorly<br />

understood than the positive symptoms, although some atypical antipsychotic drugs, with<br />

actions at receptor subtypes other than the D2 receptor, may be more effective than<br />

typical antipsychotics at improving these symptoms (Meltzer et al., 1994;Wong and Van<br />

Tol, 2003).<br />

3


Finally, impairments in cognitive domains such as pre-attentive processing,<br />

executive function, attention, learning, memory, and general intellectual function are an<br />

important component of schizophrenia (Elvevag and Goldberg, 2000;Sharma and<br />

Antonova, 2003;Lewis, 2004). Intense interest has focused recently on these symptoms<br />

because of studies showing that the cognitive symptoms are the most enduring and<br />

consistently debilitating of the disorder (Elvevag and Goldberg, 2000). Additionally,<br />

impairments in cognition may precede the development of the positive symptoms of the<br />

disorder, there<strong>by</strong> providing a potential tool with which to identify those individuals at<br />

risk for expressing full-blown psychosis in early adulthood (Walker, 1994;Walker et al.,<br />

1994;Jones et al., 1994;Lewis, 2004).<br />

In addition to the behavioral changes observed in those suffering from the<br />

disorder, many alterations in the normal structure and function of the brain have been<br />

reported. For example, an overwhelming array of neuroanatomical changes is reported to<br />

exist in schizophrenia, many of which occur in the frontal and temporal lobes (McCarley<br />

et al, 1999;Wright et al., 2000). Ventricular enlargement (Stevens, 1997;Harrison, 1999)<br />

and reductions in temporal (Lawrie and Abukmeil, 1998;Nelson et al., 1998) or frontal<br />

lobe volumes (Wright et al., 1999) are observed in the brains of schizophrenic patients.<br />

Reduced volume of the hippocampus (Suddath et al., 1990;Noga et al., 1996;Bogerts,<br />

1997;Heckers, 2001), reduced cell numbers in the hippocampus (Gothelf et al.,<br />

2000;Heckers, 2001), nucleus accumbens (NAc; Pakkenberg, 1990), and thalamus<br />

(Jones, 1997;Clinton and Meador-Woodruff, 2004), and altered neuronal size and density<br />

in certain frontal cortical areas (Harrison, 1999) have been observed in patients with<br />

schizophrenia. Abnormalities in patterns of hippocampal (Heckers et al., 1998;Benes,


2000;Heckers, 2001;Holt et al., 2005), striatal (Meyer-Lindenberg et al., 2002) and<br />

prefrontal activation (McClure and Weinberger, 2001;Meyer-Lindenberg et al., 2002)<br />

during the performance of a variety of cognitive tasks are also characteristic of<br />

schizophrenic patients (Wong and Van Tol, 2003). Taken together, these<br />

neuroanatomical changes may be considered an enduring trait for schizophrenia in<br />

adulthood. Interestingly, relatives of patients with schizophrenia may also have reduced<br />

cortical volumes (Cannon et al., 1993), altered hippocampi (Seidman et al., 2002) and<br />

enlarged ventricles (Honer et al., 1994;Lawrie et al., 1999), there<strong>by</strong> supporting the<br />

assertion that an underlying genetic abnormality may predispose individuals to develop<br />

the disorder, although this underlying 'trait' for schizophrenia may have to interact with<br />

other environmental factors to result in the expression of a schizophrenic 'state' in early<br />

adulthood. Such conceptions of the etiology of schizophrenia are termed 'two-hit'<br />

hypotheses <strong>by</strong> some authors (McCarley et al., 1999;Bayer et al., 1999;Wong and Van<br />

Tol, 2003;Ellenbroek, 2003).<br />

The neurochemical changes present in schizophrenia are also complex and<br />

incompletely understood. The affinity of typical antipsychotics for the D2 receptor and<br />

psychotomimetic agents such as amphetamine and phencyclidine for the dopamine<br />

transporter and the N-methyl-D-aspartate (NMDA) receptor, respectively, are suggestive<br />

of the importance of dopamine and glutamate neurotransmission in schizophrenia<br />

(Angrist et al., 1974;Jentsch and Roth, 1999). More direct evidence of abnormal<br />

dopamine-glutamate interactions is gained from studies suggesting that alterations in<br />

glutamate and glutamate receptor levels may be present in schizophrenia (for review, see<br />

Goff and Coyle, 2001) and that schizophrenic patients show elevated synaptic levels of<br />

5


dopamine (Abi-Dargham et al., 2000) and potentiated increases in striatal dopamine<br />

release in response to amphetamine administration during episodes of acute psychosis<br />

(Breier et al, 1997;Abi-Dargham et al., 1998;Laruelle et al., 1999;Laruelle et al., 2003)<br />

and periods of relative remission (McGowan et al., 2004). Current theories of cortico-<br />

striatal-limbic function and schizophrenia have begun to delineate mechanisms <strong>by</strong> which<br />

these neurochemical changes and developmental abnormalities may manifest themselves<br />

as the symptoms of schizophrenia. One such example is the tonic-phasic model of<br />

ventral striatal dopamine release proposed <strong>by</strong> Grace (Grace, 1991). The tonic-phasic<br />

model posits that in schizophrenia, reduced activity from cortical glutamatergic afferents<br />

to the ventral striatum reduces basal (or tonic) levels of dopamine in the ventral striatum.<br />

Tonic levels of dopamine are thought to regulate the responsivity of the ventral striatum<br />

to short-lasting (or phasic) changes in dopamine efflux in response to environmental<br />

stimuli. As a result, patients with schizophrenia are predicted to exhibit increased<br />

dopaminergic responsivity to salient environmental stimuli, which may be a partial<br />

explanation of the positive symptoms (Grace, 1991). This model, along with others like<br />

it (Carlsson et al., 1999), illustrates the importance of dopamine-glutamate interactions in<br />

the neurobiology of schizophrenia.<br />

Additionally, the therapeutic efficacy of atypical antipsychotic drugs (Emsley and<br />

Oosthuizen, 2003; A wad and Voruganti, 2004) and a variety of additional<br />

neuropathological findings (Costa et al., 2004;Lewis et al., 2005) underlie the importance<br />

of other neurotransmitter systems in schizophrenia. In particular, alterations in the<br />

GABA and serotonin systems may exist in schizophrenia (Costa et al., 2004;Roth et al.,<br />

2004), although they will not be the focus of the present review.<br />

6


Etiological Theories<br />

Currently, the cause(s) of schizophrenia remain poorly defined. Twin and gene<br />

linkage studies provide convincing evidence that a genetic predisposition to develop<br />

schizophrenia exists, although environmental factors must also contribute as concordance<br />

rates between monozygotic twins are approximately 50 percent, whereas for dizygotic<br />

twins and siblings, the risk is approximately 10 percent (Kennedy et al., 2003;Owen et<br />

al., 2004). One particularly prominent etiological theory of the last 15 to 20 years is the<br />

neurodevelopmental theory (Weinberger, 1987;Lewis and Levitt, 2002;Church et al.,<br />

2002;Eastwood, 2004;Rapoport et al., 2005). In its original form, the theory suggests that<br />

genetic abnormalities may alter early brain development, or make the brain more<br />

susceptible to disruption. This 'compromised brain' then interacts with environmental<br />

factors or later occurring events of brain development and ultimately results in the<br />

expression of the symptoms typically observed in schizophrenic patients (Benes,<br />

2000;Benes et al., 2000;McClure and Weinberger, 2001;Lewis and Levitt, 2002).<br />

The neurodevelopmental theory is supported <strong>by</strong> evidence suggesting that people<br />

who develop schizophrenia have an increased incidence of adverse gestational and<br />

perinatal events, cognitive and behavioral abnormalities before diagnosis, congenital<br />

physical abnormalities, and alterations in brain morphology (for reviews see McClure and<br />

Weinberger, 2001;Lewis and Levitt, 2002). Further support is gained from evidence<br />

suggesting that the neuroanatomical changes observed in schizophrenia are present from<br />

the first episode diagnosis and their severity usually does not increase over time<br />

(Weinberger, 1987), although recent data challenges this notion (Weinberger,<br />

1996;Rapoport et al., 1999;Church et al., 2002;Rapoport et al., 2005). Additionally, signs<br />

7


of active neurodegeneration, such as gliosis, are generally absent from the brains of<br />

schizophrenic patients (Roberts et al., 1986;McClure and Weinberger, 2001), although<br />

such observations do not exclude the possibility that active neurodegenerative processes<br />

such as increased rates of apoptosis occur in patients with schizophrenia (Church et al.,<br />

2002).<br />

Animal Models<br />

Animal models have been used extensively in medicine to simplify complex and<br />

poorly understood pathologies. In psychiatry, and specifically in regards to<br />

schizophrenia, the development of reliable and valid animal models has proven difficult.<br />

Obviously, many symptoms of psychiatric disorders are inherently human, and thus can<br />

never be fully modeled in animals. As a result, attempts at modeling disorders such as<br />

schizophrenia in animals have been met with skepticism. Recently, a number of authors<br />

(Geyer and Markou, 1995;Lipska and Weinberger, 2000;Robbins, 2004) have advanced<br />

the notion that valuable and unique insights can be gained from modeling psychiatric<br />

disorders in animals, including rodents. Paramount to this undertaking is a realistic<br />

notion of the goal of the animal model, and the criteria which will be used to verify<br />

whether or not this goal has been achieved.<br />

With respect to schizophrenia, animal models have been developed for at least<br />

three main goals: furthering our understanding of its etiology, advancing our<br />

understanding of the symptoms (both behavioral and biological), and finally, aiding in the<br />

development and understanding of pharmaceutical therapies (Lipska and Weinberger,<br />

2000;Robbins, 2004). The evaluation criteria (or dependent measures) used to validate<br />

models of schizophrenia are quite varied, mostly due to the inherent difficulties of<br />

8


measuring the disorder in animals. Generally, models are designed to replicate the<br />

abnormal neurochemistry/neuropathology or behavioral changes described for<br />

schizophrenia. Within the present thesis, two behavioral tests - prepulse inhibition and<br />

locomotor behavior - are used extensively to validate the experimental manipulations<br />

used to model schizophrenia.<br />

Prepulse inhibition (PPI) of the acoustic startle response is an operational measure<br />

of sensorimotor gating, and is a normal pre-attentive behavioral response in which a weak<br />

sensory event (or prepulse) inhibits, or gates, the motor response to a starding stimulus<br />

(or pulse) (Fig. 1-1; Swerdlow et al., 2000a). PPI is disrupted in patients with<br />

schizophrenia or schizotypal personality disorder (Braff et al., 2001;Hamm et al.,<br />

2001 ;Ludewig et al., 2003) and in the first degree relatives of patients with schizophrenia<br />

(Cadenhead et al., 2000). These data suggest that PPI deficits are not a result of acute<br />

psychosis (Braff et al., 2001; but see also Meincke et al., 2004). Typical antipsychotic<br />

drugs do not ameliorate PPI deficits in schizophrenic patients (Mackeprang et al.,<br />

2002;Duncan et al., 2003a;Duncan et al., 2003b), although some beneficial effects of<br />

atypical antipsychotic drugs have been reported (Braff et al., 2001;Leumann et al.,<br />

2002;Kumari et al., 2002). More sophisticated longitudinal designs are necessary to<br />

further address the efficacy of antipsychotic drugs in reversing the PPI deficits observed<br />

in patients with schizophrenia (Kumari and Sharma, 2002). Importantly, PPI deficits are<br />

correlated with cognitive and behavioral symptoms such as distractibility, social<br />

perception, and thought disorder (Perry and Braff, 1994;Karper et al., 1996;Braff et al.,<br />

1999;Perry et al., 1999;Swerdlow et al., 2000a;Meincke et al., 2004;Wynn et al., 2005).<br />

Deficits in information processing capabilities, such as those measured <strong>by</strong> PPI, are core<br />

9


cognitive symptoms of schizophrenia and may relate to deficits in stimulus filtering<br />

commonly proposed to exist in the disorder (Swerdlow et al., 2000a).<br />

As PPI is easily and reliably measured in animals, it has been extensively used in<br />

cross-species work as valid behavioral test with relevance to schizophrenia (Swerdlow et<br />

al., 2000a). The use of PPI as an "index" of schizophrenia in animals is validated <strong>by</strong><br />

pharmacological experiments demonstrating that DA agonists such as apomorphine and<br />

NMDA antagonists such as phencyclidine alter PPI responding in animals and healthy<br />

humans, and antipsychotic drugs ameliorate these deficits in animals (Braff et al.,<br />

2001;Geyer et al., 2001). Extensive experimentation has been conducted to understand<br />

the neural substrates underlying the dopaminergic and glutamatergic regulation of PPI<br />

(Swerdlow et al., 2001). These experiments reveal a distributed circuitry in cortical,<br />

limbic, striatal, and pallidal areas important in the regulation of PPI. The pertinent details<br />

of this literature are reviewed in Chapter Two, and therefore will not be elaborated upon<br />

here.<br />

Locomotor responses to a variety of arousing stimuli such as novel environments<br />

or stimulant drugs are commonly measured in rodents as a measure of mesolimbic<br />

dopamine activity (Kelly et al., 1975;Castall et al., 1977;Porrino et al., 1984). More<br />

vigorous locomotor behavior in response to these treatments correlates positively with<br />

elevated ventral striatal dopamine levels (Kelly et al., 1975;Castall et al., 1977;Hooks et<br />

al., 1992). As previously reviewed, ventral striatal dopamine levels are higher in patients<br />

with schizophrenia than controls (Laruelle et al., 2003). As a result, locomotor activity<br />

levels are often used as an indirect measure of ventral striatal dopamine levels in animal<br />

models of schizophrenia (Lipska and Weinberger, 2000;Bast and Feldon, 2003;Boksa,<br />

10


2004). Additionally, some authors have argued that increased locomotor activity may be<br />

homologous to some cognitive abnormalities in schizophrenia patients during acute<br />

episodes (Bast and Feldon, 2003), although these accounts are debatable (Marcotte et al.,<br />

2001).<br />

In the present thesis, PPI and locomotor activity levels were used as the main<br />

endpoints to assess whether the novel experimental manipulations employed would<br />

produce effects resembling aspects of schizophrenia. These tests were selected for the<br />

following reasons: (1) Both tests have been used frequendy to assess previous preclinical<br />

models of schizophrenia. Thus, comparisons could easily be rnade between existing<br />

models and those developed within the present work. (2) These tests can be reliably<br />

conducted before and after puberty, enabling the developmental course of the behavioral<br />

effects of the neonatal manipulations performed in Chapters Three and Four to be<br />

assessed. (3) Pharmacological challenges are easily conducted in conjunction with these<br />

tests there<strong>by</strong> allowing the neurochemical mechanisms of observed behavioral changes to<br />

be evaluated.<br />

Given the multi-dimensional etiology of schizophrenia, researchers have<br />

advanced numerous strategies to model the disorder in animals, each with its own<br />

strengths and limitations. A brief review of the models pertinent to the present discussion<br />

of PPI and locomotor activity will now be presented.<br />

Genetic Models<br />

As previously discussed, the etiology of schizophrenia is at least partly dependent<br />

on complex genetic risk factors, likely involving a number of different genes. Therefore,<br />

there have been numerous attempts to model the disorder with genetic strategies.<br />

11


Knockout and knockdown strategies typically target genes for neurotransmitter systems<br />

implicated in schizophrenia. One well-known example includes dopamine transporter<br />

knockout mice, which show hyperlocomotion and deficits in PPI that are reversed with<br />

antipsychotics (Gainetdinov et al., 2001). Similar results have been gained with the NR-1<br />

knockdown mouse, which has reduced NMDA receptor function (Mohn et al.,<br />

1999;Duncan et al, 2004).<br />

Genetic models targeting genes involved in neurodevelopment and implicated in<br />

schizophrenia have also been developed. Reduced expression of reelin, a protein<br />

important for normal cell positioning in the brain, has been described in schizophrenia<br />

(Impagnatiello et al., 1998). Heterozygous reeler mice, which exhibit a down regulation<br />

of reelin similar to levels observed in schizophrenic patients, also have a variety of<br />

behavioral abnormalities similar to those in schizophrenia (Costa et al., 2002). These<br />

genetic strategies are useful in understanding the impact of genetic alterations on<br />

behavior, neurochemistry, and histology at a systems level, although these single gene<br />

approaches only model part of the complex etiology of schizophrenia. Clearly, the future<br />

challenge for genetic strategies lies in modeling the complex multi-gene abnormalities<br />

that will likely be implicated in schizophrenia (Wong and Van Tol, 2003). Additionally,<br />

combining genetic approaches with environmental factors in a 'two-hit' approach may be<br />

especially valuable in the future (Ellenbroek, 2003).<br />

Adult Pharmacological Models<br />

These models generally rely on producing acute or long-lasting behavioral<br />

changes relevant to schizophrenia <strong>by</strong> administering psychoactive drugs to animals.<br />

Dopamine agonists such as amphetamine and NMDA antagonists such as MK-801,<br />

12


phencyclidine or ketamine are most commonly used. Acute administration of these drugs<br />

induces deficits in PPI and increased locomotor activity (for review see Marcotte et al.,<br />

2001). Not surprisingly, the changes induced <strong>by</strong> dopamine agonists are reversed <strong>by</strong><br />

typical antipsychotics (Marcotte et al., 2001). Additionally, sensitization with<br />

amphetamine, but not phencyclidine, in rats induces long-lasting deficits in PPI, which<br />

may more faithfully model the stable PPI deficits in schizophrenia (Tenn et al., 2005).<br />

Interestingly, administration of NMDA antagonists may more accurately model<br />

schizophrenia <strong>by</strong> inducing a broader range of symptoms, including cognitive deficits and<br />

negative symptoms (Jentsch and Roth, 1999). The effects of NMDA antagonism are also<br />

reversed <strong>by</strong> atypical antipsychotics, there<strong>by</strong> improving the potential of the model to<br />

identify novel antipsychotic compounds.<br />

The data gained from adult pharmacological models is limited for a number of<br />

reasons. First, the effects are typically induced in normally developed adult animals. A<br />

wide range of evidence indicates that people who develop schizophrenia have<br />

abnormalities before adulthood. Importantly, combining neonatal perturbations of brain<br />

development with pharmacological challenges has provided valuable data, and will be<br />

discussed in the next section. Secondly, the changes in the activity of neurotransmitters<br />

such as dopamine are also more regionally specific than can be modeled with systemic<br />

administration of drugs (Kilts, 2001). Nevertheless, some treatment paradigms have<br />

proven to model more faithfully the neurochemical and behavioral changes in<br />

schizophrenia. For example, chronic exposure to phencyclidine in non-human primates<br />

results in cognitive deficits and reduced dopamine turn-over in the prefrontal cortex, both<br />

of which persist after drug treatment in discontinued (Jentsch et al., 1997). These effects<br />

13


can also be reversed following treatment with the atypical antipsychotic clozapine<br />

(Jentsch et al, 1997). Finally, some researchers argue that the development of novel<br />

antipsychotic drugs is precluded <strong>by</strong> dopamine agonist models because the therapeutic<br />

drugs that have emerged from these models block dopamine receptors, there<strong>by</strong> leading to<br />

a circular pattern of discovery (Lipska and Weinberger, 2000).<br />

Adult Lesion Models<br />

Adult lesion models focus on the effects of lesioning limbic and cortical areas<br />

commonly implicated in both subcortical dopamine regulation and schizophrenia, such as<br />

the prefrontal cortex, hippocampus, and thalamus (Lipska and Weinberger,<br />

2000;Marcotte et al., 2001). Obviously, the construct validity of adult lesion models is<br />

compromised <strong>by</strong> their adult initiation and large degree of damage that occurs (at least<br />

compared to that observed in schizophrenic patients); however, such models do provide<br />

unique insights into the neural substrates that may underlie many of the behavioral and<br />

neurochemical abnormalities in schizophrenia.<br />

Adult lesions of the medial prefrontal cortex in rodents result in hyper-<br />

responsiveness to stress (Jaskiw et al., 1990b), increases in spontaneous locomotor<br />

activity (Braun et al., 1993), increased activity in response to amphetamine (Jaskiw et al.,<br />

1990a), and deficits in PPI after challenge with the dopamine agonist apomorphine<br />

(Swerdlow et al, 1995). Similarly, lesions of the ventral, but not dorsal, hippocampus<br />

result in increased spontaneous activity and hyperactivity and PPI deficits following<br />

administration of dopamine agonists (Lipska et al., 1991 ;Lipska et al., 1992;Swerdlow et<br />

al., 1995;Mittleman et al., 1998). These data enable a detailed understanding of the<br />

neural substrates mediating PPI and locomotor activity to be developed, and will be<br />

14


eturned to in greater detail in Chapter Two. Importantly, they can be effectively<br />

integrated with neuroanatomical and neurochemical data, leading to new hypotheses<br />

regarding the mechanisms underlying the behavioral effects of the lesions. For example,<br />

the connectivity of the ventral, but not dorsal, hippocampus with areas such as the<br />

prefrontal cortex, amygdala, nucleus accumbens, and ventral tegmental area of the<br />

midbrain may be linked causally to the behavioral effects observed following its removal<br />

(Bast and Feldon, 2003).<br />

OBJECTIVE ONE: Numerous well-documented shortcomings of permanent lesions<br />

exist, such as compensatory alterations in brain areas other than the lesioned area<br />

(Schoenfeld and Hamilton, 1977;Stein, 1979;Bast and Feldon, 2003). Additionally,<br />

animals may develop strategies that aid in coping with the permanent loss of function<br />

normally carried out <strong>by</strong> the lesioned area (Lomber, 1999;Bast and Feldon, 2003).<br />

Specifically in the context of PPI and locomotor behavior, differences between<br />

permanent and temporary manipulations of the hippocampus have been described<br />

(Swerdlow et al., 1995;Pouzet et al., 1999;Swerdlow et al., 2000b;Zhang et al., 2002).<br />

Therefore, in Chapter Two, I examine the effect of brief stimulation of discrete sub-<br />

regions of the hippocampus on PPI. Given the important role of the hippocampus in<br />

schizophrenia, these experiments may provide further insights into the pathophysiology<br />

of the disorder.<br />

15


Developmental Animal Models<br />

As previously discussed, the neurodevelopmental hypothesis of schizophrenia has<br />

received considerable attention. Therefore, it is not surprising that numerous strategies<br />

have been developed to model schizophrenia using manipulations at distinct periods from<br />

embryological development until the time of weaning. As the present experiments focus<br />

on manipulations in the early neonatal period, most of the studies detailed below will<br />

focus on this time point. Numerous excellent reviews of experiments examining the<br />

effects of prenatal stress, obstetric complications, prenatal infection, and other<br />

developmental manipulations are available (Van den Buuse et al., 2003;Boksa, 2004).<br />

Before discussing developmental models, it is important to note that rodents are<br />

born at later stage of development than humans. Thus, the first week of postnatal life in<br />

rodents is typically described as corresponding to the third trimester of human fetal brain<br />

development (Lipska and Weinberger, 2000). Additionally, the surge of gonadal<br />

hormones that coincide with the onset of puberty occurs during the 5 th<br />

and 6 th<br />

postnatal<br />

weeks of the rodent life span. Thus, assessments of prepubertal function are generally<br />

conducted at approximately postnatal day 35 and assessments of adult function are<br />

performed after postnatal day 56.<br />

Maternal Separation and Social Isolation Models<br />

These two types of models involve removal of developing rats from their normal<br />

sources of social interaction, whether it be from their mothers early in development<br />

(maternal separation) or from their peers immediately following weaning (social<br />

isolation). Stress likely plays a major role in producing the behavioral changes observed<br />

in both models (Hall, 1998;Lipska and Weinberger, 2000;Weiss and Feldon, 2001 ;Van<br />

16


den et al., 2003). In one example of a maternal separation paradigm, a 24 hour separation<br />

period on postnatal day three, six, or nine is sufficient to produce deficits in PPI and<br />

increased sensitivity to dopamine agonists that occur only after puberty (Ellenbroek et al.,<br />

1998;Husum et al., 2002). Interestingly, these deficits can be reversed with typical or<br />

atypical antipsychotics (Ellenbroek et al., 1998). Strain differences are also observed,<br />

with effects present in Wistar rats, but not the Lewis or Fisher 344 strains (Ellenbroek<br />

and Cools, 2000). These data provide important support for the assertion that early life<br />

experiences interact with genetic background to alter adult patterns of behavior with<br />

relevance to schizophrenia (Lipska and Weinberger, 2000;Ellenbroek and Cools, 2000).<br />

The social isolation model involves rearing weanling rat pups individually<br />

without access to their peers, there<strong>by</strong> depriving them of social contact and considerable<br />

sensory stimulation (Hall, 1998;Weiss and Feldon, 2001 ;Van den et al., 2003). This<br />

model is distinct from maternal separation discussed above in that early development of<br />

the animals is not disturbed. Considerable research has been conducted using the social<br />

isolation model showing fairly consistent reductions in PPI, alterations in spontaneous<br />

locomotor activity, and locomotor responses to dopamine agonists and NMDA<br />

antagonists (Hall, 1998;Lipska and Weinberger, 2000;Weiss and Feldon, 2001).<br />

Parametric studies of the duration of rearing reveal that the pups must be in isolation for<br />

greater than three weeks starting at weaning (postnatal day 21 to 28) for behavioral<br />

alterations to be observed (Bakshi and Geyer, 1999;Varty et al., 1999). The behavioral<br />

effects are also sensitive to the strain of rats used (Weiss and Feldon, 2001), and are<br />

usually reversed with either typical or atypical antipsychotic drug treatment (Varty and<br />

Higgins, 1995;Bakshi et al., 1998). Thus, the social isolation model is an important<br />

17


nonpharmacological tool that can be used to identify novel antipsychotic compounds<br />

(Weiss and Feldon, 2001).<br />

With respect to both the maternal separation and isolation rearing paradigms,<br />

reliability is a concern. For example, some groups have failed to observe PPI deficits in<br />

maternally separated rats using similar, but not identical, methods (Lehmann et al.,<br />

2000;Weiss and Feldon, 2001). Factors such as the temperature at which the pups are<br />

maintained and the exact conditions under which they are separated (i.e. separated and<br />

kept in a group versus kept individually) may help explain the conflicting results (Weiss<br />

and Feldon, 2001). With regard to isolation rearing, the robustness of the behavioral<br />

effects has been questioned, as some reports indicate that the model is quite sensitive to<br />

handling or other testing procedures (Weiss and Feldon, 2001). However, a long-term<br />

evaluation of the reliability of PPI deficits in the social isolation model indicate that PPI<br />

deficits occur in approximately 85 percent of groups tested against matched controls<br />

(Cilia et al., 2005).<br />

Neonatal Lesion Models<br />

A great deal of work in the last ten to fifteen years has examined the utility of<br />

performing neonatal lesions of cortical and limbic areas in modeling schizophrenia<br />

(Lipska and Weinberger, 2000). This procedure has a number of distinct advantages over<br />

those discussed above: (1) The normal maturation of specific brain areas is altered and<br />

this simplifies the search for mechanisms underlying observed changes in the model. (2)<br />

The consequences of the lesion can be examined over the course of development there<strong>by</strong><br />

allowing the timing of any changes to be compared to the normal course of the disease.<br />

(3) Manipulating the brain at an early age is consistent with the neurodevelopmental<br />

18


theory of schizophrenia. (4) Further manipulations can be performed later in development<br />

to test the 'two-hit' hypothesis of schizophrenia.<br />

Most prominent among these models is the ventral hippocampal lesion model<br />

initially described <strong>by</strong> Lipska and colleagues (Lipska et al., 1993). In this model, rats are<br />

subjected to bilateral lesions of the ventral hippocampus with the excitotoxin ibotenic<br />

acid on postnatal day seven. The animals are then allowed to develop normally and can<br />

be tested before or after puberty on an array of behavioral tests relevant to schizophrenia.<br />

Numerous reports indicate that rats with neonatal ventral hippocampal lesions display<br />

PPI deficits and increased spontaneous locomotor activity (Lipska et al., 1993;Lipska et<br />

al., 1995). The PPI deficits are potentiated <strong>by</strong> a low dose of apomorphine (Lipska et al.,<br />

1995) and reversed <strong>by</strong> atypical antipsychotic drugs, but not <strong>by</strong> haloperidol (Le Pen and<br />

Moreau, 2002). Locomotor activity is also increased following stress (Lipska et al.,<br />

1993), dopamine agonist administration (Lipska et al., 1993), or treatment with NMDA<br />

antagonists (Al Amin et al., 2000;A1 Amin et al., 2001). The alterations in locomotor<br />

behavior can be normalized with haloperidol, but not atypical antipsychotic drugs (Al<br />

Amin et al., 2000). Treatment with ionotropic glutamate receptor antagonists (Al Amin<br />

et al., 2000) and increasing glycine availability (Le Pen et al., 2003) also reverses some<br />

of the behavioral impairments, there<strong>by</strong> indicating a role of glutamate transmission in the<br />

underlying effects of neonatal ventral hippocampal lesions.<br />

Consistent with the typical course of schizophrenia, the PPI deficits and<br />

locomotor activity changes are not expressed until after the rats reach puberty and persist<br />

at least until mid-adulthood (Lipska and Weinberger, 1993;Lipska et al., 1995;Lipska and<br />

Weinberger, 2000;A1 Amin et al., 2001). Interestingly, lesioned rats show reduced social<br />

19


interaction before and after puberty, a test which may have some relevance to the<br />

abnormalities observed in schizophrenic patients before their first episode (Sams-Dodd et<br />

al., 1997). Parametric experiments also suggest that the delayed onset of behavioral<br />

changes in the animals only occurs if the lesions are created during the first postnatal<br />

week of life (Wood et al., 1997), which corresponds to the third trimester in human<br />

development (Lipska and Weinberger, 2000). Finally, in an important paper, Lipska and<br />

Weinberger (Lipska and Weinberger, 1995) demonstrated that the effects of the neonatal<br />

lesion depend on the genetic background of the animals. Rats highly responsive to stress<br />

(the Fischer 344 strain) showed exaggerated effects of the lesion, whereas rats hypo-<br />

responsive to stress (the Lewis strain) showed reduced lesion effects. Thus, this model<br />

supports the theory that schizophrenia may develop from an interaction between genetic<br />

predisposition to the disorder and adverse environmental events (Lipska and Weinberger,<br />

1995;Benes, 2000;Benes et al., 2000;McClure and Weinberger, 2001;Ellenbroek, 2003).<br />

The results gained from the ventral hippocampal model have led others to<br />

examine the effects of neonatally lesioning other areas such as the medial prefrontal<br />

cortex (Flores et al., 1996;Schneider and Koch, 2005), amygdala (Hanlon and Sutherland,<br />

2000;Daenen et al., 2001;Daenen et al., 2002a;Daenen et al, 2002b;Daenen et al.,<br />

2003a;Daenen et al., 2003b) and thalamus (Lipska et al., 2003) on a similar range of<br />

behaviors. Many of these experiments have yielded results similar to, or less consistent<br />

than, those already discussed. As a result, they will not be elaborated on further.<br />

While it is clear that many characteristics of schizophrenia have been modeled<br />

with great success using neonatal ventral hippocampal lesions, the severity and<br />

permanent nature of the hippocampal lesion limits the construct validity of this model for<br />

20


schizophrenia (Lipska and Weinberger, 2000). As a result, Lipska and colleagues (2002)<br />

demonstrated that reversible temporary inactivation of the ventral hippocampus with<br />

tetrodotoxin on postnatal day seven is sufficient to induce significant changes in<br />

locomotor behavior in adulthood, but not before puberty. This manipulation produces<br />

limited gross morphological damage to the ventral hippocampus, and is short-lasting (24-<br />

48 hr) (Lipska et al., 2002). These data clearly highlight the fragility of the hippocampal-<br />

cortical circuit during this developmental period. Additional data demonstrate that a<br />

single infusion of the NMDA antagonist (±)-2-amino-5-phosphonopentanoic acid (APV)<br />

into the rat medial prefrontal cortex on postnatal day seven results in exaggerated<br />

amphetamine-induced locomotor activity in adulthood (Lafleur et al., 2001). These data<br />

support the hypothesis that an abnormality in activity-dependent plasticity involving<br />

glutamatergic hippocampal-prefrontal interactions may underlie some of the symptoms<br />

observed following neonatal ventral hippocampal lesions (Lipska et al., 2002).<br />

Additionally, these data support the hypothesis that the first postnatal week is a critical<br />

period for the development of cortico-limbic circuits mediating behaviors such as<br />

locomotor activity and PPI.<br />

Although the neonatal ventral hippocampal lesion model provides unique insight<br />

into the consequences of damaging a particular area early in development, it is unlikely<br />

that schizophrenia results from damage to a single brain area. Thus, the efficacy of<br />

altering the activity of the brain pharmacologically during the first postnatal week has<br />

also been tested in an effort to model schizophrenia. These experiments are discussed in<br />

the following section.<br />

21


Neonatal Pharmacological Models<br />

As previously discussed, alterations of the glutamatergic system are likely to be<br />

involved in the expression of the symptoms of schizophrenia. A number of lines of<br />

evidence support the hypothesis that the glutamatergic system may also be involved in<br />

the development of the disorder. For example, adverse events during early development<br />

such as febrile seizures, fetal alcohol exposure, and hypoxia/ischemia involve the<br />

glutamatergic system (Morimoto et al., 1995;01ney et al., 1999;Zornberg et al.,<br />

. 2000;Olney, 2004) and are risk factors for psychosis and/or schizophrenia (Cannon,<br />

1997;Famy et al., 1998;01ney et al., 1999;Kanemoto et al., 2001;Vestergaard et al.,<br />

2005). The normal maturation and development of limbic and cortical areas relevant to<br />

schizophrenia is also dependent on normal glutamatergic function. For example,<br />

processes such as cell birth, survival and synaptogenesis are critically dependent on<br />

glutamate transmission, particularly at NMDA receptors (Rabacchi et al., 1992;Gould et<br />

al., 1994;Fox et al., 1996;Vallano, 1998;Ikonomidou et al., 1999;Luthi et al., 2001).<br />

Given the established role of glutamate neurotransmission in neonatal<br />

development during periods implicated in schizophrenia, a number of studies have<br />

examined the behavioral effects of manipulating glutamate transmission<br />

pharmacologically early in life. Most of these experiments have tested the effects of<br />

systemically administered NMDA receptor antagonists such as MK-801, ketamine or<br />

phencyclidine during the neonatal period. A number of groups have demonstrated<br />

deficits in PPI and alterations in locomotor behavior following treatment with these drugs<br />

(Facchinetti et al., 1993;Wang et al., 2001;Ffarris et al., 2003;Fredriksson et al., 2004).<br />

As will be elaborated further in Chapter Four of the present thesis, the long-lasting<br />

22


morphological effects of administering non-competitive NMDA antagonists early in the<br />

neonatal period are also relevant to schizophrenia (Wang et al., 2001 ;Harris et al., 2003).<br />

OBJECTIVE TWO: Given that the end of the first postnatal week is a sensitive period for<br />

the development of neural circuits mediating PPI and locomotor activity (Wood et al.,<br />

1997;Wang et al., 2001;Lipska et al., 2002;Harris et al, 2003), the second objective of<br />

this thesis was to extend these findings in two novel directions. In Chapter Three, a<br />

series of experiments examined the long-term behavioral effects of administering the<br />

glutamate receptor agonist kainic acid to postnatal day seven rats. Chapter Four<br />

summarizes an independent set of experiments designed to test the effects of<br />

administration of the highly specific NMDA receptor antagonist [(+/-)-(R*,S*)-alpha-(4-<br />

hydroxyphenyl)-beta-methyl-4-(phenylmethyl)-l-piperidine propanol] (Ro25-6981)<br />

during the same time period. These pharmacological manipulations were designed to<br />

subtly alter the normal development of the cortico-limbic circuitry involved in<br />

schizophrenia - kainic acid <strong>by</strong> selectively activating the hippocampus and Ro25-6981 <strong>by</strong><br />

blocking NR2B subunit-containing NMDA receptors, which are highly expressed during<br />

the first postnatal week in rodents.<br />

23


References<br />

Abi-Dargham A, Gil R, Krystal J, Baldwin RM, Sei<strong>by</strong>l JP, Bowers M, van Dyck CH,<br />

Charney DS, Innis RB, Laruelle M (1998) Increased striatal dopamine transmission in<br />

schizophrenia: confirmation in a second cohort. Am J Psychiatry 155: 761-767.<br />

Abi-Dargham A, Rodenhiser J, Printz D, Zea-Ponce Y, Gil R, Kegeles LS, Weiss R,<br />

Cooper TB, Mann JJ, Van Heertum RL, Gorman JM, Laruelle M (2000) Increased<br />

baseline occupancy of D2 receptors <strong>by</strong> dopamine in schizophrenia. Proc Natl Acad Sci U<br />

S A 97: 8104-8109.<br />

Al Amin HA, Shannon WC, Weinberger DR, Lipska BK (2001) Delayed onset of<br />

enhanced MK-801-induced motor hyperactivity after neonatal lesions of the rat ventral<br />

hippocampus. Biol Psychiatry 49: 528-539.<br />

Al Amin HA, Weinberger DR, Lipska BK (2000) Exaggerated MK-801-induced motor<br />

hyperactivity in rats with the neonatal lesion of the ventral hippocampus. Behav<br />

Pharmacol 11: 269-278.<br />

Angrist B, Sathananthan G, Wilk S, Gershon S (1974) Amphetamine psychosis:<br />

behavioral and biochemical aspects. J Psychiatr Res 11: 13-23.<br />

Awad AG, Voruganti LN (2004) Impact of atypical antipsychotics on quality of life in<br />

patients with schizophrenia. CNS Drugs 18: 877-893.<br />

Bakshi VP, Geyer MA (1999) Ontogeny of isolation rearing-induced deficits in<br />

sensorimotor gating in rats. Physiol Behav 67: 385-392.<br />

Bakshi VP, Swerdlow NR, Braff DL, Geyer MA (1998) Reversal of isolation rearinginduced<br />

deficits in prepulse inhibition <strong>by</strong> Seroquel and olanzapine. Biol Psychiatry 43:<br />

436-445.<br />

Bast T, Feldon J (2003) Hippocampal modulation of sensorimotor processes. Prog<br />

Neurobiol 70: 319-345.<br />

Bayer TA, Falkai P, Maier W (1999) Genetic and non-genetic vulnerability factors in<br />

schizophrenia: the basis of the "Two hit hypothesis". Journal of Psychiatric Research 33:<br />

543-548.<br />

Benes FM (2000) Emerging principles of altered neural circuitry in schizophrenia. Brain<br />

Res Brain Res Rev 31: 251-269.<br />

Benes FM, Taylor JB, Cunningham MC (2000) Convergence and plasticity of<br />

monoaminergic systems in the medial prefrontal cortex during the postnatal period:<br />

implications for the development of psychopathology. Cereb Cortex 10: 1014-1027.<br />

Bogerts B (1997) The temporolimbic system theory of positive schizophrenic symptoms.<br />

Schizophr Bull 23: 423-435.<br />

24


Boksa P (2004) Animal models of obstetric complications in relation to schizophrenia.<br />

Brain Res Brain Res Rev 45: 1-17.<br />

Braff DL, Geyer MA, Swerdlow NR (2001) Human studies of prepulse inhibition of<br />

startle: normal subjects, patient groups, and pharmacological studies.<br />

Psychopharmacology (Berl) 156: 234-258.<br />

Braff DL, Swerdlow NR, Geyer MA (1999) Symptom correlates of prepulse inhibition<br />

deficits in male schizophrenic patients. Am J Psychiatry 156: 596-602.<br />

Braun AR, Jaskiw GE, Vladar K, Sexton RH, Kolachana BS, Weinberger DR (1993)<br />

Effects of ibotenic acid lesion of the medial prefrontal cortex on dopamine agonistrelated<br />

behaviors in the rat. Pharmacol Biochem Behav 46: 51-60.<br />

Breier A, Su TP, Saunders R, Carson RE, Kolachana BS, de Bartolomeis A, Weinberger<br />

DR, Weisenfeld N, Malhotra AK, Eckelman WC, Pickar D (1997) Schizophrenia is<br />

associated with elevated amphetamine-induced synaptic dopamine concentrations:<br />

evidence from a novel positron emission tomography method. Proc Natl Acad Sci U S A<br />

94: 2569-2574.<br />

Cadenhead KS, Swerdlow NR, Shafer KM, Diaz M, Braff DL (2000) Modulation of the<br />

startle response and startle laterality in relatives of schizophrenic patients and in subjects<br />

with schizotypal personality disorder: evidence of inhibitory deficits. Am J Psychiatry<br />

157: 1660-1668.<br />

Cannon TD (1997) On the nature and mechanisms of obstetric influences in<br />

schizophrenia: a review and synthesis of epidemiologic studies. International Review of<br />

Psychiatry 9: 387-397.<br />

Cannon TD, Mednick SA, Parnas J, Schulsinger F, Praestholm J, Vestergaard A (1993)<br />

Developmental brain abnormalities in the offspring of schizophrenic mothers. I.<br />

Contributions of genetic and perinatal factors. Arch Gen Psychiatry 50: 551-564.<br />

Carlsson A, Waters N, Carlsson ML (1999) Neurotransmitter interactions in<br />

schizophrenia—therapeutic implications. Biol Psychiatry 46: 1388-1395.<br />

Castall B, Marsden CD, Naylor RJ, Pycock CJ (1977) Stereotyped behaviour patterns and<br />

hyperactivity induced <strong>by</strong> amphetamine and apomorphine after discrete 6hydroxydopamine<br />

lesions of extrapyramidal and mesolimbic nuclei. Brain Res 123: 89-<br />

111.<br />

Church SM, Cotter D, Bramon E, Murray RM (2002) Does schizophrenia result from<br />

developmental or degenerative processes? Journal of Neural Transmission-Supplement<br />

129-147.<br />

Cilia J, Hatcher PD, Reavill C, Jones DN (2005) Long-term evaluation of isolationrearing<br />

induced prepulse inhibition deficits in rats: an update. Psychopharmacology<br />

(Berl) 180: 57-62.<br />

25


Clinton SM, Meador-Woodruff JH (2004) Thalamic dysfunction in schizophrenia:<br />

neurochemical, neuropathological, and in vivo imaging abnormalities. Schizophrenia<br />

Research 69: 237-253.<br />

Costa E, Davis J, Pesold C, Tueting P, Guidotti A (2002) The heterozygote reeler mouse<br />

as a model for the development of a new generation of antipsychotics. Curr Opin<br />

Pharmacol 2: 56-62.<br />

Costa E, Davis JM, Dong E, Grayson DR, Guidotti A, Tremolizzo L, Veldic M (2004) A<br />

GABAergic cortical deficit dominates schizophrenia pathophysiology. Crit Rev<br />

Neurobiol 16: 1-23.<br />

Daenen EW, Van Der Heyden JA, Kruse CG, Wolterink G, Van Ree JM (2001)<br />

Adaptation and habituation to an open field and responses to various stressful events in<br />

animals with neonatal lesions in the amygdala or ventral hippocampus. Brain Res 918:<br />

153-165.<br />

Daenen EW, Wolterink G, Gerrits MA, Van Ree JM (2002a) Amygdala or ventral<br />

hippocampal lesions at two early stages of life differentially affect open field behaviour<br />

later in life; an animal model of neurodevelopmental psychopathological disorders.<br />

Behav Brain Res 131: 67-78.<br />

Daenen EW, Wolterink G, Gerrits MA, Van Ree JM (2002b) The effects of neonatal<br />

lesions in the amygdala or ventral hippocampus on social behaviour later in life. Behav<br />

Brain Res 136: 571-582.<br />

Daenen EW, Wolterink G, Van Der Heyden JA, Kruse CG, Van Ree JM (2003a)<br />

Neonatal lesions in the amygdala or ventral hippocampus disrupt prepulse inhibition of<br />

the acoustic startle response; implications for an animal model of neurodevelopmental<br />

disorders like schizophrenia. Eur Neuropsychopharmacol 13: 187-197.<br />

Daenen EW, Wolterink G, Van Ree JM (2003b) Hyperresponsiveness to phencyclidine in<br />

animals lesioned in the amygdala on day 7 of life. Implications for an animal model of<br />

schizophrenia. Eur Neuropsychopharmacol 13: 273-279.<br />

Duncan E, Szilagyi S, Schwartz M, Kunzova A, Negi S, Efferen T, Peselow E,<br />

Chakravorty S, Stephanides M, Harmon J, Bugarski-Kirola D, Gonzenbach S, Rotrosen J<br />

(2003a) Prepulse inhibition of acoustic startle in subjects with schizophrenia treated with<br />

olanzapine or haloperidol. Psychiatry Res 120: 1-12.<br />

Duncan EJ, Szilagyi S, Efferen TR, Schwartz MP, Parwani A, Chakravorty S, Madonick<br />

SH, Kunzova A, Harmon JW, Angrist B, Gonzenbach S, Rotrosen JP (2003b) Effect of<br />

treatment status on prepulse inhibition of acoustic startle in schizophrenia.<br />

Psychopharmacology (Berl) 167: 63-71.<br />

Duncan GE, Moy SS, Perez A, Eddy DM, Zinzow WM, Lieberman JA, Snouwaert JN,<br />

Roller BH (2004) Deficits in sensorimotor gating and tests of social behavior in a genetic<br />

model of reduced NMDA receptor function. Behav Brain Res 153: 507-519.<br />

26


Eastwood SL (2004) The synaptic pathology of schizophrenia: Is aberrant<br />

neurodevelopment and plasticity to blame? Disorders of Synaptic Plasticity and<br />

Schizophrenia 59: 47-72.<br />

Ellenbroek BA (2003) Animal models in the genomic era: possibilities and limitations<br />

with special emphasis on schizophrenia. Behav Pharmacol 14: 409-417.<br />

Ellenbroek BA, Cools AR (2000) The long-term effects of maternal deprivation depend<br />

on the genetic background. Neuropsychopharmacology 23: 99-106.<br />

Ellenbroek BA, van den Kroonenberg PT, Cools AR (1998) The effects of an early<br />

stressful life event on sensorimotor gating in adult rats. Schizophr Res 30: 251-260.<br />

Elvevag B, Goldberg TE (2000) Cognitive impairment in schizophrenia is the core of the<br />

disorder. Crit Rev Neurobiol 14: 1-21.<br />

Emsley R, Oosthuizen P (2003) The new and evolving pharmacotherapy of<br />

schizophrenia. Psychiatr Clin North Am 26: 141-163.<br />

Facchinetti F, Ciani E, Dallolio R, Virgili M, Contestabile A, Fonnum F (1993)<br />

Structural, Neurochemical and Behavioral Consequences of Neonatal Blockade of Nmda<br />

Receptor Through Chronic Treatment with Cgp-39551 Or Mk-801. Developmental Brain<br />

Research 74: 219-224.<br />

Famy C, Streissguth AP, Unis AS (1998) Mental illness in adults with fetal alcohol<br />

syndrome or fetal alcohol effects. Am J Psychiatry 155: 552-554.<br />

Flores G, Wood GK, Liang JJ, Quirion R, Srivastava LK (1996) Enhanced amphetamine<br />

sensitivity and increased expression of dopamine D2 receptors in postpubertal rats after<br />

neonatal excitotoxic lesions of the medial prefrontal cortex. J Neurosci 16: 7366-7375.<br />

Fox K, Schlaggar BL, Glazewski S, O'Leary DD (1996) Glutamate receptor blockade at<br />

cortical synapses disrupts development of thalamocortical and columnar organization in<br />

somatosensory cortex. Proc Nad Acad Sci U S A 93: 5584-5589.<br />

Fredriksson A, Archer T, Aim H, Gordh T, Eriksson P (2004) Neurofunctional deficits<br />

and potentiated apoptosis <strong>by</strong> neonatal NMDA antagonist administration. Behavioural<br />

Brain Research 153: 367-376.<br />

Gainetdinov RR, Mohn AR, Caron MG (2001) Genetic animal models: focus on<br />

schizophrenia. Trends Neurosci 24: 527-533.<br />

Geyer MA, Krebs-Thomson K, Braff DL, Swerdlow NR (2001) Pharmacological studies<br />

of prepulse inhibition models of sensorimotor gating deficits in schizophrenia: a decade<br />

in review. Psychopharmacology (Berl) 156: 117-154.<br />

27


Geyer MA, Markou A (1995) Animal models of psychiatric disorders. In:<br />

Psychopharmacology: The Fourth Generation of Progress (Bloom FE, Kupfer DJ, eds),<br />

pp 787-798. New York: Raven Press, Ltd.<br />

Goff DC, Coyle JT (2001) The emerging role of glutamate in the pathophysiology and<br />

treatment of schizophrenia. Am J Psychiatry 158: 1367-1377.<br />

Gothelf D, Soreni N, Nachman RP, Tyano S, Hiss Y, Reiner O, Weizman A (2000)<br />

Evidence for the involvement of the hippocampus in the pathophysiology of<br />

schizophrenia. Eur Neuropsychopharmacol 10: 389-395.<br />

Gould E, Cameron HA, McEwen BS (1994) Blockade of NMDA receptors increases cell<br />

death and birth in the developing rat dentate gyrus. J Comp Neurol 340: 551-565.<br />

Grace AA (1991) Phasic versus tonic dopamine release and the modulation of dopamine<br />

system responsivity: a hypothesis for the etiology of schizophrenia. Neuroscience 41: 1-<br />

24.<br />

Hall FS (1998) Social deprivation of neonatal, adolescent, and adult rats has distinct<br />

neurochemical and behavioral consequences. Crit Rev Neurobiol 12: 129-162.<br />

Hamm AO, Weike Al, Schupp HT (2001) The effect of neuroleptic medication on<br />

prepulse inhibition in schizophrenia patients: current status and future issues.<br />

Psychopharmacology (Berl) 156: 259-265.<br />

Hanlon FM, Sutherland RJ (2000) Changes in adult brain and behavior caused <strong>by</strong><br />

neonatal limbic damage: implications for the etiology of schizophrenia. Behav Brain Res<br />

107:71-83.<br />

Harris LW, Sharp T, Gartlon J, Jones DNC, Harrison PJ (2003) Long-term behavioural,<br />

molecular and morphological effects of neonatal NMDA receptor antagonism. European<br />

Journal of Neuroscience 18: 1706-1710.<br />

Harrison PJ (1999) The neuropathology of schizophrenia. A critical review of the data<br />

and their interpretation. Brain 122 (Pt 4): 593-624.<br />

Heckers S (2001) Neuroimaging studies of the hippocampus in schizophrenia.<br />

Hippocampus 11: 520-528. ;<br />

Heckers S, Rauch SL, Goff D, Savage CR, Schacter DL, Fischman AJ, Alpert NM (1998)<br />

Impaired recruitment of the hippocampus during conscious recollection in schizophrenia.<br />

Nat Neurosci 1: 318-323.<br />

Holt DJ, Weiss AP, Rauch SL, Wright CI, Zalesak M, Goff DC, Ditman T, Welsh RC,<br />

Heckers S (2005) Sustained activation of the hippocampus in response to fearful faces in<br />

schizophrenia. Biological Psychiatry 57: 1011-1019.<br />

28


Honer WG, Bassett AS, Smith GN, Lapointe JS, Falkai P (1994) Temporal lobe<br />

abnormalities in multigenerational families with schizophrenia. Biol Psychiatry 36: 737-<br />

743.<br />

Hooks MS, Colvin AC, Juncos JL, Justice JB, Jr. (1992) Individual differences in basal<br />

and cocaine-stimulated extracellular dopamine in the nucleus accumbens using<br />

quantitative microdialysis. Brain Res 587: 306-312.<br />

Husum H, Termeer E, Mathe AA, Bolwig TG, Ellenbroek BA (2002) Early maternal<br />

deprivation alters hippocampal levels of neuropeptide Y and calcitonin-gene related<br />

peptide in adult rats. Neuropharmacology 42: 798-806.<br />

Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vockler J, Dikranian K, Tenkova TI,<br />

Stefovska V, Turski L, Olney JW (1999) Blockade of NMDA receptors and apoptotic<br />

neurodegeneration in the developing brain. Science 283: 70-74.<br />

Impagnatiello F, Guidotti AR, Pesold C, Dwivedi Y, Caruncho H, Pisu MG, Uzunov DP,<br />

Smalheiser NR, Davis JM, Pandey GN, Pappas GD, Tueting P, Sharma RP, Costa E<br />

(1998) A decrease of reelin expression as a putative vulnerability factor in schizophrenia.<br />

Proc Natl Acad Sci U S A 95: 15718-15723.<br />

Jablensky A (2000) Epidemiology of schizophrenia: the global burden of disease and<br />

disability. Eur Arch Psychiatry Clin Neurosci 250: 274-285.<br />

Jaskiw GE, Karoum F, Freed WJ, Phillips I, Kleinman JE, Weinberger DR (1990a) Effect<br />

of ibotenic acid lesions of the medial prefrontal cortex on amphetamine-induced<br />

locomotion and regional brain catecholamine concentrations in the rat. Brain Res 534:<br />

263-272.<br />

Jaskiw GE, Karoum FK, Weinberger DR (1990b) Persistent elevations in dopamine and<br />

its metabolites in the nucleus accumbens after mild subchronic stress in rats with ibotenic<br />

acid lesions of the medial prefrontal cortex. Brain Res 534: 321-323.<br />

Jentsch JD, Redmond DE, Jr., Elsworth JD, Taylor JR, Youngren KD, Roth RH (1997)<br />

Enduring cognitive deficits and cortical dopamine dysfunction in monkeys after longterm<br />

administration of phencyclidine. Science 277: 953-955.<br />

Jentsch JD, Roth RH (1999) The neuropsychopharmacology of phencyclidine: from<br />

NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia.<br />

Neuropsychopharmacology 20: 201-225.<br />

Jones EG (1997) Cortical development and thalamic pathology in schizophrenia.<br />

Schizophr Bull 23: 483-501.<br />

Jones P, Rodgers B, Murray R, Marmot M (1994) Child development risk factors for<br />

adult schizophrenia in the British 1946 birth cohort. Lancet 344: 1398-1402.<br />

29


Kanemoto K, Tsuji T, Kawasaki J (2001) Reexamination of interictal psychoses based on<br />

DSM IV psychosis classification and international epilepsy classification. Epilepsia 42:<br />

98-103.<br />

Kapur S, Mamo D (2003) Half a century of antipsychotics and still a central role for<br />

dopamine D2 receptors. Prog Neuropsychopharmacol Biol Psychiatry 27: 1081-1090.<br />

Karper LP, Freeman GK, Grillon C, Morgan CA, UI, Charney DS, Krystal JH (1996)<br />

Preliminary evidence of an association between sensorimotor gating and distractibility in<br />

psychosis. J Neuropsychiatry Clin Neurosci 8: 60-66.<br />

Kelly PH, Seviour PW, Iversen SD (1975) Amphetamine and apomorphine responses in<br />

the rat following 6-OHDA lesions of the nucleus accumbens septi and corpus striatum.<br />

Brain Res 94: 507-522.<br />

Kennedy JL, Farrer LA, Andreasen NC, Mayeux R, George-Hyslop P (2003) The<br />

genetics of adult-onset neuropsychiatric disease: complexities and conundra? Science<br />

302: 822-826.<br />

Kilts CD (2001) The changing roles and targets for animal models of schizophrenia. Biol<br />

Psychiatry 50: 845-855.<br />

Kumari V, Sharma T (2002) Effects of typical and atypical antipsychotics on prepulse<br />

inhibition in schizophrenia: a critical evaluation of current evidence and directions for<br />

future research. Psychopharmacology (Berl) 162: 97-101.<br />

Kumari V, Soni W, Sharma T (2002) Prepulse inhibition of the startle response in<br />

risperidone-treated patients: comparison with typical antipsychotics. Schizophr Res 55:<br />

139-146.<br />

Lafleur, I, Marcotte, E. R., Quirion, R., and Srivastava, L. K. Increased adult locomotor<br />

activity following a single neonatal infusion of the NMDA antagonist AP-5 in the rat<br />

prefrontal cortex. Society for Neuroscience Abstracts , 876.10. 2001.<br />

Ref Type: Abstract<br />

Laruelle M, Abi-Dargham A, Gil R, Kegeles L, Innis R (1999) Increased dopamine<br />

transmission in schizophrenia: relationship to illness phases. Biol Psychiatry 46: 56-72.<br />

Laruelle M, Kegeles LS, Abi-Dargham A (2003) Glutamate, dopamine, and<br />

schizophrenia: from pathophysiology to treatment. Ann N Y Acad Sci 1003: 138-158.<br />

Lawrie SM, Abukmeil SS (1998) Brain abnormality in schizophrenia. A systematic and<br />

quantitative review of volumetric magnetic resonance imaging studies. Br J Psychiatry<br />

172: 110-120.<br />

Lawrie SM, Whalley H, Kestelman JN, Abukmeil SS, Byrne M, Hodges A, Rimmington<br />

JE, Best JJ, Owens DG, Johnstone EC (1999) Magnetic resonance imaging of brain in<br />

people at high risk of developing schizophrenia. Lancet 353: 30-33.<br />

30


Le Pen G, Kew J, Alberati D, Borroni E, Heitz MP, Moreau JL (2003) Prepulse inhibition<br />

deficits of the startle reflex in neonatal ventral hippocampal-lesioned rats: reversal <strong>by</strong><br />

glycine and a glycine transporter inhibitor. Biol Psychiatry 54: 1162-1170.<br />

Le Pen G, Moreau JL (2002) Disruption of prepulse inhibition of startle reflex in a<br />

neurodevelopmental model of schizophrenia: reversal <strong>by</strong> clozapine, olanzapine and<br />

risperidone but not <strong>by</strong> haloperidol. Neuropsychopharmacology 27: 1-11.<br />

Lehmann J, Pryce CR, Feldon J (2000) Lack of effect of an early stressful life event on<br />

sensorimotor gating in adult rats. Schizophr Res 41: 365-371. ,<br />

Leumann L, Feldon J, Vollenweider FX, Ludewig K (2002) Effects of typical and<br />

atypical antipsychotics on prepulse inhibition and latent inhibition in chronic<br />

schizophrenia. Biol Psychiatry 52: 729-739.<br />

Lewis DA, Hashimoto T, Volk DW (2005) Cortical inhibitory neurons and<br />

schizophrenia. Nat Rev Neurosci 6: 312-324.<br />

Lewis DA, Levitt P (2002) Schizophrenia as a disorder of neurodevelopment. Annu Rev<br />

Neurosci 25: 409-432.<br />

Lewis R (2004) Should cognitive deficit be a diagnostic criterion for schizophrenia?<br />

Journal of Psychiatry & Neuroscience 29: 102-113.<br />

Lipska BK, Halim ND, Segal PN, Weinberger DR (2002) Effects of reversible<br />

inactivation of the neonatal ventral hippocampus on behavior in the adult rat. J Neurosci<br />

22: 2835-2842.<br />

Lipska BK, Jaskiw GE, Chrapusta S, Karoum F, Weinberger DR (1992) Ibotenic acid<br />

lesion of the ventral hippocampus differentially affects dopamine and its metabolites in<br />

the nucleus accumbens and prefrontal cortex in the rat. Brain Res 585: 1-6.<br />

Lipska BK, Jaskiw GE, Karoum F, Phillips I, Kleinman JE, Weinberger DR (1991)<br />

Dorsal hippocampal lesion does not affect dopaminergic indices in the basal ganglia.<br />

Pharmacol Biochem Behav 40: 181-184.<br />

Lipska BK, Jaskiw GE, Weinberger DR (1993) Postpubertal emergence of<br />

hyperresponsiveness to stress and to amphetamine after neonatal excitotoxic hippocampal<br />

damage: a potential animal model of schizophrenia. Neuropsychopharmacology 9: 67-75.<br />

Lipska BK, Luu S, Halim ND, Weinberger DR (2003) Behavioral effects of neonatal and<br />

adult excitotoxic lesions of the mediodorsal thalamus in the adult rat. Behav Brain Res<br />

141: 105-111.<br />

Lipska BK, Swerdlow NR, Geyer MA, Jaskiw GE, Braff DL, Weinberger DR (1995)<br />

Neonatal excitotoxic hippocampal damage in rats causes post-pubertal changes in<br />

prepulse inhibition of startle and its disruption <strong>by</strong> apomorphine. Psychopharmacology<br />

(Berl) 122: 35-43.<br />

31


Lipska BK, Weinberger DR (1993) Delayed effects of neonatal hippocampal damage on<br />

haloperidol-induced catalepsy and apomorphine-induced stereotypic behaviors in the rat.<br />

Brain Res Dev Brain Res 75: 213-222.<br />

Lipska BK, Weinberger DR (1995) Genetic variation in vulnerability to the behavioral<br />

effects of neonatal hippocampal damage in rats. Proc Natl Acad Sci U S A 92: 8906-<br />

8910.<br />

Lipska BK, Weinberger DR (2000) To model a psychiatric disorder in animals:<br />

schizophrenia as a reality test. Neuropsychopharmacology 23: 223-239.<br />

Lomber SG (1999) The advantages and limitations of permanent or reversible<br />

deactivation techniques in the assessment of neural function. Journal of Neuroscience<br />

Methods 86: 109-117.<br />

Ludewig K, Geyer MA, Vollenweider FX (2003) Deficits in prepulse inhibition and<br />

habituation in never-medicated, first-episode schizophrenia. Biol Psychiatry 54: 121-128.<br />

Luthi A, Schwyzer L, Mateos JM, Gahwiler BH, McKinney RA (2001) NMDA receptor<br />

activation limits the number of synaptic connections during hippocampal development.<br />

Nat Neurosci 4: 1102-1107.<br />

Mackeprang T, Kristiansen KT, Glenthoj BY (2002) Effects of antipsychotics on<br />

prepulse inhibition of the startle response in drug-naive schizophrenic patients. Biol<br />

Psychiatry 52: 863-873.<br />

Marcotte ER, Pearson DM, Srivastava LK (2001) Animal models of schizophrenia: a<br />

critical review. J Psychiatry Neurosci 26: 395-410.<br />

McCarley RW, Wible CG, Frumin M, Hirayasu Y, Levitt JJ, Fischer IA, Shenton ME<br />

(1999) MRI anatomy of schizophrenia. Biol Psychiatry 45: 1099-1119.<br />

McClure RK, Weinberger DR (2001) The neurodevelopmental hypothesis of<br />

schizophrenia: A review of the evidence. In: Current Issues in the Psychopharmacology<br />

of Schizophrenia. (Breier A, Tran PV, Herrea JM, Tollefson GD, Bymaster FP, eds), pp<br />

27-56. New York: Lippincott Williams & Wilkins.<br />

McGowan S, Lawrence AD, Sales T, Quested D, Gras<strong>by</strong> P (2004) Presynaptic<br />

dopaminergic dysfunction in schizophrenia: a positron emission tomographic<br />

[18F]fluorodopa study. Arch Gen Psychiatry 61: 134-142.<br />

Meincke U, Morth D, Voss T, Thelen B, Geyer MA, Gouzoulis-Mayfrank E (2004)<br />

Prepulse inhibition of the acoustically evoked startle reflex in patients with an acute<br />

schizophrenic psychosis-a longitudinal study. Eur Arch Psychiatry Clin Neurosci 254:<br />

415-421.<br />

Meltzer HY, Lee MA, Ranjan R (1994) Recent advances in the pharmacotherapy of<br />

schizophrenia. Acta Psychiatr Scand Suppl 384: 95-101.<br />

32


Meyer-Lindenberg A, Miletich RS, Kohn PD, Esposito G, Carson RE, Quarantelli M,<br />

Weinberger DR, Berman KF (2002) Reduced prefrontal activity predicts exaggerated<br />

striatal dopaminergic function in schizophrenia. Nat Neurosci 5: 267-271.<br />

Mitdeman G, Bratt AM, Chase R (1998) Heterogeneity of the hippocampus: effects of<br />

subfield lesions on locomotion elicited <strong>by</strong> dopaminergic agonists. Behav Brain Res 92:<br />

31-45.<br />

Mohn AR, Gainetdinov RR, Caron MG, Roller BH (1999) Mice with reduced NMDA<br />

receptor expression display behaviors related to schizophrenia. Cell 98: 427-436.<br />

Morimoto T, Kida K, Nagao H, Yoshida K, Fukuda M, Takashima S (1995) The<br />

pathogenic role of the NMDA receptor in hyperthermia-induced seizures in developing<br />

rats. Brain Res Dev Brain Res 84: 204-207.<br />

Nelson MD, Saykin AJ, Flashman LA, Riordan HJ (1998) Hippocampal volume<br />

reduction in schizophrenia as assessed <strong>by</strong> magnetic resonance imaging: a meta-analytic<br />

study. Arch Gen Psychiatry 55: 433-440.<br />

Noga JT, Bartley AJ, Jones DW, Torrey EF, Weinberger DR (1996) Cortical gyral<br />

anatomy and gross brain dimensions in monozygotic twins discordant for schizophrenia.<br />

Schizophr Res 22: 27-40.<br />

Olney JW (2004) Fetal alcohol syndrome at the cellular level. Addict Biol 9: 137-149.<br />

Olney JW, Newcomer JW, Farber NB (1999) NMDA receptor hypofunction model of<br />

schizophrenia. J Psychiatr Res 33: 523-533.<br />

Owen MJ, Williams NM, O'Donovan MC (2004) The molecular genetics of<br />

schizophrenia: new findings promise new insights. Mol Psychiatry 9: 14-27.<br />

Pakkenberg B (1990) Pronounced reduction of total neuron number in mediodorsal<br />

thalamic nucleus and nucleus accumbens in schizophrenics. Arch Gen Psychiatry 47:<br />

1023-1028.<br />

Perry W, Braff DL (1994) Information-processing deficits and thought disorder in<br />

schizophrenia. Am J Psychiatry 151: 363-367.<br />

Perry W, Geyer MA, Braff DL (1999) Sensorimotor gating and thought disturbance<br />

measured in close temporal proximity in schizophrenic patients. Arch Gen Psychiatry 56:<br />

277-281.<br />

Porrino LJ, Lucignani G, Dow-Edwards D, Sokoloff L (1984) Correlation of dosedependent<br />

effects of acute amphetamine administration on behavior and local cerebral<br />

metabolism in rats. Brain Res 307: 311-320.<br />

33


Pouzet B, Feldon J, Veenman CL, Yee BK, Richmond M, Nicholas J, Rawlins P, Weiner<br />

I (1999) The effects of hippocampal and fimbria-fornix lesions on prepulse inhibition.<br />

Behav Neurosci 113: 968-981.<br />

Rabacchi S, Bailly Y, Delhaye-Bouchaud N, Mariani J (1992) Involvement of the Nmethyl<br />

D-aspartate (NMDA) receptor in synapse elimination during cerebellar<br />

development. Science 256: 1823-1825.<br />

Rapoport JL, Addington AM, Frangou S, Psych M (2005) The neurodevelopmental<br />

model of schizophrenia: update 2005. Molecular Psychiatry 10: 434-449.<br />

Rapoport JL, Giedd JN, Blumenthal J, Hamburger S, Jeffries N, Fernandez T, Nicolson<br />

R, Bedwell J, Lenane M, Zijdenbos A, Paus T, Evans A (1999) Progressive cortical<br />

change during adolescence in childhood-onset schizophrenia. A longitudinal magnetic<br />

resonance imaging study. Arch Gen Psychiatry 56: 649-654.<br />

Robbins TW (2004) Animal models of the psychoses. In: Neurobiology of Mental Illness,<br />

2nd. Ed. (Charney DS, Nestler EJ, eds), pp 263-286. New York: Oxford University Press.<br />

Roberts GW, Colter N, Lofthouse R, Bogerts B, Zech M, Crow TJ (1986) Gliosis in<br />

schizophrenia: a survey. Biol Psychiatry 21: 1043-1050.<br />

Roth BL, Hanizavareh SM, Blum AE (2004) Serotonin receptors represent highly<br />

favorable molecular targets for cognitive enhancement in schizophrenia and other<br />

disorders. Psychopharmacology 174: 17-24.<br />

Sams-Dodd F, Lipska BK, Weinberger DR (1997) Neonatal lesions of the rat ventral<br />

hippocampus result in hyperlocomotion and deficits in social behaviour in adulthood.<br />

Psychopharmacology (Berl) 132: 303-310.<br />

Schneider M, Koch M (2005) Behavioral and morphological alterations following<br />

neonatal excitotoxic lesions of the medial prefrontal cortex in rats. Exp Neurol.<br />

Schoenfeld TA, Hamilton LW (1977) Secondary Brain Changes Following Lesions -<br />

New Paradigm for Lesion Experimentation. Physiology & Behavior 18: 951-967.<br />

Seidman LJ, Faraone SV, Goldstein JM, Kremen WS, Horton NJ, Makris N, Toomey R,<br />

Kennedy D, Caviness VS, Tsuang MT (2002) Left hippocampal volume as a<br />

vulnerability indicator for schizophrenia: a magnetic resonance imaging morphometric<br />

study of nonpsychotic first-degree relatives. Arch Gen Psychiatry 59: 839-849.<br />

Sharma T, Antonova L (2003) Cognitive function in schizophrenia - Deficits, functional<br />

consequences, and future treatment. Psychiatric Clinics of North America 26: 25-+.<br />

Snyder SH (1973) Amphetamine psychosis: a "model" schizophrenia mediated <strong>by</strong><br />

catecholamines. Am J Psychiatry 130: 61-67.<br />

34


Stein DG (1979) The Ghost in the Machine Is Still There. Behavioral and Brain Sciences<br />

2: 346-348.<br />

Stevens JR (1997) Anatomy of schizophrenia revisited. Schizophr Bull 23: 373-383.<br />

Suddarh RL, Christison GW, Torrey EF, Casanova MF, Weinberger DR (1990)<br />

Anatomical abnormalities in the brains of monozygotic twins discordant for<br />

schizophrenia. N Engl J Med 322: 789-794.<br />

Swerdlow NR, Braff DL, Geyer MA (2000a) Animal models of deficient sensorimotor<br />

gating: what we know, what we think we know, and what we hope to know soon. Behav<br />

Pharmacol 11: 185-204.<br />

Swerdlow NR, Geyer MA, Braff DL (2001) Neural circuit regulation of prepulse<br />

inhibition of startle in the rat: current knowledge and future challenges.<br />

Psychopharmacology (Berl) 156: 194-215.<br />

Swerdlow NR, Lipska BK, Weinberger DR, Braff DL, Jaskiw GE, Geyer MA (1995)<br />

Increased sensitivity to the sensorimotor gating-disruptive effects of apomorphine after<br />

lesions of medial prefrontal cortex or ventral hippocampus in adult rats.<br />

Psychopharmacology (Berl) 122: 27-34.<br />

Swerdlow NR, Taaid N, Halim N, Randolph E, Kim YK, Auerbach P (2000b)<br />

Hippocampal lesions enhance startle gating-disruptive effects of apomorphine in rats: a<br />

parametric assessment. Neuroscience 96: 523-536.<br />

Tenn CC, Kapur S, Fletcher PJ (2005) Sensitization to amphetamine, but not<br />

phencyclidine, disrupts prepulse inhibition and latent inhibition. Psychopharmacology<br />

(Berl) 180: 366-376.<br />

Vallano ML (1998) Developmental aspects of NMDA receptor function. Crit Rev<br />

Neurobiol 12: 177-204.<br />

Van den BM, Garner B, Koch M (2003) Neurodevelopmental animal models of<br />

schizophrenia: effects on prepulse inhibition. Curr Mol Med 3: 459-471.<br />

Varty GB, Braff DL, Geyer MA (1999) Is there a critical developmental 'window' for<br />

isolation rearing-induced changes in prepulse inhibition of the acoustic startle response?<br />

Behav Brain Res 100: 177-183.<br />

Varty GB, Higgins GA (1995) Examination of drug-induced and isolation-induced<br />

disruptions of prepulse inhibition as models to screen antipsychotic drugs.<br />

Psychopharmacology (Berl) 122: 15-26.<br />

Vestergaard M, Pedersen CB, Christensen J, Madsen KM, Olsen J, Mortensen PB (2005)<br />

Febrile seizures and risk of schizophrenia. Schizophr Res 73: 343-349.<br />

35


Walker EF (1994) Developmentally moderated expressions of the neuropathology<br />

underlying schizophrenia. Schizophr Bull 20: 453-480.<br />

Walker EF, Savoie T, Davis D (1994) Neuromotor precursors of schizophrenia.<br />

Schizophr Bull 20: 441-451.<br />

Wang C, Mclnnis J, Ross-Sanchez M, Shinnick-Gallagher P, Wiley JL, Johnson KM<br />

(2001) Long-term behavioral and neurodegenerative effects of perinatal phencyclidine<br />

administration: Implications for schizophrenia. Neuroscience 107: 535-550.<br />

Weinberger DR (1987) Implications of normal brain development for the pathogenesis of<br />

schizophrenia. Arch Gen Psychiatry 44: 660-669.<br />

Weinberger DR (1996) On the plausibility of "the neurodevelopmental hypothesis" of<br />

schizophrenia. Neuropsychopharmacology 14: 1S-11S.<br />

Weiss IC, Feldon J (2001) Environmental animal models for sensorimotor gating<br />

deficiencies in schizophrenia: a review. Psychopharmacology (Berl) 156: 305-326.<br />

Wong AH, Van Tol HH (2003) Schizophrenia: from phenomenology to neurobiology.<br />

Neurosci Biobehav Rev 27: 269-306.<br />

Wood GK, Lipska BK, Weinberger DR (1997) Behavioral changes in rats with early<br />

ventral hippocampal damage vary with age at damage. Brain Res Dev Brain Res 101: 17-<br />

25.<br />

Wright IC, Ellison ZR, Sharma T, Friston KJ, Murray RM, McGuire PK (1999) Mapping<br />

of grey matter changes in schizophrenia. Schizophr Res 35: 1-14.<br />

Wright IC, Rabe-Hesketh S, Woodruff PWR, David AS, Murray RM, Bullmore ET<br />

(2000) Meta-analysis of regional brain volumes in schizophrenia. American Journal of<br />

Psychiatry 157: 16-25.<br />

Wynn JK, Sergi MJ, Dawson ME, Schell AM, Green MF (2005) Sensorimotor gating,<br />

orienting and social perception in schizophrenia. Schizophr Res 73: 319-325.<br />

Zhang WN, Bast T, Feldon J (2002) Prepulse inhibition in rats with temporary<br />

inhibition/inactivation of ventral or dorsal hippocampus. Pharmacol Biochem Behav 73:<br />

929-940.<br />

Zornberg GL, Buka SL, Tsuang MT (2000) Hypoxic-ischemia-related fetal/neonatal<br />

complications and risk of schizophrenia and other nonaffective psychoses: A 19-year<br />

longitudinal study. American Journal of Psychiatry 157: 196-202.<br />

36


CHAPTER TWO: ELECTRICAL STIMULATION OF <strong>THE</strong> HIPPOCAMPUS<br />

DISRUPTS PREPULSE INHIBITION IN <strong>RAT</strong>S: FREQUENCY AND SITE<br />

DEPENDENT EFFECTS. 1<br />

Introduction<br />

Prepulse inhibition (PPI) is the normal reduction in an acoustic startle response<br />

produced when a brief, quieter stimulus or prepulse is presented 30-500 ms prior to<br />

presentation of the starde-evoking stimulus (Koch, 1999;Swerdlow et al., 2000a). PPI<br />

provides a measure of sensorimotor gating, the inhibitory process <strong>by</strong> which responses to<br />

sensory stimuli are suppressed, there<strong>by</strong> facilitating appropriate responding in complex,<br />

sensory rich environments (Swerdlow, 1996). Interest in the neural mechanisms<br />

underlying PPI has increased because it is often disrupted in humans diagnosed with a<br />

variety of psychiatric disorders including schizophrenia (Weike et al., 2000;Braff et al.,<br />

2001). As similar stimuli elicit PPI in rodents and humans, interventions in rodents that<br />

disrupt PPI may reveal aspects of neuronal dysfunction that underlie disrupted<br />

sensorimotor gating in schizophrenic patients (Swerdlow, 1996;Weike et al., 2000).<br />

Similarities exist in the neuroanatomical and neurochemical substrates related to<br />

both schizophrenia and PPI. A large body of research suggests that schizophrenic<br />

patients show abnormalities in cortico-limbic-striatal circuitry, in areas such as the frontal<br />

cortex, nucleus accumbens (NAc), and hippocampus (Pakkenberg, 1990;Bogerts,<br />

1997;Harrison, 1999;Wright et al., 1999) likely involving alterations in dopamine (DA)<br />

and glutamate (Glu) transmission (Breier et al., 1997;Carlsson et al., 1999;Laruelle et al.,<br />

1999;Goff and Coyle, 2001;Meyer-Lindenberg et al., 2002). Consistent with these<br />

1<br />

A version of the chapter has been published: Howland JG, Mackenzie EM, Yim TT, Taepavarapruk P,<br />

Phillips AG (2004). Electrical stimulation of the hippocampus disrupts prepulse inhibition in rats:<br />

frequency and site dependent effects. Behavioural Brain Research 152:187-197.<br />

37


findings is evidence indicating that the regulation of PPI is also dependent on a complex<br />

array of structures commonly termed the cortico-striatal-pallidal-pontine (CSPP) circuitry<br />

(see Swerdlow et al., 2001a for review), including the medial prefrontal cortex (mPFC)<br />

(Koch and Bubser, 1994;Lacroix et al., 2000), amygdala (Wan and Swerdlow,<br />

1997;Fendt et al., 2000), NAc (Swerdlow et al.,1986;1990;1994;Wan and Swerdlow,<br />

1996), and hippocampus (Caine et al., 1992;Swerdlow et al., 1995;2001b;Wan et al.,<br />

1996;Bakshi and Geyer, 1998;Klarner et al, 1998;Zhang et al., 1999;2002a;Bast et al.,<br />

2001;Caine et al., 2001). Importantly, the systemic administration of DA or DA agonists<br />

(Mansbach et al, 1988;Swerdlow et al., 1990;Zhang et al., 2000a) and N-Methyl-D-<br />

Aspartate (NMDA) antagonists (Mansbach and Geyer, 1989;Bakshi and Geyer, 1998),<br />

likely acting in areas of the CSPP circuitry, also disrupt PPI in animals (Geyer et al.,<br />

2001). Recently, a complex role of NAc DA and Glu via D2, NMDA, and (±)-a-amino-<br />

3-hydroxy-5-methylisoxazole-4-propionic acid hydrobromide (AMPA) receptors in the<br />

modulation of PPI has been described (Mansbach et al., 1988;Swerdlow et al, 1990;Wan<br />

and Swerdlow, 1993;1996;Reijmers et al., 1995;Wan et al., 1995).<br />

Hippocampal dysfunction has been linked to symptoms of schizophrenia<br />

(Silbersweig et al., 1995;Heckers et al., 1998;Harrison, 1999;Dierks et al., 1999;Grace,<br />

2000), and consequently the roles of distinct hippocampal subregions in the modulation<br />

of PPI have been examined extensively. Although permanent lesions of the ventral<br />

hippocampus (vHip) generally do not disrupt PPI (Swerdlow et al., 1995;2000b;Pouzet et<br />

al., 1999 but see also (Caine et al., 2001), temporary inactivation of vHip or dorsal<br />

hippocampal (dHip) with either the GABA A agonist muscimol or the sodium channel<br />

blocker tetrodotoxin disrupts PPI (Zhang et al., 2002b). Debate also exists as to whether<br />

38


infusion of glutamatergic antagonists, such as MK801, into the dHip, but not the vHip,<br />

disrupts PPI (Bakshi and Geyer, 1998;1999;Bast et al., 2000;Zhang et al., 2000b).<br />

Nevertheless, compelling evidence exists for the disruption of PPI following chemical<br />

stimulation of the vHip (Wan et al., 1996;Klarner et al., 1998;Zhang et al., 1999;Bast et<br />

al., 2001;Swerdlow et al., 2001b;Zhang et al., 2002a), but not dHip (Swerdlow et al.,<br />

2001b;Zhang et al., 2002a) with infusions of the ionotropic Glu agonist NMDA. As<br />

these studies suggest, various manipulations of the vHip and dHip can impair<br />

sensorimotor gating behavior, and regional differences clearly exist.<br />

Both the vHip and dHip are well positioned neuroanatomically to interact with the<br />

CSPP circuitry to modulate PPI. The vHip projects densely to the NAc shell, amygdala,<br />

and mPFC (Kelley and Domesick, 1982;Groenewegen et al., 1987;Brog et al.,<br />

1993;Conde et al., 1995) whereas the dHip projects to the NAc core (Swanson and<br />

Cowan, 1977;Whitter, 1986), and also communicates with the vHip via intra-<br />

hippocampal connections (Amaral and Whitter, 1995). At the level of the ventral<br />

striatum, projections from the vHip exert powerful control over DA efflux in the NAc<br />

shell and firing of NAc medium spiny projection neurons (O'Donnell and Grace,<br />

1995;Taepavarapruk et al., 2000;Floresco et al., 2001). More specifically, stimulation of<br />

the vHip with NMDA (Brudzynski and Gibson, 1997;Legault et al., 2000) or brief trains<br />

of higher frequency electrical current (20 Hz, 10 s) (Blaha et al., 1997;Taepavarapruk et<br />

al., 2000) cause a long-lasting increase in NAc DA efflux and an increase in locomotor<br />

activity. Activation of ionotropic Glu receptors (iGluR's) in the NAc underlies the<br />

changes in NAc DA efflux observed following electrical stimulation of the vHip (Blaha<br />

et al., 1997;Taepavarapruk et al., 2000). In contrast, preliminary studies performed in our<br />

39


laboratory demonstrate that lower frequency (2 Hz) stimulation of the vHip for 100 s can<br />

significantly reduce NAc DA efflux for at least 2 hr, an effect that may result from the<br />

activation of intra-NAc group 2 and 3 metabotropic Glu receptors (Taepavarapruk et al.,<br />

1998). The consequences of stimulating the dHip on NAc DA efflux have been poorly<br />

characterized, although stimulation of the previously kindled dHip results in a brief<br />

increase in NAc DA efflux (Strecker and Moneta, 1994).<br />

In the following experiments, the effects of electrical stimulation of the Hip on<br />

PPI were examined. Electrical stimulation was chosen because it has a number of<br />

distinct advantages over chemical stimulation, including precise control over the duration<br />

and intensity of the stimulation. Additionally, electrical stimulation parameters can<br />

approximate the physiological firing activity of the neurons that are stimulated. Given<br />

that intra-vHip NMDA infusions and 20 Hz electrical stimulation of the vHip produce<br />

similar increases in NAc DA efflux (Brudzynski and Gibson, 1997;Legault et al.,<br />

2000;Taepavarapruk et al., 2000), and that vHip NMDA infusions disrupt PPI (Wan et<br />

al., 1996;Klarner et al, 1998;Zhang et al, 1999;2002a;Bast et al., 2001;Swerdlow et al.,<br />

2001b), the first objective of the present study was to assess whether 20 Hz stimulation of<br />

the vHip would induce a reversible disruption of PPI. In a complementary experiment,<br />

we examined the effect of 20 Hz electrical stimulation to the dHip on PPI, hypothesizing<br />

that if dHip NMDA infusions fail to disrupt PPI (Swerdlow et al., 2001b;Zhang et al,<br />

2002a), then electrical stimulation at this frequency should also fail to reduce PPI. The<br />

final behavioral experiment examined the effect of 2 Hz vHip stimulation on PPI.<br />

Additional microdialysis experiments addressed two issues related to the effects of 20 Hz<br />

stimulation of the hippocampus. The first assessed whether unilateral vHip stimulation<br />

40


would result in an increase in DA efflux bilaterally in the NAc. Secondly, as the effects<br />

of 20 Hz dHip stimulation on NAc DA are unknown, a similar microdialysis experiment<br />

assessed the effects of these stimulation parameters on NAc DA efflux. Male Long-<br />

Evans (LE) rats were used in all experiments in an effort to maintain consistency between<br />

the neurochemical data collected previously and the present behavioral experiments.<br />

Methods<br />

Subjects<br />

Male LE rats (Charles River Canada, St. Constant, Quebec, Canada) were pah-<br />

housed in Plexiglas cages and handled daily until surgery. The colony was kept on a<br />

12/12 hour light/dark cycle (lights on at 0700), at a temperature of 22±1°C. Rats were<br />

given food (Purina Rat Chow) and tap water ad libitum. Experiments were conducted in<br />

accordance with the standards of the Canadian Council on Animal Care and the<br />

Committee on Animal Care at the University of British Columbia approved all<br />

procedures.<br />

Surgery<br />

At the time of surgery, subjects weighed 330 to 370 g. Rats were anaesthetized<br />

with ketamine hydrochloride (100 mg/kg, i.p., MTC Pharmaceuticals) and xylazine (10<br />

mg/kg, i.p., Rompun), and placed in a stereotaxic frame. The dorsal surface of the skull<br />

was exposed, and holes were drilled. A bipolar stimulating electrode (Plastics One,<br />

Roanoke, VA) was implanted into either the vHip (AP -5.8 mm from bregma, ML ±5.5<br />

mm from midline, DV -6.0 mm from dura) or dHip (AP -3.0 mm, ML ±1.5 mm, DV -<br />

3.0 mm) of all rats. Guide cannulae were also implanted dorsal to the NAc in those rats<br />

used in the microdialysis experiments (AP +1.7 mm, ML ±1.1 mm, DV -1.0 mm).


Cannulae and the electrode were secured to the skull with four jeweler's screws and<br />

dental acrylic. Wire obdurators were inserted into the cannula to keep them patent.<br />

Animals were allowed at least 7 days to recover from surgery before testing began, and<br />

were regularly handled beginning three days following surgery.<br />

Prepulse Inhibition Testing<br />

Testing was conducted in a single sound-attenuating startle chamber (ambient<br />

noise level 62 dB), containing a transparent Plexiglas tube (8.2 cm in diameter, 20 cm in<br />

length), mounted on a Plexiglas frame (SR-LAB, San Diego Instruments, San Diego,<br />

CA). Noise bursts were presented through a speaker mounted 24 cm above the tube. An<br />

accelerometer below the frame of the apparatus measured whole body startle amplitude,<br />

defined as the average of 100 1 ms accelerometer readings collected beginning at<br />

stimulus onset. Each PPI test session began with a 5 min acclimatization period during<br />

which a 70 dB background noise level was presented, which remained constant for the<br />

entire test session. Following the acclimatization period, six pulse alone trials (120 dB,<br />

40 ms) were presented to achieve a relatively stable level of startle amplitude before<br />

presentation of the prepulse + pulse trials. The data from these pulse alone trials was not<br />

considered in the analysis of PPI. Immediately following the six initial pulse alone trials,<br />

presentation of the trials that were used in the calculation of PPI levels began. A total of<br />

84 trials of five different types were presented. Trials presented were of three types:<br />

pulse alone (12 trials; 120 dB, 40 ms), prepulse + pulse (10 trials X 3 prepulse intensities<br />

- discussed below), or no stimulus (42 trials). Prepulse + pulse trials consisted of the<br />

presentation of a 20 ms prepulse of 73, 76, or 82 dB 80 ms before the presentation of the<br />

pulse. The pulse and prepulse + pulse trials were presented in a pseudorandom order.<br />

42


One no stimulus trial was presented between each pair of pulse and prepulse + pulse<br />

trials. The inter-trial interval varied randomly from 3 to 12 s (average 7.5 s). The total<br />

length of each PPI session was approximately 18 min. Calibration of the apparatus was<br />

performed using a RadioShack Digital Sound Level Meter and adjustments were made as<br />

necessary.<br />

A within-subjects design was used in all PPI experiments. Experiments were<br />

conducted in two successive weeks; all animals were tested four times during these two<br />

weeks. The four PPI sessions were referred to as follows. The 'stim' PPI session<br />

occurred 2 min following electrical stimulation of either the vHip or dHip, 'stim+48' was<br />

the session 48 hr following stimulation, 'control' was the PPI session 2 min following<br />

exposure to the stimulation box without stimulation, and 'control+48' followed the<br />

'control' session <strong>by</strong> 48 hrs. Animals were randomly assigned to one of two sequences to<br />

control for potential order effects. Rats in sequence one received their PPI sessions in the<br />

following order: stim, stim+48, control, and control+48. Rats in sequence two received<br />

their PPI sessions in the control, control+48, stim, and stim+48 order.<br />

Following recovery from surgery, animals were habituated to the stimulation<br />

environment on the two days immediately preceding their first PPI test. Each rat was<br />

removed from the colony and taken to the stimulation room. This small room was<br />

immediately adjacent to the room used for PPI testing, and contained two transparent<br />

Plexiglas chambers (32 cm X 32 cm X 41 cm) used for stimulation. In the stimulation<br />

room, each rat was connected to a stimulation lead and placed in a Plexiglas chamber for<br />

10 min, following which they were returned to the colony. On the day of their first PPI<br />

test, the animals were treated exactly as on the two previous days except that after 10 min<br />

43


in the stimulation box, electrical stimulation (20 Hz: 200 cathodal, constant current<br />

pulses, 300 uA, 20 Hz, pulse width 0.5 ms; 2 Hz: identical except frequency of pulses<br />

was 2 Hz) was applied to half the rats, while the other half were given no current.<br />

Stimulation was delivered through an isolator (Iso-flex, A.M.P.I, Israel) via a Master-8<br />

stimulator (A.M.P.I.). The experimenter observed the animals for 90 sec following<br />

stimulation, disconnected them, and quickly placed them in the PPI chamber. This<br />

procedure took a total of 2 min. Immediately following the PPI session, the animals were<br />

returned to the colony. All rats were then tested for PPI 48 hours later. For this test, the<br />

rats were removed from the colony and immediately placed in the PPI apparatus.<br />

Following the second PPI session, the animals were handled daily for five days. On the<br />

sixth day following the first PPI test session, all rats were given one habituation session<br />

in the stimulation chamber for 10 min. The following day, the PPI testing procedure<br />

described above was repeated, with control animals receiving electrical stimulation and<br />

previously stimulated animals receiving no current. Forty-eight hours later, all rats<br />

received their last PPI session.<br />

Neurochemical Experiments<br />

The microdialysis procedure commonly employed in our laboratory has been<br />

described in detail elsewhere (Taepavarapruk et al, 2000;Howland et al., 2002). Briefly,<br />

concentric-style microdialysis probes (2 mm of exposed membrane) were constructed in<br />

our laboratory. Probes were attached to gas-tight syringes via a liquid swivel containing<br />

perfusion medium (147 mM NaCl, 3.0 mM KC1,1.3 mM CaCl 2.H 20, 1.0 mM<br />

MgCl 2.6H 20, 0.01 sodium phosphate buffer; pH 7.3-7.4) and flushed for 10 to 20 min<br />

using a syringe pump. Probes were then secured in a copper collar and inserted into the<br />

44


NAc (7.8 mm ventral to dura) and the rats were allowed to move freely in a Plexiglas box<br />

(32 cm x 32 cm x 41 cm high) with access to food and water for 12 to 18 hours before<br />

experimental testing began the following morning. The probes were continuously<br />

perfused at 1 pL/min overnight. Probes were inserted bilaterally for the vHip stimulation<br />

experiment and unilaterally ipsilateral to the electrode for the dHip stimulation<br />

experiments.<br />

Two high-performance liquid chromatography systems with electrochemical<br />

detection (HPLC-ED) consisting of an ESA 582 pump (ESA Inc., Bedford, MA),<br />

Rheodyne Inert manual injector (Rheodyne, Rohnert Park, CA), an Ultrasphere column<br />

(Beckmann, Fullerton, CA.; ODS 5 pm, 15 cm x 4.6 mm), an ESA 5011 analytical cell,<br />

and a Coulochem II EC detector (ESA Inc.) were used to quantify DA levels in all<br />

experiments. The working potentials were: +450 mV (electrode 1), -300 mV (electrode<br />

2), and +450 mV (guard cell). The mobile phase (pH 3.5) consisted of 6 g/L sodium<br />

acetate, 10 mg/L ethylenediaminetetra-acetic acid (EDTA), 150 mg/L octyl sulfate<br />

(adjustable), 35 ml/L glacial acetic acid and 865 mL Milli Q purified water.<br />

Chromatograms were registered on a dual-pen chart recorder (Kipp and Zonen, Bohemia,<br />

NY). All samples were injected immediately after collection and DA peak heights were<br />

measured manually.<br />

Dialysate samples were collected at 10 min intervals throughout the experiment.<br />

Once four baseline samples were collected that did not differ <strong>by</strong> more than ± 10%, the<br />

rats received 20 Hz electrical stimulation of either the vHip or dHip (same parameters<br />

described for the PPI experiments). Stimulation was timed to ensure that the next<br />

dialysis sample reflected only "stimulation-evoked" changes in DA efflux. The<br />

45


experimenter remained in the testing room following the stimulation to record any<br />

behavioral effects of the stimulation. Nine samples were collected following stimulation,<br />

after which the animals were disconnected and sacrificed.<br />

Histology<br />

Upon completion of each experiment, animals were sacrificed with an overdose of<br />

pentobarbital, and perfused transcardially with 0.9% saline followed <strong>by</strong> 10%<br />

formaldehyde. Brains were stored in 10% sucrose in 10% formaldehyde for at least 1<br />

week, after which they were sectioned (50 pm) using a cryostat and stained with cresyl<br />

violet. Placements of the electrodes and probes were confirmed under a light microscope<br />

with the assistance of a rat brain atlas (Paxinos and Watson, 1997).<br />

Data Analysis<br />

Data were analysed using repeated measures analyses of variance with the aid of<br />

SPSS (version 10.0). All post-hoc tests were performed separately. Variance is indicated<br />

on all graphs with the standard error of the mean (SEM). Significance levels for all<br />

statistical tests were set at 0.05.<br />

For the PPI experiments, two measures were calculated for each animal. The<br />

startle amplitude represented the mean startle amplitude of the 12 pulse alone trials<br />

presented after the six habituation trials. For calculation of PPI, startle amplitudes were<br />

averaged for each trial type. The percent PPI for each prepulse intensity was calculated<br />

using the formula: [100 - (100 X startle amplitude on prepulse + pulse trials) (startle<br />

amplitude on pulse alone trials)]. Two-way (prepulse intensity, test session as factors)<br />

repeated measures ANOVAs were performed on the data obtained from each stimulation<br />

site. As the prepulse intensity factor did not significantly interact with any of the other<br />

46


factors, percent PPI data was averaged across the three prepulse intensities for each rat,<br />

there<strong>by</strong> creating a global PPI score (Wan et al., 1995;Bakshi and Geyer, 1998;1999).<br />

Both startle amplitude and percent PPI were then analyzed using separate one-way<br />

ANOVAs with condition as a repeated measures factor. The repeated measures<br />

assumption of sphericity was met in all cases. Post-hoc analyses were performed using<br />

the Neuman-Keul's test where appropriate.<br />

Following the microdialysis experiments, baseline DA levels for each rat were<br />

calculated <strong>by</strong> averaging the four baseline samples. In all figures, DA levels are expressed<br />

as a percentage change from baseline (baseline is expressed as 0%). One-way repeated<br />

measures analyses of variance with time as a within-subjects factor were performed on all<br />

data and Dunnett's post-hoc tests were performed where appropriate. The baseline<br />

sample taken immediately before the first stimulation was used as the critical value<br />

during computation of the Dunnett's post-hoc tests.<br />

Results<br />

Immediate Behavioral Effects of Stimulation<br />

During the 2 min that the rats remained in the stimulation apparatus after<br />

stimulation was applied, a variety of behavioral effects were noted. The rats were<br />

generally awake and alert before administration of stimulation. At the onset of<br />

stimulation, behavioral arrest was observed in all animals. Higher frequency (20 Hz)<br />

stimulation of the vHip resulted in 5 to 20 wet dog shakes (WDS) that were restricted to<br />

45 s following initiation of the stimulation in 20 of the 21 animals (rats used in the<br />

dialysis studies are included in this description). Additionally, all rats engaged in<br />

vigorous forward locomotor behavior following cessation of the 20 Hz stimulation, as has<br />

47


een previously quantified (Taepavarapruk et al., 2000). Stimulation of the dHip resulted<br />

in an initial period of WDS activity in 4 of the 28 animals. All rats displayed a period of<br />

increased activity and rearing within 30 sec of the cessation of stimulation. Between 70<br />

and 100 s following the cessation of stimulation, 18 of the 28 rats had a second bout of<br />

WDS activity. One to 20 WDS were displayed during this period. Two Hz stimulation<br />

of the vHip caused an initial period of behavioral arrest followed <strong>by</strong> behavioral activation<br />

manifested <strong>by</strong> grooming and some rearing. Wet dog shakes were observed in 3 of 15<br />

animals during application of the 2 Hz stimulation.<br />

Differential Effects of 20 Hz Stimulation of the vHip or dHip on PPI and Startle<br />

Amplitude<br />

Twenty Hz stimulation of the vHip (n=14) resulted in a significant and reversible<br />

disruption of PPI (Fig. 2-1 A). A one-way repeated measures ANOVA performed on the<br />

PPI data for the vHip stimulation group revealed a significant main effect for the test<br />

condition factor (F(3, 39) = 9.32, p< 0.001). Post-hoc analyses revealed that percent PPI<br />

levels were significantly lower in the stim session (20.19 ± 6 %) than in the stim+48<br />

(41.73 ± 4%), control (42.39 ± 4 %), or control+48 (44.47 ± 4 %) sessions. This effect<br />

was replicated with rats tested previously with 2 Hz stimulation (data discussed in section<br />

3.3 below). In contrast, 20 Hz stimulation of the dHip (n=13) failed to induce any<br />

significant changes in percent PPI (Fig. 2-1C; F(3, 36) = 2.01, N.S.). Percent PPI levels<br />

were similar in the stim (39.40 ± 3%), control (46.94 ± 5 %), stim+48 (51.40 ± 4 %), and<br />

control+48 (48.67 ± 5 %) sessions.<br />

48


stim control stim control<br />

+48 +48<br />

Condition<br />

stim control stim: control<br />

+48 +48<br />

Condition<br />

startle<br />

stim control stim control<br />

+48 +48<br />

Condition<br />

dHip - startle<br />

stim control stim control<br />

+48 +48<br />

Condition<br />

Figure 2-1. Effects of 20 Hz stimulation of the hippocampus on PPI and acoustic startle<br />

amplitude. A, Percent PPI scores 2 min (stim) or 48 hr (stim+48) following 20 Hz<br />

stimulation (10 s) of the vHip (n=14), and respective control PPI scores (control and<br />

control+48). The asterisk denotes a significant reduction in PPI in the stim group when<br />

compared to all other groups (p


Presentation of the auditory pulse alone induced robust startle in all rats (Fig. 2-<br />

1B, D). A one-way repeated measure ANOVA revealed that average startle amplitudes<br />

for the 4 PPI test sessions of the vHip 20 Hz stimulation group did not differ significantly<br />

(F(3, 39) = 1.40, N.S.). Thus, stimulation of the vHip did not significantly affect startle<br />

amplitude (average startle amplitude 116.61 ± 20 arbitrary startle units). Although<br />

stimulation of the dHip did not alter percent PPI levels (Fig. 2-1C), a significant main<br />

effect of test session was observed when startle amplitude was examined (Fig. 2-1D, F(3,<br />

36) = 3.37, p < 0.03). Post-hoc analyses revealed that when tested in the stim+48<br />

condition, rats had significantly lower startle amplitudes (74.24 ± 10) than when tested in<br />

the control condition (101.30 ± 13). Although a similar trend was noted between the stim<br />

and stim+48 test sessions, it was not significant.<br />

Effects of 2 Hz Stimulation of the vHip on PPI and Startle Amplitude<br />

Two hertz stimulation of the vHip (n=15) did not significantly alter PPI levels<br />

(Fig. 2-2A, F(3, 42) = 0.34, N.S.). When tested in the stim (46.93 ± 4 %), control (42.35<br />

± 4 %), stim+48 (43.03 ± 4 %), and control+48 (43.52 ± 3 %) conditions, the 2 Hz group<br />

demonstrated similar levels of PPI. In order to verify that 20 Hz stimulation would<br />

disrupt PPI in this group of rats, one week following their last PPI session, nine of the<br />

rats received 20 Hz stimulation of the vHip, and were tested for PPI 2 min and 24 hr later.<br />

Results revealed that 2 min following stimulation, PPI scores were significantly lower<br />

(30.01 ± 6 %) than 24 hr following stimulation (46.96 ± 4 %, F(l, 8) = 5.88, p < 0.05,<br />

data not shown). These data confirmed the short-term disruptive effect of 20 Hz<br />

stimulation of the vHip on PPI previously described.<br />

Stimulation (2 Hz) of the vHip produced an unexpected significant increase in<br />

50


A<br />

vHip-PPI<br />

stim control stim control<br />

+48 +48<br />

Condition<br />

B<br />

300, vHip - startle<br />

stim control stim control:<br />

+48 +48<br />

Condition<br />

Figure 2-2. Effects of 2 Hz stimulation of the vHip on PPI and acoustic startle<br />

amplitude. A, Percent PPI scores resulting from 2 Hz stimulation (100 s) of the vHip<br />

(n=15) either 2 min (stim) or 48 hr (stim+48) prior to testing, and respective control PPI<br />

scores (control and control+48). Control testing sessions were run exactly the same as<br />

'stim' sessions; however, no stimulation was applied to the vHip. B, Amplitude of the<br />

startle response during the same test conditions depicted in panel A. The cross denotes a<br />

significant difference between the stim condition and the stim+48 and control+48 groups<br />

(p


startle amplitude (Fig. 2-2B, F(3, 42) = 9.04, p < 0.001). Post-hoc analyses revealed that<br />

following 2 Hz stimulation, startle amplitudes were significantly higher (213.68 ± 29)<br />

than during the stim+48 (137.04 ± 15) or control+48 (134.71 ± 19) test sessions.<br />

Effects of Stimulation of the vHip and dHip on NAc DA Efflux<br />

As depicted in Fig. 2-3, baseline levels of DA in the NAc were relatively stable in<br />

all groups prior to stimulation. Stimulation of the vHip (20 Hz, n=7) induced a robust,<br />

long-lasting 35.41 ±4% increase in DA efflux in the NAc that was restricted to the<br />

hemisphere ipsilateral to the stimulating electrode. Separate one-way repeated measures<br />

ANOVA's confirmed that the increase in DA efflux in the ipsilateral hemisphere was<br />

significantly greater than baseline for 20 min (F(12, 72) = 4.04, p < 0.001, Dunnett's p <<br />

0.05), whereas no significant changes in DA efflux were observed in the contralateral<br />

hemisphere following vHip stimulation (F(12, 72) = 1.23, N.S.). Stimulation of the dHip<br />

(n=7) failed to evoke an increase in DA efflux in the NAc (F(12, 72) = 1.52, N.S.).<br />

Histology<br />

The locations of representative stimulation electrodes and the active tips of<br />

microdialysis probes are depicted in Fig. 2-4A-C. Ventral hippocampal placements (Fig.<br />

4A) were similar to those previously reported in the CAl/ventral subiculum (Wan et al.,<br />

1996;Blaha et al., 1997;Klarner et al, 1998;Zhang et al., 1999;2002a;Taepavarapruk et<br />

al, 2000;Bast et al., 2001;Swerdlow et al., 2001b). Electrodes aimed at the dHip (Fig.<br />

4B) were similarly positioned to dHip cannulae reported <strong>by</strong> a number of different groups<br />

(Swerdlow et al., 2001b;Zhang et al., 2002a) in the CA3/dentate gyrus region of the<br />

dHip. The placements of the active tips of the microdialysis probes aimed at the NAc did<br />

52


^ -20 -I 1 1 1 1 1 1 1 1 1 1 1 1<br />

1 2 3 4 5 6 7 8 9 10 11 12 13<br />

Time (X 10 min)<br />

Figure 2-3. Unilateral stimulation (20 Hz, 10 s) of either the vHip (n=7, stim) or dHip<br />

(n=7), and its effect on NAc DA efflux in the ipsilateral (vHip - black diamonds; dHip -<br />

white triangles) or contralateral (vHip - white squares) hemisphere. Data points<br />

represent the mean level of DA obtained over a 10 min sampling period, and error bars<br />

represent SEM. Asterisks denote a significant difference from baseline (p


Figure 2-4. Schematic diagram of the placements of stimulating electrodes and<br />

microdialysis probes in all experiments. A, Representative placements of electrode tips<br />

(black circles) aimed at the vHip. B, Representative placements of electrode tips (black<br />

circles) aimed at the dHip. C, Representative placements of microdialysis probes aimed<br />

at the NAc. The black bars illustrate the position of the probes and are scaled to 2 mm in<br />

length to accurately reflect only the area of the brain from which each probe sampled.<br />

Numbers correspond to the anterior or posterior distance (in mm) of each plate from<br />

bregma. Plates adapted from Paxinos and Watson (Paxinos and Watson, 1997).<br />

54


not substantially differ between the vHip and dHip stimulation experiments and are<br />

depicted on the same panels (Fig. 2-4C).<br />

Discussion<br />

In the present study, the effects of electrical stimulation of the hippocampus on<br />

PPI were site and frequency specific. Unilateral 20 Hz stimulation of the vHip, but not<br />

the dHip, caused a significant reduction in PPI at all prepulse intensities when testing<br />

began 2 min, but not 24 or 48 hr, after stimulation. Two Hz stimulation of the vHip<br />

failed to produce any changes in PPI. Changes in startie amplitude were unpredictable<br />

and difficult to interpret. Higher frequency stimulation of the vHip did not change startle<br />

amplitude significantly, whereas 2 Hz stimulation significantly increased startle<br />

amplitude 2 min following stimulation. When tested in the control condition, rats in the<br />

dHip group showed significantly higher startle amplitudes than when tested in the<br />

stim+48 condition. Experiments performed with microdialysis in freely moving rats<br />

confirmed our previous report that 20 Hz stimulation of the vHip results in a significant<br />

increase in NAc DA efflux (Taepavarapruk et al., 2000;Bast et al., 2001), and showed<br />

further that this increase was restricted to the hemisphere ipsilateral to the stimulating<br />

electrode. Finally, 20 Hz stimulation of the dHip failed to significantly increase NAc DA<br />

efflux.<br />

Potent Disruption of Sensorimotor Gating Induced <strong>by</strong> 20 Hz Stimulation of the vHip, but<br />

not dHip<br />

Numerous studies have reported that prolonged activation of the vHip, but not<br />

dHip, induced <strong>by</strong> bilateral infusion of NMDA, disrupts PPI in both Sprauge-Dawley (SD)<br />

and Wistar (W) rat strains (Wan et al., 1996;Klarner et al., 1998;Zhang et al,<br />

55


1999;2002a;Bast et al., 2001;Swerdlow et al, 2001b). The present study demonstrated a<br />

similar disruption of PPI following 20 Hz electrical stimulation of the vHip in the LE<br />

strain. Although hooded rats, such as the LE strain, are less commonly used in PPI<br />

experiments than the SD and W strains, they have been employed in studies of the<br />

genetic determinants (Swerdlow et al., 2001c;2003) and other pharmacological aspects of<br />

PPI (Rasmussen et al., 1997;Faraday et al., 1999;Binder et al., 2001). Recent<br />

experiments suggest that the three strains are differentially sensitive to the disruptive<br />

effects of DA agonists (Swerdlow et al., 2001c;2003), which is interesting given the role<br />

of the vHip in modulating NAc DA efflux discussed below.<br />

The degree (~ 50 %) and duration (< 24 hr) of PPI disruption following the brief<br />

20 Hz electrical stimulation employed in these studies were similar to that observed<br />

following NMDA infusion (Wan et al, 1996;Zhang et al., 1999;2002a;Bast et al.,<br />

2001;Swerdlow et al., 2001b), although some inconsistencies have been reported<br />

(Klarner et al., 1998). By using electrical stimulation of the vHip, we have demonstrated<br />

the brevity (10 s) and specificity (20 Hz vs. 2 Hz) <strong>by</strong> which aberrant activity in the vHip<br />

results in disrupted PPI. Clearly, the mechanism <strong>by</strong> which vHip stimulation disrupts PPI<br />

must significantly outlast the duration of stimulation. Analysis of the startle amplitude<br />

data revealed that no significant changes in startle occurred following 20 Hz stimulation<br />

of the vHip. Inspection of the startle amplitude data following NMDA infusion into the<br />

vHip suggests that startle amplitude is typically either unchanged (Wan et al.,<br />

1996;Klarner et al., 1998;Bast et al., 2001) or reduced (Zhang et al.,<br />

1999;2002a;Swerdlow et al., 2001b) following chemical stimulation of the vHip. Given<br />

the inconsistent changes in startie amplitude relative to consistent disruption of PPI<br />

56


following stimulation of the vHip, it is unlikely that the reduction in PPI observed here is<br />

due to changes in startle amplitude.<br />

The role of the hippocampus in the modulation of PPI has also been assessed with<br />

a variety of other manipulations. Permanent lesions of either the vHip or dHip do not<br />

affect PPI (Swerdlow et al., 1995;2000b;Pouzet et al., 1999 but see also Caine et al.,<br />

2001), although lesions of the vHip increase the sensitivity of rats to apomorphine-<br />

induced disruption of PPI (Swerdlow et al., 1995;2000b). Interestingly, temporary<br />

inactivation of either the vHip or dHip reversibly disrupts PPI (Zhang et al., 2002b).<br />

Data regarding modulation of PPI <strong>by</strong> the dHip are inconsistent and difficult to interpret.<br />

Infusion of the noncompetitive NMDA antagonist MK-801 into the vHip does not affect<br />

PPI (Bakshi and Geyer, 1998), whereas MK-801 infusions into the dHip may (Bakshi and<br />

Geyer, 1998;1999) or may not (Bast et al., 2000;Zhang et al., 2000b) alter PPI<br />

responding. In summary, these previous data demonstrate a subtle role for the vHip in<br />

the modulation of PPI under normal conditions. However, the present 20 Hz stimulation<br />

data, and those of others groups using NMDA (Wan et al., 1996;Zhang et al.,<br />

1999;2002a;Bast et al., 2001;Swerdlow et al., 2001b), indicate that overactivity in the<br />

vHip can cause a potent reduction in PPI. Although the dHip may also modulate PPI<br />

under certain conditions, data from this study and others (Swerdlow et al., 2001b;Zhang<br />

et al., 2002a) clearly indicate that induced overactivity of this structure does not<br />

significantly disrupt PPI.<br />

Frequency Dependence of the vHip Stimulation-induced Disruption of PPI<br />

Two Hz stimulation of the vHip, which causes a significant reduction in NAc DA<br />

efflux that may be mediated <strong>by</strong> the activation of group 2/3 mGluR's (Taepavarapruk et<br />

57


al., 1998), failed to significantly affect PPI in this study (Fig. 2-2). Available<br />

pharmacological evidence is consistent with the observed lack of effect on PPI following<br />

2 Hz vHip stimulation. For instance, blockade of DA receptors with DA antagonists,<br />

such as haloperidol, does not alter levels of PPI in rats (Mansbach et al., 1988;Depoortere<br />

et al., 1997;Hart et al., 1998;Feifel and Priebe, 1999). Additionally, systemic<br />

administration of the group 2/3 mGluR agonist LY354740 has no effect on PPI levels<br />

(Schreiber et al., 2000). Thus, pharmacological treatments that mimic some of the effects<br />

of 2 Hz vHip stimulation are also without effect on PPI responding.<br />

Unexpectedly, 2 Hz stimulation of the vHip resulted in a significant increase in<br />

startle amplitude immediately following the stimulation when compared to the stim+48<br />

and control+48 conditions. Treatment with group 2/3 mGluR agonists or haloperidol<br />

decreased or had no effect on startle magnitude respectively (Mansbach et al.,<br />

1988;Depoortere et al., 1997;Hart et al., 1998;Feifel and Priebe, 1999;Schreiber et al.,<br />

2000). Given that these drugs were administered systemically, it is possible that their<br />

effects may result from actions in brain areas other than those affected <strong>by</strong> 2 Hz<br />

stimulation. However, it is interesting that stimulation of the vHip with two different<br />

frequencies had different effects on both startle and PPI. Given the inconsistencies in<br />

startle changes observed following NMDA stimulation (Wan et al., 1996;Zhang et al.,<br />

1999;2002a;Legault et al., 2000;Taepavarapruk et al., 2000;Bast et al., 2001;Swerdlow et<br />

al., 2001b), it will be important to replicate these findings in separate groups and/or<br />

strains of rats to ensure their reliability.<br />

58


Increases in NAc DA Efflux following Hip Stimulation are Site and Hemisphere Specific<br />

Stimulation of the vHip with either NMDA or brief periods of 20 Hz electrical<br />

stimulation produces a long-lasting increase in NAc DA efflux (Legault et al.,<br />

2000;Taepavarapruk et al., 2000). The effect of 20 Hz stimulation is mediated <strong>by</strong><br />

iGluR's in the NAc, there<strong>by</strong> implicating the direct glutamatergic projection from the<br />

vHip to NAc in this effect (Blaha et al, 1997;Taepavarapruk et al., 2000). The present<br />

neurochemical experiments (Fig. 3) extend our previous findings, as the increase in NAc<br />

DA efflux following 20 Hz electrical stimulation of the vHip was restricted to the<br />

hemisphere ipsilateral to the stimulating electrode. This finding is consistent with the<br />

fact that the vHip projections to forebrain areas such as the NAc are primarily unilateral<br />

(Kelley and Domesick, 1982;Groenewegen et al, 1987;Brog et al., 1993).<br />

In contrast to stimulation of the vHip, 20 Hz stimulation of the dHip failed to<br />

increase NAc DA efflux significantly. Although the dHip sends glutamatergic efferents<br />

to the NAc, it is important to note that the vHip projects primarily to the NAc shell<br />

whereas the dHip projects primarily to the NAc core (Swanson and Cowan, 1977;Kelley<br />

and Domesick, 1982;Whitter, 1986;Groenewegen et al., 1987;Brog et al., 1993). Probe<br />

placements adjacent to both the shell and core subregions of the NAc were employed in<br />

studies in which stimulation of the vHip increased NAc DA efflux (Fig. 2-4C); therefore,<br />

we monitored DA efflux in a similar region of the NAc following stimulation of the<br />

dHip. This raises the possibility that any effect of the dHip stimulation on NAc DA<br />

efflux was restricted to the NAc core. However, unpublished observations failed to<br />

confirm an increase in NAc DA efflux with dialysis probes located in the core of the NAc<br />

(Howland, Taepavararpuk, and Phillips).<br />

59


Microdialysis experiments have examined dynamic changes in NAc DA efflux<br />

during PPI test sessions. Zhang and colleagues (Zhang et al., 2000a) demonstrated that<br />

systemic administration of amphetamine disrupted PPI, whereas treatment with cocaine<br />

had no effect (but also see Martinez et al., 1999). Interestingly, both drugs increased<br />

NAc DA efflux <strong>by</strong> at least 100 % of baseline during the PPI test sessions. Consequently,<br />

these authors suggest that the behavioral effects of DA agonists on PPI are critically<br />

dependent on their mechanism of action. Other experiments have shown that<br />

presentation of startle pulses alone cause a significant decrease in NAc DA efflux that is<br />

not observed when prepulses of a moderate intensity are presented prior to the startle<br />

pulses (Hum<strong>by</strong> et al., 1996). These data suggest that during PPI, the presentation of the<br />

prepulses prevents perturbation of the mesoaccumbens DA system in response to the<br />

startling stimuli. Together, these data imply that certain drugs, such as amphetamine, can<br />

interfere with the effect of a prepulse on DA efflux in the NAc, there<strong>by</strong> disrupting PPI.<br />

Given these data, it is intriguing that two different methods of stimulating the<br />

vHip, both of which increase DA efflux in the NAc, can disrupt PPI. The fact that 20 Hz<br />

stimulation of the dHip or 2 Hz stimulation of the vHip failed to disrupt PPI and did not<br />

increase NAc DA efflux is consistent with the hypothesis that increased NAc DA efflux<br />

following 20 Hz vHip stimulation may be a factor in the disruption of PPI. However,<br />

systemic administration of the D 2 antagonist haloperidol (Wan et al., 1996;Zhang et al.,<br />

1999;Bast et al., 2001) or intra-NAc infusions of the non-NMDA receptor antagonist<br />

CNQX (Wan et al., 1996) fail to block the disruptive effects of intra-vHip NMDA<br />

infusion on PPI responding. Therefore, it is unlikely that the disruptive effects of<br />

60


stimulation of the vHip on PPI are mediated solely <strong>by</strong> activation of the mesoaccumbens<br />

DA system or non-NMDA receptors in the NAc (Wan et al., 1996;Bast et al., 2001).<br />

Potential Mechanisms Underlying vHip Stimulation-induced Disruption in PPI<br />

Normal DA and Glu transmission appears to be important for PPI (Zhang et al.,<br />

2000a;Geyer et al., 2001). For example, infusions of DA or DA agonists into the NAc<br />

disrupt PPI (Mansbach et al., 1988;Swerdlow et al., 1990;Wan et al., 1995), and infusions<br />

of Glu agonists and antagonists such as AMPA, APV, CNQX, and MK-801 into the NAc<br />

have complex and regionally specific effects (Reijmers et al., 1995;Wan et al., 1995;Wan<br />

and Swerdlow, 1996). Recent physiological experiments raise the possibility that the<br />

effects of vHip stimulation on PPI may involve interactions between multiple receptor<br />

subtypes in the NAc. Stimulation of the vHip shifts NAc neurons to an activated or 'up'<br />

state (O'Donnell and Grace, 1995) and recent experiments from our laboratory indicate<br />

that 20 Hz stimulation (10 s) of the vHip alters the responses of medium spiny neurons in<br />

the NAc shell to afferent input from the vHip and BLA (Floresco et al., 2001). These<br />

effects are dependent on the 20 Hz stimulation-induced increase in NAc DA and a<br />

complex pharmacology involving activation of both D t and NMDA receptors, but not D 2<br />

and non-NMDA receptors (Floresco et al., 2001). Importantly, the potentiating effect of<br />

20 Hz stimulation of the vHip on NAc neurons is maintained for at least 20 min, which<br />

corresponds to the duration of PPI trials in the present study.<br />

Stimulation of the vHip may also disrupt PPI via the widespread connections of<br />

the vHip to a number of areas in the CSPP circuitry critical for normal PPI (Klarner et al.,<br />

1998). Data obtained in the present study do not refute this hypothesis. The vHip is<br />

reciprocally connected to a number of areas other than the NAc, such as the mPFC,<br />

61


amygdala, thalamus, septum, and ventral pallidum (Groenewegen et al, 1987;Amaral and<br />

Whitter, 1995;Conde et al., 1995), all of which are important for the regulation of PPI<br />

(Swerdlow et al., 2001a). Stimulation of the vHip with doses of NMDA that disrupt PPI<br />

cause increased fos-protein expression in a number of limbic and cortical areas such as<br />

the NAc, lateral septum, mPFC, and dorsomedial striatum (Klarner et al., 1998).<br />

Stimulation of the vHip also induces various types of plasticity in the mPFC such as<br />

long-term potentiation and depression (Laroche et al., 2000). Therefore, subtle changes<br />

in multiple areas induced <strong>by</strong> vHip stimulation may disrupt PPI (Klarner et al., 1998).<br />

Although the dHip projects to the NAc core, it is not directly connected to other areas<br />

implicated in PPI (Swanson and Cowan, 1977;Kelley and Domesick, 1982;Whitter,<br />

1986;Amaral and Whitter, 1995), and therefore aberrant activity generated in the dHip<br />

may not have access to the neural substrates critical to disrupt PPI. This conjecture is<br />

consistent with the failure of 20 Hz stimulation of the dHip to increase NAc DA efflux in<br />

the NAc or to disrupt PPI in the present study.<br />

Conclusion<br />

Our data clearly demonstrate that brief, higher frequency stimulation of vHip, but<br />

not dHip, disrupts PPI and increases DA efflux in the NAc. Given the anatomical and<br />

functional abnormalities frequently observed in the hippocampal formation of patients<br />

with schizophrenia, these data support the assertion that abnormalities in limbic-cortico-<br />

striatal interactions may underlie deficits in sensorimotor gating in schizophrenia.<br />

Identification of the mechanism underlying the disruptive effects of vHip stimulation on<br />

PPI may provide insight into potential targets for pharmacological treatment strategies for<br />

schizophrenia.<br />

62


\<br />

References<br />

Amaral DG, Whitter MP (1995) Hippocampal formation. In: The Rat Nervous System<br />

(Paxinos G, ed), pp 443-493. San Diego: Academic Press.<br />

Bakshi VP, Geyer MA (1998) Multiple limbic regions mediate the disruption of prepulse<br />

inhibition produced in rats <strong>by</strong> the noncompetitive NMDA antagonist dizocilpine. J<br />

Neurosci 18: 8394-8401.<br />

Bakshi VP, Geyer MA (1999) Alpha-1-adrenergic receptors mediate sensorimotor gating<br />

deficits produced <strong>by</strong> intracerebral dizocilpine administration in rats. Neuroscience 92:<br />

113-121.<br />

Bast T, Zhang W, Feldon J, White JJVI (2000) Effects of MK801 and neuroleptics on<br />

prepulse inhibition: re-examination in two strains of rats. Pharmacol Biochem Behav 67:<br />

647-658.<br />

Bast T, Zhang WN, Heidbreder C, Feldon J (2001) Hyperactivity and disruption of<br />

prepulse inhibition induced <strong>by</strong> N-methyl-D-aspartate stimulation of the ventral<br />

hippocampus and the effects of pretreatment with haloperidol and clozapine.<br />

Neuroscience 103: 325-335.<br />

Binder EB, Kinkead B, Owens MJ, Kilts CD, Nemeroff CB (2001) Enhanced neurotensin<br />

neurotransmission is involved in the clinically relevant behavioral effects of antipsychotic<br />

drugs: evidence from animal models of sensorimotor gating. J Neurosci 21: 601-608.<br />

Blaha CD, Yang CR, Floresco SB, Barr AM, Phillips AG (1997) Stimulation of the<br />

ventral subiculum of the hippocampus evokes glutamate receptor-mediated changes in<br />

dopamine efflux in the rat nucleus accumbens. Eur J Neurosci 9: 902-911.<br />

Bogerts B (1997) The temporolimbic system theory of positive schizophrenic symptoms.<br />

Schizophr Bull 23: 423-435.<br />

Braff DL, Geyer MA, Swerdlow NR (2001) Human studies of prepulse inhibition of<br />

starde: normal subjects, patient groups, and pharmacological studies.<br />

Psychopharmacology (Berl) 156: 234-258.<br />

Breier A, Su TP, Saunders R, Carson RE, Kolachana BS, de Bartolomeis A, Weinberger<br />

DR, Weisenfeld N, Malhotra AK, Eckelman WC, Pickar D (1997) Schizophrenia is<br />

associated with elevated amphetamine-induced synaptic dopamine concentrations:<br />

evidence from a novel positron emission tomography method. Proc Natl Acad Sci USA<br />

94: 2569-2574.<br />

Brog JS, Salyapongse A, Deutch AY, Zahm DS (1993) The patterns of afferent<br />

innervation of the core and shell in the "accumbens" part of the rat ventral striatum:<br />

immunohistochemical detection of retrogradely transported fluoro-gold. J Comp Neurol<br />

338: 255-278.<br />

63


Bradzynski SM, Gibson CJ (1997) Release of dopamine in the nucleus accumbens<br />

caused <strong>by</strong> stimulation of the subiculum in freely moving rats. Brain Res Bull 42: SOS-<br />

SOS.<br />

Caine SB, Geyer MA, Swerdlow NR (1992) Hippocampal modulation of acoustic startle<br />

and prepulse inhibition in the rat. Pharmacol Biochem Behav 43: 1201-1208.<br />

Caine SB, Hum<strong>by</strong> T, Robbins TW, Everitt BJ (2001) Behavioral effects of psychomotor<br />

stimulants in rats with dorsal or ventral subiculum lesions: locomotion, cocaine selfadministration,<br />

and prepulse inhibition of startle. Behav Neurosci 115: 880-894.<br />

Carlsson A, Waters N, Carlsson ML (1999) Neurotransmitter interactions in<br />

schizophrenia—therapeutic implications. Biol Psychiatry 46: 1388-1395.<br />

Conde F, Maire-Lepoivre E, Audinat E, Crepel F (1995) Afferent connections of the<br />

medial frontal cortex of the rat. II. Cortical and subcortical afferents. J Comp Neurol 352:<br />

567-593.<br />

Depoortere R, Perrault G, Sanger DJ (1997) Potentiation of prepulse inhibition of the<br />

startle reflex in rats: pharmacological evaluation of the procedure as a model for<br />

detecting antipsychotic activity. Psychopharmacology (Berl) 132: 366-374.<br />

Dierks T, Linden DE, Jandl M, Formisano E, Goebel R, Lanfermann H, Singer W (1999)<br />

Activation of Heschl's gyrus during auditory hallucinations. Neuron 22: 615-621.<br />

Faraday MM, O'Donoghue VA, Grunberg NE (1999) Effects of nicotine and stress on<br />

startle amplitude and sensory gating depend on rat strain and sex. Pharmacol Biochem<br />

Behav 62: 273-284.<br />

Feifel D, Priebe K (1999) The effects of subchronic haloperidol on intact and dizocilpinedisrupted<br />

sensorimotor gating. Psychopharmacology (Berl) 146: 175-179.<br />

Fendt M, Schwienbacher I, Koch M (2000) Amygdaloid N-methyl-D-aspartate and<br />

gamma-aminobutyric acid(A) receptors regulate sensorimotor gating in a dopaminedependent<br />

way in rats. Neuroscience 98: 55-60.<br />

Floresco SB, Blaha CD, Yang CR, Phillips AG (2001) Modulation of hippocampal and<br />

amygdalar-evoked activity of nucleus accumbens neurons <strong>by</strong> dopamine: cellular<br />

mechanisms of input selection. J Neurosci 21: 2851-2860.<br />

Geyer MA, Krebs-Thomson K, Braff DL, Swerdlow NR (2001) Pharmacological studies<br />

of prepulse inhibition models of sensorimotor gating deficits in schizophrenia: a decade<br />

in review. Psychopharmacology (Berl) 156: 117-154.<br />

Goff DC, Coyle JT (2001) The emerging role of glutamate in the pathophysiology and<br />

treatment of schizophrenia. Am J Psychiatry 158: 1367-1377.<br />

64


Grace AA (2000) Gating of information flow within the limbic system and the<br />

pathophysiology of schizophrenia. Brain Res Brain Res Rev 31: 330-341.<br />

Groenewegen HJ, Vermeulen-Van der Zee E, te KA, Witter MP (1987) Organization of<br />

the projections from the subiculum to the ventral striatum in the rat. A study using<br />

anterograde transport of Phaseolus vulgaris leucoagglutinin. Neuroscience 23: 103-120.<br />

Harrison PJ (1999) The neuropathology of schizophrenia. A critical review of the data<br />

and their interpretation. Brain 122 (Pt 4): 593-624.<br />

Hart S, Zreik M, Carper R, Swerdlow NR (1998) Localizing haloperidol effects on<br />

sensorimotor gating in a predictive model of antipsychotic potency. Pharmacol Biochem<br />

Behav 61: 113-119.<br />

Heckers S, Rauch SL, Goff D, Savage CR, Schacter DL, Fischman AJ, Alpert NM (1998)<br />

Impaired recruitment of the hippocampus during conscious recollection in schizophrenia.<br />

Nat Neurosci 1: 318-323.<br />

Howland JG, Taepavarapruk P, Phillips AG (2002) Glutamate receptor-dependent<br />

modulation of dopamine efflux in the nucleus accumbens <strong>by</strong> basolateral, but not central,<br />

nucleus of the amygdala in rats. J Neurosci 22: 1137-1145.<br />

Hum<strong>by</strong> T, Wilkinson LS, Robbins TW, Geyer MA (1996) Prepulses inhibit startleinduced<br />

reductions of extracellular dopamine in the nucleus accumbens of rat. J Neurosci<br />

16: 2149-2156.<br />

Kelley AE, Domesick VB (1982) The distribution of the projection from the hippocampal<br />

formation to the nucleus accumbens in the rat: an anterograde- and retrogradehorseradish<br />

peroxidase study. Neuroscience 7: 2321-2335.<br />

Klarner A, Koch M, Schnitzler HU (1998) Induction of Fos-protein in the forebrain and<br />

disruption of sensorimotor gating following N-methyl-D-aspartate infusion into the<br />

ventral hippocampus of the rat. Neuroscience 84: 443-452.<br />

Koch M (1999) The neurobiology of startle. Prog Neurobiol 59: 107-128.<br />

Koch M, Bubser M (1994) Deficient sensorimotor gating after 6-hydroxydopamine lesion<br />

of the rat medial prefrontal cortex is reversed <strong>by</strong> haloperidol. Eur J Neurosci 6: 1837-<br />

1845.<br />

Lacroix L, Spinelli S, White W, Feldon J (2000) The effects of ibotenic acid lesions of<br />

the medial and lateral prefrontal cortex on latent inhibition, prepulse inhibition and<br />

amphetamine-induced hyperlocomotion. Neuroscience 97: 459-468.<br />

Laroche S, Davis S, Jay TM (2000) Plasticity at hippocampal to prefrontal cortex<br />

synapses: dual roles in working memory and consolidation. Hippocampus 10: 438-446.<br />

65


Laruelle M, Abi-Dargham A, Gil R, Kegeles L, Innis R (1999) Increased dopamine<br />

transmission in schizophrenia: relationship to illness phases. Biol Psychiatry 46: 56-72.<br />

Legault M, Rompre PP, Wise RA (2000) Chemical stimulation of the ventral<br />

hippocampus elevates nucleus accumbens dopamine <strong>by</strong> activating dopaminergic neurons<br />

of the ventral tegmental area. J Neurosci 20: 1635-1642.<br />

Mansbach RS, Geyer MA (1989) Effects of phencyclidine and phencyclidine biologs on<br />

sensorimotor gating in the rat. Neuropsychopharmacology 2: 299-308.<br />

Mansbach RS, Geyer MA, Braff DL (1988) Dopaminergic stimulation disrupts<br />

sensorimotor gating in the rat. Psychopharmacology (Berl) 94: 507-514.<br />

Martinez ZA, Ellison GD, Geyer MA, Swerdlow NR (1999) Effects of sustained cocaine<br />

exposure on sensorimotor gating of startle in rats. Psychopharmacology (Berl) 142: 253-<br />

260.<br />

Meyer-Lindenberg A, Miletich RS, Kohn PD, Esposito G, Carson RE, Quarantelli M,<br />

Weinberger DR, Berman KF (2002) Reduced prefrontal activity predicts exaggerated<br />

striatal dopaminergic function in schizophrenia. Nat Neurosci 5: 267-271.<br />

O'Donnell P, Grace AA (1995) Synaptic interactions among excitatory afferents to<br />

nucleus accumbens neurons: hippocampal gating of prefrontal cortical input. J Neurosci<br />

15: 3622-3639.<br />

Pakkenberg B (1990) Pronounced reduction of total neuron number in mediodorsal<br />

thalamic nucleus and nucleus accumbens in schizophrenics. Arch Gen Psychiatry 47:<br />

1023-1028.<br />

Paxinos G, Watson C (1997) The rat brain in stereotaxic co-ordinates. San Diego:<br />

Academic Press.<br />

Pouzet B, Feldon J, Veenman CL, Yee BK, Richmond M, Nicholas J, Rawlins P, Weiner<br />

I (1999) The effects of hippocampal and fimbria-fornix lesions on prepulse inhibition.<br />

Behav Neurosci 113: 968-981.<br />

Rasmussen K, Gates MR, Burger JE, Czachura JF (1997) The novel atypical<br />

antipsychotic olanzapine, but not the CCK-B antagonist LY288513, blocks apomorphineinduced<br />

disruption of pre-pulse inhibition. Neurosci Lett 222: 61-64.<br />

Reijmers LG, Vanderheyden PM, Peeters BW (1995) Changes in prepulse inhibition after<br />

local administration of NMDA receptor ligands in the core region of the rat nucleus<br />

accumbens. Eur J Pharmacol 272: 131-138.<br />

Schreiber R, Lowe D, Voerste A, De Vry J (2000) LY354740 affects startle responding<br />

but not sensorimotor gating or discriminative effects of phencyclidine. Eur J Pharmacol<br />

388: R3-R4.<br />

66


Silbersweig DA, Stern E, Frith C, Cahill C, Holmes A, Grootoonk S, Seaward J,<br />

McKenna P, Chua SE, Schnorr L,. (1995) A functional neuroanatomy of hallucinations<br />

in schizophrenia. Nature 378: 176-179.<br />

Strecker RE, Moneta ME (1994) Electrical stimulation of the kindled hippocampus<br />

briefly increases extracellular dopamine in the nucleus accumbens. Neurosci Lett 176:<br />

173-177.<br />

Swanson LW, Cowan WM (1977) An autoradiographic study of the organization of the<br />

efferent connections of the hippocampal formation in the rat. J Comp Neurol 172: 49-84.<br />

Swerdlow NR (1996) Cortico-striatal substrates of cognitive, motor, and sensory gating:<br />

speculations and implications for psychological dysfunction. Advances in Biological<br />

Psychiatry 2: 179-207.<br />

Swerdlow NR, Braff DL, Geyer MA (2000a) Animal models of deficient sensorimotor<br />

gating: what we know, what we think we know, and what we hope to know soon. Behav<br />

Pharmacol 11: 185-204.<br />

Swerdlow NR, Braff DL, Geyer MA, Koob GF (1986) Central dopamine hyperactivity in<br />

rats mimics abnormal acoustic startle response in schizophrenics. Biol Psychiatry 21: 23-<br />

33.<br />

Swerdlow NR, Braff DL, Masten VL, Geyer MA (1990) Schizophrenic-like sensorimotor<br />

gating abnormalities in rats following dopamine infusion into the nucleus accumbens.<br />

Psychopharmacology (Berl) 101: 414-420.<br />

Swerdlow NR, Braff DL, Taaid N, Geyer MA (1994) Assessing the validity of an animal<br />

model of deficient sensorimotor gating in schizophrenic patients. Arch Gen Psychiatry<br />

51: 139-154.<br />

Swerdlow NR, Geyer MA, Braff DL (2001a) Neural circuit regulation of prepulse<br />

inhibition of startle in the rat: current knowledge and future challenges.<br />

Psychopharmacology (Berl) 156: 194-215.<br />

Swerdlow NR, Hanlon FM, Henning L, Kim YK, Gaudet I, Halim ND (2001b)<br />

Regulation of sensorimotor gating in rats <strong>by</strong> hippocampal NMDA: anatomical<br />

localization. Brain Res 898: 195-203.<br />

Swerdlow NR, Lipska BK, Weinberger DR, Braff DL, Jaskiw GE, Geyer MA (1995)<br />

Increased sensitivity to the sensorimotor gating-disruptive effects of apomorphine after<br />

lesions of medial prefrontal cortex or ventral hippocampus in adult rats.<br />

Psychopharmacology (Berl) 122: 27-34.<br />

Swerdlow NR, Platten A, Kim YK, Gaudet I, Shoemaker J, Pitcher L, Auerbach P<br />

(2001c) Sensitivity to the dopaminergic regulation of prepulse inhibition in rats: evidence<br />

for genetic, but not environmental determinants. Pharmacol Biochem Behav 70: 219-226.<br />

67


Swerdlow NR, Shoemaker JM, Platten A, Pitcher L, Goins J, Crain S (2003) Heritable<br />

differences in the effects of amphetamine but not DOI on startle gating in albino and<br />

hooded outbred rat strains. Pharmacol Biochem Behav 75: 191-197.<br />

Swerdlow NR, Taaid N, Halim N, Randolph E, Kim YK, Auerbach P (2000b)<br />

Hippocampal lesions enhance startle gating-disruptive effects of apomorphine in rats: a<br />

parametric assessment. Neuroscience 96: 523-536.<br />

Taepavarapruk P, Blaha CD, Phillips AG (1998) Decrease in extracellular dopamine<br />

levels in the nucleus accumbens induced <strong>by</strong> low frequency stimulation of the ventral<br />

subiculum/CAl region of the rat hippocampus. SfN Abstracts 24, 823.2.<br />

Taepavarapruk P, Floresco SB, Phillips AG (2000) Hyperlocomotion and increased<br />

dopamine efflux in the rat nucleus accumbens evoked <strong>by</strong> electrical stimulation of the<br />

ventral subiculum: role of ionotropic glutamate and dopamine Dl receptors.<br />

Psychopharmacology (Berl) 151: 242-251.<br />

Wan FJ, Caine SB, Swerdlow NR (1996) The ventral subiculum modulation of prepulse<br />

inhibition is not mediated via dopamine D2 or nucleus accumbens non-NMDA glutamate<br />

receptor activity. Eur J Pharmacol 314: 9-18.<br />

Wan FJ, Geyer MA, Swerdlow NR (1995) Presynaptic dopamine-glutamate interactions<br />

in the nucleus accumbens regulate sensorimotor gating. Psychopharmacology (Berl) 120:<br />

433-441.<br />

Wan FJ, Swerdlow NR (1993) Intra-accumbens infusion of quinpirole impairs<br />

sensorimotor gating of acoustic startle in rats. Psychopharmacology (Berl) 113: 103-109.<br />

Wan FJ, Swerdlow NR (1996) Sensorimotor gating in rats is regulated <strong>by</strong> different<br />

dopamine-glutamate interactions in the nucleus accumbens core and shell subregions.<br />

Brain Res 722: 168-176.<br />

Wan FJ, Swerdlow NR (1997) The basolateral amygdala regulates sensorimotor gating of<br />

acoustic startle in the rat. Neuroscience 76: 715-724.<br />

Weike Al, Bauer U, Hamm AO (2000) Effective neuroleptic medication removes<br />

prepulse inhibition deficits in schizophrenia patients. Biol Psychiatry 47: 61-70.<br />

Whitter MP (1986) A survey of the anatomy of the hippocampal formation, with<br />

emphasis on the septotemporal organization of its intrinsic and extrinsic connections. In:<br />

Advances in experimental and medical biology: Vol. 203. Excitatory amino acids and<br />

epilepsy. (Schwarcz R, Ben-Ari Y, eds), pp 67-82. New York: Plenum.<br />

Wright IC, Ellison ZR, Sharma T, Friston KJ, Murray RM, McGuire PK (1999) Mapping<br />

of grey matter changes in schizophrenia. Schizophr Res 35: 1-14.<br />

Zhang J, Forkstam C, Engel JA, Svensson L (2000a) Role of dopamine in prepulse<br />

inhibition of acoustic startle. Psychopharmacology (Berl) 149: 181-188.<br />

68


Zhang W, Pouzet B, Jongen-Relo AL, Weiner I, Feldon J (1999) Disruption of prepulse<br />

inhibition following N-methyl-D-aspartate infusion into the ventral hippocampus is<br />

antagonized <strong>by</strong> clozapine but not <strong>by</strong> haloperidol: a possible model for the screening of<br />

atypical antipsychotics. Neuroreport 10: 2533-2538.<br />

Zhang WN, Bast T, Feldon J (2000b) Microinfusion of the non-competitive N-methyl-Daspartate<br />

receptor antagonist MK-801 (dizocilpine) into the dorsal hippocampus of wistar<br />

rats does not affect latent inhibition and prepulse inhibition, but increases startle reaction<br />

and locomotor activity. Neuroscience 101: 589-599.<br />

Zhang WN, Bast T, Feldon J (2002a) Effects of hippocampal N-methyl-D-aspartate<br />

infusion on locomotor activity and prepulse inhibition: differences between the dorsal<br />

and ventral hippocampus. Behav Neurosci 116: 72-84.<br />

Zhang WN, Bast T, Feldon J (2002b) Prepulse inhibition in rats with temporary<br />

inhibition/inactivation of ventral or dorsal hippocampus. Pharmacol Biochem Behav 73:<br />

929-940.<br />

69


CHAPTER THREE: DELAYED ONSET OF PREPULSE INHIBITION DEFICITS<br />

FOLLOWING KAINIC ACID TREATMENT ON POSTNATAL DAY SEVEN IN<br />

<strong>RAT</strong>S. 2<br />

Introduction<br />

Given the heterogeneity and complexity of schizophrenia, its etiology has been<br />

difficult to define. The neurodevelopmental hypothesis of schizophrenia proposes that<br />

adverse events or genetic abnormalities may disrupt early brain development, and interact<br />

with environmental factors or influence subsequent brain development to cause the<br />

disorder (Weinberger, 1995;Benes et al., 2000;McClure and Weinberger, 2001). Once<br />

the illness is expressed, a diverse array of subde changes are found in temporal and<br />

frontal brain areas such as the hippocampus, striatum, thalamus, and frontal cortex<br />

(Pakkenberg, 1990;Suddath et al., 1990;Bogerts, 1997;Jones, 1997;Heckers etal.,<br />

1998;Harrison, 1999;Gothelf et al., 2000;McClure and Weinberger, 2001;Meyer-<br />

Lindenberg et al., 2002). Neurodegeneration in these areas is rarely observed in the<br />

brains of patients with schizophrenia (but see Stevens, 1994), therefore alterations in<br />

normal patterns of connectivity between affected brain areas may underlie the expression<br />

of some symptoms of schizophrenia (Harrison, 1999;Friston, 1999;Benes, 2000;Penn,<br />

2001).<br />

Accordingly, the consequences of disrupting the normal development of limbic<br />

and cortical areas on behaviors in rodents related to schizophrenia have been examined<br />

(for reviews see Lipska and Weinberger, 2000;Van den et al., 2003). Specific behavioral<br />

abnormalities have been observed in adult, but not prepubescent rats, that received<br />

2<br />

A version of this chapter has been published: Howland JG, Hannesson DK, Phillips AG (2004). Delayed<br />

onset of prepulse inhibition deficits following kainic acid treatment on postnatal day seven in rats.<br />

European Journal of Neuroscience 20:2639-2648.<br />

70


ventral hippocampal (vHip) lesions on postnatal day (PND) 7 (Lipska and Weinberger,<br />

2000). Behavioral changes in prepulse inhibition (PPI) and locomotor responses to<br />

novelty, which resemble certain positive symptoms in schizophrenia, are robust and can<br />

be exacerbated <strong>by</strong> acute administration of dopamine (DA) agonists immediately before<br />

testing in adulthood (Lipska et al., 1993; 1995;Le Pen and Moreau, 2002). Importantly,<br />

temporary reduction of neural activity within the vHip on PND7, with the sodium<br />

channel blocker tetrodotoxin (TTX) increased locomotor activity in response to novelty,<br />

injection stress, and amphetamine after puberty (Lipska et al., 2002). Similar results have<br />

been observed following neonatal lesions of the amygdala (Daenen et al., 2001; 2002;<br />

2003), but not the medial prefrontal cortex (Lipska et al., 1998;Brake et al., 2000;Van<br />

den et al., 2003 but see Flores et al., 1996). Additionally, systemic administration of the<br />

non-competitive NMDA receptor antagonist dizocilpine (MK-801) on PND7 can reduce<br />

PPI levels and increase locomotor activity in female rats (Harris et al., 2003; but see<br />

Beninger et al., 2002).<br />

The apparent sensitivity of limbic-cortical circuits on PND7 to functional<br />

disruption raises the possibility that increased neural activity induced <strong>by</strong> the ionotropic<br />

glutamate agonist kainic acid (KA) at this developmental age would result in behavioral<br />

changes in rats similar to those discussed above. Kainic acid has been used extensively<br />

to study limbic-motor seizures and excitotoxicity in adult rats (for reviews see Sperk,<br />

1994;Ben Ari and Cossart, 2000); however, its effects during early development are not<br />

as well characterized. During the first two postnatal weeks, systemic administration of<br />

KA results in tonic-clonic seizures and the selective activation of the hippocampus and<br />

lateral septum without the high levels of cell death normally observed in limbic and<br />

71


cortical areas following administration during or after the 3 r<br />

postnatal week (Nitecka et<br />

al., 1984;Tremblay et al., 1984;Stafstrom et al., 1992;Khalilov et al., 1999;Koh et al.,<br />

1999;Lynch et al., 2000;Silveira et al., 2002). Although few studies have analyzed<br />

behavioral changes in adulthood following a single administration of KA during the first<br />

week of life, there are reports of deficits in conditioned avoidance (de Feo et al., 1986)<br />

and spatial learning using the radial arm maze (Lynch et al., 2000), but not the water<br />

maze (Stafstrom et al., 1993;Koh et al., 1999).<br />

The present study sought to examine whether systemic KA administration on<br />

PND7 would alter the behavior of rats in a pattern similar to that observed in other animal<br />

models of schizophrenia (Lipska et al., 1993;Harris et al., 2003). Kainic acid or saline<br />

was administered to rat pups on PND7 and their PPI levels and locomotor responses to a<br />

novel environment were measured before and after puberty. To assess potential<br />

alterations of the DA system following KA administration, we tested the potential<br />

interactive effects of KA treatment and apomorphine, a direct DA agonist, on PPI and<br />

also with amphetamine-induced locomotion. Subgroups of KA- and saline-treated rats<br />

were also tested on a standard hippocampal-dependent Morris Water Maze task in<br />

adulthood to examine aspects of spatial learning and memory that are dependent on<br />

normal hippocampal function (Morris et al., 1990).<br />

Methods<br />

Subjects<br />

Two independent groups of animals (i.e. Group 1 and Group 2) were tested in this<br />

study. Testing protocols for both groups were similar with small procedural differences<br />

noted in the methods section where appropriate. Pregnant Long-Evans rats were obtained<br />

72


from Charles River (Quebec, Canada) at 13 to 15 days of gestation. They were singly<br />

housed and left undisturbed until giving birth. The colony was maintained on a 12/12<br />

hour light/dark cycle (lights on at 0700), at a temperature of 22+1 °C. All rats were given<br />

food (Purina Rat Chow) and tap water ad libitum. Experiments were conducted in<br />

accordance with the standards of the Canadian Council on Animal Care and were<br />

approved <strong>by</strong> the Committee on Animal Care at the University of British Columbia.<br />

Kainic Acid Administration<br />

The day of birth of the pups was designated post-natal day (PND) 0. On PND3,<br />

the litters were sexed and culled to include only males (6-9 rats per litter). On PND7, all<br />

but one of the pups were removed from the nest, weighed and individually placed in<br />

small compartments of a cardboard box for KA administration. They were then removed<br />

from the colony and taken to a small heated room. KA (1.5 mg/kg, Tocris) or saline was<br />

injected (i.p.) with a 30-gauge needle (10 ml/kg). Care was taken to ensure that both KA<br />

and saline was administered to members from each litter. Previous reports indicated that<br />

KA administration of 1-2 mg/kg on PND7 causes behavioral seizures in most pups<br />

without high mortality rates (Stafstrom et al., 1993;Lynch et al., 2000). The behavior of<br />

all rats was recorded with an overhead video camera for 180 minutes. Before being<br />

returned to their mothers, the pups were earmarked according to treatment condition.<br />

The litters were then left undisturbed (except for normal cage changing) until weaning on<br />

PND25. Weanling rats were housed in cages of 2 or 3 with members of their litter. All<br />

rats were handled for at least 5 days before behavioral testing.


Prepulse Inhibition<br />

On PND35 and 56, rats were removed individually from the colony and taken<br />

immediately to the PPI apparatus. Testing was conducted in a single sound-attenuating<br />

startle chamber (ambient noise level 64 dB), containing a transparent Plexiglas tube (8.2<br />

cm in diameter, 20 cm in length), mounted on a Plexiglas frame (SR-LAB, San Diego<br />

Instruments, San Diego). Noise bursts were presented through a speaker mounted 24-cm<br />

above the tube. An accelerometer below the frame of the apparatus measured whole<br />

body startle amplitude, defined as the average of 100 1-ms accelerometer readings<br />

collected from stimulus onset. Each PPI test session began with a 5-min acclimatization<br />

period during which a 70-dB background noise level was presented, which remained<br />

constant for the entire test session. Following the acclimatization period, six pulse alone<br />

trials (120 dB, 40 ms) were presented to achieve a relatively stable startle amplitude<br />

before PPI testing. Data from these pulse-alone trials was not considered in the analysis<br />

of PPI. Immediately following the six initial pulse-alone trials, presentation of the trials<br />

that were used in the calculation of PPI levels were initiated. Trials presented were of<br />

three types: pulse alone (12 trials, 120 dB, 40 ms), prepulse + pulse (10 trials X 3<br />

prepulse intensities - discussed below), or no stimulus (42 trials). Prepulse + pulse trials<br />

consisted of the presentation of a 20 ms prepulse of 73, 76, or 82 dB 80 ms before the<br />

presentation of the pulse. The pulse and prepulse + pulse trials were presented in a<br />

pseudorandom order. One no-stimulus trial was presented between each pair of pulse and<br />

prepulse + pulse trials. The inter-trial interval varied randomly from 3 to 12 s (average<br />

7.5 s). Calibration of the apparatus was performed using a RadioShack Digital Sound<br />

Level Meter and adjustments were made as necessary.<br />

74


Between PND98 and 105, a randomly selected subset of the rats from Group 2<br />

were re-tested on PPI following injection with vehicle (0.1% ascorbic acid) and<br />

apomorphine (Sigma; 0.2 mg/kg; s.e.) 72 hr later to assess: 1) the consistency of the PPI<br />

reduction in KA-treated animals and 2) the potential role of dopaminergic mechanisms in<br />

the observed PPI decreases in the KA-treated animals. The PPI sessions were conducted<br />

in a manner identical to that described above, except that immediately before the PPI<br />

session, all rats were weighed and injected with the appropriate volume of vehicle or<br />

drug.<br />

Locomotor Activity<br />

On PND36 and 57, rats were weighed, removed from the colony, and<br />

immediately placed in locomotor boxes. For Group 1, the locomotor boxes were in two<br />

similar, small adjacent rooms (4 boxes in one, 3 in the other) constructed from clear<br />

Plexiglas (32 cm X 32 cm X height 41 cm) and fitted with four pairs of infra-red beams<br />

positioned 10 cm apart and 2.5 cm above a metal grid floor. All sensors were interfaced<br />

to a computer-controlled system (MANX). Motor activity counts were collected in 10-<br />

min bins during testing. Care was taken to ensure that rats from each group were tested<br />

in both rooms and in all boxes. Spontaneous locomotor activity levels in a novel<br />

environment were measured for 60 min. Immediately following the spontaneous<br />

locomotor activity test, all rats were injected with D-amphetamine (1.5 mg/kg, i.p.) and<br />

returned to the locomotor boxes. Locomotor activity was then monitored for an<br />

additional 90 min before the rats were returned to the colony room. For Group 2, testing<br />

procedures were identical, except that a Med Associates System was used to collect the<br />

data. Eight Med Associates Test Chambers (ENV-008; 30.5 cm X 24.1cm X height 21.0<br />

75


cm) were fitted with 4 pairs of infrared photocells 3.5 cm from the floor evenly spaced on<br />

the long walls. The chambers had metal grid floors and two operant levers (which were<br />

retracted during locomotor activity testing). Each chamber was contained within a Med<br />

Associates sound attenuating cubicle with a house light that was illuminated during<br />

testing.<br />

Water Maze Testing<br />

A subgroup of the animals from Group 1 was tested between PND65 and 75 in a<br />

water maze. Testing was performed in a circular, white swimming pool 180 cm in<br />

diameter, 54 cm in height. The escape platform was 34 cm in height and 14 cm in<br />

diameter. The pool was filled with water such that the top of the platform was 3 cm<br />

below the surface of the water. For visible platform trials, an additional piece of wood<br />

and wire mesh was secured to the top of the invisible platform such that it extended 3 cm<br />

out of the water. The water was rendered opaque <strong>by</strong> the addition of white Tempura<br />

Powder paint. Data were recorded using a computerized tracking system (HVS Image,<br />

Hampton, England). Rats were transported to the pool room in groups and testing was<br />

conducted over 3 consecutive days. Rats were released facing the wall of the pool from<br />

one of the geographic poles in a pseudorandom order. Each rat was allowed 60 s to swim<br />

to the platform, after which they were left on the platform for 10 s. They were then<br />

removed from the water maze, and the next rat was tested. The intertrial interval for each<br />

rat was approximately 7 min. On the first day, rats were trained to swim to the visible<br />

platform over 5 trials to allow habituation to the testing environment and swimming. The<br />

location of the platform was varied for each trial, and was never in the location of the<br />

hidden platform used on day 2. Hidden platform training was performed on day 2. Rats<br />

76


were given 10 trials to learn to find the constant location of the hidden platform. Finally,<br />

on day 3, the rats were tested in a 30-s probe trial without the platform present. The<br />

percent time spent in the quadrant that contained the hidden platform on day 2 was taken<br />

as a measure of spatial memory.<br />

Histology<br />

Following behavioral testing, a subgroup of animals (n=5 from each group) was<br />

randomly selected, sacrificed with an overdose of pentobarbital, and perfused<br />

transcardially with 0.9% saline followed <strong>by</strong> 10% formalin. Brains were stored in 10%<br />

sucrose/10% formalin for at least 1 week, after which coronal sections (40 u.m)<br />

containing the hippocampus were taken using a vibratome. Every fourth section was<br />

saved and stained with cresyl violet. An experimenter blind to the treatment group<br />

examined each section using a light microscope for gross cell loss in the hippocampus<br />

with particular attention paid to the CA3 region due to its established sensitivity to the<br />

excitotoxic effects of KA in adult rats (Ben Ari and Cossart, 2000).<br />

Data Analysis<br />

For the PPI experiments, two measures were calculated for each animal. The<br />

startle amplitude represented the mean startle amplitude of the 12 pulse-alone trials<br />

presented after the 6 habituation trials. Startle amplitude data were compared with an<br />

independent samples t-test for each age. PPI was calculated <strong>by</strong> averaging startle<br />

amplitudes for each trial type. The percent PPI for each prepulse intensity was calculated<br />

using the formula: [100 - (100 X startle amplitude on prepulse + pulse trials) -s- (startle<br />

amplitude on pulse alone trials)]. A repeated measures ANOVA was performed on the<br />

data obtained from each age with prepulse intensity as a within subjects factor and<br />

77


treatment on PND7 as a between subjects factor. Results from the apomorphine<br />

experiment were also analyzed with a mixed ANOVA (prepulse intensity and test as<br />

within subjects factors, and treatment at PND7 as a between subjects factor). Locomotor<br />

activity data were compared using repeated measures ANOVA at each age with test type<br />

as a within subjects factor and treatment on PND7 as a between subjects factor. For the<br />

water maze task, trials were blocked into pairs to reduce variance; latencies and path<br />

lengths to find the hidden platform were analyzed using a repeated measures ANOVA<br />

(trial block as a within subjects factor, treatment on PND7 as a between subjects factor).<br />

Additionally, swim speeds were measured for all rats and compared using a repeated<br />

measures ANOVA. After the probe trials, the percent time spent in each quadrant was<br />

calculated <strong>by</strong> dividing the time spent in that quadrant <strong>by</strong> the total length of the probe trial<br />

(30 s). A repeated measures ANOVA was performed on the data with quadrant as a<br />

within subjects factor and treatment on PND7 as a between subjects factor. For all<br />

ANOVA's, post-hoc analyses were performed using the Neuman-Keuls test where<br />

appropriate. The significance level for all statistical tests was p < 0.05.<br />

Results<br />

Immediate Behavioral Effects ofPND7 KA Administration<br />

Ten to 20 min following KA administration, the majority of pups began to show<br />

behavioral signs of seizure activity as previously reported (Tremblay et al.,<br />

1984;Stafstrom et al, 1992;Lynch et al., 2000;Silveira et al, 2002). Initially, the<br />

majority of the pups showed evidence of a hunched posture and heavy breathing followed<br />

<strong>by</strong> the appearance of a continuous, clonic 'scratching' activity of the rear limbs. Many of<br />

the rats developed swimming seizures during which they rolled onto their sides during<br />

78


outs of forelimb and hindlimb clonus, which continued for 30 to 120 min. Some of the<br />

pups developed more severe tonic seizures in the second hour after KA administration,<br />

which lasted up to 60 min. Only data from pups that displayed behavioral signs of<br />

seizure activity for at least 30 minutes were analyzed in these experiments, and no<br />

differences in seizure severity between pups from Group 1 and Group 2 could be readily<br />

quantified. Six pups (11%) died as a result of the KA administration, while 10 (18%)<br />

were not used for the behavioral experiments due to mild, intermittent seizures. None of<br />

the saline-treated animals displayed seizure-related behaviors.<br />

Effects ofPND7 KA Administration on Body Weight<br />

On PND7, pups randomly selected for KA administration weighed 18.13 ± 0.37 g<br />

while those administered saline weighed 17.86 ± 0.46 g. On PND8, the pups were<br />

weighed again to ensure that suckling behavior was not altered between the groups after<br />

treatment on PND7. The KA- and saline-treated rats weighted 20.45 ± 0.45 g and 20.77<br />

± 0.55 g on PND8 respectively. An independent samples t-test revealed that both groups<br />

of animals gained a similar amount of weight during the 24 hr after treatment (t(50) = -<br />

1.28, N.S.). On PND36, the rats treated with KA on PND7 weighed slightly more than<br />

those treated with saline (KA: 181.90 ± 2 g; saline: 175.90 ± 2 g); however, when tested<br />

on PND57, the rats treated with KA weighed significantly more than those treated with<br />

saline (KA: 395.33 ± 5 g; saline: 379.66 ± 5 g) as revealed <strong>by</strong> a significant age <strong>by</strong> weight<br />

interaction (F(l, 68) = 3.98, p = 0.05).<br />

79


70-,<br />

A PND35<br />

PP3 PP6 PP12 average<br />

Prepulse Intensity<br />

PP3 PP6 PP12 average<br />

Prepulse Intensity<br />

B PND35<br />

140 i<br />

•g 120 H<br />

100 -<br />

Q.<br />

E 80 -<br />

ii<br />

• 60 -i<br />

CO OJ 40<br />

20<br />

0<br />

CO oq 40<br />

20 A<br />

saline kainic<br />

Treatment<br />

D PND56<br />

140<br />

-o 120-1<br />

Q.<br />

E<br />

<<br />

100<br />

80<br />

a) 60 H<br />

1<br />

saline kainic<br />

Treatment<br />

Figure 3-1. Prepulse inhibition (PPI) scores from rats in Group 1 treated on postnatal day<br />

(PND) 7 with saline (white bars, n=17) or kainic acid (KA; black bars, n=20). A, Effects<br />

of.PND 7 KA treatment on PPI at PND35. B, Average starde amplitudes of the rats on<br />

PND35. C, Percent PPI scores for those rats tested at PND56. D, Average startle<br />

amplitudes of the rats on PND56. Asterisks indicate a significant difference between<br />

groups (p


Prepulse Inhibition is Reduced in Post-Pubescent, but not Pre-pubescent Rats, Following<br />

PND7 KA Administration<br />

Group 1: As shown in Fig. 3-1 A, rats administered either KA (n=20) or saline<br />

(n=17) on PND7 had similar levels of PPI on PND35. Percent PPI scores were similar in<br />

response to trials with 3 and 6 dB prepulses, whereas PPI was substantially increased for<br />

trials during which a 12 dB prepulse was presented.' A repeated measures ANOVA<br />

revealed a significant effect of prepulse (F(2, 70) = 78.92, p< 0.001), but no significant<br />

prepulse <strong>by</strong> treatment interaction (F(2, 70) = 0.26, N.S.) or main effect of treatment (F(l,<br />

35) = 2.04, N.S.). On PND35, rats in the KA-treated group demonstrated slightly lower<br />

starde amplitudes than those in the saline treated group to the pulse alone trials (55.27 ± 5<br />

vs. 67.51 ± 7 arbitrary startle units) groups (Fig. 3-1B). However, this difference was not<br />

significant (t(35) = -1.41, N.S.).<br />

In contrast to PND35, PPI levels obtained from KA- and saline-treated rats in<br />

young adulthood (PND56) were significantly different (Fig. 3-1C). A repeated measures<br />

ANOVA revealed a significant effect of prepulse intensity (F(2, 70) = 33.05, p < 0.001),<br />

as well as a significant prepulse <strong>by</strong> treatment interaction (F(2, 70) = 5.57, p < 0.01). Rats<br />

in the saline-treated condition showed a typical pattern of increasing percent PPI in<br />

response to prepulses of increasing intensity (24.80 ± 5% for PP3, 41.77 ± 4% for PP6,<br />

and 54.38 ± 4% for PP12 trials). When compared to the saline-treated animals, KA-<br />

treated rats responded with significantly lower percent PPI scores for the PP6 trials<br />

(19.17 ± 6% PPI, p < 0.05), and attenuated PPI in response to PP12 trials (47.15 ± 4%).<br />

As a result, animals in the KA-treated group demonstrated a 25% reduction in average<br />

percent PPI scores when compared with the saline-treated group (KA-treated = 30.18 ±<br />

81


3%; saline-treated = 40.32 ± 4%), which approached significance (F(l, 35) = 3.93, p =<br />

0.055). The average startle amplitude of the KA-treated rats (77.20 ± 12 arbitrary startle<br />

units) was lower than that of the saline-treated group (102.11 ± 18), although this<br />

difference was not significant (t(35) = -1.17, N.S.).<br />

Group 2: As in Group 1, rats in both the KA- (n=19) and saline- (n=14) treated<br />

groups tested on PND35 had similar levels of PPI (Fig. 3-2A). A repeated measures<br />

ANOVA revealed a significant main effect of prepulse intensity (F(2, 62) = 28.62, p<br />


£-•30<br />

CD<br />

OL 20 A<br />

A PND35<br />

PP3 PP6 PP12 average<br />

C PND56<br />

Prepulse Intensity<br />

PP3 PP6 PP 12 average<br />

Prepulse Intensity<br />

140<br />

. 120<br />

"§ 100<br />

CO<br />

co<br />

80<br />

60<br />

40<br />

20<br />

B PND35<br />

saline kainic<br />

Treatment<br />

D PND56<br />

140<br />

„ 120<br />

"§ 100<br />

|- 80<br />

tr.<br />

•a-<br />

CO<br />

60<br />

40<br />

20<br />

0-<br />

saline kainic<br />

Treatment<br />

Figure 3-2. Prepulse inhibition (PPI) scores from rats in Group 2 treated on postnatal day<br />

(PND) 7 with saline (white bars, n=14) or kainic acid (black bars, n=19). A, Effects of<br />

postnatal day (PND) 7 KA treatment on PPI at PND35. B, Average startle amplitudes of<br />

the rats on PND35. C, Percent PPI scores for those rats tested at PND56. D, Average<br />

startle amplitudes of the rats on PND56. The asterisk indicates a significant difference<br />

between groups (p


shown). Additionally, a significant main effect of test (F(l, 16) = 16.94, p < 0.005) and a<br />

test <strong>by</strong> prepulse interaction (F(2,32) = 4.82, p < 0.02) were observed. Inspection of the<br />

data confirmed that apomorphine treatment significantly disrupted PPI in both groups of<br />

rats (Fig. 3-3A) while post-hoc analyses revealed that apomorphine disrupted PPI<br />

significantly more during the 6 dB prepulse trials than during either the 3 or 12 dB trials<br />

(p < 0.05, data not shown).<br />

Importantly, the absence of a significant test <strong>by</strong> treatment interaction (F(l,16) =<br />

0.09, N.S.) does not support the hypothesis that animals treated with KA on PND7 are<br />

more sensitive to the disruptive effects of low doses of apomorphine on PPI. A separate<br />

analysis of the data obtained following the vehicle injection revealed that those animals<br />

treated with KA (n=10) on PND7 had lower PPI scores (38.36 ± 4%) than those treated<br />

with saline (n=8; 50.00 ± 6%, Fig. 3-3 A). Although this difference was of a similar<br />

magnitude to the differences in PPI reported at PND56 for groups 1 and 2 above, it failed<br />

to reach significance perhaps due to insufficient power (t(16) = -1.65, p = 0.12).<br />

Apomorphine treatment significantly decreased startle amplitude in both groups (Fig. 3-<br />

3B). This is reflected <strong>by</strong> a significant main effect of test (F(l,16) = 6.15, p < 0.05), but<br />

not group (F(l, 16) = 0.13, N.S.) or a group <strong>by</strong> test interaction (F(l, 16) = 0.07, N.S.).<br />

Locomotor Activity in Response to Novelty and Amphetamine in Rats Following PND7<br />

KA Administration<br />

Group 1: As depicted in Fig. 3-4A and B, KA- (n=20) and saline- (n=17) treated<br />

rats in this group showed similar levels of spontaneous locomotor activity in a novel<br />

environment (60 min) or following amphetamine administration (1.5 mg/kg, 90 min) on<br />

84


S-V K-V S-A K-A<br />

Treatment<br />

S-V K-V S-A K-A<br />

VTreatment<br />

Figure 3-3. A, Effects of pretreatment with vehicle (0.1% ascorbic acid, A) or<br />

apomorphine (0.2 mg/kg, B) on average percent PPI scores in adulthood. Animals were<br />

administered either saline (n=8, white bars) or kainic acid (n=10, black bars) on postnatal<br />

day 7. B, Average startle amplitudes of those rats depicted in panel A. In both panels,<br />

asterisks indicate a significant difference between groups. The cross indicates a between<br />

group difference that approached significance (p


1 4<br />

o 12<br />

X 10<br />

8 £<br />

< 4 •<br />

<strong>TO</strong> 2,<br />

o<br />

A Nov PND36<br />

1 4<br />

0 12<br />

>< 10<br />

1 8<br />

£ 6 4<br />

< 4 -<br />

iS 2<br />

S-1 K-1 S-2 K-2<br />

Treatment<br />

S-1 K-1 S-2 K-2<br />

Treatment<br />

3 5<br />

o -<br />

°30-<br />

X;25-<br />

^20-<br />

•^•'1.5o<br />

< 10-<br />

I 5<br />

0<br />

B Amph PND36<br />

S-1 K-1 S-2 K-2<br />

Treatment<br />

C Nov PND57 D Amph PND57<br />

S-1 K-1 S-2 K-2<br />

: Treatment<br />

Figure 3-4. Locomotor activity levels in response to novelty (A, C) or amphetamine (1.5<br />

mg/kg; B, D) of all rats tested on either postnatal day (PND) 36 (A, B) or PND57 (C, D).<br />

Group labels on the x-axis of each panel reflect the treatment of the rats on PND7 (5 =<br />

saline, K - kainic acid) and their group (1 = group 1,2 = group 2). The asterisk indicates<br />

a significant difference between the denoted groups (p


PND36. A repeated measures ANOVA failed to confirm a significant treatment effect<br />

(F(l, 35) = 0.16, N.S.) or treatment <strong>by</strong> test interaction (F(l, 35) = 0.12, N.S.). At PND57,<br />

a repeated measures ANOVA did not reveal a main effect of treatment (F(l, 35) = 2.01,<br />

N.S.), but did reveal a significant test <strong>by</strong> treatment interaction (F(l, 35) = 4.10, p = 0.05).<br />

Post-hoc analyses revealed that although both groups showed similar locomotor<br />

responses in the novelty condition (Fig. 3-4C), rats treated with KA on PND7 had<br />

significantly higher locomotor responses (2640.25 ± 225 beam crosses) following<br />

injection with amphetamine than rats treated with saline (2145.06 ± 143; Fig. 3-4D).<br />

Group 2: Similar to the data presented for Group 1, the locomotor responses of<br />

rats treated with either KA (n=19) or saline (n=14) on PND7 did not differ in either the<br />

spontaneous activity or amphetamine conditions on PND36 (Fig. 3-4A and B). This is<br />

reflected in an insignificant main effect of treatment (F(l, 31) = 1.30, N.S.) and treatment<br />

<strong>by</strong> test interaction (F(l, 31) = 2.29, N.S.). In contrast to the data obtained from Group 1,<br />

when tested on PND57, rats in Group 2 failed to show significant differences in<br />

locomotor activity in either condition (main effect of treatment: F(l, 31) = 1.70, N.S.;<br />

treatment <strong>by</strong> test interaction: F(l, 31) = 1.66, N.S.).<br />

Spatial Learning and Memory in the Morris Water Maze is not Altered in Rats Following<br />

PND7 KA Administration<br />

On the first test day, all rats learned to escape <strong>by</strong> swimming to the visible<br />

platform (data not shown). On the second day of testing, rats in both groups learnt to<br />

swim to the hidden platform (Fig. 3-5A). A repeated measures ANOVA performed on<br />

the latency data revealed a significant main effect of trial block (F(4, 52) = 17.43, p <<br />

0.001), but not a significant effect of group (F(l, 13) = 0.279, N.S.) or a significant block<br />

87


E<br />

F<br />

50<br />

40<br />

30<br />

20<br />

10<br />

A<br />

0 -n———r———i •'•.- i-<br />

B<br />

Trial Block<br />

1 • ' / .2 . . 3 ;<br />

Quadrant<br />

Figure 3-5. A, Average latencies (s) to locate the hidden platform in the water maze of<br />

rats treated with saline (white diamonds, n=7) or kainic acid (black squares, n=8) on<br />

postnatal day 7. Trial blocks represent the average search time of 2 trials for each rat.<br />

The asterisk indicates a significant difference between trial blocks (p


y group interaction (F(4, 52) = 0.487, N.S.). Post-hoc analyses revealed that both the<br />

KA- (n=8) and saline-treated (n=7) rats were significantly slower at finding the platform<br />

during block 1 (i.e. trials 1 and 2) than the subsequent 4 blocks (average search time -<br />

block 1: 38.66 ± 6 s; blocks 2 to 5: 14.38 ± 3 s; p < 0.05). Path lengths were significantly<br />

reduced across trial blocks in both groups (F(4, 52) = 14.33, p < 0.01; data not shown);<br />

however, neither total path length (KA-treated = 4,029.10 ± 483 cm, saline-treated =<br />

4597.04 ± 609 cm; F(l, 13) = 1.56, N.S.) nor swim speed (KA-treated = 22.33 ± 1 cm/s,<br />

saline-treated = 25.14 ± 1 cm/s; F(l, 13) = 3.21, N.S.) differed significandy between the<br />

groups. When retention of the platform location was tested 24 hr later using a 30 s probe<br />

trial, both groups spent most of their time searching in the quadrant which had contained<br />

the platform the previous day (Fig. 3-5B). A repeated measures ANOVA revealed a<br />

significant main effect of quadrant (F(3, 39) = 20.88, p < 0.001), but no significant group<br />

<strong>by</strong> quadrant interaction (F(3, 39) = 0.581, N.S.). Post-hoc analyses revealed that both<br />

groups spent significandy more time in quadrant 2 (41.65 ± 5 %), which contained the<br />

platform, than in either quadrants 3 (19.22 ± 4 %) or 4 (8.24 ± 3 %). Inspection of the<br />

data presented in Fig. 3-5B reveals that the animals also spent more time in quadrant 2<br />

than quadrant 1 (30.89 ± 3 %), although this difference failed to reach significance.<br />

Histology<br />

In agreement with previous studies (Nitecka et al., 1984;Stafstrom et al.,<br />

1992;Koh et al., 1999;Lynch et al., 2000), histological examination of the hippocampi of<br />

rats which had received either KA or saline on PND7 revealed no signs of gross cell loss<br />

or hippocampal damage (Fig. 3-6).<br />

89


Figure 3-6. A representative cresyl violet stained section of the dorsal hippocampus of a<br />

KA-treated (A) and saline-treated rat (B).<br />

90


Discussion<br />

The present study demonstrates that treatment with KA on PND7 results in the<br />

delayed emergence of behavioral changes in adulthood not seen before puberty.<br />

Following treatment with KA on PND7, rats in two independent groups showed disrupted<br />

PPI on PND56, but not PND35. When re-tested 6 weeks later, a subgroup of the KA-<br />

treated animals continued to exhibit relatively lower PPI than saline-treated controls.<br />

Additionally, administration of a low dose of apomorphine (0.2 mg/kg) disrupted PPI in<br />

both KA- and saline-treated rats; however, KA-treated rats failed to show enhanced<br />

sensitivity to the disruptive effects of apomorphine. When the locomotor responses of<br />

these animals in response to a novel environment and amphetamine challenge were<br />

analyzed, an inconsistent pattern emerged. KA- and saline-treated rats had similar<br />

locomotor responses to both novelty and amphetamine on PND36, as well as novelty on<br />

PND57. Following amphetamine administration (1.5 mg/kg) on PND57, locomotor<br />

activity was significantly increased in the KA-treated rats from Group 1, but not from<br />

Group 2. Furthermore, neither spatial learning nor memory were affected <strong>by</strong> the<br />

administration of KA on PND7 as assessed in the Morris water-maze, confirming<br />

previous studies (Stafstrom et al., 1993;Koh et al., 1999).<br />

Effects of Neonatal KA Administration on PPI<br />

In two separate groups of rats, we observed that KA treatment on PND7<br />

significantly disrupted PPI in postpubescence, but not before puberty. It is important to<br />

note that PPI has been reliably measured in rat pups as young as 16 days of age (Martinez<br />

et al., 2000;Swerdlow et al., 2000), and that the rats tested in the present experiments on<br />

PND35 showed consistent levels of PPI with relatively low variance. Post-pubescent rats<br />

91


treated with KA on PND7 had mean PPI levels that were 75 to 80% of those shown <strong>by</strong><br />

saline-treated animals. This effect was observed consistently in two groups of animals<br />

tested independendy (Fig. 3-1C and 3-2C) and was evident without changes in startle<br />

amplitude (Fig. 3-1D and 3-2D). Previous studies have reported the effects of neonatal<br />

manipulations on PPI in adult rats. Data presented <strong>by</strong> Lipska and colleagues (Lipska et<br />

al, 1995; Fig. 4) from rats that had sustained permanent lesions of the vHip on PND7,<br />

indicate PPI levels that were approximately 70% of their control littermates, although a<br />

significant difference between groups was noted only at the lowest prepulse intensity (4<br />

dB above baseline). Other studies report slightiy larger effects over a wider range of<br />

prepulse intensities following neonatal lesions of the vHip (Le Pen and Moreau,<br />

2002;Daenen et al., 2003) and the amygdala (Daenen et al., 2003) or treatment with MK-<br />

801 on PND7 (Harris et al., 2003). Given that these treatments result in relatively subtle<br />

disruptions in PPI under baseline testing conditions, it is perhaps not surprising that KA<br />

treatment on PND7, which does not cause gross brain damage (Nitecka et al.,<br />

1984;Stafstrom et al., 1992;Koh et al., 1999;Lynch et al, 2000; present data), causes<br />

relatively small disruptions in PPI.<br />

Rats with neonatal vHip lesions also show heightened sensitivity to the disruptive<br />

effects of apomorphine (0.1 mg/kg), there<strong>by</strong> supporting the hypothesis that mesolimbic<br />

DA transmission is altered in the lesioned rats (Lipska et al., 1995). In the present study,<br />

KA treatment on PND7 failed to enhance the disruptive effects of apomorphine treatment<br />

on PPI. However, administration of 0.2 mg/kg of apomorphine immediately before<br />

testing disrupted PPI in both the KA- and saline-treated animals (Fig. 3-3A). This result<br />

is surprising because Long-Evans rats are reported to be less sensitive than Sprague<br />

92


Dawley rats (the strain used <strong>by</strong> Lipska et al., 1995) to the disruptive effects of<br />

apomorphine on PPI. Indeed, it has been demonstrated that Long-Evans fats fail to show<br />

disrupted PPI following administration of doses of apomorphine as high as 0.5 mg/kg<br />

(Swerdlow et al., 2001). Although no clear explanation exists for these discrepant data,<br />

complex interactions between rat strain and PPI have been studied extensively (Varty and<br />

Geyer, 1998;Ellenbroek and Cools, 2000;Swerdlow et al., 2000). In this context, further<br />

examination of the effect of early KA-treatment's potentially interactive effects with<br />

apomorphine may be profitable in other rat strains.<br />

Using C57BL6 mice, Yee and colleagues (Yee et al., 2004a; 2004b) elegandy<br />

demonstrated that pretreatment with dopamine agonists such as apomorphine or non­<br />

competitive A/-methyl-D-aspartate antagonists, such as dizocilpine and phencyclidine,<br />

disrupts PPI, a finding that has previously been well documented (Geyer et al., 2001).<br />

However, when prepulse-elicited reactivity was examined, a dissociation between the two<br />

classes of drugs was observed where<strong>by</strong> apomorphine increases and dizocilpine and<br />

phencyclidine decrease prepulse-elicited reactivity (Yee et al., 2004a; 2004b).<br />

Incorporation of a prepulse alone condition in future experiments may allow for parallels<br />

to be drawn between these pharmacological studies and KA treatment. For example, if<br />

KA administration on PND7 decreased prepulse-elicited startle, it could be argued that<br />

KA administration has effects similar to dizocilpine or phencyclidine pretreatment, but<br />

not apomorphine pretreatment.<br />

Effects of Neonatal KA on Locomotor Activity<br />

Increased locomotor activity in response to treatments that activate the<br />

mesolimbic DA system is a cardinal feature of many animal models of schizophrenia<br />

93


(Lipska et al., 1993;Hall, 1998;Weiss et al., 2000;Lipska et al., 2002;Harris et al, 2003).<br />

Neonatal KA treatment in the present study failed to increase locomotor behavior<br />

consistently following exposure to a novel environment or an amphetamine injection,<br />

although Group 1 did show a significant increase in locomotor activity following<br />

treatment with amphetamine (Fig. 3-4D). It is unclear why KA-treated rats in Group 2<br />

failed to show elevated locomotor responses following amphetamine treatment on<br />

PND57 given that both groups displayed similar seizure-related behaviors following KA<br />

administration on PND7 and similar deficits in PPI when tested at PND56. These results,<br />

considered in conjunction with the failure of apomorphine to affect PPI, question the role<br />

of postsynaptic alterations in the mesolimbic DA system in the observed effects of KA<br />

treatment on PPI. Furthermore, the absence of consistent changes in locomotor behavior<br />

in early adulthood following KA-induced aberrant activity in developing cortico-limbic<br />

circuits on PND7, underscores important differences between these data and the<br />

decreased activity induced <strong>by</strong> the TTX administration into the vHip (Lipska et al., 2002)<br />

and systemic MK-801 administration (Harris et al., 2003).<br />

Potential Mechanisms Underlying the Observed PPI Changes<br />

TTX infusions into the vHip on PND7 potentiate locomotor activity in early<br />

adulthood (Lipska et al., 2002) and systemic administration of the NMDA antagonist<br />

MK-801 on PND7 may also increase locomotor activity and disrupt PPI (Harris et al.,<br />

2003, but see Beninger et al., 2002). Lipska and colleagues failed to detect any gross<br />

morphological changes in the adult brains of animals injected with TTX on PND7<br />

(Lipska et al., 2002). In contrast, administration of MK-801 on PND7 increases<br />

apoptosis in limbic and cortical brain areas such as the hippocampus and thalamus<br />

94


(Harris et al., 2003), and this effect may underlie the observed behavioral changes in<br />

adult animals. Taken together, these data suggest that neural circuits subserving<br />

locomotor behavior and PPI are particularly sensitive to alterations in neural activity<br />

during the first postnatal week.<br />

All rats included in this study displayed generalized tonic-clonic seizures for 1 to<br />

2 hr following KA treatment on PND7, therefore it is reasonable to assume that aberrant<br />

activity would have occurred in neural circuits that regulate PPI and locomotor activity in<br />

adulthood (i.e. the hippocampus and septum; (Tremblay et al., 1984;Khalilov et al.,<br />

1999;Silveira et al., 2002). This raises the question of whether the behavioral effects<br />

observed in this study are the result of the activation of glutamate receptors and/or the<br />

consequence of seizures early in postnatal development. Obviously, these possibilities<br />

cannot be resolved based on the present experiments. However, it is well known that<br />

ionotropic glutamate receptor activity is critical for the normal formation of cortico-<br />

limbic circuits (Ben Ari et al., 1997;Feldman and Knudsen, 1998;Luthi et al., 2001).<br />

Although most of this research has focused on NMDA and AMPA receptor mediated<br />

events, some recent studies suggest that kainate receptors are dynamically regulated<br />

during critical periods of experience-dependent plasticity in thalamocortical synapses<br />

(Kidd and Isaac, 1999;Kidd et al., 2002). As a result, it is conceivable that<br />

hyperstimulation of kainate receptors could result in developmental alterations of the<br />

neural circuitry known to regulate PPI. Given that KA was administered systemically in<br />

the present study and genes encoding kainate receptor subunits are widely expressed in<br />

the brain during early postnatal development (Bahn et al., 1994), KA administration<br />

95


could have subtly altered a number of brain regions, there<strong>by</strong> producing the observed<br />

behavioral effects.<br />

The dose of KA (1.5 mg/kg) used in the present study induced seizures in the<br />

neonatal rats, therefore the effects of seizures, and not ionotropic glutamate receptor<br />

hyperstimulation per se, may also underlie the behavioral effects observed. Seizures<br />

induced <strong>by</strong> systemic administration of KA on PND7 increase glucose metabolism<br />

primarily in the dorsal hippocampus and lateral septum (Tremblay et al., 1984) and<br />

transiendy increase c-fos expression in the hippocampus (Silveira et al., 2002).<br />

Significant changes in the morphology of the hippocampus observed routinely following<br />

KA administration post-weaning are not evident after acute systemic treatment with KA<br />

during postnatal weeks 1 and 2 (Nitecka et al., 1984;Stafstrom et al., 1992;Koh et al.,<br />

1999;Lynch et al., 2000).<br />

More subtle alterations in the excitability of limbic circuits have been noted<br />

following administration of KA on PND7 including a reduction in long-term potentiation<br />

in the dentate gyrus, slower kindling rates, and increased paired pulse inhibition in the<br />

perforant path in adult rats, accompanied <strong>by</strong> impairments of spatial learning as assessed<br />

on a radial arm maze (Lynch et al., 2000). Exposure to KA on PND15 may also<br />

exacerbate cognitive impairment and cell loss in the hippocampus induced <strong>by</strong> seizures in<br />

adulthood (Koh et al., 1999). Recently, i.c.v. administration of KA (10-50 nmol) on<br />

PND7 has been demonstrated to cause a relatively subtle loss of hippocampal neurons,<br />

particularly in the CA3 and CA1 subregions of the hippocampus (Montgomery et al.,<br />

1999;Dong et al., 2003a). Although some cell loss is observed immediately following<br />

KA administration on PND7, greater cell loss occurs in adulthood (Humphrey et al.,<br />

96


2002). Interestingly, dynamic alterations in neurogenesis are correlated with this cell<br />

loss; when more hippocampal cells are lost, the rate of neurogenesis in the dentate gyrus<br />

is increased (Dong et al., 2003b). These results obtained with i.e.v. administration<br />

suggest that hippocampal circuits may be particularly vulnerable to alterations caused <strong>by</strong><br />

KA during the end of the first postnatal week. It is important to note that the histological<br />

methods used in the present study preclude a detailed analysis of subtle changes in<br />

hippocampal circuits such as those discussed above following i.e.v. administration.<br />

However, given the findings of Lynch and colleagues (2000) and those using i.e.v.<br />

administration of KA, the changes in PPI observed in the present study may be attributed<br />

to alterations in the development of the hippocampus, particularly the dentate gyrus.<br />

This hypothesis receives further support from studies in adult rats that confirm a<br />

role for the hippocampus and dentate gyrus in the neural circuitry that regulates PPI. For<br />

example, stimulation or deactivation of the hippocampus disrupts PPI (for review see<br />

Bast and Feldon, 2003), and infusion of the cholinergic agonist carbachol into the dentate<br />

gyrus disrupts PPI (Caine et al., 1991; 1992), an effect that is not reversed <strong>by</strong><br />

pretreatment with the D2 antagonist spiperone (Caine et al., 1991). Additionally, reduced<br />

synaptophysin immunoreactivity (Varty et al., 1999) and alterations in specific<br />

interneurons (Greene et al., 2001) are observed in the dentate gyri of socially-isolated rats<br />

that exhibit disrupted PPI.<br />

Conclusion<br />

Attempts to model the etiologies of psychiatric disorders such as schizophrenia in<br />

animals are challenging. All current models have drawbacks, mainly attributable to the<br />

specific brain manipulations performed (Lipska and Weinberger, 2000;Penn, 2001 ;Van<br />

97


den Buuse et al., 2003). The data reported here demonstrate that a single period of<br />

intense cortico-limbic activity induced <strong>by</strong> the systemic administration of KA on PND7<br />

can cause a selective disruption of PPI that is only observed in adult rats. This provides<br />

further support for the hypothesis that in rats, the end of the first postnatal week is a<br />

critical period for the development of cortico-limbic circuits that mediate behaviors such<br />

as PPI and locomotor activity (Lipska et al., 1993; 1995; Wood et al., 1997;Daenen et al.,<br />

2002; 2003;Harris et al., 2003). However, the present apomorphine-PPI experiment and<br />

locomotor activity experiments do not support the notion that acute administration of KA<br />

on PND7 results in the expression of an array of behavioral changes consistent with<br />

symptoms of schizophrenia.<br />

98


References<br />

Bahn S, Volk B, Wisden W (1994) Kainate Receptor Gene-Expression in the Developing<br />

Rat-Brain. J Neurosci 14: 5525-5547.<br />

Bast T, Feldon J (2003) Hippocampal modulation of sensorimotor processes. Prog<br />

Neurobiol 70: 319-345.<br />

Ben Ari Y, Cossart R (2000) Kainate, a double agent that generates seizures: two decades<br />

of progress. Trends Neurosci 23: 580-587.<br />

Ben Ari Y, Khazipov R, Leinekugel X, Caillard O, Gaiarsa JL (1997) GABAA, NMDA<br />

and AMPA receptors: a developmentally regulated 'menage a trois'. Trends Neurosci 20:<br />

523-529.<br />

Benes FM (2000) Emerging principles of altered neural circuitry in schizophrenia. Brain<br />

Res Brain Res Rev 31: 251-269.<br />

Benes FM, Taylor JB, Cunningham MC (2000) Convergence and plasticity of<br />

monoaminergic systems in the medial prefrontal cortex during the postnatal period:<br />

implications for the development of psychopathology. Cereb Cortex 10: 1014-1027.<br />

Beninger RJ, Jhamandas A, Aujla H, Xue L, Dagnone RV, Boegman RJ, Jhamandas K<br />

(2002) Neonatal exposure to the glutamate receptor antagonist MK-801: effects on<br />

locomotor activity and pre-pulse inhibition before and after sexual maturity in rats.<br />

Neurotox Res 4: 477-488.<br />

Bogerts B (1997) The temporolimbic system theory of positive schizophrenic symptoms.<br />

Schizophr Bull 23: 423-435.<br />

Brake WG, Flores G, Francis D, Meaney MJ, Srivastava LK, Gratton A (2000) Enhanced<br />

nucleus accumbens dopamine and plasma corticosterone stress responses in adult rats<br />

with neonatal excitotoxic lesions to the medial prefrontal cortex. Neuroscience 96: 687-<br />

695.<br />

Caine SB, Geyer MA, Swerdlow NR (1991) Carbachol infusion into the dentate gyrus<br />

disrupts sensorimotor gating of startle in the rat. Psychopharmacology (Berl) 105: 347-<br />

354.<br />

Caine SB, Geyer MA, Swerdlow NR (1992) Hippocampal modulation of acoustic startle<br />

and prepulse inhibition in the rat. Pharmacol Biochem Behav 43: 1201-1208.<br />

Daenen EW, Van Der Heyden JA, Kruse CG, Wolterink G, Van Ree JM (2001)<br />

Adaptation and habituation to an open field and responses to various stressful events in<br />

animals with neonatal lesions in the amygdala or ventral hippocampus. Brain Res 918:<br />

153-165.<br />

99


Daenen EW, Wolterink G, Gerrits MA, Van Ree JM (2002) Amygdala or ventral<br />

hippocampal lesions at two early stages of life differentially affect open field behaviour<br />

later in life; an animal model of neurodevelopmental psychopathological disorders.<br />

Behav Brain Res 131: 67-78.<br />

Daenen EW, Wolterink G, Van Der Heyden JA, Kruse CG, Van Ree JM (2003) Neonatal<br />

lesions in the amygdala or ventral hippocampus disrupt prepulse inhibition of the acoustic<br />

startle response; implications for an animal model of neurodevelopmental disorders like<br />

schizophrenia. Eur Neuropsychopharmacol 13: 187-197.<br />

de Feo MR, Mecarelli O, Palladini G, Ricci GF (1986) Long-term effects of early status<br />

epilepticus on the acquisition of conditioned avoidance behavior in rats. Epilepsia 27:<br />

476-482.<br />

Dong H, Csernansky CA, Chu Y, Csernansky JG (2003a) Intracerebroventricular kainic<br />

acid administration to neonatal rats alters interneuron development in the hippocampus.<br />

Brain Res Dev Brain Res 145: 81-92.<br />

Dong H, Csernansky CA, Goico B, Csernansky JG (2003b) Hippocampal neurogenesis<br />

follows kainic acid-induced apoptosis in neonatal rats. J Neurosci 23: 1742-1749.<br />

Ellenbroek BA, Cools AR (2000) The long-term effects of maternal deprivation depend<br />

on the genetic background. Neuropsychopharmacology 23: 99-106.<br />

Feldman DE, Knudsen EI (1998) Experience-dependent plasticity and the maturation of<br />

glutamatergic synapses. Neuron 20: 1067-1071.<br />

Flores G, Wood GK, Liang JJ, Quirion R, Srivastava LK (1996) Enhanced amphetamine<br />

sensitivity and increased expression of dopamine D2 receptors in postpubertal rats after<br />

neonatal excitotoxic lesions of the medial prefrontal cortex. J Neurosci 16: 7366-7375.<br />

Friston KJ (1999) Schizophrenia and the disconnection hypothesis. Acta Psychiatr Scand<br />

Suppl 395: 68-79.<br />

Geyer MA, Krebs-Thomson K, Braff DL, Swerdlow NR (2001) Pharmacological studies<br />

of prepulse inhibition models of sensorimotor gating deficits in schizophrenia: a decade<br />

in review. Psychopharmacology (Berl) 156: 117-154.<br />

Gothelf D, Soreni N, Nachman RP, Tyano S, Hiss Y, Reiner O, Weizman A (2000)<br />

Evidence for the involvement of the hippocampus in the pathophysiology of<br />

schizophrenia. Eur Neuropsychopharmacol 10: 389-395.<br />

Greene JR, Kerkhoff JE, Guiver L, Totterdell S (2001) Structural and functional<br />

abnormalities of the hippocampal formation in rats with environmentally induced<br />

reductions in prepulse inhibition of acoustic startle. Neuroscience 103: 315-323.<br />

Hall FS (1998) Social deprivation of neonatal, adolescent, and adult rats has distinct<br />

neurochemical and behavioral consequences. CritRev Neurobiol 12: 129-162.<br />

100


Harris LW, Sharp T, Gartlon J, Jones DN, Harrison PJ (2003) Long-term behavioural,<br />

molecular and morphological effects of neonatal NMDA receptor antagonism. Eur J<br />

Neurosci 18: 1706-1710.<br />

Harrison PJ (1999) The neuropathology of schizophrenia. A critical review of the data<br />

and their interpretation. Brain 122 (Pt 4): 593-624.<br />

Heckers S, Rauch SL, Goff D, Savage CR, Schacter DL, Fischman AJ, Alpert NM (1998)<br />

Impaired recruitment of the hippocampus during conscious recollection in schizophrenia.<br />

Nat Neurosci 1: 318-323.<br />

Humphrey WM, Dong H, Csernansky CA, Csernansky JG (2002) Immediate and delayed<br />

hippocampal neuronal loss induced <strong>by</strong> kainic acid during early postnatal development in<br />

the rat. Brain Res Dev Brain Res 137: 1-12.<br />

Jones EG (1997) Cortical development and thalamic pathology in schizophrenia.<br />

Schizophr Bull 23: 483-501.<br />

Khalilov I, Dzhala V, Medina I, Leinekugel X, Melyan Z, Lamsa K, Khazipov R, Ben<br />

Ari Y (1999) Maturation of kainate-induced epileptiform activities in interconnected<br />

intact neonatal limbic structures in vitro. Eur J Neurosci 11: 3468-3480.<br />

Kidd FL, Coumis U, Collingridge GL, Crabtree JW, Isaac JT (2002) A presynaptic<br />

kainate receptor is involved in regulating the dynamic properties of thalamocortical<br />

synapses during development. Neuron 34: 635-646.<br />

Kidd FL, Isaac JT (1999) Developmental and activity-dependent regulation of kainate<br />

receptors at thalamocortical synapses. Nature 400: 569-573.<br />

Koh S, Storey TW, Santos TC, Mian AY, Cole AJ (1999) Early-life seizures in rats<br />

increase susceptibility to seizure-induced brain injury in adulthood. Neurology 53: 915-<br />

921.<br />

Le Pen G, Moreau JL (2002) Disruption of prepulse inhibition of startle reflex in a<br />

neurodevelopmental model of schizophrenia: reversal <strong>by</strong> clozapine, olanzapine and<br />

risperidone but not <strong>by</strong> haloperidol. Neuropsychopharmacology 27: 1-11.<br />

Lipska BK, al Amin HA, Weinberger DR (1998) Excitotoxic lesions of the rat medial<br />

prefrontal cortex. Effects on abnormal behaviors associated with neonatal hippocampal<br />

damage. Neuropsychopharmacology 19: 451-464.<br />

Lipska BK, Halim ND, Segal PN, Weinberger DR (2002) Effects of reversible<br />

inactivation of the neonatal ventral hippocampus on behavior in the adult rat. J Neurosci<br />

22: 2835-2842.<br />

Lipska BK, Jaskiw GE, Weinberger DR (1993) Postpubertal emergence of<br />

hyperresponsiveness to stress and to amphetamine after neonatal excitotoxic hippocampal<br />

damage: a potential animal model of schizophrenia. Neuropsychopharmacology 9: 67-75.<br />

101


Lipska BK, Swerdlow NR, Geyer MA, Jaskiw GE, Braff DL, Weinberger DR (1995)<br />

Neonatal excitotoxic hippocampal damage in rats causes post-pubertal changes in<br />

prepulse inhibition of startle and its disruption <strong>by</strong> apomorphine. Psychopharmacology<br />

(Berl) 122: 35-43.<br />

Lipska BK, Weinberger DR (2000) To model a psychiatric disorder in animals:<br />

schizophrenia as a reality test. Neuropsychopharmacology 23: 223-239.<br />

Luthi A, Schwyzer L, Mateos JM, Gahwiler BH, McKinney RA (2001) NMDA receptor<br />

activation limits the number of synaptic connections during hippocampal development.<br />

Nat Neurosci 4: 1102-1107.<br />

Lynch M, Sayin U, Bownds J, Janumpalli S, Sutula T (2000) Long-term consequences of<br />

early postnatal seizures on hippocampal learning and plasticity. Eur J Neurosci 12: 2252-<br />

2264.<br />

Martinez ZA, Halim ND, Oostwegel JL, Geyer MA, Swerdlow NR (2000) Ontogeny of<br />

phencyclidine and apomorphine-induced startle gating deficits in rats. Pharmacol<br />

Biochem Behav 65: 449-457.<br />

McClure RK, Weinberger DR (2001) The neurodevelopmental hypothesis of<br />

schizophrenia: A review of the evidence. In: Current Issues in the Psychopharmacology<br />

of Schizophrenia. (Breier A, Tran PV, Herrea JM, Tollefson GD, Bymaster FP, eds), pp<br />

27-56. New York: Lippincott Williams & Wilkins.<br />

Meyer-Lindenberg A, Miletich RS, Kohn PD, Esposito G, Carson RE, Quarantelli M,<br />

Weinberger DR, Berman KF (2002) Reduced prefrontal activity predicts exaggerated<br />

striatal dopaminergic function in schizophrenia. Nat Neurosci 5: 267-271.<br />

Montgomery EM, Bardgett ME, Lall B, Csernansky CA, Csernansky JG (1999) Delayed<br />

neuronal loss after administration of intracerebroventricular kainic acid to preweanling<br />

rats. Brain Res Dev Brain Res 112: 107-116.<br />

Morris RG, Schenk F, Tweedie F, Jarrard LE (1990) Ibotenate Lesions of Hippocampus<br />

and/or Subiculum: Dissociating Components of Allocentric Spatial Learning. Eur J<br />

Neurosci 2: 1016-1028.<br />

Nitecka L, Tremblay E, Charton G, Bouillot JP, Berger ML, Ben Ari Y (1984)<br />

Maturation of kainic acid seizure-brain damage syndrome in the rat. II. Histopathological<br />

sequelae. Neuroscience 13: 1073-1094.<br />

Pakkenberg B (1990) Pronounced reduction of total neuron number in mediodorsal<br />

thalamic nucleus and nucleus accumbens in schizophrenics. Arch Gen Psychiatry 47:<br />

1023-1028.<br />

Penn AA (2001) Early brain wiring: activity-dependent processes. Schizophr Bull 27:<br />

337-347.<br />

102


Silveira DC, Sogawa Y, Holmes GL (2002) The expression of Fos following kainic acidinduced<br />

seizures is age-dependent. Eur J Neurosci 15: 329-344.<br />

Sperk G (1994) Kainic acid seizures in the rat. Prog Neurobiol 42: 1-32.<br />

Stafstrom CE, Chronopoulos A, Thurber S, Thompson JL, Holmes GL (1993) Agedependent<br />

cognitive and behavioral deficits after kainic acid seizures. Epilepsia 34: 420-<br />

432.<br />

Staf strom CE, Thompson JL, Holmes GL (1992) Kainic acid seizures in the developing<br />

brain: status epilepticus and spontaneous recurrent seizures. Brain Res Dev Brain Res 65:<br />

227-236.<br />

Stevens JR (1994) Brain atrophy or dystrophy in schizophrenia: when did it happen?<br />

Arch Gen Psychiatry 51: 927.<br />

Suddath RL, Christison GW, Torrey EF, Casanova MF, Weinberger DR (1990)<br />

Anatomical abnormalities in the brains of monozygotic twins discordant for<br />

schizophrenia. N Engl J Med 322: 789-794.<br />

Swerdlow NR, Martinez ZA, Hanlon FM, Platten A, Farid M, Auerbach P, Braff DL,<br />

Geyer MA (2000) Toward understanding the biology of a complex phenotype: rat strain<br />

and substrain differences in the sensorimotor gating-disruptive effects of dopamine<br />

agonists. J Neurosci 20: 4325-4336.<br />

Swerdlow NR, Platten A, Kim YK, Gaudet I, Shoemaker J, Pitcher L, Auerbach P (2001)<br />

Sensitivity to the dopaminergic regulation of prepulse inhibition in rats: evidence for<br />

genetic, but not environmental determinants. Pharmacol Biochem Behav 70: 219-226.<br />

Tremblay E, Nitecka L, Berger ML, Ben Ari Y (1984) Maturation of kainic acid seizurebrain<br />

damage syndrome in the rat. I. Clinical, electrographic and metabolic observations.<br />

Neuroscience 13: 1051-1072.<br />

Van den BM, Garner B, Koch M (2003) Neurodevelopmental animal models of<br />

schizophrenia: effects on prepulse inhibition. Curr Mol Med 3: 459-471.<br />

Varty GB, Geyer MA (1998) Effects of isolation rearing on startle reactivity, habituation,<br />

and prepulse inhibition in male Lewis, Sprague-Dawley, and Fischer F344 rats. Behav<br />

Neurosci 112: 1450-1457.<br />

Varty GB, Marsden CA, Higgins GA (1999) Reduced synaptophysin immunoreactivity in<br />

the dentate gyrus of prepulse inhibition-impaired isolation-reared rats. Brain Res 824:<br />

197-203.<br />

Weinberger DR (1995) Schizophrenia as a neurodevelopmental disorder: A review of the<br />

concept. In: Schizophrenia (Hirsch SR, Weinberger DR, eds), pp 293-323. London:<br />

Blackwood.<br />

103


Weiss IC, Di Iorio L, Feldon J, Domeney AM (2000) Strain differences in the isolationinduced<br />

effects on prepulse inhibition of the acoustic startle response and on locomotor<br />

activity. Behav Neurosci 114: 364-373.<br />

Wood GK, Lipska BK, Weinberger DR (1997) Behavioral changes in rats with early<br />

ventral hippocampal damage vary with age at damage. Brain Res Dev Brain Res 101: 17-<br />

25.<br />

Yee BK, Chang DL, Feldon J (2004a) The Effects of dizocilpine and phencyclidine on<br />

prepulse inhibition of the acoustic startle reflex and on prepulse-elicited reactivity in<br />

C57BL6 mice. Neuropsychopharmacology 29: 1865-1877.<br />

Yee BK, Russig H, Feldon J (2004b) Apomorphine-induced prepulse inhibition<br />

disruption is associated with a paradoxical enhancement of prepulse stimulus reactivity.<br />

Neuropsychopharmacology 29: 240-248.<br />

104


CHAPTER FOUR: BEHAVIORAL CONVULSIONS INDUCED BY EARLY<br />

POSTNATAL ADMINIST<strong>RAT</strong>ION OF <strong>THE</strong> NR2B ANTAGONIST R025-6981<br />

FAIL <strong>TO</strong> AFFECT SENSORIMO<strong>TO</strong>R GATING OR LOCOMO<strong>TO</strong>R BEHAVIOR<br />

IN PRE-AND POST-PUBESCENT <strong>RAT</strong>S. 3<br />

Introduction<br />

Schizophrenia is a complex disorder resulting from multiple etiological risk<br />

factors. Adverse events during early development have been proposed to contribute to<br />

the etiology of schizophrenia (Weinberger, 1987). Supporting evidence includes<br />

increased risk of developing schizophrenia following perinatal events such as maternal<br />

infection and obstetrical complications (for review see Lewis and Levitt, 2002).<br />

Additionally, subtle signs of cognitive and behavioral impairment are present during<br />

childhood and early adolescence in those who go on to develop schizophrenia in<br />

adulthood (Lewis and Levitt, 2002). These observations, among others, led to the<br />

proposal that the primary pathology of schizophrenia occurs during early development<br />

and its effects somehow remain largely silent until adulthood (Weinberger, 1987;Lewis<br />

and Levitt, 2002). The effects of disturbing the early neural development of rats on<br />

behavioral measures relevant to schizophrenia are being examined to test this hypothesis.<br />

During early postnatal life, glutamate neurotransmission, particularly via NMDA<br />

receptors, is critical for the formation of neural circuits due to its established role in<br />

processes such as synaptogenesis (Rabacchi et al., 1992;Fox et al., 1996;Luthi et al.,<br />

2001), cell birth (Gould et al., 1994), and cell survival (Vallano, 1998;Ikonomidou et al,<br />

1999). Abnormalities of the glutamate system, with an emphasis On NMDA<br />

hypofunction, have been linked to patients with schizophrenia (Jentsch and Roth,<br />

3<br />

A version of this Chapter will be submitted for publication: Howland JG, Choi FY, Phillips AG (2005)<br />

Behavioral convulsions induced <strong>by</strong> early postnatal administration of the NR2B antagonist Ro25-<br />

6981 fail to affect sensorimotor gating or locomotor behavior in pre- and post-pubescent rats.<br />

105


1999;01ney et al., 1999;Goff and Coyle, 2001;Coyle and Tsai, 2004). Accordingly,<br />

previous studies have administered non-competitive NMDA antagonists such as<br />

phencyclidine, ketamine, and MK-801 to neonatal rats and demonstrated alterations in<br />

several behavioral paradigms including prepulse inhibition (PPI; (Wang et al.,<br />

2001;Harris et al., 2003), locomotor activity (Facchinetti et al, 1993;Wang et al.,<br />

2001;Harris et al., 2003;Fredriksson et al., 2004), stereotypy (Semba et al., 2001), set-<br />

shifting (Stefani and Moghaddam, 2005), and spatial memory (Gorter and de Bruin,<br />

1992;Wang et al., 2001;Fredriksson et al., 2004;Stefani and Moghaddam, 2005).<br />

Importandy, blocking NMDA receptor function at the end of the first postnatal week<br />

causes extensive apoptosis throughout forebrain areas implicated in schizophrenia such as<br />

the hippocampus, prefrontal cortex, striatum, and thalamus (Ikonomidou et al.,<br />

1999;Wang et al., 2001;Harris et al., 2003;Fredriksson et al, 2004) there<strong>by</strong> providing a<br />

potential mechanism that may underlie the behavioral changes observed in adulthood.<br />

Recently, an explosion in knowledge surrounding the composition of NMDA<br />

receptors and their subunit composition has emerged (Cull-Candy et al, 2001). This<br />

provides a unique opportunity to examine the potential role of various subclasses of the<br />

receptors in disorders such as schizophrenia. NMDA receptors are heterodimers<br />

composed of a variety of subunits (Fig. 4-1) and over the course of development, the<br />

expression levels of some subunits undergo dynamic change. At birth, NR2B subunits<br />

are expressed at high levels, whereas expression of NR2A subunit levels is low. Over the<br />

first two to three weeks of development, this pattern reverses, and during late adolescence<br />

and into adulthood, NMDA receptors containing NR2A subunits predominate (Monyer et<br />

al, 1994;Guilarte and McGlothan, 1998;Ritter et al., 2002;Zhang et al., 2002). As the<br />

106


Figure 4-1. Cartoon illustrating an NMDA receptor within the cell membrane. NMDA<br />

receptors are assemblies of various subunits including NR1, NR2A to NR2D, and NR3A<br />

and NR3B. Functional receptors contain two NR1 subunits and some combination of two<br />

additional subunits. As is depicted, the binding site for non-competitive NMDA<br />

antagonists such as MK-801 and phencyclidine is located within the pore of the receptor.<br />

Thus, the receptor must be open in order for these antagonists to bind. In the present<br />

experiments, the NR2B-specific antagonists Ro25-6981 and ifenprodil were used, which<br />

have been reported to bind in the region of the polyamine site. Interestingly, these drugs<br />

only bind to receptors termed diheteromers that contain two NR1 subunits and two NR2B<br />

subunits. In this manner, their high degree of specificity is achieved.<br />

107


subunit composition of NMDA receptors determine their precise kinetics, the unique<br />

subunit composition of NMDA receptors during the neonatal period is likely to be an<br />

important factor in normal development (Cull-Candy et al., 2001).<br />

Interestingly, numerous lines of evidence suggest that subunit-specific changes of<br />

the NMDA receptor may exist in schizophrenia: (1) significant increases in the<br />

expression of NR2B subunits have been noted in the hippocampus (Gao et al., 2000),<br />

thalamus (Clinton and Meador-Woodruff, 2004), and temporal cortex (Grimwood et al.,<br />

1999) of schizophrenic patients, (2) increased expression of NR2D in the prefrontal<br />

cortex (Akbarian et al., 1996) and decreases in NR1 expression in the hippocampus (Gao<br />

et al., 2000) have been demonstrated, (3) novel polymorphisms and variants in the<br />

promoter regions of the NR2A (Itokawa et al, 2003) and NR2B genes (Miyatake et al.,<br />

2002; Qin et al., 2005) may also exist in schizophrenia, (4) behavioral and neurochemical<br />

abnormalities consistent with the disorder are observed in mice with genetically altered<br />

NR1 (Mohn et al., 1999;Duncan et al., 2004) and NR2A (Miyamoto et al., 2001)<br />

subunits. Therefore, in an effort to further understand the effects of transiently disrupting<br />

glutamate receptors containing specifically NR2B subunits, we administered the subunit-<br />

selective NMDA antagonist [(+/-)-(R*,S*)-alpha-(4-hydroxyphenyl)-beta-methyl-4-<br />

(phenylmethyl)-l-piperidine propanol] (Ro25-6981) to neonatal rats on postnatal days<br />

(PND) 6 and 7.<br />

Ro25-6981 is a high affinity non-competitive antagonist of receptors containing<br />

the NR2B subunit (Fischer et al., 1997). As NR2B-containing NMDA receptors are<br />

overexpressed in the neonatal period, administration of Ro25-6981 provides a unique<br />

opportunity to assess the importance of these specific receptors in the maturation of the<br />

108


neural circuits that mediate behaviors with potential relevance to schizophrenia.<br />

Importantly, Ro25-6981 has low affinities for a-adrenergic and serotoninergic receptors,<br />

a problem recognized for other NR2B antagonists such as ifenprodil (Mutel et al., 1998).<br />

Following neonatal treatment, rats were tested for PPI and locomotor responses to a novel<br />

environment and amphetamine challenge both before and after puberty. These behavioral<br />

tests are extensively used to model the information processing deficits and striatal<br />

dopamine elevations observed in patients with schizophrenia (Lipska and Weinberger,<br />

2000;Braff et al., 2001 ;van den et al., 2003;Bast and Feldon, 2003). Finally, the<br />

disruptive effect of the dopamine agonist apomorphine on PPI was also assessed in a sub­<br />

group of adult rats.<br />

Unexpectedly, the dose of the Ro25-6981 used (15 mg/kg) caused behavioral<br />

convulsions when administered on PND 6. As is detailed in the methods section, this<br />

dose has been used extensively in adult rats without reports of convulsive activity, and<br />

was chosen based on "affinity estimates for NR2B-containing receptors. Due to this<br />

observation, we also performed a series of parametric studies to further examine the<br />

convulsive effects of a number of NMDA antagonists during early development.<br />

Additionally, previous work performed in our laboratory demonstrated that seizures<br />

induced <strong>by</strong> kainic acid on PND 7 resulted in the delayed expression of PPI deficits in rats<br />

(Howland et al., 2004). From this standpoint, the long-term behavioral effects of<br />

convulsions following Ro25-6981 were of considerable interest.<br />

109


Methods<br />

Experiment 1 - Incidence and Characteristics of Behavioral Convulsions in Postnatal<br />

Rats Following Antagonism of NMDA Receptors Containing the NR2B Subunit<br />

Subjects<br />

Long-Evans or Sprague-Dawley rat pups were used in all experiments. Pregnant<br />

Long-Evans rats were obtained from Charles River (Quebec, Canada) at 13 to 15 days of<br />

gestation. They were singly housed and left undisturbed until giving birth. The colony<br />

was maintained on a 12/12 hour light/dark cycle (lights on at 0700), at a temperature of<br />

22±1°C. All rats were given food (Purina Rat Chow) and tap water ad libitum. Sprague-<br />

Dawley rat pups were obtained from the University of British Columbia Animal Care<br />

Center. The pups were delivered to our animal colony on the desired postnatal day<br />

without their mother. All experiments were initiated within 60 min of the arrival of the<br />

rat pups in the laboratory. Experiments were conducted in accordance with the standards<br />

of the Canadian Council on Animal Care and were approved <strong>by</strong> the Committee on<br />

Animal Care at the University of British Columbia.<br />

Experimental Procedures<br />

The day of birth of the pups was designated PND 0. Rat pups of the appropriate<br />

age (PND 6, 9, 12,15) were weighed and separated into individual compartments of a<br />

cardboard box for drug administration. The pups were removed from the colony and<br />

taken to a room where the behavioral effects of the administered drugs were videotaped<br />

and scored <strong>by</strong> an observer. The subunit-selective NMDA-NR2B antagonists Ro25-6981<br />

(7.5, 15, and 30 mg/kg) and ifenprodil (15 and 30 mg/kg) were injected (10 ml/kg; i.p.)<br />

and the behavior of the animals was videotaped for 60 minutes. Reports indicate that 15<br />

110


mg/kg of Ro25-6981 is in the dose range necessary to block most NR2B-containing<br />

receptors in the adult rodent brain (Murray et al., 2000;Lee and Rajakumar, 2003), and<br />

doses of 20 to 30 mg/kg have been used in vivo (Chaperon et al., 2003;Higgins et al.,<br />

2005). Similar data exists for ifenprodil (Murray et al., 2000). Following the<br />

experiment, the pups were sacrificed with carbon dioxide. All drugs were obtained from<br />

Sigma.<br />

Experiment 2 - Long-term Behavioral Effects of Convulsions Resulting from Antagonism<br />

of NMDA Receptors Containing the NR2B Subunit<br />

Subjects<br />

Two independent groups of Long-Evans rats (i.e. Group 1 and Group 2) were<br />

tested in this study. The animals were obtained, housed and cared for in a manner<br />

identical to that described in Experiment 1. Testing protocols for both groups were<br />

similar with small procedural differences noted in the methods section where appropriate.<br />

Ro25-6981 Administration<br />

On PND3, the litters were sexed and culled to include only males (4-9 rats per<br />

litter). At 4:00 p.m. on the afternoon of PND 6 and 9:00 a.m. and 4:00 p.m. of PND 7, all<br />

pups were removed from the nest, weighed and placed individually in small<br />

compartments of a cardboard box. They were then removed from the colony and taken to<br />

a small heated room. Ro25-6981 (15 mg/kg) or saline was injected (i.p.) with a 30-gauge<br />

needle (10 ml/kg). Care was taken to ensure that both treatments were administered to<br />

members of each litter. The behavior of all rats was recorded with an overhead video<br />

camera for 60 min. Before being returned to their mothers, the pups were earmarked<br />

according to treatment condition. The litters were then left undisturbed (except for<br />

111


normal cage changing) until weaning on PND 25. Weanling rats were housed in cages of<br />

2 or 3 with members of their litter and these groups remained together for the duration of<br />

the experiment. All rats were handled before behavioral testing.<br />

Prepulse Inhibition<br />

On PND 35 and 56, rats were removed individually from the colony and taken<br />

immediately to the PPI apparatus. Testing was conducted in a single sound-attenuating<br />

startle chamber (ambient noise level 64 dB), containing a transparent Plexiglas tube (8.2<br />

cm in diameter, 20 cm in length), mounted on a Plexiglas frame (SR-LAB, San Diego<br />

Instruments, San Diego). Noise bursts were presented through a speaker mounted 24-cm<br />

above the tube. An accelerometer below the frame of the apparatus measured whole<br />

body startle amplitude, defined as the average of 100 1-ms accelerometer readings<br />

collected from stimulus onset. Each PPI test session began with a 5-min acclimatization<br />

period during which a 70-dB background noise level was presented, which remained<br />

constant for the entire test session. Following the acclimatization period, six pulse alone<br />

trials (120 dB, 40 ms) were presented to achieve a relatively stable startle amplitude<br />

before PPI testing. Data from these pulse-alone trials was not considered in the analysis<br />

of PPI. Immediately following the six initial pulse-alone trials, presentation of the trials<br />

that were used in the calculation of PPI levels were initiated. Trials presented were of<br />

four types: pulse alone (12 trials, 120 dB, 40 ms), prepulse + pulse (12 trials X 3 prepulse<br />

intensities - discussed below), prepulse alone (12 trials X 3 prepulse intensities) or no<br />

stimulus (12 trials). Prepulse + pulse trials consisted of the presentation of a 20 ms<br />

prepulse of 73, 76, or 82 dB 80 ms before the presentation of the pulse. Prepulse alone<br />

trials consisted only of a 20 ms prepulse (73, 76, 82 dB). All trials were presented in a<br />

112


pseudorandom order. After the trials used for calculation of PPI were presented, 6<br />

additional pulse-alone trials were presented. These, along with the first 6 pulse-alone<br />

trials, were used to calculate the level of habituation over the testing period. As no<br />

significant differences were observed between the groups in habituation and or for the<br />

prepulse-alone trials, these data are not presented. The inter-trial interval varied<br />

randomly from 3 to 12 s (average 7.5 s). Calibration of the apparatus was performed<br />

using a RadioShack Digital Sound Level Meter and adjustments were made as necessary.<br />

Spontaneous Locomotor Activity<br />

On PND 36, rats were weighed, removed from the colony, and immediately<br />

placed in 1 of 8 Med Associates Test Chambers (ENV-008; 30.5 cm X 24.1cm X height<br />

21.0 cm) to measure spontaneous locomotor activity for 60 min. The chambers were<br />

fitted with 4 pairs of infrared photocells 3.5 cm from the floor evenly spaced on the long<br />

walls and had metal grid floors and two operant levers (which were retracted during<br />

locomotor activity testing). Each chamber was contained within a Med Associates sound<br />

attenuating cubicle with a house light illuminated during testing.<br />

Dopaminergic Challenges of PPI and Locomotor Activity<br />

For those rats in Group 1, 7 to 14 days after PPI testing in early adulthood, the<br />

responses of the animals to administration of dopamine agonists were measured in both<br />

the PPI and locomotor activity tests. Some rats were tested for PPI first followed <strong>by</strong><br />

locomotor activity while others were tested in the reverse order. A minimum of 7 days<br />

separated locomotor activity and PPI testing. The PPI sessions were conducted in a<br />

manner identical to that described above, except that immediately before the PPI session,<br />

all rats were weighed and injected with the appropriate volume of vehicle (ascorbic acid,<br />

113


0.1%) or drug (apomorphine, Sigma, 0.2 mg/kg; s.e.). Each rat was tested twice (i.e.<br />

following either vehicle or apomorphine injection), and the PPI tests were at least 5 days<br />

apart. For locomotor activity testing, the rats were weighed, removed from the colony,<br />

and placed in the locomotor activity boxes for 60 min (as described above for PND 36).<br />

All rats were then immediately injected with D-amphetamine (1.5 mg/kg, i.p.) and<br />

returned to the locomotor boxes. Locomotor activity was monitored for an additional 90<br />

min before the rats were returned to the colony room.<br />

For rats in Group 2, PPI responses following apomorphine challenge were not<br />

measured. However, locomotor responses to D-amphetamine were examined at both<br />

PND 36 and PND 57. Rats were weighed and placed in the locomotor boxes as<br />

previously described, however, at both ages, they were injected with D-amphetamine (1.5<br />

mg/kg) immediately following the spontaneous locomotor activity test. Locomotor<br />

activity following D-amphetamine was measured for 90 min in all cases.<br />

Data Analysis<br />

For the PPI experiments, two measures were calculated for each animal. The<br />

startle amplitude represented the mean startle amplitude of the 12 pulse-alone trials<br />

presented after the 6 habituation trials. Startle amplitude data were compared using an<br />

independent samples t-test for each age. PPI was calculated <strong>by</strong> averaging startle<br />

amplitudes for each trial type. The percent PPI for each prepulse intensity was calculated<br />

using the formula: [100 - (100 X startle amplitude on prepulse + pulse trials) (startle<br />

amplitude on pulse alone trials)]. A repeated measures ANOVA was performed on the<br />

data obtained from each age with prepulse intensity as a within-subjects factor and<br />

treatment on PND 6 and 7 as a between-subjects factor. Results from the apomorphine<br />

114


experiment were also analyzed using a repeated measures ANOVA (prepulse intensity<br />

and test as within-subjects factors, and treatment at PND 7 as a between-subjects factor).<br />

Locomotor activity data were compared using repeated measures ANOVA at each age<br />

with test type as a within-subjects factor and treatment on PND 7 as a between-subjects<br />

factor. For all ANOVA's, post-hoc analyses were performed using the Neuman-Keuls<br />

test where appropriate. The significance level for all statistical tests was p < 0.05.<br />

Results<br />

Experiment 1 - Behavioral Effects of Ro25-6981 Administration in the Early Postnatal<br />

Period<br />

Approximately 5 min following injection with Ro25-6981, the majority of PND 6<br />

pups displayed behavioral convulsions. The convulsions were similar those typically<br />

described following administration of kainic acid (Howland et al., 2004) or bicuculline<br />

(Lai et al., 2002) in neonatal rats. Generally, they consisted of loss of posture,<br />

vocalizations, and rapid bursts of motor activity (especially with the hind legs). The<br />

majority of the animals showed bouts of bilateral forelimb and hindlimb clonus and/or<br />

tonus on their backs, in what has been called a 'swimming' posture <strong>by</strong> other authors<br />

(Stafstrom et al., 1993). In most cases, the behavioral convulsions subsided within 30 to<br />

60 min following injection. Interestingly, following their initial bout of convulsions,<br />

some animals would have another period of convulsive activity if they were handled <strong>by</strong><br />

the experimenter. Those pups injected repeatedly with Ro25-6981 (see Experiment 2)<br />

showed no evidence of tolerance to the convulsive effects of the drug.<br />

As depicted in Table 4-1, the convulsive effects of Ro25-6981 were age and dose<br />

dependent. Whereas all pups treated with 15 mg/kg of the drug on PND 6 had<br />

115


convulsions, only 25% of PND 9 pups, and 0% of PND 12 or 15 (using either 15 or 30<br />

mg/kg) pups displayed evidence of convulsions. However, pups injected with Ro25-<br />

6981 were more active than saline-injected controls. Additionally, none of the rats<br />

treated with 7.5 mg/kg of Ro25-6981 displayed behavioral convulsions. In an effort to<br />

rule out the possibility that the convulsions observed in the PND 6 pups were caused <strong>by</strong><br />

an unknown property of Ro25-6981 unrelated to NR2B antagonism, ifenprodil was also<br />

administered to some animals on PND 6. Similarly to the results obtained with Ro25-<br />

6981, almost all pups (89%) treated with ifenprodil had behavioral convulsions.<br />

Experiment 2 - Effects of Neonatal Ro25-6981 Administration on Body Weight<br />

As described in detail for experiment 1, administration of Ro25-6981 (15 mg/kg;<br />

Group 1: n=25; Group 2: n=16) resulted in behavioral convulsions for 30 to 60 minutes.<br />

Some individual differences in the severity of the convulsions were noted; although these<br />

did not correlate with performance on the behavioral tests performed. As a result, data<br />

from all rats is included.<br />

All rats were weighed on PND 6, twice on PND 7, and once on PND 9, 36 and 57.<br />

Repeated measures ANOVA's performed on these data indicate that administration of<br />

Ro25-6981 to the pups had no significant effect on body weight at any age (Group 1: F(l,<br />

41)=0.90, N.S.-; Group 2: F(l, 30)=0.49, N.S.). However, comparing the average weights<br />

of rats in Group 1 and Group 2 revealed that the animals in Group 1 were significandy<br />

lighter than those in Group 2 (F(l, 73)=48.97, p < 0.001). On PND 6 and 7, rats in Group<br />

1 weighed approximately 70% of those in Group 2 (PND 6: Group 1 mean = 12.16 g;<br />

Group 2 = 17.00 g). During puberty and early adulthood, the difference in weights<br />

116


Drug Postnatal Dose (i.p., Total Number Total Number<br />

Day mg/kg) Injected Exhibiting<br />

Convulsions<br />

Ro25-6981 6 7.5 8 0<br />

Ro25-6981 6 15 8 8<br />

Ro25-6981 9 15 4 1<br />

Ro25-6981 12 15 5 0(A)<br />

Ro25-6981 15 15 4 0(A)<br />

Ro25-6981 15 30 3 0(A)<br />

Ifenprodil 6 15 4 4<br />

Ifenprodil 6 30 5 4<br />

Table 4-1. The incidence of behavioral convulsions in rats administered NR2B-selective<br />

NMDA antagonists at various postnatal ages. i.p. = intraperitoneal.<br />

117


narrowed to a 10 to 15% difference (PND 36: Group 1 = 159.77 g; Group 2 = 173.58 g;<br />

PND 57: Group 1 = 339.26 g; Group 2 = 398.96 g).<br />

Neonatal Ro25-6981 Administration Does not Alter PPI Responding Either before<br />

Puberty or in Early Adulthood<br />

Before Puberty (PND 35): As is shown in Figure 4-2A and 4-3A, comparable<br />

levels of PPI was elicited in both saline- and Ro25-6981-treated rats for Groups 1 (saline:<br />

n=19, average PPI: 27.86±2%; Ro25-6981: n=25, average PPI: 26.53±3%) and 2 (saline<br />

n=16, average PPI: 32.24±3%; Ro25-6981: n=16, average PPI: 26.38±4%). Repeated<br />

measures ANOVA's performed separately for each Group revealed a significant effect of<br />

prepulse (Group 1: F(2, 84)=96.62, p < 0.001; Group 2: F(2, 60)=49.92, p < 0.001), but<br />

no significant treatment effects (Group 1: F(l, 43)=0.13, N.S.; Group 2: F(l, 30)=1.45,<br />

N.S.) or treatment <strong>by</strong> prepulse interactions (Group 1: F(2, 84)=0.08, N.S.; Group 2: F(2,<br />

60)=0.14, N.S.). Figures 4-2B and 4-3B depict the average startie amplitudes for all<br />

animals tested in Groups 1 and 2. Independent samples t-tests revealed no significant<br />

differences between saline- and Ro25-6981-treated rats in either Group (Group 1:<br />

t(42)=1.35, N.S.; Group 2: t(30)=0.03, N.S.).<br />

Early Adulthood (PND 56): Similar to before puberty, all groups tested<br />

demonstrated robust PPI in early adulthood (Fig. 4-2C and 4-3C), and no significant<br />

differences were observed between animals treated with either saline (average PPI, Group<br />

1: 30.60±3%; Group 2: 31.74±3%) or Ro25-6981 (average PPI, Group 1: 28.33±3%;<br />

Group 2: 37.00±5%). Repeated measures ANOVA's confirmed these observations with<br />

no significant treatment (Group 1: F(l, 42)=0.29, N.S.; Group 2: F(l, 30)=0.68, N.S.) or<br />

treatment <strong>by</strong> prepulse interactions (Group 1: F(2, 84)=0.63, N.S.; Group 2: F(2, 60)=0.69,<br />

118


Q_<br />

Q.<br />

:


pp3 pp6 pp12 average<br />

Prepulse Intensity :<br />

pp3 pp6 pp12<br />

Prepulse Intensity average<br />

B<br />

160i<br />

D<br />

160!<br />

1120<br />

I 80]<br />

I 40]<br />

saline Ro<br />

Group<br />

saline Ro<br />

Group<br />

Figure 4-3. Prepulse inhibition (PPI) scores from rats in Group 2 treated on postnatal day<br />

(PND) 6 and 7 with saline (white bars, n=16) or Ro25-6981 (Ro, 15mg/kg; black bars,<br />

n=16). A, Effects of postnatal day (PND) 7 KA treatment on PPI at PND 35. B, Average<br />

starde amplitudes of the rats on PND 35. C, Percent PPI scores for those rats tested at<br />

PND 56. D, Average startle amplitudes of the rats on PND 56.<br />

120


N.S.)- As expected, significant main effects for prepulse intensity were found for both<br />

groups (Group 1: F(2, 84)=70.90, p < 0.001; Group 2: F(2, 60)=32.71, p < 0.001).<br />

Additionally, startle amplitudes did not differ as a result of treatment in either Group 1<br />

(Fig. 4-2D; t(42)=l .05, N.S.) or Group 2 (Fig. 4-3D; t(30)=0.73, N.S.)<br />

Apomorphine Challenge in Adulthood has Similar Effects on PPI in Neonatally Saline- or<br />

Ro25-6981 -treated Rats<br />

To examine the potential effects of neonatal Ro25-6981 treatment on the<br />

dopaminergic regulation of PPI, 16 saline-treated and 18 Ro25-6981 -treated rats from<br />

Group 1 were retested for PPI following injection with either vehicle (0.1% ascorbic acid<br />

in saline) or apomorphine (0.2 mg/kg). Figure 4-4A summarizes the results of these tests.<br />

Following vehicle injection, PPI levels in both saline- and Ro25-6981-treated rats were<br />

similar to those observed when PPI was tested on PND 56. Apomorphine (0.2 mg/kg)<br />

significantly disrupted PPI in both the saline- and Ro25-6981-treated rats, as reflected <strong>by</strong><br />

a significant main effect of test (vehicle versus apomorphine; F(l, 32)=31.18, p < 0.01)<br />

and a significant test <strong>by</strong> prepulse interaction (F(2, 64)=10.75, p < 0.001, post-hoc, p <<br />

0.05). Additional analyses revealed that significant effects only existed for trials with 6<br />

and 12 dB prepulses in both groups (p < 0.05). As shown in Fig. 4-4B, startle amplitude<br />

was also significantly reduced <strong>by</strong> apomorphine administration in both groups (F(l,<br />

32)=8.59, p < 0.01).<br />

Neonatal Treatment with Ro25-6981 and Amphetamine-Induced Locomotor Activity in<br />

Adult Rats<br />

Group 1: Spontaneous locomotor activity in response to a novel environment was<br />

assessed for 60 min in the saline- and Ro25-6981-treated rats. Both before puberty (Fig.<br />

121


GL<br />

D-:<br />

a><br />

70-<br />

60-<br />

50-<br />

40-<br />

30-<br />

20 \<br />

lO-<br />

CI<br />

A<br />

asal+veh<br />

•Ro+veh<br />

•sal+apol<br />

oRo+apo<br />

Condition<br />

average<br />

sal+ Ro+ sal+ Ro+<br />

veh veh apo apo<br />

Group<br />

Figure 4-4. A, Effects of pretreatment with vehicle (0.1% ascorbic acid) or apomorphine<br />

(0.2 mg/kg) on percent PPI scores in adulthood. Animals were administered either saline<br />

(n=16) or Ro25-6981 (15 mg/kg; n=18) on postnatal day 6 and 7. B, Average starde<br />

amplitudes of those rats depicted in panel A. In both panels, asterisks indicate a<br />

significant difference between groups.<br />

122


Figure 4-5. Locomotor activity levels (photobeam breaks) in response to novelty (A, B<br />

left side) or amphetamine (1.5 mg/kg; B right side) of rats tested on either postnatal day<br />

(PND) 36 (A) or PND 57 (B) in Group 1. Rats were treated with either saline (white<br />

bars) or Ro25-6981 (black bars, 15 mg/kg) on PND 6 and 7. The asterisk denotes a<br />

significant difference between amphetamine-treated groups (p


Figure 4-6. Locomotor activity levels (photobeam breaks) in response to novelty or<br />

amphetamine (1.5 mg/kg) of rats tested on either postnatal day (PND) 36 (A) or PND 57<br />

(B) in Group 2. Rats were treated with either saline (white bars, n = 12) or Ro25-6981<br />

(black bars, 15 mg/kg, n = 12) on PND 6 and 7. Note the difference in scale for the units<br />

of activity levels (y-axis) for each panel.<br />

124


4-5A) and in early adulthood (Fig. 4-5B), exploration levels were similar in both groups<br />

(before puberty: t(42)=0.45, N.S.; early adulthood: see ANOVA below). Interestingly,<br />

administration of the dopamine agonist D-amphetamine (1.5 mg/kg) further increased<br />

locomotor activity <strong>by</strong> approximately 28% in rats neonatally treated with Ro25-6981<br />

(2193.75 ± 143 beam breaks) than saline (1712.67 ± 133 beam breaks) during the 90 min<br />

test (Fig. 4-5B). This effect was confirmed <strong>by</strong> a significant treatment <strong>by</strong> test interaction<br />

(F(l, 22)=7.83, p < 0.05), and post-hoc analyses (p < 0.05).<br />

Group 2: Spontaneous and amphetamine-induced locomotor activity was compared<br />

between treatment groups both before puberty (Fig. 4-6A) and in early adulthood (Fig. 4-<br />

6B) for those rats in Group 2. Unexpectedly, the groups did not significandy differ on<br />

either test at either age. Statistical analyses revealed insignificant main effects of<br />

treatment (PND 36: F(l, 22)= 0.27, N.S.; PND 57: 0.18, N.S.) and treatment <strong>by</strong> test<br />

interactions (PND 36: F(l, 22)=0.12, N.S.; PND 57: F(l, 22)=0.18, N.S.).<br />

Discussion<br />

The present experiments assessed the acute and delayed behavioral effects of the<br />

administration of Ro25-6981, an antagonist of NR2B-containing NMDA receptors, in<br />

rats. During PND 6 or 7, administration of Ro25-6981 resulted in behavioral convulsions<br />

that typically lasted 30 to 60 minutes. The convulsions were dose-dependent and could<br />

be elicited with a second NR2B antagonist, ifenprodil. Parametric experiments<br />

demonstrated that convulsions could not be reliably elicited in rats older than PND 7.<br />

The delayed behavioral effects of administering Ro25-6981 on PND 6 and 7 were<br />

inconsistent. Ro25-6981-treated rats in Group 1 performed similarly to saline-treated<br />

animals on PPI and spontaneous locomotion tests both before and after puberty.<br />

125


However, the Ro25-6981 -treated animals were significantly more active when treated<br />

with amphetamine in early adulthood, but failed to show increased sensitivity to the<br />

disruptive effects of apomorphine on PPI. In contrast, Ro25-6981-treated rats tested in<br />

Group 2 failed to show significant differences from saline-treated animals in any of the<br />

behavioral tests conducted.<br />

Antagonism of NR2B-containing NMDA Receptors Induces Behavioral Convulsions<br />

The convulsive effects of Ro25-6981 were unexpected in the present experiments;<br />

however, they appear to be reliable as they were dose-dependently elicited in two strains<br />

of rats <strong>by</strong> both Ro25-6981 and ifenprodil. Interestingly, some groups (Fischer et al.,<br />

1997;Mutel et al., 1998;Mallon et al., 2005) have described agonist-like properties of<br />

Ro25-6981 when the drug is applied in conjunction with low concentrations of NMDA.<br />

In Xenopus oocytes, Ro25-6981 (1 uM) profoundly inhibits the current elicited <strong>by</strong> 100<br />

u.M of NMDA, but surprisingly potentiates the current elicited <strong>by</strong> 1 uM of NMDA<br />

(Fischer et al., 1997). In another study using hippocampal slices from pubescent rats,<br />

Ro25-6981 (3 uM) potentiated the effects of NMDA (4 to 10 uM) on CA1 field<br />

potentials and paired-pulse interactions (Mallon et al., 2005). As also reported <strong>by</strong> Fisher<br />

et al. (1997), these effects were more significant with lower concentrations of NMDA<br />

(Mallon et al., 2005). Interestingly, the NR2A specific antagonist NVP-AAM077<br />

(Auberson et al., 2002) effectively blocked the effects of Ro25-6981 in the hippocampal<br />

slice suggesting that the NR2B subunit may reduce NMDA receptor activity <strong>by</strong> exerting<br />

tonic inhibitory effects on receptors containing the NR2A subunit (Mallon et al., 2005).<br />

A number of issues make integrating the present data with other reports in the<br />

literature challenging. Most importantly, the subunit composition of NMDA receptors<br />

126


dynamically changes over development; therefore, care must be taken when comparing<br />

data sets gathered from animals of different ages (for example, comparing the present<br />

data to those of Mallon et al., 2005). During the first postnatal week, NR2A subunits are<br />

expressed at very low levels in the rodent brain, whereas in adulthood NR2A subunits<br />

predominate (Monyer et al., 1994;Guilarte and McGlothan, 1998;Cull-Candy et al.,<br />

2001;Ritter et al., 2002;Zhang et al., 2002). Given these expression differences, it is<br />

unlikely that the observations made <strong>by</strong> Mallon and colleagues (2005) apply during the<br />

early neonatal period since very few NR2A subunits are expressed at that age.<br />

Additionally, numerous reports indicate that in vivo Ro25-6981 antagonizes NR2B<br />

containing NMDA receptors at doses within the range used in the present study (Lee and<br />

Rajakumar, 2003;Chaperon et al., 2003;De Vry and Jentzsch, 2003;Boyce-Rustay and<br />

Holmes, 2005). Therefore, the most likely explanation of the present data is that the<br />

behavioral convulsions observed resulted from the antagonist action of Ro25-6981 on<br />

NR2B-containing NMDA receptors.<br />

Although convulsions resulting from NMDA receptor antagonism are<br />

counterintuitive, they are not without precedence in the literature. For example, systemic<br />

administration of the non-competitive NMDA antagonist MK801 (0.1, 0.5, 1.0 mg/kg) to<br />

neonatal rat pups induces behavioral and electrographic seizures and exacerbates seizures<br />

caused <strong>by</strong> kainic acid (Stafstrom et al., 1997). Given that MK-801 would primarily act<br />

on NMDA receptors containing NR2B subunits at this developmental age, these results<br />

support our assertion that the neonatal brain may be prone to convulsions following the<br />

blockade of receptors containing NR2B subunits.<br />

127


Further experiments are necessary to understand the mechanism underlying the<br />

convulsive effects of neonatal NMDA receptor antagonism. Seizures are more common<br />

neonatally than later in development, an effect that may be related to differences in the<br />

net ratio of excitation and inhibition early in development (Ben Ari et al., 1997;Holmes et<br />

al, 2002). As previously described, intricate developmental regulation of NMDA<br />

receptor expression exists during the first few weeks of life. The expression patterns and<br />

function of other receptors, including GABA A and AMPA receptors, are also<br />

developmentally regulated. For instance, GABA A receptors are expressed early in the<br />

embryonic period and provide much of the excitatory drive in the developing brain until<br />

the end of the first postnatal week when NMDA and AMPA currents begin to<br />

predominate (Ben Ari et al., 1997;Holmes et al., 2002;Khazipov et al., 2004). The<br />

excitatory action of GABA is primarily due to delayed expression of the K +<br />

/Cf co-<br />

transporter KCC2, resulting in increased intracellular Cf concentrations. Thus, when<br />

GABA A channels open under these conditions, Cl" ions exit the neuron resulting in<br />

depolarization (Holmes et al., 2002). However, under some conditions, GABA A<br />

activation may also increase the resting membrane conductance sufficiently that the net<br />

effect of opening these channels is inhibitory (Leinekugel et al., 1999;Khalilov et al.,<br />

1999a;Holmes et al., 2002). As resting membrane conductance at this age is also<br />

critically dependent on glutamate receptor conductances (Khalilov et al., 1999a), one<br />

possible explanation of our observations is that antagonism of NMDA receptors with<br />

Ro25-6981 may 'tip the balance' in favor of excitation, there<strong>by</strong> triggering a convulsion.<br />

128


The Long-Term Behavioral Effects of Ro25-6981 on PPI and Locomotor Activity<br />

The levels of PPI and locomotor activity observed before and after puberty in the<br />

present study are similar to previous experiments from our group (Howland et al., 2004).<br />

Accordingly, percent PPI increased with higher prepulse intensities (Fig. 4-2 and 4-3).<br />

Average PPI levels were also lower in prepubescence than in early adulthood.<br />

Interestingly, a relatively low dose of apomorphine (0.2 mg/kg) disrupted PPI in the<br />

present study (Fig. 4-4A). This effect is consistent with previous results using the Long-<br />

Evans strain in our laboratory (Howland et al., 2004), but not others (Swerdlow et al.,<br />

2001).<br />

Although the significantly potentiated locomotor activity following amphetamine<br />

administration in the Ro25-6981-treated rats of Group 1 (Fig. 4-5B) is intriguing, the<br />

failure to replicate this effect in Group 2 (Fig. 4-6B) questions the reliability of these<br />

data. However, an interaction between Ro25-6981 administration and the factors that led<br />

to significantly reduced body weight in Group 1 may explain these behavioral changes.<br />

When the present body weights are compared to previous studies from our laboratory<br />

(Howland et al., 2004) in which Long-Evans pups were reared under identical conditions<br />

to those described herein, it is clear that the pups tested in Group 1 had abnormally low<br />

body weights. The cause of these differences cannot be confirmed, although some<br />

renovations to our facility occurred during the period which the pregnant females arrived<br />

and gave birth to the pups in Group 1. Thus, the mothers of the rats tested in Group 1<br />

may have been abnormally stressed during the pre- and postnatal period. Indeed, stress<br />

during these developmental periods has been shown to regulate dopamine-dependent<br />

behaviors such as locomotor activity in adult rats (Kofman, 2002;Brake et al., 2004).<br />

129


The present experiments fail to provide convincing evidence that neonatal<br />

treatment with Ro25-6981 results in specific behavioral changes related to schizophrenia.<br />

However, these data stand in contrast to reports of significant behavioral effects of<br />

neonatal NMDA antagonism in adulthood using non-competitive antagonists. For<br />

example, neonatal treatment with phencyclidine (PND 7, 9, and 11) reduces PPI and<br />

significantly increases locomotor activity in response to a phencyclidine challenge, both<br />

of which were reversed following antipsychotic treatment (Wang et al., 2001). Increased<br />

spontaneous locomotor activity is also observed following ketamine administration to<br />

mice on PND 10 (Fredriksson et al., 2004). Finally, neonatal administration of MK-801<br />

alters PPI (Harris et al., 2003), locomotor activity (Harris et al., 2003), and set-shifting<br />

(Stefani and Moghaddam, 2005), although the PPI and locomotor effects were not<br />

independently replicated (Beninger et al., 2002) and were only found in female animals<br />

(Harris et al, 2003).<br />

A number of differences between non-competitive NMDA antagonists and Ro25-<br />

6981 may underlie the different behavioral effects observed following their<br />

administration. For example, non-competitive NMDA antagonists have actions at<br />

dopamine D 2 and serotonin 5-HT 2 receptors, in addition to their effects of NMDA<br />

receptors (Kapur and Seeman, 2002). Importantly, the neonatal administration of non­<br />

competitive NMDA antagonists also causes significant apoptosis in areas such as the<br />

hippocampus, thalamus, cortex, and striatum 24 to 48 hours after treatment (Wang et al.,<br />

2001 ;Lai et al., 2002;Beninger et al., 2002;Harris et al., 2003-/Fredriksson et al., 2004).<br />

Given that cell loss or disarray in these areas is reported in schizophrenia (Pakkenberg,<br />

1990;Jones, 1997;Harrison, 1999;Gothelf et al., 2000), these findings strengthen the<br />

130


validity of neonatal non-competitive NMDA antagonist administration as a model of<br />

schizophrenia, and may explain their behavioral effects. It is not known whether Ro25-<br />

6981 administration causes similar neuropathological effects, although the potential<br />

exists given that various competitive and non-competitive NMDA antagonists cause<br />

apoptosis in the developing brain (Dconomidou et al., 1999).<br />

Conclusion<br />

The convulsions caused <strong>by</strong> Ro25-6981 on PND 6 and 7 failed to affect PPI or<br />

locomotor responding in the present experiments. In contrast, febrile seizures early in<br />

life are associated with increased risk for schizophrenia (Vestergaard et al., 2005) and<br />

psychosis (Kanemoto et al., 2001). Additionally, kainic acid-induced seizures on PND 7<br />

in rats result in the delayed emergence of subtle PPI disruptions in early adulthood<br />

(Howland et al., 2004) and have short and long-term effects on temporal lobe<br />

electrophysiology, especially within the hippocampus (Tremblay et al., 1984;Stafstrom et<br />

al., 1992;Khalilov et al., 1999b;Lynch et al., 2000;Silveira et al., 2002). As the<br />

electrographic characteristics of the convulsions induced <strong>by</strong> Ro25-6981 are presently<br />

unknown, experiments designed to address this issue may enable more specific<br />

hypotheses to be generated regarding the brain areas important for long-term behavioral<br />

changes following neonatal seizures and their relationship to schizophrenia.<br />

131


References<br />

Akbarian S, Sucher NJ, Bradley D, Tafazzoli A, Trinh D, Hetrick WP, Potkin SG,<br />

Sandman CA, Bunney WE, Jr., Jones EG (1996) Selective alterations in gene expression<br />

for NMDA receptor subunits in prefrontal cortex of schizophrenics. J Neurosci 16: 19-30.<br />

Auberson YP, Allgeier H, Bischoff S, Lingenhoehl K, Moretti R, Schmutz M (2002) 5-<br />

Phosphonomethylquinoxalinediones as competitive NMDA receptor antagonists with a<br />

preference for the human 1A/2A, rather than 1A/2B receptor composition. Bioorg Med<br />

Chem Lett 12: 1099-1102.<br />

Bast T, Feldon J (2003) Hippocampal modulation of sensorimotor processes. Prog<br />

Neurobiol 70: 319-345.<br />

Ben Ari Y, Khazipov R, Leinekugel X, Caillard O, Gaiarsa JL (1997) GABAA, NMDA<br />

and AMPA receptors: a developmentally regulated 'menage a trois'. Trends Neurosci 20:<br />

523-529.<br />

Beninger RJ, Jhamandas A, Aujla H, Xue L, Dagnone RV, Boegman RJ, Jhamandas K<br />

(2002) Neonatal exposure to the glutamate receptor antagonist MK-801: effects on<br />

locomotor activity and pre-pulse inhibition before and after sexual maturity in rats.<br />

Neurotox Res 4: 477-488.<br />

Boyce-Rustay JM, Holmes A (2005) Functional roles of NMDA receptor NR2A and<br />

NR2B subunits in the acute intoxicating effects of ethanol in mice. Synapse 56: 222-225.<br />

Braff DL, Geyer MA, Swerdlow NR (2001) Human studies of prepulse inhibition of<br />

startle: normal subjects, patient groups, and pharmacological studies.<br />

Psychopharmacology (Berl) 156: 234-258.<br />

Brake WG, Zhang TY, Diorio J, Meaney MJ, Gratton A (2004) Influence of early<br />

postnatal rearing conditions on mesocorticolimbic dopamine and behavioural responses<br />

to psychostimulants and stressors in adult rats. Eur J Neurosci 19: 1863-1874.<br />

Chaperon F, Muller W, Auberson YP, Tricklebank MD, Neijt HC (2003) Substitution for<br />

PCP, disruption of prepulse inhibition and hyperactivity induced <strong>by</strong> N-methyl-Daspartate<br />

receptor antagonists: preferential involvement of the NR2B rather than NR2A<br />

subunit. Behav Pharmacol 14: 477-487.<br />

Clinton SM, Meador-Woodruff JH (2004) Abnormalities of the NMDA Receptor and<br />

Associated Intracellular Molecules in the Thalamus in Schizophrenia and Bipolar<br />

Disorder. Neuropsychopharmacology 29: 1353-1362.<br />

Coyle JT, Tsai G (2004) NMDA receptor function, neuroplasticity, and the<br />

pathophysiology of schizophrenia. Int Rev Neurobiol 59: 491-515.<br />

Cull-Candy S, Brickley S, Farrant M (2001) NMDA receptor subunits: diversity,<br />

development and disease. Curr Opin Neurobiol 11: 327-335.<br />

132


De Vry J, Jentzsch KR (2003) Role of the NMDA receptor NR2B subunit in the<br />

discriminative stimulus effects of ketamine. Behav Pharmacol 14: 229-235.<br />

Duncan GE, Moy SS, Perez A, Eddy DM, Zinzow WM, Lieberman JA, Snouwaert JN,<br />

Roller BH (2004) Deficits in sensorimotor gating and tests of social behavior in a genetic<br />

model of reduced NMDA receptor function. Behav Brain Res 153: 507-519.<br />

Facchinetti F, Ciani E, Dall'Olio R, Virgili M, Contestable A, Fonnum F (1993)<br />

Structural, neurochemical and behavioural consequences of neonatal blockade of NMDA<br />

receptor through chronic treatment with CGP 39551 or MK-801. Brain Res Dev Brain<br />

Res 74: 219-224.<br />

Fischer G, Mutel V, Trube G, Malherbe P, Kew JN, Mohacsi E, Heitz MP, Kemp JA<br />

(1997) Ro 25-6981, a highly potent and selective blocker of N-methyl-D-aspartate<br />

receptors containing the NR2B subunit. Characterization in vitro. J Pharmacol Exp Ther<br />

283: 1285-1292.<br />

Fox K, Schlaggar BL, Glazewski S, O'Leary DD (1996) Glutamate receptor blockade at<br />

cortical synapses disrupts development of thalamocortical and columnar organization in<br />

somatosensory cortex. Proc Natl Acad Sci U S A 93: 5584-5589.<br />

Fredriksson A, Archer T, Aim H, Gordh T, Eriksson P (2004) Neurofunctional deficits<br />

and potentiated apoptosis <strong>by</strong> neonatal NMDA antagonist administration. Behav Brain<br />

Res 153: 367-376.<br />

Gao XM, Sakai K, Roberts RC, Conley RR, Dean B, Tamminga CA (2000) Ionotropic<br />

glutamate receptors and expression of N-methyl-D-aspartate receptor subunits in<br />

subregions of human hippocampus: effects of schizophrenia. Am J Psychiatry 157: 1141-<br />

1149.<br />

Goff DC, Coyle JT (2001) The emerging role of glutamate in the pathophysiology and<br />

treatment of schizophrenia. Am J Psychiatry 158: 1367-1377.<br />

Gorter JA, de Bruin JP (1992) Chronic neonatal MK-801 treatment results in an<br />

impairment of spatial learning in the adult rat. Brain Res 580: 12-17.<br />

Gothelf D, Soreni N, Nachman RP, Tyano S, Hiss Y, Reiner O, Weizman A (2000)<br />

Evidence for the involvement of the hippocampus in the pathophysiology of<br />

schizophrenia. Eur Neuropsychopharmacol 10: 389-395.<br />

Gould E, Cameron HA, McEwen BS (1994) Blockade of NMDA receptors increases cell<br />

death and birth in the developing rat dentate gyrus. J Comp Neurol 340: 551-565.<br />

Grimwood S, Slater P, Deakin JF, Hutson PH (1999) NR2B-containing NMDA receptors<br />

are up-regulated in temporal cortex in schizophrenia. Neuroreport 10: 461-465.<br />

Guilarte TR, McGlothan JL (1998) Hippocampal NMDA receptor mRNA undergoes<br />

subunit specific changes during developmental lead exposure. Brain Res 790: 98-107.<br />

133


Harris LW, Sharp T, Gartlon J, Jones DN, Harrison PJ (2003) Long-term behavioural,<br />

molecular and morphological effects of neonatal NMDA receptor antagonism. Eur J<br />

Neurosci 18: 1706-1710.<br />

Harrison PJ (1999) The neuropathology of schizophrenia. A critical review of the data<br />

and their interpretation. Brain 122 (Pt 4): 593-624.<br />

Higgins GA, Ballard TM, Enderlin M, Haman M, Kemp JA (2005) Evidence for<br />

improved performance in cognitive tasks following selective NR2B NMDA receptor<br />

antagonist pre-treatment in the rat. Psychopharmacology (Berl) 179: 85-98.<br />

Holmes GL, Khazipov R, Ben Ari Y (2002) New concepts in neonatal seizures.<br />

Neuroreport 13: A3-A8.<br />

Howland JG, Hannesson DK, Phillips AG (2004) Delayed onset of prepulse inhibition<br />

deficits following kainic acid treatment on postnatal day 7 in rats. Eur J Neurosci 20:<br />

2639-2648. ' .<br />

Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vockler J, Dikranian K, Tenkova TI,<br />

Stefovska V, Turski L, Olney JW (1999) Blockade of NMDA receptors and apoptotic<br />

neurodegeneration in the developing brain. Science 283: 70-74.<br />

Itokawa M, Yamada K, Yoshitsugu K, Toyota T, Suga T, Ohba H, Watanabe A, Hattori<br />

E, Shimizu H, Kumakura T, Ebihara M, Meerabux JM, Toru M, Yoshikawa T (2003) A<br />

microsatellite repeat in the promoter of the N-methyl-D-aspartate receptor 2A subunit<br />

(GRIN2A) gene suppresses transcriptional activity and correlates with chronic outcome<br />

in schizophrenia. Pharmacogenetics 13: 271-278.<br />

Jentsch JD, Roth RH (1999) The neuropsychopharmacology of phencyclidine: from<br />

NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia.<br />

Neuropsychopharmacology 20: 201-225.<br />

Jones EG (1997) Cortical development and thalamic pathology in schizophrenia.<br />

Schizophr Bull 23: 483-501.<br />

Kanemoto K, Tsuji T, Kawasaki J (2001) Reexamination of interictal psychoses based on<br />

DSM IV psychosis classification and international epilepsy classification. Epilepsia 42:<br />

98-103.<br />

Kapur S, Seeman P (2002) NMDA receptor antagonists ketamine and PCP have direct<br />

effects on the dopamine D(2) and serotonin 5-HT(2)receptors-implications for models of<br />

schizophrenia. Mol Psychiatry 7: 837-844.<br />

Khalilov I, Dzhala V, Ben Ari Y, Khazipov R (1999a) Dual role of GABA in the<br />

neonatal rat hippocampus. Dev Neurosci 21: 310-319.<br />

134


Khalilov I, Dzhala V, Medina I, Leinekugel X, Melyan Z, Lamsa K, Khazipov R, Ben<br />

Ari Y (1999b) Maturation of kainate-induced epileptiform activities in interconnected<br />

intact neonatal limbic structures in vitro. Eur J Neurosci 11: 3468-3480.<br />

Khazipov R, Khalilov I, Tyzio R, Morozova E, Ben Ari Y, Holmes GL (2004)<br />

Developmental changes in GABAergic actions and seizure susceptibility in the rat<br />

hippocampus. Eur J Neurosci 19: 590-600.<br />

Kofman O (2002) The role of prenatal stress in the etiology of developmental<br />

behavioural disorders. Neurosci Biobehav Rev 26: 457-470.<br />

Lai MC, Liou CW, Yang SN, Wang CL, Hung PL, Wu CL, Tung YR, Huang LT (2002)<br />

Recurrent Bicuculline-Induced Seizures in Rat Pups Cause Long-Term Motor Deficits<br />

and Increase Vulnerability to a Subsequent Insult. Epilepsy Behav 3: 60-66.<br />

Lee J, Rajakumar N (2003) Role of NR2B-containing N-methyl-D-aspartate receptors in<br />

haloperidol-induced c-Fos expression in the striatum and nucleus accumbens.<br />

Neuroscience 122: 739-745.<br />

Leinekugel X, Khalilov I, McLean H, Caillard O, Gaiarsa JL, Ben Ari Y, Khazipov R<br />

(1999) GAB A is the principal fast-acting excitatory transmitter in the neonatal brain. Adv<br />

Neurol 79: 189-201.<br />

Lewis DA, Levitt P (2002) Schizophrenia as a disorder of neurodevelopment. Annu Rev<br />

Neurosci 25: 409-432.<br />

Lipska BK, Weinberger DR (2000) To model a psychiatric disorder in animals:<br />

schizophrenia as a reality test. Neuropsychopharmacology 23: 223-239.<br />

Luthi A, Schwyzer L, Mateos JM, Gahwiler BH, McKinney RA (2001) NMDA receptor<br />

activation limits the number of synaptic connections during hippocampal development.<br />

Nat Neurosci 4: 1102-1107.<br />

Lynch M, Sayin U, Bownds J, Janumpalli S, Sutula T (2000) Long-term consequences of<br />

early postnatal seizures on hippocampal learning and plasticity. Eur J Neurosci 12: 2252-<br />

2264.<br />

Mallon AP, Auberson YP, Stone TW (2005) Selective subunit antagonists suggest an<br />

inhibitory relationship between NR2B and NR2A-subunit containing N-methyl-D: -<br />

aspartate receptors in hippocampal slices. Exp Brain Res 162: 374-383.<br />

Miyamoto Y, Yamada K, Noda Y, Mori H, Mishina M, Nabeshima T (2001)<br />

Hyperfunction of dopaminergic and serotonergic neuronal systems in mice lacking the<br />

NMDA receptor epsilonl subunit. J Neurosci 21: 750-757.<br />

Miyatake R, Furukawa A, Suwaki H (2002) Identification of a novel variant of the human<br />

NR2B gene promoter region and its possible association with schizophrenia. Mol<br />

Psychiatry 7: 1101-1106.<br />

135


Mohn AR, Gainetdinov RR, Caron MG, Roller BH (1999) Mice with reduced NMDA<br />

receptor expression display behaviors related to schizophrenia. Cell 98: 427-436.<br />

Monyer H, Burnashev N, Laurie DJ, Sakmann B, Seeburg PH (1994) Developmental and<br />

regional expression in the rat brain and functional properties of four NMDA receptors.<br />

Neuron 12: 529-540.<br />

Murray F, Kennedy J, Hutson PH, Elliot J, Huscroft I, Mohnen K, Russell MG,<br />

Grimwood S (2000) Modulation of [3H]MK-801 binding to NMDA receptors in vivo and<br />

in vitro. Eur J Pharmacol 397: 263-270.<br />

Mutel V, Buchy D, Klingelschmidt A, Messer J, Bleuel Z, Kemp JA, Richards JG (1998)<br />

In vitro binding properties in rat brain of [3H]Ro 25-6981, a potent and selective<br />

antagonist of NMDA receptors containing NR2B subunits. J Neurochem 70: 2147-2155.<br />

Olney JW, Newcomer JW, Farber NB (1999) NMDA receptor hypofunction model of<br />

schizophrenia. J Psychiatr Res 33: 523-533.<br />

Pakkenberg B (1990) Pronounced reduction of total neuron number in mediodorsal<br />

thalamic nucleus and nucleus accumbens in schizophrenics. Arch Gen Psychiatry 47:<br />

1023-1028.<br />

Qin S, Zhao X, Pan Y, Liu J, Feng G, Fu J, Bao J, Zhang Z, He L (2005) An association<br />

study of the N-methyl-D-aspartate receptor NR1 subunit gene (GRIN1) and NR2B<br />

subunit gene (GRIN2B) in schizophrenia with universal DNA microarray. Eur J Hum<br />

Genet., in press.<br />

Rabacchi S, Bailly Y, Delhaye-Bouchaud N, Mariani J (1992) Involvement of the Nmethyl<br />

D-aspartate (NMDA) receptor in synapse elimination during cerebellar<br />

development. Science 256: 1823-1825.<br />

Ritter LM, Vazquez DM, Meador-Woodruff JH (2002) Ontogeny of ionotropic glutamate<br />

receptor subunit expression in the rat hippocampus. Brain Res Dev Brain Res 139: 227-<br />

236.<br />

Semba J, Tanaka N, Wakuta M, Suhara T (2001) Neonatal phencyclidine treatment<br />

selectively attenuates mesolimbic dopamine function in adult rats as revealed <strong>by</strong><br />

methamphetamine-induced behavior and c-fos mRNA expression in the brain. Synapse<br />

40: 11-18.<br />

Silveira DC, Sogawa Y, Holmes GL (2002) The expression of Fos following kainic acidinduced<br />

seizures is age-dependent. Eur J Neurosci 15: 329-344.<br />

Stafstrom CE, Chronopoulos A, Thurber S, Thompson JL, Holmes GL (1993) Agedependent<br />

cognitive and behavioral deficits after kainic acid seizures. Epilepsia 34: 420-<br />

432.<br />

136


StafStrom CE, Tandon P, Hori A, Liu Z, Mikati MA, Holmes GL (1997) Acute effects of<br />

MK801 on kainic acid-induced seizures in neonatal rats. Epilepsy Res 26: 335-344.<br />

Stafstrom CE, Thompson JL, Holmes GL (1992) Kainic acid seizures in the developing<br />

brain: status epilepticus and spontaneous recurrent seizures. Brain Res Dev Brain Res 65:<br />

227-236.<br />

Stefani MR, Moghaddam B (2005) Transient N-methyl-D-aspartate receptor blockade in<br />

early development causes lasting cognitive deficits relevant to schizophrenia. Biol<br />

Psychiatry 57: 433-436.<br />

Swerdlow NR, Platten A, Kim YK, Gaudet I, Shoemaker J, Pitcher L, Auerbach P (2001)<br />

Sensitivity to the dopaminergic regulation of prepulse inhibition in rats: evidence for<br />

genetic, but not environmental determinants. Pharmacol Biochem Behav 70: 219-226.<br />

Tremblay E, Nitecka L, Berger ML, Ben Ari Y (1984) Maturation of kainic acid seizurebrain<br />

damage syndrome in the rat. I. Clinical, electrographic and metabolic observations.<br />

Neuroscience 13: 1051-1072.<br />

Vallano ML (1998) Developmental aspects of NMDA receptor function. Crit Rev<br />

Neurobiol 12: 177-204.<br />

van den BM, Garner B, Koch M (2003) Neurodevelopmental animal models of<br />

schizophrenia: effects on prepulse inhibition. Curr Mol Med 3: 459-471.<br />

Vestergaard M, Pedersen CB, Christensen J, Madsen KM, Olsen J, Mortensen PB (2005)<br />

Febrile seizures and risk of schizophrenia. Schizophr Res 73: 343-349.<br />

Wang C, Mclnnis J, Ross-Sanchez M, Shinnick-Gallagher P, Wiley JL, Johnson KM<br />

(2001) Long-term behavioral and neurodegenerative effects of perinatal phencyclidine<br />

administration: implications for schizophrenia. Neuroscience 107: 535-550.<br />

Weinberger DR (1987) Implications of normal brain development for the pathogenesis of<br />

schizophrenia. Arch Gen Psychiatry 44: 660-669.<br />

Zhang XY, Liu AP, Ruan DY, Liu J (2002) Effect of developmental lead exposure on the<br />

expression of specific NMDA receptor subunit mRNAs in the hippocampus of neonatal<br />

rats <strong>by</strong> digoxigenin-labeled in situ hybridization histochemistry. Neurotoxicol Teratol 24:<br />

149-160.<br />

137


CHAPTER FIVE: GENERAL DISCUSSION<br />

The experiments contained within the present dissertation were designed to<br />

address two main objectives. First, Chapter Two detailed a number of experiments<br />

conducted to test the effects of electrical stimulation of discrete sub-regions of the<br />

hippocampus on PPI. Results of these experiments suggest brief periods of higher<br />

frequency activity that result in increased locomotor activity and ventral striatal<br />

dopamine efflux reversibly disrupt PPI when applied to the ventral, but not dorsal,<br />

hippocampus. The experiments described in Chapters Three and Four were designed to<br />

further the hypothesis that a period at the end of the first postnatal week is particularly<br />

sensitive for the development of the circuits mediating PPI and locomotor activity<br />

(Lipska and Weinberger, 2000). The experiments summarized in Chapter Three support<br />

this hypothesis <strong>by</strong> demonstrating that a single exposure to the kainate receptor agonist<br />

kainic acid on postnatal day seven disrupted PPI in early adulthood, but not before<br />

puberty. In Chapter Four, the effects of administration of the NR2B-subunit selective<br />

NMDA receptor antagonist Ro25-6981 on postnatal day six and seven were also<br />

assessed. Serendipitously, Ro25-6981 was observed to induce convulsions at this age;<br />

however, no consistent effects of this treatment on PPI or locomotor activity were noted<br />

either before puberty or in early adolescence. Although the implications of these results<br />

have been discussed at some length in the proceeding chapters, the present chapter will<br />

serve to further integrate these data with contemporary strategies of modeling<br />

schizophrenia in rodents.<br />

138


The Role of the Hippocampus in the Regulation of Prepulse Inhibition and Locomotor<br />

Activity<br />

As detailed in the discussion section of Chapter Two, hippocampal regulation of<br />

PPI is complex and incompletely understood. However, a great deal of evidence using<br />

both NMDA (Wan et al., 1996;Klarner et al., 1998;Zhang et al, 1999;Swerdlow et al.,<br />

2001b;Zhang et al., 2002) and higher frequency electrical (Howland et al., 2004b)<br />

stimulation protocols, indicates that over-activity of the ventral, but not dorsal,<br />

hippocampus disrupts PPI. Numerous studies have also assessed the role of hippocampal<br />

activity in locomotor behavior patterns of adult rats. Similar to the effects described for<br />

PPI, stimulation of the ventral hippocampus with either NMDA (Mogenson and Nielsen,<br />

1984;Bardgett and Henry, 1999;Bast et al., 2001;Zhang et al., 2002) or higher frequency<br />

electrical current (Taepavarapruk et al., 2000) increases locomotion, whereas comparable<br />

stimulation of the dorsal hippocampus has no effect (Zhang et al., 2002). Here it is<br />

important to note that the dorsal hippocampus can influence locomotor activity under<br />

some conditions as dorsal hippocampal infusions of carbachol (Flicker and Geyer,<br />

1982a;Mogenson and Nielsen, 1984), picrotoxin (Flicker and Geyer, 1982b) or other<br />

agents (Bast and Feldon, 2003), can alter the behavior. Thus, although the regulation of<br />

PPI and locomotor activity <strong>by</strong> the dorsal and ventral hippocampus is complex, both<br />

behaviors are clearly more sensitive to stimulation of the ventral than the dorsal region<br />

(Zhang et al., 2002;Bast and Feldon, 2003;Howland et al., 2004b). Previous research<br />

supports functional differentiation within the hippocampus, especially in regards to<br />

spatial learning and memory (supported <strong>by</strong> the dorsal hippocampus) and fear-related<br />

behavior (ventral hippocampus) (Moser and Moser, 1998; Kjelstrup et al., 2002).<br />

139


When the roles of dopamine and glutamate in mediating the effects of either<br />

NMDA or electrical stimulation of the ventral hippocampus on PPI or locomotor activity<br />

are considered, an interesting dissociation has emerged (Bast and Feldon, 2003). As was<br />

previously described for PPI, neither dopamine antagonists (systemically administered)<br />

nor glutamate antagonists (infused into the nucleus accumbens) effectively block the<br />

disruption of PPI induced <strong>by</strong> infusion of NMDA into the ventral hippocampus (Wan et<br />

al., 1996;Bast et al., 2001). In contrast, both dopamine and glutamate antagonists block<br />

the increases in locomotion observed following stimulation of the ventral hippocampus<br />

(Bardgett and Henry, 1999;Taepavarapruk et al., 2000;Bast et al., 2001). Additionally,<br />

locomotor activity in a novel environment increases dopamine efflux in the nucleus<br />

accumbens, an effect that can be reversed <strong>by</strong> inactivation of the ventral hippocampus or<br />

administration of a glutamate receptor antagonist into the ventral tegmental area (Legault<br />

and Wise, 2001). Thus, these findings indicate that dopamine and glutamate transmission<br />

is critical for the ventral hippocampal modulation of locomotor activity, but not for PPI.<br />

Although the circuitry underlying the role of dopamine and glutamate in the<br />

ventral hippocampal stimulation-elicited increases in locomotor behavior is still being<br />

investigated, evidence suggests that dopamine and glutamate receptors in the nucleus<br />

accumbens (Taepavarapruk et al., 2000) and ventral tegmental area (Legault and Wise,<br />

2001) play an important role, although other brain regions may also be involved.<br />

Stimulation of the ventral hippocampus with NMDA increases dopamine efflux in the<br />

medial prefrontal cortex (Peleg-Raibstein et al., 2005), and co-administration of<br />

dopamine antagonists with NMDA into the ventral hippocampus blocks the expected<br />

increase in locomotor behavior observed when NMDA is administered alone (Gimenez-<br />

140


Llort et al., 2002). Thus, strong parallels exist between the neural circuitry and<br />

pharmacology underlying the regulation of locomotor activity <strong>by</strong> the ventral<br />

hippocampus and that commonly described for schizophrenia.<br />

On the other hand, the mechanisms underlying the disruption of PPI following<br />

stimulation of the ventral hippocampus are less clear. As neither dopamine nor glutamate<br />

antagonists reverse the PPI disruptions following ventral hippocampal stimulation (Wan<br />

et al., 1996;Bast et al., 2001), direct activation of glutamatergic afferents from the ventral<br />

hippocampus to the nucleus accumbens is unlikely to underlie these effects. A recent<br />

study provides further support for this notion <strong>by</strong> demonstrating that lesions of the fornix,<br />

a tract containing the ventral hippocampal efferents to the nucleus accumbens, fail to<br />

block the disruption of PPI following stimulation of the ventral hippocampus with<br />

NMDA (Swerdlow et al., 2004). Therefore, the modulation of PPI <strong>by</strong> the ventral<br />

hippocampus likely involves either (1) interactions with brain areas other than those<br />

directly implicated in locomotor control (i.e., areas other than the nucleus accumbens) or<br />

(2) poly-synaptic pathways to the nucleus accumbens that do not include projections via<br />

the fornix.<br />

One potential circuit that may underlie the effects of stimulation of the ventral<br />

hippocampus includes the ventral hippocampal projections to the perirhinal and<br />

entorhinal cortex (Swerdlow et al., 2004). These areas have been implicated in the<br />

modulation of PPI (Swerdlow et al., 2001b;Goto et al, 2002) and are connected, via non-<br />

fornical routes, to structures such as the nucleus accumbens (Totterdell and Meredith,<br />

1997), basolateral amygdala (Pitkanen et al., 2000), and medial prefrontal cortex (Insausti<br />

et al., 1997) that are also involved in the modulation of PPI (Swerdlow et al., 2001a).<br />

141


Importantly, given that the mechanisms through which stimulation of the ventral<br />

hippocampus disrupts PPI are currendy unknown, it has been suggested that further<br />

understanding of this phenomenon may aid in the development of novel therapeutic<br />

strategies for schizophrenia (Bast and Feldon, 2003).<br />

The Utility of Adult Animal Models of Schizophrenia<br />

As was discussed in Chapter One, animal models of schizophrenia created <strong>by</strong><br />

manipulations performed during adulthood generally have poor construct validity given<br />

numerous lines of evidence that support a developmental component to the etiology of<br />

the disorder. However, adult models of schizophrenia can still contribute to a better<br />

understanding of the disorder in a number of different ways. For example, novel insights<br />

into the neural substrates underlying the behavioral symptoms of schizophrenia can be<br />

gained <strong>by</strong> performing experiments using adult animals. Support for this assertion is<br />

provided <strong>by</strong> data presented in Chapter Two that suggest increased ventral hippocampal<br />

activity in adulthood may underlie some symptoms of schizophrenia. Interestingly,<br />

results from brain imaging studies confirm the importance of these preclinical findings <strong>by</strong><br />

demonstrating that basal levels of hippocampal activity are increased in schizophrenia<br />

(Heckers et al., 1998;Benes, 2000;Heckers, 2001).<br />

Additionally, the present results (Howland et al., 2004b), and those of other<br />

researchers (Zhang et al., 2002), suggest that the ventral hippocampus may be more<br />

involved in the regulation of sensorimotor processes such as PPI and locomotor activity<br />

than the dorsal hippocampus. As a result, examining alterations in specific sub-regions<br />

of the hippocampus in patients with schizophrenia may reveal previously unappreciated<br />

characteristics of the neurobiology of the disorder (Goldman and Mitchell, 2004). In<br />

142


humans, the anterior portion of the hippocampus corresponds to the rodent ventral<br />

hippocampus, and some studies indicate that this region may be preferentially altered in<br />

schizophrenia (Csernansky et al., 1998;Szeszko et al., 2003;Narr et al., 2004; but see also<br />

Narr et al., 2001;Velakoulis et al., 2001).<br />

Furthermore, experiments with adult rats enable basic knowledge of the neural<br />

mechanisms underlying performance of behavioral tasks to be generated quickly. This<br />

knowledge can then be used to assess the potential mechanisms underlying behavioral<br />

deficits following developmental manipulations. This type of strategy was used to<br />

demonstrate that the PPI disruptions observed in socially isolated rats are dependent on<br />

dopamine levels in the nucleus accumbens (Powell et al., 2003) and extensively <strong>by</strong><br />

Lipska and colleagues regarding various aspects of the altered neural circuitry and<br />

pharmacology underlying changes in the neonatal ventral hippocampal lesion model<br />

(Lipska et al., 1998;Lipska and Weinberger, 2000). Thus, combining information gained<br />

from both adult and developmental approaches will likely enable a more rapid and<br />

thorough understanding of the disorder (Floresco et al., 2005).<br />

Developmental Models of Schizophrenia - Effects of Early Postnatal Glutamate<br />

Manipulations<br />

Clearly, the results gained from both the neonatal kainic acid and Ro25-6981<br />

manipulations are not as strong as was desired at the outset of the experiments. However,<br />

a number of interesting observations have been gained from these data, many of which<br />

should be verified in future experiments. The most important result was the subtle and<br />

reliable decrease in PPI observed following kainic acid administration in Chapter Three.<br />

Consistent with the ventral hippocampal lesion model, the PPI deficits were observed<br />

143


only after the animals had reached early adulthood. However, these deficits were not<br />

augmented <strong>by</strong> apomorphine administration or accompanied consistently <strong>by</strong> changes in<br />

locomotor activity, as is the case with the PPI deficits following neonatal ventral<br />

hippocampal lesions (Lipska et al., 1993;Lipska et al., 1995).<br />

As was discussed in the introduction, some first degree relatives of patients with<br />

schizophrenia exhibit lower PPI responses than matched controls (Cadenhead et al.,<br />

2000). Thus, disrupted PPI may be considered a candidate marker for a 'trait' (or<br />

underlying predisposition) to develop schizophrenia. Accordingly, the kainic acid model,<br />

while not reproducing the full-blown expression of schizophrenia, may be useful as a<br />

method of producing an underlying predisposition for the disorder in rodents. Further<br />

support for this assertion is gained from studies suggesting that the hippocampus is also<br />

altered in first degree relatives of schizophrenic patients (Seidman et al., 2002) and in<br />

adult rats <strong>by</strong> neonatal kainic acid administration (Lynch et al., 2000).<br />

Assessing the interaction of neonatal kainic acid administration with additional<br />

manipulations may support the notion that schizophrenia results from multiple 'hits' over<br />

the course of development (McCarley et al., 1999;Bayer et al., 1999;Lewis and Levitt,<br />

2002;Wong and Van Tol, 2003;Ellenbroek, 2003). To date, such strategies have been<br />

rarely used in an attempt to model the disorder although in one report, rats with neonatal<br />

ventral hippocampal lesions treated repeatedly with PCP in early adulthood displayed<br />

enhanced locomotor responses when compared to lesioned rats that had not been treated<br />

with PCP (Hori et al., 2000). Support for this experimental design is provided <strong>by</strong><br />

observations indicating that neonatal kainic acid administration renders rats more<br />

144


sensitive to morphological damage and behavioral impairments following kainic acid<br />

administration in adulthood (Koh et al., 1999).<br />

It is disappointing that the long-term behavioral experiments performed with<br />

Ro25-6981 failed to yield positive results. However, investigating novel mechanisms of<br />

brain development in the context of schizophrenia will improve understanding of the<br />

disorder. The rational for the use of Ro25-6981 was conceived from detailed knowledge<br />

surrounding the dynamic changes of specific NMDA subunits during early development,<br />

and similar experiments developed to test mechanistically-driven hypotheses will likely<br />

be an important component of preclinical schizophrenia research in the future.<br />

Further consideration of the null results of the experiments assessing the<br />

behavioral effects of neonatal administration of the NR2B antagonist Ro25-6981 can be<br />

focused in two main directions: (1) why the experiments performed fail to yield<br />

significant results and (2) what conclusions may be drawn from these experiments with<br />

respect to the existing literature regarding early developmental animal models of<br />

schizophrenia.<br />

Although the experiments using Ro25-6981 were carefully designed, the effects<br />

of this drug have not been extensively studied, especially in regard to the developing<br />

mammalian brain. In contrast, the acute effects of kainic acid administration are well<br />

known during development, and fit logically with some conceptions of the etiology of<br />

schizophrenia (Howland et al., 2004a). For example, kainic acid administration early in<br />

the neonatal period alters the activity of the hippocampus (Stafstrom et al., 1992;Khalilov<br />

et al., 1999;Lynch et al., 2000;Silveira et al., 2002) and additional limbic and cortical<br />

areas including the entorhinal cortex (Khalilov et al., 1999) implicated in schizophrenia.<br />

145


Additionally, rats treated neonatally with kainic acid exhibit subtle physiological<br />

alterations in the hippocampus during adulthood, which may underlie some of the<br />

behavioral alterations observed in these animals (Lynch et al., 2000). To my knowledge,<br />

the present experiments are the first to assess the effects of Ro25-6981 administration in<br />

neonatal rats. In retrospect, it may have been profitable to more carefully assess the acute<br />

effects of Ro25-6981 administration on the neonatal brain before the long-term<br />

behavioral experiments described herein were performed.<br />

Two main avenues will likely be especially profitable for understanding both the<br />

acute and long-term effects of neonatal Ro25-6981 administration on brain and behavior.<br />

Initially, determining which brain areas are activated with either convulsive or sub-<br />

convulsive doses of the drug will enable the development of a more detailed hypothesis-<br />

driven approach regarding the potential long-term behavioral effects of the convulsions<br />

induced <strong>by</strong> Ro25-6981. I expect that the activity of structures such as the hippocampus,<br />

cortex, and striatum will be altered <strong>by</strong> administration of Ro25-6981 due to high levels of<br />

NR2B expression in those areas neonatally (Monyer et al., 1994;Loftis and Janowsky,<br />

2003), and the well documented role of these areas in seizures (Ben Ari and Cossart,<br />

2000). However, high levels of NR2B-containing NMDA receptors are also expressed in<br />

other areas including the thalamus and cerebellum at this developmental age (Loftis and<br />

Janowsky, 2003). Interestingly, connections between these areas and the frontal cortex<br />

have been implicated in schizophrenia (Andreasen et al., 1999;Konarski et al., 2005).<br />

Thus, altered activity patterns in distributed neural circuits may underlie the convulsions<br />

produced <strong>by</strong> Ro25-6981.<br />

146


Secondly, an assessment of the effects of Ro25-6981 administration on cell<br />

survival in the developing brain is essential. As detailed in Chapter Four, a number of<br />

competitive and non-competitive NMDA receptor antagonists induce massive cell death<br />

in the developing brain, a factor which may be important in the long-term behavioral<br />

effects of their administration (Ikonomidou et al., 1999;Wang et al, 2001;Harris et al.,<br />

2003;Fredriksson et al., 2004). The influence of Ro25-6981 on cell survival in the<br />

developing brain is currently unknown, however the high proportion of NR2J3-containing<br />

NMDA receptors early in development supports the hypothesis that treatment with Ro25-<br />

6981 may cause apoptosis during this developmental age.<br />

Although a detailed understanding of the effects of Ro25-6981 administration is<br />

lacking, the results of the behavioral experiments conducted prior to and after puberty are<br />

still surprising as the induction of three strong convulsions during the end of the postnatal<br />

week failed to alter PPI or locomotor responding. One factor that could be especially<br />

important in the different behavioral effects observed between the convulsions elicited <strong>by</strong><br />

kainic acid and Ro25-6981 is that convulsions elicited <strong>by</strong> kainic acid are significantly<br />

longer in duration than those elicited with Ro25-6981. Longer convulsions may disturb<br />

developing neural networks to a significantly greater extent than shorter ones (Jensen and<br />

Baram, 2000;Holmes, 2004). Previous studies using other convulsive agents such as<br />

flourothyl, which have a short duration of action (10 to 15 minutes), support this<br />

assertion. These experiments indicate that a series of 25 to 50 convulsions during the<br />

first week of life must be elicited for long-term alterations in behavior to be observed<br />

(Holmes et al., 1998;Huang et al., 1999). Further support for this hypothesis is gained<br />

from data suggesting that a history of prolonged febrile seizures is a significant risk factor<br />

147


for psychosis in adulthood epileptics (Kanemoto et al., 2001) and that prolonged febrile<br />

seizures result in long-term changes in hippocampal excitability in rodents (Chen et al.,<br />

1999;Jensen and Baram, 2000). Therefore, it is tempting to speculate that repeated<br />

convulsions induced <strong>by</strong> Ro25-6981 during the first postnatal week would be related to<br />

alterations in PPI, locomotor activity, and cognitive deficits in adult rats.<br />

Developmental Models of Schizophrenia - The Time Course of Symptom Emergence<br />

Although developmental animal models of schizophrenia are likely more<br />

ecologically valid than adult models, they are much more difficult to establish. One of<br />

the cardinal features of the majority of patients with schizophrenia is the delayed onset of<br />

many symptoms of the disorder (Marcotte et al., 2001;Wong and Van Tol, 2003),<br />

however, this feature of the disorder remains elusive in animal models (Marcotte et al.,<br />

2001). Research examining recovery of function after brain injury generally supports the<br />

assertion that behavioral impairment is less severe if the injury is suffered early in life - a<br />

phenomenon commonly termed the Kennard principle (Kolb et al., 2000;Marcotte et al.,<br />

2001). However, it is important to note that the pattern of recovery from injuries<br />

sustained during early brain development show some exceptions to the Kennard<br />

principle. For example, lesions of the frontal cortex induced at specific developmental<br />

stages (i.e. late during the embryologic period or after the first postnatal week) result in<br />

significant recovery over time, while other periods exist (i.e. the first postnatal week)<br />

where recovery from similar injuries is significantly poorer than expected (Kolb and<br />

Whishaw, 1989;Kolb et al., 2000).<br />

In the case of schizophrenia, the relation between adverse events early in<br />

development and subsequent manifestation of symptoms suggest a delayed response in<br />

148


which behavioral deficits emerge after a period of relative normality. The neonatal<br />

ventral hippocampal lesion model provides important proof that alterations in the neural<br />

substrates implicated in schizophrenia during certain developmental stages may result in<br />

the delayed onset of various behavioral and electrophysiological alterations related to the<br />

disorder (Lipska et al., 1993;Lipska et al., 1995;Wood et al., 1997;Lipska and<br />

Weinberger, 2000;Lipska et al, 2002;Goto and O'Donnell, 2002). Additional research<br />

suggests that manipulations other than ventral hippocampal lesions such as immune<br />

activation (Zuckerman et al., 2003), administration of the anti-mitotic agent arabinoside<br />

(Elmer et al., 2004), or kainic acid (Howland et al., 2004a) result in the delayed onset of<br />

behavioral abnormalities associated with schizophrenia (unfortunately, unlike the design<br />

employed in this thesis, the behavioral effects of many early neonatal treatments<br />

developed have not been tested before adulthood). Taken together, these data suggest<br />

that certain critical periods exist in the neonatal brain for the normal development of the<br />

neural circuits mediating behaviors commonly ascribed to schizophrenia in animals<br />

(Lipska and Weinberger, 2000;Marcotte et al., 2001;Howland et al., 2004a). Future work<br />

related to the mechanisms underlying these changes will likely provide novel insights<br />

into the etiology of the disorder.<br />

Criteria for Establishing Validity in Behavioral Models of Schizophrenia<br />

The present experiments relied heavily on PPI and locomotor behavior in<br />

assessing the experimental manipulations performed in modeling schizophrenia.<br />

Although these behaviors have been used extensively <strong>by</strong> researchers in the field, they<br />

have a number of shortcomings. Importantly, PPI is disrupted in a number of<br />

neurological and psychiatric disorders in addition to schizophrenia including<br />

149


Huntington's disease, Tourette's syndrome, obsessive-compulsive disorder, and<br />

pathological gambling (Braff et al., 2001). Given the complexity of the neural circuitry<br />

mediating PPI this is not particularly surprising; however, it is problematic in that<br />

disrupted PPI may not be characteristic of schizophrenia per se, but rather of alterations<br />

in the distributed circuitry mediating it (Swerdlow et al., 2001a).<br />

As previously discussed, increased locomotor activity is often used as an indirect<br />

measure of ventral striatal dopamine activity, especially when combined with the<br />

administration of dopamine agonists (Kelly et al., 1975;Castall et al., 1977;Porrino et al.,<br />

1984;Lipska and Weinberger, 2000;Marcotte et al., 2001). Unfortunately, altered<br />

locomotor behavior is a very general phenomenon and can be influenced <strong>by</strong> a number of<br />

factors including anxiety levels, alterations in sensory processing, and memory<br />

disruptions (Bast and Feldon, 2003). Although some parallels between increased<br />

locomotor activity and certain symptoms of schizophrenia have been suggested (Bast and<br />

Feldon, 2003), these assertions are clearly speculative (Marcotte et al., 2001).<br />

As a result of these shortcomings, testing PPI and locomotor activity levels should<br />

be viewed as an important 'first-pass' in the assessment of potential animal models of<br />

schizophrenia. Future experiments with an array of behaviors relevant to schizophrenia<br />

will serve to strengthen the validity of putative models of schizophrenia (Lipska and<br />

Weinberger, 2000). Recent interest has been generated around preclinical correlates of<br />

the negative and cognitive symptoms of schizophrenia as they have traditionally been<br />

ignored in much of the literature (Ellenbroek and Cools, 2000;Floresco et al., 2005).<br />

Tests assessing social interaction, reward sensitivity, and executive functions such as<br />

cognitive set shifting and working memory are likely to be especially useful in further<br />

150


understanding the more complicated and treatment-resistant symptoms of the disorder<br />

(Ellenbroek and Cools, 2000;Robbins, 2004;Floresco et al., 2005). It is worth pointing<br />

out, however, that many individuals with schizophrenia do not exhibit all symptoms of<br />

the disorder (Wong and Van Tol, 2003). Therefore, reliance on strategies that<br />

successfully model many aspects of the disorder in animals may be counterproductive in<br />

some instances.<br />

Interestingly, some manipulations, such as neonatal ventral hippocampal lesions,<br />

produce changes in many behaviors resembling schizophrenia (Lipska and Weinberger,<br />

2000), while others, such as certain neonatal NMDA antagonist treatment regimes,<br />

produce selective deficits on certain behavioral tasks relevant to one class of symptoms<br />

(e.g. cognitive symptoms) (Stefani and Moghaddam, 2005). The present data do not<br />

indicate how animals treated with either kainic acid or Ro25-6981 may respond if further<br />

testing was conducted. However, in some studies, rats treated neonatally with kainic acid<br />

display deficits in learning and memory (Lynch et al., 2000), although null findings have<br />

also been reported (Stafstrom et al., 1993). Therefore, in the future, it may be profitable<br />

to greatly extend the test battery beyond the tests presently employed.<br />

Finally, testing the efficacy of antipsychotic drugs at reversing the behavioral<br />

disturbances observed following neonatal kainic acid administration could greatly aid in<br />

assessing its validity as a model of schizophrenia. As has been noted throughout the<br />

present dissertation, both typical and atypical antipsychotics are effective at ameliorating<br />

the behavioral disturbances, including PPI deficits, in numerous animal models of<br />

schizophrenia (Lipska and Weinberger, 2000;Marcotte et al., 2001;Bast et al., 2001 ;Le<br />

Pen and Moreau, 2002;Bast and Feldon, 2003;Van den et al., 2003;Le Pen et al., 2003).<br />

151


Conclusion<br />

In the introduction, three main goals for developing animal models of<br />

schizophrenia were introduced: (1) advancing the understanding of the symptoms of the<br />

disorder, (2) furthering the understanding of schizophrenia's etiology, and (3) aiding in<br />

the development of pharmaceutical therapies (Lipska and Weinberger, 2000). The<br />

present dissertation explored two general strategies for developing behaviorally-oriented<br />

animal models of schizophrenia. Although the results were somewhat mixed, the<br />

experiments were successful at providing novel insights into the symptoms and etiology<br />

of the disorder. In general, they support the assertion that short periods of altered activity<br />

in the limbic system, and hippocampus in particular, at different points during<br />

development may underlie the expression of some of the most basic symptoms of<br />

schizophrenia. These data also suggest that the nature and anatomical location of these<br />

alterations critically determines their long-term functional effects.<br />

152


References<br />

Andreasen NC, Nopoulos P, O'Leary DS, Miller DD, Wassink T, Flaum M (1999)<br />

Defining the phenotype of schizophrenia: cognitive dysmetria and its neural mechanisms.<br />

Biol Psychiatry 46: 908-920.<br />

Bardgett ME, Henry JD (1999) Locomotor activity and accumbens Fos expression driven<br />

<strong>by</strong> ventral hippocampal stimulation require Dl and D2 receptors. Neuroscience 94: 59-<br />

70.<br />

Bast T, Feldon J (2003) Hippocampal modulation of sensorimotor processes. Prog<br />

Neurobiol 70: 319-345.<br />

Bast T, Zhang WN, Heidbreder C, Feldon J (2001) Hyperactivity and disruption of<br />

prepulse inhibition induced <strong>by</strong> N-methyl-D-aspartate stimulation of the ventral<br />

hippocampus and the effects of pretreatment with haloperidol and clozapine.<br />

Neuroscience 103: 325-335.<br />

Bayer TA, Falkai P, Maier W (1999) Genetic and non-genetic vulnerability factors in<br />

schizophrenia: the basis of the "Two hit hypothesis". Journal of Psychiatric Research 33:<br />

543-548.<br />

Ben Ari Y, Cossart R (2000) Kainate, a double agent that generates seizures: two decades<br />

of progress. Trends in Neurosciences 23: 580-587.<br />

Benes FM (2000) Emerging principles of altered neural circuitry in schizophrenia. Brain<br />

Res Brain Res Rev 31: 251-269.<br />

Braff DL, Geyer MA, Swerdlow NR (2001) Human studies of prepulse inhibition of<br />

startle: normal subjects, patient groups, and pharmacological studies.<br />

Psychopharmacology (Berl) 156: 234-258.<br />

Cadenhead KS, Swerdlow NR, Shafer KM, Diaz M, Braff DL (2000) Modulation of the<br />

startle response and startle laterality in relatives of schizophrenic patients and in subjects<br />

with schizotypal personality disorder: evidence of inhibitory deficits. Am J Psychiatry<br />

157: 1660-1668.<br />

Castall B, Marsden CD, Naylor RJ, Pycock CJ (1977) Stereotyped behaviour patterns and<br />

hyperactivity induced <strong>by</strong> amphetamine and apomorphine after discrete 6hydroxydopamine<br />

lesions of extrapyramidal and mesolimbic nuclei. Brain Res 123: 89-<br />

111.<br />

Chen K, Baram TZ, Soltesz I (1999) Febrile seizures in the developing brain result in<br />

persistent modification of neuronal excitability in limbic circuits. Nat Med 5: 888-894.<br />

Csernansky JG, Joshi S, Wang L, Haller JW, Gado M, Miller JP, Grenander U, Miller MI<br />

(1998) Hippocampal morphometry in schizophrenia <strong>by</strong> high dimensional brain mapping.<br />

Proc Natl Acad Sci U S A 95: 11406-11411.<br />

153


Ellenbroek BA (2003) Animal models in the genomic era: possibilities and limitations<br />

with special emphasis on schizophrenia. Behav Pharmacol 14: 409-417.<br />

Ellenbroek BA, Cools AR (2000) Animal models for the negative symptoms of<br />

schizophrenia. Behav Pharmacol 11: 223-233.<br />

Elmer Gl, Sydnor J, Guard H, Hercher E, Vogel MW (2004) Altered prepulse inhibition<br />

in rats treated prenatally with the antimitotic Ara-C: an animal model for sensorimotor<br />

gating deficits in schizophrenia. Psychopharmacology (Berl) 174: 177-189.<br />

Flicker C, Geyer MA (1982a) Behavior during hippocampal microinfusions. II.<br />

Muscarinic locomotor activation. Brain Res 257: 105-127.<br />

Flicker C, Geyer MA (1982b) Behavior during hippocampal microinfusions. III.<br />

Lidocaine versus picrotoxin. Brain Res 257: 129-136.<br />

Floresco SB, Geyer MA, Gold LH, Grace AA (2005) Developing Predictive Animal<br />

Models and Establishing a Preclinical Trials Network for Assessing Treatment Effects on<br />

Cognition in Schizophrenia. Schizophr Bull.<br />

Fredriksson A, Archer T, Aim H, Gordh T, Eriksson P (2004) Neurofunctional deficits<br />

and potentiated apoptosis <strong>by</strong> neonatal NMDA antagonist administration. Behavioural<br />

Brain Research 153: 367-376.<br />

Gimenez-Llort L, Wang FH, Ogren SO, Ferre S (2002) Local dopaminergic modulation<br />

of the motor activity induced <strong>by</strong> N-methyl-D-aspartate receptor stimulation in the ventral<br />

hippocampus. Neuropsychopharmacology 26: 737-743.<br />

Goldman MB, Mitchell CP (2004) What is the functional significance of hippocampal<br />

pathology in schizophrenia? Schizophr Bull 30: 367-392.<br />

Goto K, Ueki A, Iso H, Morita Y (2002) Reduced prepulse inhibition in rats with<br />

entorhinal cortex lesions. Behav Brain Res 134: 201-207.<br />

Goto Y, O'Donnell P (2002) Delayed mesolimbic system alteration in a developmental<br />

animal model of schizophrenia. J Neurosci 22: 9070-9077.<br />

Harris LW, Sharp T, Gartlon J, Jones DNC, Harrison PJ (2003) Long-term behavioural,<br />

molecular and morphological effects of neonatal NMDA receptor antagonism. European<br />

Journal of Neuroscience 18: 1706-1710.<br />

Heckers S (2001) Neuroimaging studies of the hippocampus in schizophrenia.<br />

Hippocampus 11: 520-528.<br />

Heckers S, Rauch SL, Goff D, Savage CR, Schacter DL, Fischman AJ, Alpert NM (1998)<br />

Impaired recruitment of the hippocampus during conscious recollection in schizophrenia.<br />

Nat Neurosci 1: 318-323.<br />

154


Holmes GL (2004) Effects of early seizures on later behavior and epileptogenicity. Ment<br />

Retard Dev Disabil Res Rev 10: 101-105.<br />

Holmes GL, Gairsa JL, Chevassus-Au-Louis N, Ben Ari Y (1998) Consequences of<br />

neonatal seizures in the rat: morphological and behavioral effects. Ann Neurol 44: 845-<br />

857.<br />

Hori T, Subramaniam S, Srivastava LK, Quirion R (2000) Behavioral and neurochemical<br />

alterations following repeated phencyclidine administration in rats with neonatal ventral<br />

hippocampal lesions. Neuropharmacology 39: 2478-2491.<br />

Howland JG, Hannesson DK, Phillips AG (2004a) Delayed onset of prepulse inhibition<br />

deficits following kainic acid treatment on postnatal day 7 in rats. Eur J Neurosci 20:<br />

2639-2648.<br />

Howland JG, MacKenzie EM, Yim TT, Taepavarapruk P, Phillips AG (2004b) Electrical<br />

stimulation of the hippocampus disrupts prepulse inhibition in rats: frequency- and sitedependent<br />

effects. Behav Brain Res 152: 187-197.<br />

Huang L, Cilio MR, Silveira DC, McCabe BK, Sogawa Y, Stafstrom CE, Holmes GL<br />

(1999) Long-term effects of neonatal seizures: a behavioral, electrophysiological, and<br />

histological study. Brain Res Dev Brain Res 118: 99-107.<br />

Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vockler J, Dikranian K, Tenkova TI,<br />

Stefovska V, Turski L, Olney JW (1999) Blockade of NMDA receptors and apoptotic<br />

neurodegeneration in the developing brain. Science 283: 70-74.<br />

Insausti R, Herrero MT, Witter MP (1997) Entorhinal cortex of the rat: cytoarchitectonic<br />

subdivisions and the origin and distribution of cortical efferents. Hippocampus 7: 146-<br />

183.<br />

Jensen FE, Baram TZ (2000) Developmental seizures induced <strong>by</strong> common early-life<br />

insults: short- and long-term effects on seizure susceptibility. Ment Retard Dev Disabil<br />

Res Rev 6: 253-257.<br />

Kanemoto K, Tsuji T, Kawasaki J (2001) Reexamination of interictal psychoses based on<br />

DSM IV psychosis classification and international epilepsy classification. Epilepsia 42:<br />

98-103.<br />

Kelly PH, Seviour PW, Iversen SD (1975) Amphetamine and apomorphine responses in<br />

the rat following 6-OHDA lesions of the nucleus accumbens septi and corpus striatum.<br />

Brain Res 94: 507-522.<br />

Khalilov I, Dzhala V, Medina I, Leinekugel X, Melyan Z, Lamsa K, Khazipov R, Ben<br />

Ari Y (1999) Maturation of kainate-induced epileptiform activities in interconnected<br />

intact neonatal limbic structures in vitro. Eiir J Neurosci 11: 3468-3480.<br />

155


Kjelstrup KG, Tuvnes FA, Steffenach HA, Murison R, Moser EI, Moser MB (2002)<br />

Reduced fear expression after lesions of the ventral hippocampus. PNAS 99:10825-<br />

10830.<br />

Klarner A, Koch M, Schnitzler HU (1998) Induction of Fos-protein in the forebrain and<br />

disruption of sensorimotor gating following N-methyl-D-aspartate infusion into the<br />

ventral hippocampus of the rat. Neuroscience 84: 443-452.<br />

Koh S, Storey TW, Santos TC, Mian AY, Cole AJ (1999) Early-life seizures in rats<br />

increase susceptibility to seizure-induced brain injury in adulthood. Neurology 53: 915-<br />

921.<br />

Kolb B, Gibb R, Gorny G (2000) Cortical plasticity and the development of behavior<br />

after early frontal cortical injury. Dev Neuropsychol 18: 423-444.<br />

Kolb B, Whishaw IQ (1989) Plasticity in the neocortex: mechanisms underlying recovery<br />

from early brain damage. Prog Neurobiol 32: 235-276.<br />

Konarski JZ, Mclntyre RS, Grupp LA, Kennedy SH (2005) Is the cerebellum relevant in<br />

the circuitry of neuropsychiatric disorders? J Psychiatry Neurosci 30: 178-186.<br />

Le Pen G, Kew J, Alberati D, Borroni E, Heitz MP, Moreau JL (2003) Prepulse inhibition<br />

deficits of the starde reflex in neonatal ventral hippocampal-lesioned rats: reversal <strong>by</strong><br />

glycine and a glycine transporter inhibitor. Biol Psychiatry 54: 1162-1170.<br />

Le Pen G, Moreau JL (2002) Disruption of prepulse inhibition of startle reflex in a<br />

neurodevelopmental model of schizophrenia: reversal <strong>by</strong> clozapine, olanzapine and<br />

risperidone but not <strong>by</strong> haloperidol. Neuropsychopharmacology 27: 1-11.<br />

Legault M, Wise RA (2001) Novelty-evoked elevations of nucleus accumbens dopamine:<br />

dependence on impulse flow from the ventral subiculum and glutamatergic<br />

neurotransmission in the ventral tegmental area. Eur J Neurosci 13: 819-828.<br />

Lewis DA, Levitt P (2002) Schizophrenia as a disorder of neurodevelopment. Annu Rev<br />

Neurosci 25:409-432.<br />

Lipska BK, Al Amin HA, Weinberger DR (1998) Excitotoxic lesions of the rat medial<br />

prefrontal cortex. Effects on abnormal behaviors associated with neonatal hippocampal<br />

damage. Neuropsychopharmacology 19: 451-464.<br />

Lipska BK, Halim ND, Segal PN, Weinberger DR (2002) Effects of reversible<br />

inactivation of the neonatal ventral hippocampus on behavior in the adult rat. J Neurosci<br />

22: 2835-2842.<br />

Lipska BK, Jaskiw GE, Weinberger DR (1993) Postpubertal emergence of<br />

hyperresponsiveness to stress and to amphetamine after neonatal excitotoxic hippocampal<br />

damage: a potential animal model of schizophrenia. Neuropsychopharmacology 9: 67-75.<br />

156


Lipska BK, Swerdlow NR, Geyer MA, Jaskiw GE, Braff DL, Weinberger DR (1995)<br />

Neonatal excitotoxic hippocampal damage in rats causes post-pubertal changes in<br />

prepulse inhibition of startle and its disruption <strong>by</strong> apomorphine. Psychopharmacology<br />

(Berl) 122: 35-43.<br />

Lipska BK, Weinberger DR (2000) To model a psychiatric disorder in animals:<br />

schizophrenia as a reality test. Neuropsychopharmacology 23: 223-239.<br />

Loftis JM, Janowsky A (2003) The N-methyl-D-aspartate receptor subunit NR2B:<br />

localization, functional properties, regulation, and clinical implications. Pharmacol Ther<br />

97:55-85.<br />

Lynch M, Sayin U, Bownds J, Janumpalli S, Sutula T (2000) Long-term consequences of<br />

early postnatal seizures on hippocampal learning and plasticity. Eur J Neurosci 12: 2252-<br />

2264.<br />

Marcotte ER, Pearson DM, Srivastava LK (2001) Animal models of schizophrenia: a<br />

critical review. J Psychiatry Neurosci 26: 395-410.<br />

McCarley RW, Wible CG, Frumin M, Hirayasu Y, Levitt JJ, Fischer IA, Shenton ME<br />

(1999) MRI anatomy of schizophrenia. Biol Psychiatry 45: 1099-1119.<br />

Mogenson GJ, Nielsen M (1984) A study of the contribution of hippocampal-accumbenssubpallidal<br />

projections to locomotor activity. Behav Neural Biol 42: 38-51.<br />

Monyer H, Burnashev N, Laurie DJ, Sakmann B, Seeburg PH (1994) Developmental and<br />

regional expression in the rat brain and functional properties of four NMDA receptors.<br />

Neuron 12: 529-540.<br />

Moser MB, Moser EI (1998) Functional differentiation in the hippocampus.<br />

Hippocampus 8: 593-600.<br />

Narr KL, Thompson PM, Sharma T, Moussai J, Blanton R, Anvar B, Edris A, Krupp R,<br />

Rayman J, Khaledy M, Toga AW (2001) Three-dimensional mapping of temporo-limbic<br />

regions and the lateral ventricles in schizophrenia: gender effects. Biol Psychiatry 50: 84-<br />

97.<br />

Narr KL, Thompson PM, Szeszko P, Robinson D, Jang S, Woods RP, Kim S, Hayashi<br />

KM, Asunction D, Toga AW, Bilder RM (2004) Regional specificity of hippocampal<br />

volume reductions in first-episode schizophrenia. Neuroimage 21: 1563-1575.<br />

Peleg-Raibstein D, Pezze MA, Ferger B, Zhang WN, Murphy CA, Feldon J, Bast T<br />

(2005) Activation of dopaminergic neurotransmission in the medial prefrontal cortex <strong>by</strong><br />

N-methyl-d-aspartate stimulation of the ventral hippocampus in rats. Neuroscience 132:<br />

219-232.<br />

157


Pitkanen A, Pikkarainen M, Nurminen N, Ylinen A (2000) Reciprocal connections<br />

between the amygdala and the hippocampal formation, perirhinal cortex, and postrhinal<br />

cortex in rat. A review. Ann N Y Acad Sci 911: 369-391.<br />

Porrino LJ, Lucignani G, Dow-Edwards D, Sokoloff L (1984) Correlation of dosedependent<br />

effects of acute amphetamine administration on behavior and local cerebral<br />

metabolism in rats. Brain Res 307: 311-320.<br />

Powell SB, Geyer MA, Preece MA, Pitcher LK, Reynolds GP, Swerdlow NR (2003)<br />

Dopamine depletion of the nucleus accumbens reverses isolation-induced deficits in<br />

prepulse inhibition in rats. Neuroscience 119: 233-240.<br />

Robbins TW (2004) Animal models of the psychoses. In: Neurobiology of Mental Illness,<br />

2nd. Ed. (Charney DS, Nestler EJ, eds), pp 263-286. New York: Oxford University Press.<br />

Seidman LJ, Faraone SV, Goldstein JM, Kremen WS, Horton NJ, Makris N, Toomey R,<br />

Kennedy D, Caviness VS, Tsuang MT (2002) Left hippocampal volume as a<br />

vulnerability indicator for schizophrenia: a magnetic resonance imaging morphometric<br />

study of nonpsychotic first-degree relatives. Arch Gen Psychiatry 59: 839-849.<br />

Silveira DC, Sogawa Y, Holmes GL (2002) The expression of Fos following kainic acidinduced<br />

seizures is age-dependent. Eur J Neurosci 15: 329-344.<br />

Stafstrom CE, Chronopoulos A, Thurber S, Thompson JL, Holmes GL (1993) Agedependent<br />

cognitive and behavioral deficits after kainic acid seizures. Epilepsia 34: 420-<br />

432.<br />

Stafstrom CE, Thompson JL, Holmes GL (1992) Kainic acid seizures in the developing<br />

brain: status epilepticus and spontaneous recurrent seizures. Brain Res Dev Brain Res 65:<br />

227-236.<br />

Stefani MR, Moghaddam B (2005) Transient N-methyl-D-aspartate receptor blockade in<br />

early development causes lasting cognitive deficits relevant to schizophrenia. Biol<br />

Psychiatry 57: 433-436.<br />

Swerdlow NR, Geyer MA, Braff DL (2001a) Neural circuit regulation of prepulse<br />

inhibition of startle in the rat: current knowledge and future challenges.<br />

Psychopharmacology (Berl) 156: 194-215.<br />

Swerdlow NR, Hanlon FM, Henning L, Kim YK, Gaudet I, Halim ND (2001b)<br />

Regulation of sensorimotor gating in rats <strong>by</strong> hippocampal NMDA: anatomical<br />

localization. Brain Res 898: 195-203.<br />

Swerdlow NR, Shoemaker JM, Noh HR, Ma L, Gaudet I, Munson M, Crain S, Auerbach<br />

PP (2004) The ventral hippocampal regulation of prepulse inhibition and its disruption <strong>by</strong><br />

apomorphine in rats are not mediated via the fornix. Neuroscience 123: 675-685.<br />

158


Szeszko PR, Goldberg E, Gunduz-Bruce H, Ashtari M, Robinson D, Malhotra AK, Lencz<br />

T, Bates J, Crandall DT, Kane JM, Bilder RM (2003) Smaller anterior hippocampal<br />

formation volume in antipsychotic-naive patients with first-episode schizophrenia. Am J<br />

Psychiatry 160: 2190-2197.<br />

Taepavarapruk P, Floresco SB, Phillips AG (2000) Hyperlocomotion and increased<br />

dopamine efflux in the rat nucleus accumbens evoked <strong>by</strong> electrical stimulation of the<br />

ventral subiculum: role of ionotropic glutamate and dopamine Dl receptors.<br />

Psychopharmacology (Berl) 151: 242-251.<br />

Totterdell S, Meredith GE (1997) Topographical organization of projections from the<br />

entorhinal cortex to the striatum of the rat. Neuroscience 78: 715-729.<br />

Van den BM, Garner B, Koch M (2003) Neurodevelopmental animal models of<br />

schizophrenia: effects on prepulse inhibition. Curr Mol Med 3: 459-471.<br />

Velakoulis D, Stuart GW, Wood SJ, Smith DJ, Brewer WJ, Desmond P, Singh B,<br />

Copolov D, Pantelis C (2001) Selective bilateral hippocampal volume loss in chronic<br />

schizophrenia. Biol Psychiatry 50: 531-539.<br />

Wan FJ, Caine SB, Swerdlow NR (1996) The ventral subiculum modulation of prepulse<br />

inhibition is not mediated via dopamine D2 or nucleus accumbens non-NMDA glutamate<br />

receptor activity. Eur J Pharmacol 314: 9-18.<br />

Wang C, Mclnnis J, Ross-Sanchez M, Shinnick-Gallagher P, Wiley JL, Johnson KM<br />

(2001) Long-term behavioral and neurodegenerative effects of perinatal phencyclidine<br />

administration: Implications for schizophrenia. Neuroscience 107: 535-550.<br />

Wong AH, Van Tol HH (2003) Schizophrenia: from phenomenology to neurobiology.<br />

Neurosci Biobehav Rev 27: 269-306.<br />

Wood GK, Lipska BK, Weinberger DR (1997) Behavioral changes in rats with early<br />

ventral hippocampal damage vary with age at damage. Brain Res Dev Brain Res 101: 17-<br />

25.<br />

Zhang W, Pouzet B, Jongen-Relo AL, Weiner I, Feldon J (1999) Disruption of prepulse<br />

inhibition following N-methyl-D-aspartate infusion into the ventral hippocampus is<br />

antagonized <strong>by</strong> clozapine but not <strong>by</strong> haloperidol: a possible model for the screening of<br />

atypical antipsychotics. Neuroreport 10: 2533-2538.<br />

Zhang WN, Bast T, Feldon J (2002) Effects of hippocampal N-methyl-D-aspartate<br />

infusion on locomotor activity and prepulse inhibition: differences between the dorsal<br />

and ventral hippocampus. Behav Neurosci 116: 72-84.<br />

Zuckerman L, Rehavi M, Nachman R, Weiner I (2003) Immune activation during<br />

pregnancy in rats leads to a postpubertal emergence of disrupted latent inhibition,<br />

dopaminergic hyperfunction, and altered limbic morphology in the offspring: a novel<br />

neurodevelopmental model of schizophrenia. Neuropsychopharmacology 28: 1778-1789.<br />

159

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!