role of hippocampal neurogenesis in the etiology and

141
University of Pennsylvania University of Pennsylvania ScholarlyCommons ScholarlyCommons Publicly Accessible Penn Dissertations Summer 2011 Role Of Hippocampal Neurogenesis In The Etiology And Role Of Hippocampal Neurogenesis In The Etiology And Treatment Of Mood & Anxiety Disorders Treatment Of Mood & Anxiety Disorders Jennifer L. Onksen [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Behavioral Neurobiology Commons, Behavior and Behavior Mechanisms Commons, Mental Disorders Commons, and the Pharmacology Commons Recommended Citation Recommended Citation Onksen, Jennifer L., "Role Of Hippocampal Neurogenesis In The Etiology And Treatment Of Mood & Anxiety Disorders" (2011). Publicly Accessible Penn Dissertations. 387. https://repository.upenn.edu/edissertations/387 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/387 For more information, please contact [email protected].

Upload: others

Post on 28-Dec-2021

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Role Of Hippocampal Neurogenesis In The Etiology And

University of Pennsylvania University of Pennsylvania

ScholarlyCommons ScholarlyCommons

Publicly Accessible Penn Dissertations

Summer 2011

Role Of Hippocampal Neurogenesis In The Etiology And Role Of Hippocampal Neurogenesis In The Etiology And

Treatment Of Mood & Anxiety Disorders Treatment Of Mood & Anxiety Disorders

Jennifer L. Onksen [email protected]

Follow this and additional works at: https://repository.upenn.edu/edissertations

Part of the Behavioral Neurobiology Commons, Behavior and Behavior Mechanisms Commons,

Mental Disorders Commons, and the Pharmacology Commons

Recommended Citation Recommended Citation Onksen, Jennifer L., "Role Of Hippocampal Neurogenesis In The Etiology And Treatment Of Mood & Anxiety Disorders" (2011). Publicly Accessible Penn Dissertations. 387. https://repository.upenn.edu/edissertations/387

This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/387 For more information, please contact [email protected].

Page 2: Role Of Hippocampal Neurogenesis In The Etiology And

Role Of Hippocampal Neurogenesis In The Etiology And Treatment Of Mood & Role Of Hippocampal Neurogenesis In The Etiology And Treatment Of Mood & Anxiety Disorders Anxiety Disorders

Abstract Abstract Mood disorders are associated with deficits in hippocampal size and function. Neurogenesis in the dentate gyrus of the hippocampus has been implicated in the behavioral efficacy of some antidepressants. However, the precise function of newborn hippocampal neurons in the etiology and treatment of mood disorders remains unclear. Anxiety, which is highly comorbid with mood disorders, also appears to be influenced by neurogenesis. The intricacies of this influence are poorly understood. Here, the role of neurogenesis in the pathophysiology and treatment of mood and anxiety disorders is investigated in a transgenic mouse exhibiting partial suppression of hippocampal neurogenesis. This reduction is induced by hippocampal microinjection of a Cre-expressing adeno-associated virus to delete ATR, a cell cycle checkpoint kinase, from the hippocampus of adult mice. Subsequent to hippocampal ATR deletion, behavior is examined in mouse models of anxiety, depressive-like behavior, antidepressant efficacy, and susceptibility to stress. In addition, the necessity of heightened neurogenesis in exercise-induced anxiety is examined. ATR deletion resulted in an anhedonic phenotype in a sucrose drinking task but did not increase the likelihood of developing behavioral despair in the learned helplessness paradigm. Furthermore, mice lacking hippocampal ATR exhibited reduced anxiety in a number of behavioral tests. In the novelty-induced hypophagia task, the ability of chronic antidepressants to alleviate hyponeophagia was absent in ATR-deleted mice, and this effect was linked to an attenuation of the ability of chronic antidepressants to stimulate dendritic spine density. Finally, ATR deletion attenuated the neurogenic effect of voluntary wheel running, and concurrently blocked some of the anxiogenic effects of wheel running. In conclusion, neurogenesis appears to be an essential regulator of behaviors indicative of mood and anxiety disorders. These observations contribute to the body of work investigating the etiology and treatment of mood disorders for the purpose of guiding pharmaceutical drug discovery towards the eventual identification of antidepressant compounds with greater efficacy and fewer side effects.

Degree Type Degree Type Dissertation

Degree Name Degree Name Doctor of Philosophy (PhD)

Graduate Group Graduate Group Pharmacology

First Advisor First Advisor Dr. Julie A. Blendy

Keywords Keywords neurogenesis, antidepressants, depression, anxiety, stress, exercise

Subject Categories Subject Categories Behavioral Neurobiology | Behavior and Behavior Mechanisms | Mental Disorders | Pharmacology

This dissertation is available at ScholarlyCommons: https://repository.upenn.edu/edissertations/387

Page 3: Role Of Hippocampal Neurogenesis In The Etiology And

ROLE OF HIPPOCAMPAL NEUROGENESIS IN THE ETIOLOGY AND

TREATMENT OF MOOD & ANXIETY DISORDERS

Jennifer L Onksen

A DISSERTATION

in

Pharmacology

Presented to the Faculties of the University of Pennsylvania

in

Partial Fulfillment of the Requirements for the

Degree of Doctor of Philosophy

2011

Supervisor of Dissertation

Signature ________________________________

Julie A Blendy, Ph.D

Professor of Pharmacology

Graduate Group Chairperson

Signature_________________________________

Vladimir Muzykantov, MD, Ph.D

Professor of Pharmacology

Dissertation Committee

Steven Thomas, MD, Ph.D, Associate Professor of Pharmacology

Eric Brown, Ph.D, Assistant Professor of Cancer Biology

Irwin Lucki, Ph.D, Professor of Pharmacology

Brenda Porter, MD, Ph.D, Assistant Professor of Pediatrics and Neurology

Page 4: Role Of Hippocampal Neurogenesis In The Etiology And

ii

ABSTRACT

ROLE OF HIPPOCAMPAL NEUROGENESIS IN THE ETIOLOGY AND TREATMENT OF MOOD & ANXIETY DISORDERS

Jennifer L Onksen

Advisor: Julie A Blendy, Ph.D

Mood disorders are associated with deficits in hippocampal size and function.

Neurogenesis in the dentate gyrus of the hippocampus has been implicated in the

behavioral efficacy of some antidepressants. However, the precise function of newborn

hippocampal neurons in the etiology and treatment of mood disorders remains unclear.

Anxiety, which is highly comorbid with mood disorders, also appears to be influenced by

neurogenesis. The intricacies of this influence are poorly understood. Here, the role of

neurogenesis in the pathophysiology and treatment of mood and anxiety disorders is

investigated in a transgenic mouse exhibiting partial suppression of hippocampal

neurogenesis. This reduction is induced by hippocampal microinjection of a Cre-

expressing adeno-associated virus to delete ATR, a cell cycle checkpoint kinase, from

the hippocampus of adult mice. Subsequent to hippocampal ATR deletion, behavior is

examined in mouse models of anxiety, depressive-like behavior, antidepressant efficacy,

and susceptibility to stress. In addition, the necessity of heightened neurogenesis in

exercise-induced anxiety is examined. ATR deletion resulted in an anhedonic phenotype

in a sucrose drinking task but did not increase the likelihood of developing behavioral

despair in the learned helplessness paradigm. Furthermore, mice lacking hippocampal

ATR exhibited reduced anxiety in a number of behavioral tests. In the novelty-induced

hypophagia task, the ability of chronic antidepressants to alleviate hyponeophagia was

absent in ATR-deleted mice, and this effect was linked to an attenuation of the ability of

Page 5: Role Of Hippocampal Neurogenesis In The Etiology And

iii

chronic antidepressants to stimulate dendritic spine density. Finally, ATR deletion

attenuated the neurogenic effect of voluntary wheel running, and concurrently blocked

some of the anxiogenic effects of wheel running. In conclusion, neurogenesis appears to

be an essential regulator of behaviors indicative of mood and anxiety disorders. These

observations contribute to the body of work investigating the etiology and treatment of

mood disorders for the purpose of guiding pharmaceutical drug discovery towards the

eventual identification of antidepressant compounds with greater efficacy and fewer side

effects.

Page 6: Role Of Hippocampal Neurogenesis In The Etiology And

iv

Table of Contents

I. Introduction .................................................................................................. 1

A. Mood and anxiety disorders .................................................................................. 1

i. Mood disorders

ii. Anxiety disorders

B. Rodent models..................................................................................................... 3

i. Modeling depression

ii. Modeling anxiety

C. Hippocampal neurogenesis ................................................................................... 8

i. Characterization of hippocampal neurogenesis

ii. Neurogenesis in the etiology of mood and anxiety disorders

iii. Neurogenesis in the treatment of mood and anxiety disorders

D. Manipulating neurogenesis in rodents...................................................................12

i. Cytotoxic chemicals

ii. X-ray irradiation

iii. Genetic manipulation

E. Crelox-conditional ATR knockout ..........................................................................15

i. ATR, a cell cycle checkpoint kinase

ii. ATR deletion in the adult mouse

iii. Targeting ATR in the hippocampus

F. References..........................................................................................................17

II. Selective deletion of ATR reduces neurogenesis and alters responses in

rodent models of behavioral affect ................................................................. 29

A. Abstract..............................................................................................................30

B. Introduction........................................................................................................31

C. Materials and Methods.........................................................................................33

D. Results...............................................................................................................42

E. Figures ...............................................................................................................47

F. Discussion...........................................................................................................58

G. References .........................................................................................................64

Page 7: Role Of Hippocampal Neurogenesis In The Etiology And

v

III. Low neurogenesis does not increase susceptibility to stress-induced or

corticosterone-induced depressive behaviors in mice.................................... 96

A. Abstract..............................................................................................................70

B. Introduction........................................................................................................71

C. Materials and Methods.........................................................................................73

D. Results...............................................................................................................80

E. Figures ...............................................................................................................83

F. Discussion...........................................................................................................87

G. References .........................................................................................................91

IV. Mice deficient in hippocampal neurogenesis do not develop exercise-

induced anxiety............................................................................................... 96

A. Abstract..............................................................................................................97

B. Introduction........................................................................................................98

C. Materials and Methods.........................................................................................99

D. Results.............................................................................................................105

E. Figures .............................................................................................................110

F. Discussion.........................................................................................................117

G. References .......................................................................................................122

V. Conclusions and future directions ............................................................ 126

A. Discussion ........................................................................................................126

B. References .......................................................................................................132

Page 8: Role Of Hippocampal Neurogenesis In The Etiology And

vi

List of Illustrations

Chapter II

Figure 1. Hippocampal microinjection of AAV.Cre deletes ATR ....................................47

Figure 2. Hippocampal ATR deletion inhibits hippocampal neurogenesis ......................48

Figure 3. Hippocampal ATR deletion is associated with reduced anxiety in the

elevated zero maze (EZM) .........................................................................50

Figure 4. ATR deletion is associated with reduced anxiety ..........................................51

Figure 5. In the NIH paradigm, ATR-deleted mice exhibit altered

antidepressant response............................................................................52

Figure 6. Hippocampal CA1 dendritic spine density following DMI treatment................54

Figure s1. Hippocampal ATR deletion did not affect general locomotor activity

or HPA axis activation following an acute stress .........................................55

Figure s2. Fear conditioning is not altered by hippocampal ATR deletion .....................56

Figure s3. ATR deletion does not induce impairments in spatial learning

and memory in the Morris water maze ......................................................57

Chapter III

Figure 1. Sucrose preference and fluid intake ............................................................83

Figure 2. Learned helplessness.................................................................................84

Figure 3. Behavioral and endocrine consequences of chronic corticosterone ................85

Chapter IV

Figure 1. Experimental design ................................................................................110

Figure 2. ATRΔHipp mice exhibit deficits in running-induced neurogenesis....................111

Figure 3. Running-induced anxiety in the NIH test is attenuated in ATRΔHipp mice .......112

Figure 4. Running-induced anxiety in an open field is attenuated in ATRΔHipp mice ......113

Figure 5. Running-induced anxiety in the light-dark box is attenuated in

ATRΔHipp mice..........................................................................................115

Page 9: Role Of Hippocampal Neurogenesis In The Etiology And

1

CHAPTER I. INTRODUCTION

A. Mood and anxiety disorders

Mood and anxiety disorders are highly prevalent mental illnesses afflicting a

significant proportion of adults in the US each year. Among affected individuals, these

disorders are highly variable in their etiology and treatment outcome. A better

understanding of the pathophysiology of mood and anxiety disorders will not only

improve disease prognosis but also ease the large financial burden these illnesses impart

due to high healthcare and economic costs (Greenberg et al., 2003).

Mood disorders

Approximately 10% of adults in the US suffer from a mood disorder each year

(Kessler et al., 2005). The majority suffer from dysthymic or major depressive disorder,

both of which are characterized by persistent, depressive-like symptoms and differ in

regards to severity. The DSM-IV further subdivides depressive disorders into numerous

clinical subtypes, supporting the hypothesis that depression may represent a diverse set

of pathophysiologies presenting with similar, overlapping symptoms (Akiskal, 1989;

Fagiolini and Kupfer, 2003). Depression is defined as the presence of at least one two-

week episode of dysphoria and/or anhedonia plus a multitude of additional symptoms

including altered sleep/appetite, fatigue, agitation, inability to concentrate, and recurrent

thoughts of death or suicide (American Psychiatric Association. and American Psychiatric

Association. Task Force on DSM-IV., 2000). Depression is thought to be brought on by

an interaction between stressful life experiences and genetic susceptibility factors

(Goldberg, 2006), including alterations in the serotonin transporter gene (Caspi et al.,

Page 10: Role Of Hippocampal Neurogenesis In The Etiology And

2

2003). Treatment for depression includes cognitive behavioral therapy,

pharmacotherapy, and electroconvulsive therapy for severe, treatment resistant

depression. Current pharmacotherapies act to increase levels of monoamine transmitters

in the brain, either through blocking their reuptake at the synapse or through inhibiting

their degradation. Because these drugs require 4-6 weeks to relieve symptoms of

depression, they are thought to induce downstream changes in gene transcription and

neuronal plasticity following chronic administration (Hyman and Nestler, 1996). Current

pharmacotherapies often produce unwanted side effects and less than half of patients

show full remission of their disease, even following trials of multiple drugs (Berton and

Nestler, 2006).

Anxiety Disorders

Approximately 18% of adults in the US experience an anxiety disorder each year

(Kessler et al., 2005), including general anxiety, panic disorder, social anxiety, post-

traumatic stress disorder, and obsessive-compulsive disorder. Anxiety disorders are

marked by pervasive worry, tension and avoidance (Allgulander, 2006) and as such,

components of fear circuitry in the brain, including the amygdala, nucleus accumbens,

hippocampus and insular cortex are implicated in animal models (Kent and Rauch, 2003;

Davis, 2006; Maren, 2008). Treatment for anxiety includes benzodiazepines,

anticonvulsants, and the serotonin and norepinephrine reuptake inhibitors commonly

used in the treatment of depression (Baldwin et al., 2011). Depression and anxiety

disorders are highly comorbid; up to 60% of depressed individuals also suffer from

anxiety symptoms (Kessler et al., 2003). This comorbidity, in addition the ability of

Page 11: Role Of Hippocampal Neurogenesis In The Etiology And

3

selective serotonin reuptake inhibitors (SSRIs) to treat both disorders (Sramek et al.,

2002; Millan, 2003) suggests they may share a common underlying pathophysiology.

B. Modeling mood disorders in rodents

Extensive efforts have been made to model depression in rodents in order to

study the cellular and molecular changes which occur concurrent with the onset of

depressive-like behavior and the reversal by antidepressant drugs. Because the etiology

of depression is poorly understood, researchers often expose rodents to stressful

conditions over a period of days or weeks. Changes that occur include anhedonia,

behavioral despair, anxiety, alterations in gene transcription, dysfunctional HPA axis, and

reduced hippocampal neurogenesis.

Modeling Depression

Unpredictable Chronic Mild Stress (UCMS)

UCMS involves exposing rodents to a variety of unpredictable stressors including

dirty cages, wet bedding, food/water restriction, mild footshock, restraint, extreme

temperature, and cage tilt (Willner, 2005). Unpredictability is essential to the

development of a disease state, as predictable chronic stress has actually been shown to

enhance mood, neurogenesis, and memory (Parihar et al., 2011). UCMS results in

anhedonia, as measured by reduced interest in sucrose water, reduced brain stimulation

reward, and reduced hippocampal neurogenesis (Willner et al., 1987; Papp et al., 1991;

Moreau et al., 1993; Alonso et al., 2004). In addition to modeling some of the symptoms

of depression observed in humans, UCMS-induced behavioral alterations can be reversed

Page 12: Role Of Hippocampal Neurogenesis In The Etiology And

4

by chronic treatment with antidepressants (Alonso et al., 2004; Luo et al., 2005;

Jayatissa et al., 2006).

Learned Helplessness (LH)

The LH paradigm, first proposed by Overmier and Seligman (1967), causes

behavioral despair and deficits in coping behavior reminiscent of symptoms observed in

clinically depressed individuals. Rodents exposed to unavoidable, uncontrollable

footshocks develop escape deficits when exposed to escapable footshocks. This

behavior, as well as reduced cell proliferation which occurs as a result of acute

footshock, can be reversed by chronic treatment with an antidepressant (Malberg and

Duman, 2003). Interestingly, LH experiments in rats have demonstrated that reduced

hippocampal neurogenesis does not influence susceptibility to helplessness (Vollmayr et

al., 2003; Reif et al., 2004). Very little work has been done to examine neurogenesis in

the context of learned helplessness in mice. In addition, numerous studies have

suggested an individual susceptibility to helplessness among cohorts of animals

(Vollmayr et al., 2003; Chourbaji et al., 2005).

Chronic Social Stress

Many stressors which lead to mental illness are of a social nature (Brown and

Prudo, 1981). In primates and tree shrews, psychosocial stress leads to hippocampal

deficits in dendritic structure and cell proliferation (Uno et al., 1989; Magarinos et al.,

1996). In rodents, chronic social defeat by an aggressor induces numerous behavioral

changes representative of negative mood state, including anhedonia, anxiety, behavioral

despair, reduced motivation, social avoidance, and reduced hippocampal neurogenesis

Page 13: Role Of Hippocampal Neurogenesis In The Etiology And

5

(Avgustinovich, 2005; Rygula et al., 2005; Berton et al., 2006; Yap et al., 2006;

Schloesser et al., 2010). Stimulation of hippocampal neurogenesis may play an

important role in recovery from social stress (Schloesser et al., 2010).

Exogenous Corticosterone

In response to stress, a major risk factor for depression (Kessler, 1997), the

hypothalamic-pituitary-adrenal (HPA) axis is activated, leading to production of

glucocorticoid stress hormones which activate glucocorticoid receptors (GR) in the

hippocampus. This leads to inhibitory feedback and suppression of subsequent

glucocorticoid production (Herman et al., 1989; Jacobson and Sapolsky, 1991; van

Haarst et al., 1997). In depression, HPA axis dysfunction commonly occurs and as many

as 50% of depressed individuals exhibit impaired GR negative feedback (Gibbons, 1964).

In animal models of depression, elevated circulating corticosterone is often observed

(Czeh et al., 2002; Harvey et al., 2003). Administering exogenous corticosterone is

thought to mimic this aspect of stress and is thus used to investigate the impact of HPA

axis activation on depressive-like behaviors. Chronic low dose corticosterone reduces

hippocampal neurogenesis and induces depressive-like behavior in rodents (Murray et

al., 2008; Zhao et al., 2008; Crupi et al., 2010).

Forced Swim Test (FST), Tail Suspension Test (TST)

In the FST, a rodent is placed into a beaker of water and the time it spends

immobile is measured. In the TST, a rodent is suspended by its tail and the time

immobile is measured. Immobility is interpreted as behavioral despair, a possible

symptom of depression. While the construct and face validity of this model may be

Page 14: Role Of Hippocampal Neurogenesis In The Etiology And

6

called into question, immobility can be reduced by acute and chronic treatment with

most currently prescribed antidepressants, suggesting strong predictive validity for

identifying new potential antidepressants. Thus, these tests are often used as screens

for antidepressants (Lucki, 1997; Porsolt, 2000), rather than measures of depressive-like

behavior. However, it is not known whether only monoamine based drugs can be

detected, or if reduced immobility generalizes to antidepressant drugs with different

mechanisms of action, as compounds with non-monoamine mechanisms are not widely

used to treat depression as of yet (Nestler et al., 2002).

Modeling Anxiety

Mouse models of anxiety can be categorized as conditioned response tests or

unconditioned response tests (Belzung and Griebel, 2001, for review). Conditioned

response tests involve a stressful event and include conflict tests, active/passive

avoidance, and defensive burying. Unconditioned response tests are most commonly

used to measure anxiety and include exploration tests (maze, holeboard, dark/light

choice, open field), social tests, startle responses and separation or shock-induced

ultrasonic vocalizations. Many of these tests take advantage of the natural behavior of

mice to avoid bright, open, novel and/or aversive stimuli. Anxiety tests are validated by

the ability of anxiolytic agents such as benzodiazepines to modify behavior. This section

will cover anxiety tests relevant to the data presented in the subsequent chapters.

Exploratory-based approach/avoidance tests

The open field (Christmas and Maxwell, 1970), elevated zero maze (Shepherd et

al., 1994), and light-dark box (Crawley and Goodwin, 1980) tests all feature an

Page 15: Role Of Hippocampal Neurogenesis In The Etiology And

7

environment in which a mouse has the option of exploring an adverse environment or

remaining in a less anxiety-provoking location within the test arena. Treatment with

benzodiazepines reduces anxiety as measured by increased exploration of the open

arms of the zero maze, the light compartment of the light-dark box, and the center of

the open field.

Marble burying

Rodents will use bedding material to bury unfamiliar objects in their cage in

order to avoid what they perceive as a potential threat (Archer et al., 1987).

Benzodiazepines (Broekkamp et al., 1986; Archer et al., 1987), as well as certain

antidepressant compounds (Borsini et al., 2002), reduce marble burying without altering

locomotor activity (Nicolas et al., 2006). It has been proposed that marble burying

models components of obsessive-compulsive disorder, rather than anxiety per se

(Albelda and Joel, 2011). Nonetheless, the proven efficacy of anxiolytic agents to

change behavior in this test makes it a useful measure of anxiety-like behavior when

used in conjunction with other tests.

Hyponeophagia

Hyponeophagia paradigms serve a dual purpose: measuring anxiety state and

modeling chronic antidepressant treatment. Hyponeophagia paradigms come in two

varieties, novelty-suppressed feeding (NSF), and novelty-induced hyponeophagia (NIH)

(Bodnoff et al., 1988; Dulawa et al., 2004). In general, these paradigms measure

latency to consume food in a novel, anxiety-provoking environment. In the NSF

paradigm, mice are food-deprived and subsequently given access to a regular food

Page 16: Role Of Hippocampal Neurogenesis In The Etiology And

8

pellet in a novel environment. In the NIH paradigm, mice are trained to consume a

palatable food in their home cage, and subsequently presented with the opportunity to

consume the palatable food in a novel environment, in a fed state. In hyponeophagia

paradigms, mice face a conflict between avoiding a novel environment and the desire to

feed (Merali et al., 2003; Dulawa and Hen, 2005). Thus, this is a test of anxiety, and

latency is reduced by acute administration of a benzodiazepine. Additionally, chronic

treatment with antidepressants, but not acute treatment, will reduce latency to feed,

suggesting this is a valuable model for evaluating the cellular and molecular mechanisms

of antidepressant drugs. Of interest, ablating hippocampal neurogenesis with x-ray

irradiation blocks the efficacy of two antidepressants, the SSRI fluoxetine and the

tricyclic desipramine (Santarelli et al., 2003).

C. Hippocampal neurogenesis

The hippocampus is located in the medial temporal lobe of the brain, makes up

part of the limbic system, and is important for consolidating information into memory

and for spatial navigation. In addition, the hippocampus has been implicated in the

pathophysiology of mood disorders. Clinical studies in depressed individuals have

demonstrated reduced hippocampal volume which is reversed by antidepressant

treatment (Sheline et al., 1999). Additionally, reduced hippocampal volume may be

indicative of increased vulnerability to psychiatric illness (Gilbertson et al., 2002). The

hippocampus shares a unique feature with just one other brain region, the ability to

produce a substantial amount of new neurons through adulthood, a process called

hippocampal neurogenesis. As discussed below, extensive research has focused on

Page 17: Role Of Hippocampal Neurogenesis In The Etiology And

9

understanding the process of hippocampal neurogenesis and the function of these

newborn neurons, especially in the context of mood disorders and their treatment.

Characterizing neurogenesis

Hippocampal neurogenesis has been observed in rodents (Kaplan and Hinds,

1977), non-human primates (Gould et al., 1999), as well as in humans (Eriksson et al.,

1998). Within the dentate gyrus of the hippocampus, multipotent progenitor cells in the

subgranular zone give rise to new-born neurons which migrate to the inner granule cell

layer and mature to give rise to granule neurons (For exhaustive review, see Ming and

Song, 2005). These cells extend axonal projections to the CA3 pyramidal cell layer

(Hastings and Gould, 1999), while their dendrites extend into the molecular layer and

grow increasingly complex over a period of months (van Praag et al., 2002). Functional

integration begins as soon as one week after proliferation and the newborn neurons

continue to form additional synaptic connections in the subsequent months. Young

neurons exhibit unique properties compared to more mature granule neurons, including

a lower threshold for excitation (Schmidt-Hieber et al., 2004) and long-term potentiation

(Wang et al., 2000).

Newborn neurons are essential for certain types of hippocampal-dependent

learning, including pattern separation (Clelland et al., 2009; Aimone et al., 2010; Sahay

et al., 2011), the ability to encode and subsequently differentiate between highly similar

events or stimuli. The exact role of new neurons in emotional regulation and mood state

is not so clear.

Research into hippocampal neurogenesis has identified an extensive list of

intrinsic and extrinsic factors, ranging from behavioral (Lucassen et al., 2010) to

Page 18: Role Of Hippocampal Neurogenesis In The Etiology And

10

molecular (Hagg, 2005), which regulate all stages of neurogenesis. Factors which

increase neurogenesis include mood-improving antidepressant compounds (Malberg et

al., 2000; Manev et al., 2001; Nakagawa et al., 2002), exercise (van Praag et al., 1999),

environmental enrichment (Kempermann et al., 1997; Brown et al., 2003; Llorens-Martin

et al., 2007) and learning (Gould et al., 1999). Factors which contribute to reduced

hippocampal neurogenesis include various forms of stress (For review, Warner-Schmidt

and Duman, 2006), aging (Eckenhoff and Rakic, 1988; Kuhn et al., 1996) and disease

(Danzer, 2008). In addition to the aforementioned endogenous stimuli, numerous

proteins, neurotransmitters, hormones and epigenetic regulation modulate the

proliferation, differentiation, or survival of newborn neurons (For review, Hagg, 2005;

Hsieh and Eisch, 2010). Finally, a number of trangenic mice exist in which aspects of

hippocampal neurogenesis is altered; models relevant to the work presented in the

subsequent chapters will be covered below.

Neurogenesis in the etiology of mood and anxiety disorders

The neurogenesis hypothesis of depression was originally formulated based on

extensive evidence that stress, a major risk factor for depression, suppressed

hippocampal neurogenesis while pharmacotherapeutic antidepressants elevated

neurogenesis. However, more recent work in rodent models indicates depressed mood

can be elicited through various forms of stress without necessarily inhibiting

neurogenesis. In addition, a number of non-neurogenic phenomena have been linked to

the onset of, and recovery from, depressive-like behavior in rodents, including dentate

gyrus evoked activity (Airan et al., 2007), hippocampal circuit dynamics (Meltzer et al.,

2005), neuronal remodeling (Bessa et al., 2009) and neuroplasticity (Shors et al., 1989).

Page 19: Role Of Hippocampal Neurogenesis In The Etiology And

11

Many of these phenomena are hippocampal and may be influenced by neurogenesis, but

they nonetheless support the theory that reduced neurogenesis alone does not

guarantee depressed mood in all circumstances.

The relationship between neurogenesis and anxiety is currently not clear, as the

outcomes of studies investigating this possible relationship are inconsistent. Exercise,

which enhances neurogenesis, has been shown to both increase (Burghardt et al., 2004;

Fuss et al., 2010) and decrease anxiety (Dishman et al., 1996; Salam et al., 2009),

suggesting a complex relationship dependent on rodent strain, housing conditions,

exercise protocol, timing of tests in relation to running, and particular tasks utilized to

measure anxiety. At the other end of the spectrum, mouse models of suppressed

hippocampal neurogenesis do no better in clarifying the neurogenesis/anxiety

relationship. Some studies observe no link (Saxe et al., 2006), while others show

heightened (Bergami et al., 2009; Revest et al., 2009) or reduced (Uchida et al., 2011)

anxiety in conjunction with suppressed neurogenesis.

Thus, evaluating depressive and anxiety-like behaviors in the mouse model of

reduced neurogenesis employed in the subsequent chapters was an important aim of

this dissertation.

Neurogenesis in the treatment of mood and anxiety disorders

In contrast to the dearth of evidence supporting reduced neurogenesis as a

causal factor in the onset of depression, enhanced hippocampal neurogenesis may be

essential to the efficacy of specific antidepressant pharmacotherapies in certain

behavioral measures. Chronic administration of antidepressants with monoaminergic

mechanisms require hippocampal neurogenesis for their efficacy in a hyponeophagia

Page 20: Role Of Hippocampal Neurogenesis In The Etiology And

12

task (Santarelli et al., 2003) and for reversal of the behavioral effects of chronic mild

stress (Surget et al., 2008). Antidepressant drugs with alternative mechanisms of action,

including an MCHR1 antagonist, CRF1 antagonist and vasopressin 1b antagonist, are

effective in relieving depression and anxiety-like behaviors in mice x-ray irradiated to

block neurogenesis (David et al., 2007; Surget et al., 2008). More recently, in a chronic

corticosterone model of anxiety/depressive like state, fluoxetine was found to require

neurogenesis for mood improving actions in some, but not all behavioral tests,

suggesting particular drugs can exert both neurogenesis-dependent and neurogenesis-

independent actions (David et al., 2009).

Furthermore, not all mood-improving stimuli require neurogenesis.

Environmental enrichment, which is shown to increase hippocampal neurogenesis,

alleviates hyponeophagia and reduces anxiety in mice x-ray irradiated to block

neurogenesis (Meshi et al., 2006). The requirement of neurogenesis for antidepressant

efficacy is also strain dependent, as demonstrated in BALB/cJ mice (Holick et al., 2008).

D. Methods of manipulating neurogenesis in rodents

With the goal of identifying the functional relevance of neurogenesis and the behaviors

influenced by newborn neurons, a number of methods have been developed to suppress

neurogenesis including chemical treatment, irradiation, and genetic manipulation.

Chemical treatment

Methylazoxymethanol acetate (MAM) is a toxin which inhibits proliferating cells.

It was used in adult rats to demonstrate a role of newborn neurons in certain types of

learning and memory (Shors et al., 2002; Bruel-Jungerman et al., 2005). MAM was later

Page 21: Role Of Hippocampal Neurogenesis In The Etiology And

13

shown to alter health at the dose necessary to reduce neurogenesis and is utilized

infrequently today (Dupret et al., 2005).

X-ray irradiation

A popular approach to studying the function of neural stem/progenitor cells in

the hippocampus is to expose the brain region to focal x-ray irradiation. A single 15-gray

exposure results in at least 85% suppression of neurogenesis and lasts for up to 3

months (McGinn et al., 2008). This is a consistent, reproducible approach that allows for

the generation of a large cohort of neurogenesis-deficient mice in a short time.

However, there are some drawbacks to the x-ray irradiation approach. X-ray irradiation

can affect differentiation, increase activated microglia and disrupt angiogenesis (Tada et

al., 2000; Monje et al., 2002; Mizumatsu et al., 2003). Nonetheless, this method has

been essential in advancing the field’s understanding of the role of new neurons,

including in the first study to demonstrate a causal link between neurogenesis and the

efficacy of antidepressant drugs (Santarelli et al., 2003).

Genetic models

To compliment and confirm the findings of x-ray irradiation studies, researchers

have developed transgenic approaches to ablating neurogenesis. The first to be used

directly alongside x-ray irradiation was a transgenic mouse in which herpes simplex virus

thymidine kinase (TK) is expressed under control of the GFAP promoter (Saxe et al.,

2006; Saxe et al., 2007). Proliferating cells in the hippocampus are GFAP-expressing.

Subsequent administration of ganciclovir (GCV) kills proliferating, TK-positive cells, thus

ablating neurogenesis. To reduce toxicity and enhance specificity (Bush et al., 1998;

Page 22: Role Of Hippocampal Neurogenesis In The Etiology And

14

Bush et al., 1999), this model was later expanded to the nestin promoter, such that

nestin-expressing progenitor cells would be killed upon administration of ganciclovir,

with similar effects on neurogenesis (Singer et al., 2009). Another similar approach

involves doxycycline-inducible over-expression of the pro-apoptotic factor Bax in neural

progenitor cells through a Nestin promoter (Revest et al., 2009). This results in a 50%

reduction in neurogenesis and altered mood/anxiety-related behaviors.

A number of proteins important for cell cycle regulation, as well as those

upstream of direct cell cycle regulation, are necessary for normal hippocampal

neurogenesis, including Cdk5, Bmi1 and cyclin D2 (Kowalczyk et al., 2004; Zencak et al.,

2005; Lagace et al., 2008; Jaholkowski et al., 2009). Deletion of these various cell cycle

elements results in either partial or complete attenuation of neurogenesis, generating

models for investigating the functional relevance of neurogenesis. ATR, as a cell cycle

checkpoint kinase, falls into this category.

As previously mentioned, an extensive list of extrinsic and intrinsic factors

regulate hippocampal neurogenesis. This is exemplified by the many transgenic mice

which exhibit altered hippocampal neurogenesis and alterations in mood and anxiety-like

behaviors. Genes, and their protein products, shown to influence hippocampal

neurogenesis include p21Cip1, GABAA γ2 subunit, p75 neurotrophin receptor, AMPA

receptor GluA1 subunit, p11 (S100A10), heat shock factor 1 (HSF1), Kruppel-like factor

9 (Klf9), connexin, NeuroD1, Fmr1, and macrophage migration inhibitory factor (MMIF)

(Earnheart et al., 2007; Catts et al., 2008; Pechnick et al., 2008; Eadie et al., 2009; Gao

et al., 2009; Kunze et al., 2009; Egeland et al., 2010; Conboy et al., 2011; Procaccini et

al., 2011). These transgenic models present numerous caveats including developmental

Page 23: Role Of Hippocampal Neurogenesis In The Etiology And

15

effects and non-specific effects. Nonetheless, they provide additional clues and support

for a complex relationship between neurogenesis and behavior.

E. Crelox-conditional ATR knockout

ATR (Ataxia-telangiectasia mutated (ATM) and Rad3-related)

ATR is a cell cycle checkpoint kinase and an essential regulator of DNA

replication. It is necessary for stabilization of the replication fork during a stall in DNA

replication. In its absence, double strand breaks and subsequent loss of genomic

integrity are observed (Paulsen and Cimprich, 2007). In cell culture, cell cycle arrest and

cell death occur in murine embryonic fibroblasts and human cells lacking ATR (Cortez et

al., 2001; Brown and Baltimore, 2003). In mouse embryos, constitutive deletion of ATR

leads to chromosomal fragmentation and early embryonic lethality; cultured blastocysts

die prior to embryonic day 7 by caspase-independent apoptosis, implying loss of

genomic integrity (Brown and Baltimore, 2000).

ATR deletion from the adult mouse

Ubiquitous deletion of ATR from the adult mouse leads to aging-like phenotypes

associated with premature depletion of stem and progenitor cell pools (Ruzankina et al.,

2007). This observation made ATR a strong potential candidate for suppressing

neurogenesis in the hippocampus, as it depends on a pool of progenitor cells in the

subgranular zone. Ruzankina and colleagues also demonstrated that ATR could be

deleted from mature forebrain CNS neurons, including those in the hippocampus, by

Synapsin1 promoter-driven Cre expression, with no effects on circadian period,

Page 24: Role Of Hippocampal Neurogenesis In The Etiology And

16

coordination, strength, anxiety, or hippocampal dependent learning. These observations

suggest ATR is not necessary for the function of mature neurons in the hippocampus

and implicates loss of ATR from the proliferating cell population in the behavioral

consequences that we observe following hippocampal-specific deletion.

Targeting ATR in the hippocampus

Hippocampal ATR deletion was achieved by microinjecting an adeno-associated virus

(AAV) encoding Cre recombinase (Cre) directly into the hippocampus of mice with loxP

sequences flanking the kinase domain of ATR (ATRf/f). An AAV was selected due to its

persistent expression and minimal evoked immune response (Cearley and Wolfe, 2006).

AAV expression peaked at approximately 4 weeks and all experiments were conducted

at least 6 weeks following AAV injection to assure deletion of ATR and suppression of

neurogenesis. In addition, we treated ATRf/f mice carrying a tamoxifen-inducible Cre

transgene (ATRf/f;CreERT2) with tamoxifen to induce a ubiquitous ATR deletion. This

was done to verify that the cellular and behavioral effects of the AAV injection were due

to ATR deletion, and not an undesired side effect of the AAV. In the subsequent

chapters, ATR deletion is utilized as a model of suppressed hippocampal neurogenesis to

explore the influence of neurogenesis in the onset and treatment of mood and anxiety

disorders, as well as to examine the neurogenesis-dependent effects of exercise, which

is a potent stimulator of neurogenesis and a mood-improving stimulus in the human

population.

Page 25: Role Of Hippocampal Neurogenesis In The Etiology And

17

References Aimone JB, Deng W, Gage FH (2010). Adult neurogenesis: integrating theories and

separating functions. Trends Cogn Sci 14(7): 325-337.

Airan RD, Meltzer LA, Roy M, Gong Y, Chen H, Deisseroth K (2007). High-speed imaging reveals neurophysiological links to behavior in an animal model of depression. Science 317(5839): 819-823.

Akiskal HS (1989). New insights into the nature and heterogeneity of mood disorders. J Clin Psychiatry 50 Suppl: 6-10; discussion 11-12.

Albelda N, Joel D (2011). Animal models of obsessive-compulsive disorder: Exploring pharmacology and neural substrates. Neurosci Biobehav Rev.

Allgulander C (2006). Generalized anxiety disorder: What are we missing? Eur Neuropsychopharmacol 16 Suppl 2: S101-108.

Alonso R, Griebel G, Pavone G, Stemmelin J, Le Fur G, Soubrie P (2004). Blockade of CRF(1) or V(1b) receptors reverses stress-induced suppression of neurogenesis in a mouse model of depression. Mol Psychiatry 9(3): 278-286, 224.

American Psychiatric Association., American Psychiatric Association. Task Force on DSM-IV. (2000). Diagnostic and statistical manual of mental disorders : DSM-IV-TR. Washington, DC, American Psychiatric Association.

Archer T, Fredriksson A, Lewander T, Soderberg U (1987). Marble burying and spontaneous motor activity in mice: interactions over days and the effect of diazepam. Scand J Psychol 28(3): 242-249.

Avgustinovich DF (2005). Anxiety in females induced by long-lasting psychoemotional influences. Neurosci Behav Physiol 35(2): 193-199.

Baldwin D, Woods R, Lawson R, Taylor D (2011). Efficacy of drug treatments for generalised anxiety disorder: systematic review and meta-analysis. BMJ 342: d1199.

Belzung C, Griebel G (2001). Measuring normal and pathological anxiety-like behaviour in mice: a review. Behav Brain Res 125(1-2): 141-149.

Bergami M, Berninger B, Canossa M (2009). Conditional deletion of TrkB alters adult hippocampal neurogenesis and anxiety-related behavior. Commun Integr Biol 2(1): 14-16.

Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, Russo SJ, Graham D, Tsankova NM, Bolanos CA, Rios M, Monteggia LM, Self DW, Nestler EJ (2006). Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 311(5762): 864-868.

Page 26: Role Of Hippocampal Neurogenesis In The Etiology And

18

Berton O, Nestler EJ (2006). New approaches to antidepressant drug discovery: beyond monoamines. Nat Rev Neurosci 7(2): 137-151.

Bessa JM, Ferreira D, Melo I, Marques F, Cerqueira JJ, Palha JA, Almeida OF, Sousa N (2009). The mood-improving actions of antidepressants do not depend on neurogenesis but are associated with neuronal remodeling. Mol Psychiatry 14(8): 764-773, 739.

Bodnoff SR, Suranyi-Cadotte B, Aitken DH, Quirion R, Meaney MJ (1988). The effects of chronic antidepressant treatment in an animal model of anxiety. Psychopharmacology (Berl) 95(3): 298-302.

Borsini F, Podhorna J, Marazziti D (2002). Do animal models of anxiety predict anxiolytic-like effects of antidepressants? Psychopharmacology (Berl) 163(2): 121-141.

Broekkamp CL, Rijk HW, Joly-Gelouin D, Lloyd KL (1986). Major tranquillizers can be distinguished from minor tranquillizers on the basis of effects on marble burying and swim-induced grooming in mice. Eur J Pharmacol 126(3): 223-229.

Brown EJ, Baltimore D (2000). ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev 14(4): 397-402.

Brown EJ, Baltimore D (2003). Essential and dispensable roles of ATR in cell cycle arrest and genome maintenance. Genes Dev 17(5): 615-628.

Brown GW, Prudo R (1981). Psychiatric disorder in a rural and an urban population: 1. Aetiology of depression. Psychol Med 11(3): 581-599.

Brown J, Cooper-Kuhn CM, Kempermann G, Van Praag H, Winkler J, Gage FH, Kuhn HG (2003). Enriched environment and physical activity stimulate hippocampal but not olfactory bulb neurogenesis. Eur J Neurosci 17(10): 2042-2046.

Bruel-Jungerman E, Laroche S, Rampon C (2005). New neurons in the dentate gyrus are involved in the expression of enhanced long-term memory following environmental enrichment. Eur J Neurosci 21(2): 513-521.

Burghardt PR, Fulk LJ, Hand GA, Wilson MA (2004). The effects of chronic treadmill and wheel running on behavior in rats. Brain Res 1019(1-2): 84-96.

Bush TG, Puvanachandra N, Horner CH, Polito A, Ostenfeld T, Svendsen CN, Mucke L, Johnson MH, Sofroniew MV (1999). Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice. Neuron 23(2): 297-308.

Bush TG, Savidge TC, Freeman TC, Cox HJ, Campbell EA, Mucke L, Johnson MH, Sofroniew MV (1998). Fulminant jejuno-ileitis following ablation of enteric glia in adult transgenic mice. Cell 93(2): 189-201.

Page 27: Role Of Hippocampal Neurogenesis In The Etiology And

19

Caspi A, Sugden K, Moffitt TE, Taylor A, Craig IW, Harrington H, McClay J, Mill J, Martin J, Braithwaite A, Poulton R (2003). Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science 301(5631): 386-389.

Catts VS, Al-Menhali N, Burne TH, Colditz MJ, Coulson EJ (2008). The p75 neurotrophin receptor regulates hippocampal neurogenesis and related behaviours. Eur J Neurosci 28(5): 883-892.

Cearley CN, Wolfe JH (2006). Transduction characteristics of adeno-associated virus vectors expressing cap serotypes 7, 8, 9, and Rh10 in the mouse brain. Mol Ther 13(3): 528-537.

Chourbaji S, Zacher C, Sanchis-Segura C, Spanagel R, Gass P (2005). Social and structural housing conditions influence the development of a depressive-like phenotype in the learned helplessness paradigm in male mice. Behav Brain Res 164(1): 100-106.

Christmas AJ, Maxwell DR (1970). A comparison of the effects of some benzodiazepines and other drugs on aggressive and exploratory behaviour in mice and rats. Neuropharmacology 9(1): 17-29.

Clelland CD, Choi M, Romberg C, Clemenson GD, Jr., Fragniere A, Tyers P, Jessberger S, Saksida LM, Barker RA, Gage FH, Bussey TJ (2009). A functional role for adult hippocampal neurogenesis in spatial pattern separation. Science 325(5937): 210-213.

Conboy L, Varea E, Castro JE, Sakouhi-Ouertatani H, Calandra T, Lashuel HA, Sandi C (2011). Macrophage migration inhibitory factor is critically involved in basal and fluoxetine-stimulated adult hippocampal cell proliferation and in anxiety, depression, and memory-related behaviors. Mol Psychiatry 16(5): 533-547.

Cortez D, Guntuku S, Qin J, Elledge SJ (2001). ATR and ATRIP: partners in checkpoint signaling. Science 294(5547): 1713-1716.

Crawley J, Goodwin FK (1980). Preliminary report of a simple animal behavior model for the anxiolytic effects of benzodiazepines. Pharmacol Biochem Behav 13(2): 167-170.

Crupi R, Mazzon E, Marino A, La Spada G, Bramanti P, Cuzzocrea S, Spina E (2010). Melatonin treatment mimics the antidepressant action in chronic corticosterone-treated mice. J Pineal Res 49(2): 123-129.

Czeh B, Welt T, Fischer AK, Erhardt A, Schmitt W, Muller MB, Toschi N, Fuchs E, Keck ME (2002). Chronic psychosocial stress and concomitant repetitive transcranial magnetic stimulation: effects on stress hormone levels and adult hippocampal neurogenesis. Biol Psychiatry 52(11): 1057-1065.

Danzer SC (2008). Postnatal and adult neurogenesis in the development of human disease. Neuroscientist 14(5): 446-458.

Page 28: Role Of Hippocampal Neurogenesis In The Etiology And

20

David DJ, Klemenhagen KC, Holick KA, Saxe MD, Mendez I, Santarelli L, Craig DA, Zhong H, Swanson CJ, Hegde LG, Ping XI, Dong D, Marzabadi MR, Gerald CP, Hen R (2007). Efficacy of the MCHR1 antagonist N-[3-(1-{[4-(3,4-difluorophenoxy)phenyl]methyl}(4-piperidyl))-4-methylphen yl]-2-methylpropanamide (SNAP 94847) in mouse models of anxiety and depression following acute and chronic administration is independent of hippocampal neurogenesis. J Pharmacol Exp Ther 321(1): 237-248.

David DJ, Samuels BA, Rainer Q, Wang JW, Marsteller D, Mendez I, Drew M, Craig DA, Guiard BP, Guilloux JP, Artymyshyn RP, Gardier AM, Gerald C, Antonijevic IA, Leonardo ED, Hen R (2009). Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron 62(4): 479-493.

Davis M (2006). Neural systems involved in fear and anxiety measured with fear-potentiated startle. Am Psychol 61(8): 741-756.

Dishman RK, Dunn AL, Youngstedt SD, Davis JM, Burgess ML, Wilson SP, Wilson MA (1996). Increased open field locomotion and decreased striatal GABAA binding after activity wheel running. Physiol Behav 60(3): 699-705.

Dulawa SC, Hen R (2005). Recent advances in animal models of chronic antidepressant effects: the novelty-induced hypophagia test. Neurosci Biobehav Rev 29(4-5): 771-783.

Dulawa SC, Holick KA, Gundersen B, Hen R (2004). Effects of chronic fluoxetine in animal models of anxiety and depression. Neuropsychopharmacology 29(7): 1321-1330.

Dupret D, Montaron MF, Drapeau E, Aurousseau C, Le Moal M, Piazza PV, Abrous DN (2005). Methylazoxymethanol acetate does not fully block cell genesis in the young and aged dentate gyrus. Eur J Neurosci 22(3): 778-783.

Eadie BD, Zhang WN, Boehme F, Gil-Mohapel J, Kainer L, Simpson JM, Christie BR (2009). Fmr1 knockout mice show reduced anxiety and alterations in neurogenesis that are specific to the ventral dentate gyrus. Neurobiol Dis 36(2): 361-373.

Earnheart JC, Schweizer C, Crestani F, Iwasato T, Itohara S, Mohler H, Luscher B (2007). GABAergic control of adult hippocampal neurogenesis in relation to behavior indicative of trait anxiety and depression states. J Neurosci 27(14): 3845-3854.

Eckenhoff MF, Rakic P (1988). Nature and fate of proliferative cells in the hippocampal dentate gyrus during the life span of the rhesus monkey. J Neurosci 8(8): 2729-2747.

Page 29: Role Of Hippocampal Neurogenesis In The Etiology And

21

Egeland M, Warner-Schmidt J, Greengard P, Svenningsson P (2010). Neurogenic effects of fluoxetine are attenuated in p11 (S100A10) knockout mice. Biol Psychiatry 67(11): 1048-1056.

Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA, Gage FH (1998). Neurogenesis in the adult human hippocampus. Nat Med 4(11): 1313-1317.

Fagiolini A, Kupfer DJ (2003). Is treatment-resistant depression a unique subtype of depression? Biol Psychiatry 53(8): 640-648.

Fuss J, Ben Abdallah NM, Vogt MA, Touma C, Pacifici PG, Palme R, Witzemann V, Hellweg R, Gass P (2010). Voluntary exercise induces anxiety-like behavior in adult C57BL/6J mice correlating with hippocampal neurogenesis. Hippocampus 20(3): 364-376.

Gao Z, Ure K, Ables JL, Lagace DC, Nave KA, Goebbels S, Eisch AJ, Hsieh J (2009). Neurod1 is essential for the survival and maturation of adult-born neurons. Nat Neurosci 12(9): 1090-1092.

Gibbons JL (1964). Cortisol Secretion Rate in Depressive Illness. Arch Gen Psychiatry 10: 572-575.

Gilbertson MW, Shenton ME, Ciszewski A, Kasai K, Lasko NB, Orr SP, Pitman RK (2002). Smaller hippocampal volume predicts pathologic vulnerability to psychological trauma. Nat Neurosci 5(11): 1242-1247.

Goldberg D (2006). The aetiology of depression. Psychol Med 36(10): 1341-1347.

Gould E, Beylin A, Tanapat P, Reeves A, Shors TJ (1999). Learning enhances adult neurogenesis in the hippocampal formation. Nat Neurosci 2(3): 260-265.

Gould E, Reeves AJ, Fallah M, Tanapat P, Gross CG, Fuchs E (1999). Hippocampal neurogenesis in adult Old World primates. Proc Natl Acad Sci U S A 96(9): 5263-5267.

Greenberg PE, Kessler RC, Birnbaum HG, Leong SA, Lowe SW, Berglund PA, Corey-Lisle PK (2003). The economic burden of depression in the United States: how did it change between 1990 and 2000? J Clin Psychiatry 64(12): 1465-1475.

Hagg T (2005). Molecular regulation of adult CNS neurogenesis: an integrated view. Trends Neurosci 28(11): 589-595.

Harvey BH, Naciti C, Brand L, Stein DJ (2003). Endocrine, cognitive and hippocampal/cortical 5HT 1A/2A receptor changes evoked by a time-dependent sensitisation (TDS) stress model in rats. Brain Res 983(1-2): 97-107.

Hastings NB, Gould E (1999). Rapid extension of axons into the CA3 region by adult-generated granule cells. J Comp Neurol 413(1): 146-154.

Page 30: Role Of Hippocampal Neurogenesis In The Etiology And

22

Herman JP, Schafer MK, Young EA, Thompson R, Douglass J, Akil H, Watson SJ (1989). Evidence for hippocampal regulation of neuroendocrine neurons of the hypothalamo-pituitary-adrenocortical axis. J Neurosci 9(9): 3072-3082.

Holick KA, Lee DC, Hen R, Dulawa SC (2008). Behavioral effects of chronic fluoxetine in BALB/cJ mice do not require adult hippocampal neurogenesis or the serotonin 1A receptor. Neuropsychopharmacology 33(2): 406-417.

Hsieh J, Eisch AJ (2010). Epigenetics, hippocampal neurogenesis, and neuropsychiatric disorders: unraveling the genome to understand the mind. Neurobiol Dis 39(1): 73-84.

Hyman SE, Nestler EJ (1996). Initiation and adaptation: a paradigm for understanding psychotropic drug action. Am J Psychiatry 153(2): 151-162.

Jacobson L, Sapolsky R (1991). The role of the hippocampus in feedback regulation of the hypothalamic-pituitary-adrenocortical axis. Endocr Rev 12(2): 118-134.

Jaholkowski P, Kiryk A, Jedynak P, Ben Abdallah NM, Knapska E, Kowalczyk A, Piechal A, Blecharz-Klin K, Figiel I, Lioudyno V, Widy-Tyszkiewicz E, Wilczynski GM, Lipp HP, Kaczmarek L, Filipkowski RK (2009). New hippocampal neurons are not obligatory for memory formation; cyclin D2 knockout mice with no adult brain neurogenesis show learning. Learn Mem 16(7): 439-451.

Jayatissa MN, Bisgaard C, Tingstrom A, Papp M, Wiborg O (2006). Hippocampal cytogenesis correlates to escitalopram-mediated recovery in a chronic mild stress rat model of depression. Neuropsychopharmacology 31(11): 2395-2404.

Kaplan MS, Hinds JW (1977). Neurogenesis in the adult rat: electron microscopic analysis of light radioautographs. Science 197(4308): 1092-1094.

Kempermann G, Kuhn HG, Gage FH (1997). More hippocampal neurons in adult mice living in an enriched environment. Nature 386(6624): 493-495.

Kent JM, Rauch SL (2003). Neurocircuitry of anxiety disorders. Curr Psychiatry Rep 5(4): 266-273.

Kessler RC (1997). The effects of stressful life events on depression. Annu Rev Psychol 48: 191-214.

Kessler RC, Berglund P, Demler O, Jin R, Koretz D, Merikangas KR, Rush AJ, Walters EE, Wang PS (2003). The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R). JAMA 289(23): 3095-3105.

Kessler RC, Chiu WT, Demler O, Merikangas KR, Walters EE (2005). Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry 62(6): 617-627.

Page 31: Role Of Hippocampal Neurogenesis In The Etiology And

23

Kowalczyk A, Filipkowski RK, Rylski M, Wilczynski GM, Konopacki FA, Jaworski J, Ciemerych MA, Sicinski P, Kaczmarek L (2004). The critical role of cyclin D2 in adult neurogenesis. J Cell Biol 167(2): 209-213.

Kuhn HG, Dickinson-Anson H, Gage FH (1996). Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J Neurosci 16(6): 2027-2033.

Kunze A, Congreso MR, Hartmann C, Wallraff-Beck A, Huttmann K, Bedner P, Requardt R, Seifert G, Redecker C, Willecke K, Hofmann A, Pfeifer A, Theis M, Steinhauser C (2009). Connexin expression by radial glia-like cells is required for neurogenesis in the adult dentate gyrus. Proc Natl Acad Sci U S A 106(27): 11336-11341.

Lagace DC, Benavides DR, Kansy JW, Mapelli M, Greengard P, Bibb JA, Eisch AJ (2008). Cdk5 is essential for adult hippocampal neurogenesis. Proc Natl Acad Sci U S A 105(47): 18567-18571.

Llorens-Martin MV, Rueda N, Martinez-Cue C, Torres-Aleman I, Florez J, Trejo JL (2007). Both increases in immature dentate neuron number and decreases of immobility time in the forced swim test occurred in parallel after environmental enrichment of mice. Neuroscience 147(3): 631-638.

Lucassen PJ, Meerlo P, Naylor AS, van Dam AM, Dayer AG, Fuchs E, Oomen CA, Czeh B (2010). Regulation of adult neurogenesis by stress, sleep disruption, exercise and inflammation: Implications for depression and antidepressant action. Eur Neuropsychopharmacol 20(1): 1-17.

Lucki I (1997). The forced swimming test as a model for core and component behavioral effects of antidepressant drugs. Behav Pharmacol 8(6-7): 523-532.

Luo C, Xu H, Li XM (2005). Quetiapine reverses the suppression of hippocampal neurogenesis caused by repeated restraint stress. Brain Res 1063(1): 32-39.

Magarinos AM, McEwen BS, Flugge G, Fuchs E (1996). Chronic psychosocial stress causes apical dendritic atrophy of hippocampal CA3 pyramidal neurons in subordinate tree shrews. J Neurosci 16(10): 3534-3540.

Malberg JE, Duman RS (2003). Cell proliferation in adult hippocampus is decreased by inescapable stress: reversal by fluoxetine treatment. Neuropsychopharmacology 28(9): 1562-1571.

Malberg JE, Eisch AJ, Nestler EJ, Duman RS (2000). Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. J Neurosci 20(24): 9104-9110.

Manev H, Uz T, Smalheiser NR, Manev R (2001). Antidepressants alter cell proliferation in the adult brain in vivo and in neural cultures in vitro. Eur J Pharmacol 411(1-2): 67-70.

Page 32: Role Of Hippocampal Neurogenesis In The Etiology And

24

Maren S (2008). Pavlovian fear conditioning as a behavioral assay for hippocampus and amygdala function: cautions and caveats. Eur J Neurosci 28(8): 1661-1666.

McGinn MJ, Sun D, Colello RJ (2008). Utilizing X-irradiation to selectively eliminate neural stem/progenitor cells from neurogenic regions of the mammalian brain. J Neurosci Methods 170(1): 9-15.

Meltzer LA, Yabaluri R, Deisseroth K (2005). A role for circuit homeostasis in adult neurogenesis. Trends Neurosci 28(12): 653-660.

Merali Z, Levac C, Anisman H (2003). Validation of a simple, ethologically relevant paradigm for assessing anxiety in mice. Biol Psychiatry 54(5): 552-565.

Meshi D, Drew MR, Saxe M, Ansorge MS, David D, Santarelli L, Malapani C, Moore H, Hen R (2006). Hippocampal neurogenesis is not required for behavioral effects of environmental enrichment. Nat Neurosci 9(6): 729-731.

Millan MJ (2003). The neurobiology and control of anxious states. Prog Neurobiol 70(2): 83-244.

Ming GL, Song H (2005). Adult neurogenesis in the mammalian central nervous system. Annu Rev Neurosci 28: 223-250.

Mizumatsu S, Monje ML, Morhardt DR, Rola R, Palmer TD, Fike JR (2003). Extreme sensitivity of adult neurogenesis to low doses of X-irradiation. Cancer Res 63(14): 4021-4027.

Monje ML, Mizumatsu S, Fike JR, Palmer TD (2002). Irradiation induces neural precursor-cell dysfunction. Nat Med 8(9): 955-962.

Moreau JL, Jenck F, Martin JR, Mortas P, Haefely W (1993). Effects of moclobemide, a new generation reversible Mao-A inhibitor, in a novel animal model of depression. Pharmacopsychiatry 26(1): 30-33.

Murray F, Smith DW, Hutson PH (2008). Chronic low dose corticosterone exposure decreased hippocampal cell proliferation, volume and induced anxiety and depression like behaviours in mice. Eur J Pharmacol 583(1): 115-127.

Nakagawa S, Kim JE, Lee R, Malberg JE, Chen J, Steffen C, Zhang YJ, Nestler EJ, Duman RS (2002). Regulation of neurogenesis in adult mouse hippocampus by cAMP and the cAMP response element-binding protein. J Neurosci 22(9): 3673-3682.

Nestler EJ, Gould E, Manji H, Buncan M, Duman RS, Greshenfeld HK, Hen R, Koester S, Lederhendler I, Meaney M, Robbins T, Winsky L, Zalcman S (2002). Preclinical models: status of basic research in depression. Biol Psychiatry 52(6): 503-528.

Nicolas LB, Kolb Y, Prinssen EP (2006). A combined marble burying-locomotor activity test in mice: a practical screening test with sensitivity to different classes of anxiolytics and antidepressants. Eur J Pharmacol 547(1-3): 106-115.

Page 33: Role Of Hippocampal Neurogenesis In The Etiology And

25

Overmier JB, Seligman ME (1967). Effects of inescapable shock upon subsequent escape and avoidance responding. J Comp Physiol Psychol 63(1): 28-33.

Papp M, Willner P, Muscat R (1991). An animal model of anhedonia: attenuation of sucrose consumption and place preference conditioning by chronic unpredictable mild stress. Psychopharmacology (Berl) 104(2): 255-259.

Parihar VK, Hattiangady B, Kuruba R, Shuai B, Shetty AK (2011). Predictable chronic mild stress improves mood, hippocampal neurogenesis and memory. Mol Psychiatry 16(2): 171-183.

Paulsen RD, Cimprich KA (2007). The ATR pathway: fine-tuning the fork. DNA Repair (Amst) 6(7): 953-966.

Pechnick RN, Zonis S, Wawrowsky K, Pourmorady J, Chesnokova V (2008). p21Cip1 restricts neuronal proliferation in the subgranular zone of the dentate gyrus of the hippocampus. Proc Natl Acad Sci U S A 105(4): 1358-1363.

Porsolt RD (2000). Animal models of depression: utility for transgenic research. Rev Neurosci 11(1): 53-58.

Procaccini C, Aitta-aho T, Jaako-Movits K, Zharkovsky A, Panhelainen A, Sprengel R, Linden AM, Korpi ER (2011). Excessive novelty-induced c-Fos expression and altered neurogenesis in the hippocampus of GluA1 knockout mice. Eur J Neurosci 33(1): 161-174.

Reif A, Schmitt A, Fritzen S, Chourbaji S, Bartsch C, Urani A, Wycislo M, Mossner R, Sommer C, Gass P, Lesch KP (2004). Differential effect of endothelial nitric oxide synthase (NOS-III) on the regulation of adult neurogenesis and behaviour. Eur J Neurosci 20(4): 885-895.

Revest JM, Dupret D, Koehl M, Funk-Reiter C, Grosjean N, Piazza PV, Abrous DN (2009). Adult hippocampal neurogenesis is involved in anxiety-related behaviors. Mol Psychiatry 14(10): 959-967.

Ruzankina Y, Pinzon-Guzman C, Asare A, Ong T, Pontano L, Cotsarelis G, Zediak VP, Velez M, Bhandoola A, Brown EJ (2007). Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1(1): 113-126.

Rygula R, Abumaria N, Flugge G, Fuchs E, Ruther E, Havemann-Reinecke U (2005). Anhedonia and motivational deficits in rats: impact of chronic social stress. Behav Brain Res 162(1): 127-134.

Sahay A, Scobie KN, Hill AS, O'Carroll CM, Kheirbek MA, Burghardt NS, Fenton AA, Dranovsky A, Hen R (2011). Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature 472(7344): 466-470.

Page 34: Role Of Hippocampal Neurogenesis In The Etiology And

26

Salam JN, Fox JH, Detroy EM, Guignon MH, Wohl DF, Falls WA (2009). Voluntary exercise in C57 mice is anxiolytic across several measures of anxiety. Behav Brain Res 197(1): 31-40.

Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen R (2003). Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 301(5634): 805-809.

Saxe MD, Battaglia F, Wang JW, Malleret G, David DJ, Monckton JE, Garcia AD, Sofroniew MV, Kandel ER, Santarelli L, Hen R, Drew MR (2006). Ablation of hippocampal neurogenesis impairs contextual fear conditioning and synaptic plasticity in the dentate gyrus. Proc Natl Acad Sci U S A 103(46): 17501-17506.

Saxe MD, Malleret G, Vronskaya S, Mendez I, Garcia AD, Sofroniew MV, Kandel ER, Hen R (2007). Paradoxical influence of hippocampal neurogenesis on working memory. Proc Natl Acad Sci U S A 104(11): 4642-4646.

Schloesser RJ, Lehmann M, Martinowich K, Manji HK, Herkenham M (2010). Environmental enrichment requires adult neurogenesis to facilitate the recovery from psychosocial stress. Mol Psychiatry 15(12): 1152-1163.

Schmidt-Hieber C, Jonas P, Bischofberger J (2004). Enhanced synaptic plasticity in newly generated granule cells of the adult hippocampus. Nature 429(6988): 184-187.

Sheline YI, Sanghavi M, Mintun MA, Gado MH (1999). Depression duration but not age predicts hippocampal volume loss in medically healthy women with recurrent major depression. J Neurosci 19(12): 5034-5043.

Shepherd JK, Grewal SS, Fletcher A, Bill DJ, Dourish CT (1994). Behavioural and pharmacological characterisation of the elevated "zero-maze" as an animal model of anxiety. Psychopharmacology (Berl) 116(1): 56-64.

Shors TJ, Seib TB, Levine S, Thompson RF (1989). Inescapable versus escapable shock modulates long-term potentiation in the rat hippocampus. Science 244(4901): 224-226.

Shors TJ, Townsend DA, Zhao M, Kozorovitskiy Y, Gould E (2002). Neurogenesis may relate to some but not all types of hippocampal-dependent learning. Hippocampus 12(5): 578-584.

Singer BH, Jutkiewicz EM, Fuller CL, Lichtenwalner RJ, Zhang H, Velander AJ, Li X, Gnegy ME, Burant CF, Parent JM (2009). Conditional ablation and recovery of forebrain neurogenesis in the mouse. J Comp Neurol 514(6): 567-582.

Sramek JJ, Zarotsky V, Cutler NR (2002). Generalised anxiety disorder: treatment options. Drugs 62(11): 1635-1648.

Page 35: Role Of Hippocampal Neurogenesis In The Etiology And

27

Surget A, Saxe M, Leman S, Ibarguen-Vargas Y, Chalon S, Griebel G, Hen R, Belzung C (2008). Drug-dependent requirement of hippocampal neurogenesis in a model of depression and of antidepressant reversal. Biol Psychiatry 64(4): 293-301.

Tada E, Parent JM, Lowenstein DH, Fike JR (2000). X-irradiation causes a prolonged reduction in cell proliferation in the dentate gyrus of adult rats. Neuroscience 99(1): 33-41.

Uchida S, Hara K, Kobayashi A, Fujimoto M, Otsuki K, Yamagata H, Hobara T, Abe N, Higuchi F, Shibata T, Hasegawa S, Kida S, Nakai A, Watanabe Y (2011). Impaired hippocampal spinogenesis and neurogenesis and altered affective behavior in mice lacking heat shock factor 1. Proc Natl Acad Sci U S A 108(4): 1681-1686.

Uno H, Tarara R, Else JG, Suleman MA, Sapolsky RM (1989). Hippocampal damage associated with prolonged and fatal stress in primates. J Neurosci 9(5): 1705-1711.

van Haarst AD, Oitzl MS, de Kloet ER (1997). Facilitation of feedback inhibition through blockade of glucocorticoid receptors in the hippocampus. Neurochem Res 22(11): 1323-1328.

van Praag H, Kempermann G, Gage FH (1999). Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci 2(3): 266-270.

van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD, Gage FH (2002). Functional neurogenesis in the adult hippocampus. Nature 415(6875): 1030-1034.

Vollmayr B, Simonis C, Weber S, Gass P, Henn F (2003). Reduced cell proliferation in the dentate gyrus is not correlated with the development of learned helplessness. Biol Psychiatry 54(10): 1035-1040.

Wang S, Scott BW, Wojtowicz JM (2000). Heterogenous properties of dentate granule neurons in the adult rat. J Neurobiol 42(2): 248-257.

Warner-Schmidt JL, Duman RS (2006). Hippocampal neurogenesis: opposing effects of stress and antidepressant treatment. Hippocampus 16(3): 239-249.

Willner P (2005). Chronic mild stress (CMS) revisited: consistency and behavioural-neurobiological concordance in the effects of CMS. Neuropsychobiology 52(2): 90-110.

Willner P, Towell A, Sampson D, Sophokleous S, Muscat R (1987). Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology (Berl) 93(3): 358-364.

Yap JJ, Takase LF, Kochman LJ, Fornal CA, Miczek KA, Jacobs BL (2006). Repeated brief social defeat episodes in mice: effects on cell proliferation in the dentate gyrus. Behav Brain Res 172(2): 344-350.

Page 36: Role Of Hippocampal Neurogenesis In The Etiology And

28

Zencak D, Lingbeek M, Kostic C, Tekaya M, Tanger E, Hornfeld D, Jaquet M, Munier FL, Schorderet DF, van Lohuizen M, Arsenijevic Y (2005). Bmi1 loss produces an increase in astroglial cells and a decrease in neural stem cell population and proliferation. J Neurosci 25(24): 5774-5783.

Zhao Y, Ma R, Shen J, Su H, Xing D, Du L (2008). A mouse model of depression induced by repeated corticosterone injections. Eur J Pharmacol 581(1-2): 113-120.

Page 37: Role Of Hippocampal Neurogenesis In The Etiology And

29

CHAPTER II

Selective Deletion of a Cell Cycle Checkpoint Kinase (ATR) Reduces Neurogenesis and Alters Responses in Rodent Models of Behavioral Affect

This chapter has been published:

Onksen JL, Brown EJ, Blendy JA (2011). Selective deletion of a cell cycle checkpoint kinase (ATR) reduces neurogenesis and alters responses in rodent models of behavioral affect. Neuropsychopharmacology 36(5): 960-969.

Page 38: Role Of Hippocampal Neurogenesis In The Etiology And

30

Abstract

Hippocampal function has been implicated in mood and anxiety disorders, as well

as in the response to antidepressant (AD) treatment. However, the significance of new

neurons in the therapeutic mechanism of ADs remains unclear. In this study the

proliferation of new neurons was inhibited through conditional deletion of ataxia

telangiectasia-mutated and rad-3 related (ATR), a cell cycle checkpoint kinase, and

cellular and behavioral outcomes following AD exposure were evaluated. ATR was

conditionally deleted by microinjecting a Cre recombinase-expressing virus into the

hippocampus of floxed-ATR mice. Behavioral assessment in multiple rodent models of

affective state revealed anxiolytic-like behavior in the elevated zero maze, marble

burying test, and novelty-induced hypophagia (NIH) test. The efficacy of chronic

desipramine (DMI) treatment was evaluated in the NIH test, as this paradigm is thought

to be sensitive to increases in neurogenesis by chronic AD exposure. Chronic exposure

to DMI reduced hyponeophagia in the NIH test in control mice, whereas DMI had no

behavioral effect in ATR-deleted mice. While DMI did not alter cell proliferation in either

group, it did produce a robust increase in dendritic spine density in control mice,

indicative of enhanced neuronal plasticity. This effect of DMI on spine density was

severely attenuated following ATR deletion. These findings demonstrate that reductions

in basal neurogenesis produce an anxiolytic phenotype and reduce AD efficacy in

behaviors requiring chronic exposure. Attenuated capacity for synaptic remodeling may

underlie this reduced AD efficacy. ATR deletion may serve as a valuable model to study

the various proposed roles of newborn neurons in the hippocampus.

Page 39: Role Of Hippocampal Neurogenesis In The Etiology And

31

Introduction

Major depression is a highly prevalent psychiatric disorder characterized by a

diverse manifestation and etiology. Depression and anxiety are highly co-morbid and

pervasive (Merikangas et al., 2003). The ability of chronic antidepressant (AD) treatment

to ameliorate the symptoms of both depression and anxiety (Schoevers et al., 2008)

raises the possibility that they may share common molecular underpinnings.

Hippocampal neurogenesis has been implicated in the therapeutic efficacy of AD

treatment. This theory is supported by evidence that the maturation and functional

integration of new neurons occurs over a period of time similar to that required for ADs

to exert their therapeutic effects (Malberg et al., 2000; Jessberger and Kempermann,

2003). However, the significance of new hippocampal neurons for the onset of

depressive-like symptoms remains unclear (Balu and Lucki, 2009, for review). While

rodent models of chronic stress can reduce neurogenesis (Malberg and Duman, 2003;

Murray et al., 2008; Elizalde et al., 2010) and induce anhedonia (Papp et al., 1991),

reduced neurogenesis alone is not sufficient to induce depression and is more likely a

risk factor contributing to the onset of depressive-like symptoms (Sahay and Hen,

2007).

Despite early promising reports, the relevance of new neurons to antidepressant

effects is still unclear. Fluoxetine (FLX) was shown to be ineffective in a behavioral

model of AD efficacy following ablation of hippocampal neurogenesis by x-ray irradiation

(Santarelli et al., 2003). Additionally, in a comparison of the inbred mouse strains

MRL/MjP and C57BL/6, Balu and colleagues found behavioral efficacy following FLX and

desipramine (DMI) treatment only in the strain that exhibited a positive neurogenic

response to these drugs (Balu et al., 2009). In contrast, there is evidence that AD

Page 40: Role Of Hippocampal Neurogenesis In The Etiology And

32

treatment elicits behavioral changes independent of hippocampal neurogenesis (David et

al., 2007; Holick et al., 2008; Huang et al., 2008). Furthermore, numerous studies

implicate enhanced neuroplasticity and synaptic function in the therapeutic efficacy of

ADs (Bessa et al., 2009; Calabrese et al., 2009, for review; O'Leary et al., 2009;

Marchetti et al., 2010)

Although depression and anxiety present with high co-morbidity, little work has

been done to examine neurogenesis in the context of anxiety. Some studies find no link

(Saxe et al., 2006), while others observe an anxiogenic response associated with

reduced neurogenesis (Bergami et al., 2009; Revest et al., 2009). Voluntary exercise

leads to a robust increase in neurogenesis while also increasing anxiety-like behavior

(Fuss et al., 2010). However, it must also be noted that voluntary exercise can reduce

anxiety (Duman et al., 2008; Salam et al., 2009), suggesting a variety of factors in

addition to neurogenesis likely interact to modulate anxiety.

To address the relevance of neurogenesis to psychiatric illness, we have utilized

a genetic approach in which hippocampal neurogenesis is reduced following deletion of

ATR. ATR is necessary for genomic stability in the early embryo and cells lacking ATR

ultimately exit the cell cycle or undergo programmed cell death (Brown and Baltimore,

2000; Brown and Baltimore, 2003). Ubiquitous deletion of ATR from the adult mouse

leads to age-related phenotypes that are associated with the premature depletion of

stem/progenitor cells (Ruzankina et al., 2007). Previous research has demonstrated that

ATR can be deleted from differentiated forebrain CNS neurons by Synapsin1 promoter-

driven Cre expression with no effects on circadian period, coordination, strength,

anxiety, or hippocampal-dependent learning (Ruzankina et al., 2007). This suggests that

ATR is not necessary for the function of differentiated neurons and implicates loss of

Page 41: Role Of Hippocampal Neurogenesis In The Etiology And

33

ATR from the proliferating cell population in the behavioral consequences that we

observe following hippocampal-specific deletion. Targeting a protein that is not

necessary for the function of mature cells provides the added benefit of minimizing

possible compensatory or non-specific effects that could arise if the deleted protein were

to play a role outside of cell division.

Here, we show that deletion of ATR from the adult hippocampus leads to at least

a 50% reduction in neurogenesis. Behavioral consequences of partially inhibiting

neurogenesis have been observed previously (Revest et al., 2009; Conboy et al., 2010).

We thus evaluated behavior in rodent models of affective state and examined the

cellular and behavioral effects of AD treatment following ATR deletion. While not a

complete ablation, this model has the potential to mimic the reduced rate of

neurogenesis that is observed during aging (Bondolfi et al., 2004). Aged mice have a

reduced capacity for AD-induced neurogenesis (Couillard-Despres et al., 2009) and

treatment-resistant depression is prevalent in the aged population (Lenze et al., 2008).

Materials and Methods

Animals Mice homozygous for the Cre/lox-conditional allele of ATR (referred to as

floxed-ATR or ATRf/f) and mice expressing the tamoxifen-inducible Cre transgene

(CreERT2) on a 129SV/C57BL/6 background were generated as previously described

(Ruzankina et al., 2007). Floxed-ATR mice were used for hippocampal-specific ATR

deletion. For whole-body deletion of ATR, floxed-ATR mice carrying CreERT2 were

treated with tamoxifen as described (Ruzankina et al., 2007). For behavioral studies in

these mice, one of two control groups was utilized: (1) ATRf/f;CreERT2 + vehicle, or (2)

Page 42: Role Of Hippocampal Neurogenesis In The Etiology And

34

ATRf/f + tamoxifen. Mice (1.5-5 months old, 20-35 gm) were housed in groups of 3-4

following stereotaxic surgery and during experimental paradigms, with exceptions noted.

All behavioral experiments were conducted on males and females at least 5 weeks

following stereotaxic surgery or tamoxifen treatment. Animals were maintained on a 12

hr light/dark cycle with food and water available ad libitum in accordance with the

University of Pennsylvania Institutional Animal Care and Use Committee. In some

instances multiple behavioral tests were performed in a single cohort of mice, as

indicated here: (1) Elevated zero maze was performed 3 days following marble burying;

(2) Training in the NIH commenced 3 days following forced swim test; (3) Locomotor

activity was measured 2 days prior to the Morris water maze and restraint stress was

measured 1 week following water maze testing.

Adeno-associated virus Adeno-associated viruses (AAV) expressing Cre recombinase

(AAV2/9.CMV.PI.CRE, titer 2.84*1013 gc/ml) and eGFP (AAV2/9.CMV.eGFP, titer

3.74*1013 gc/ml) were generated by the University of Pennsylvania Vector Core. The

expression cassette consists of the AAV2 terminal repeats flanking the CMV promoter-PI-

Cre recombinase/eGFP sequences packaged into AAV9. The vectors were purified using

CsCl sedimentation method. Quantification of vector genome copies (gc) was performed

by Q-PCR. AAVs were diluted in sterile PBS for microinjections.

Hippocampal Injections Floxed-ATR mice (6-8 weeks) were anesthetized with

isoflurane and secured in a stereotaxic frame (Kopf, Tujuna, CA). Holes were drilled

bilaterally in the skull at sites of injection. Stereotaxic coordinates for the hippocampus

are (from Bregma) anterior-posterior -2.1, lateral +/- 1.4, dorso-ventral -2.0, and

Page 43: Role Of Hippocampal Neurogenesis In The Etiology And

35

anterior-posterior -2.9, lateral +/- 3.0, dorso-ventral -3.8. 0.5 ul of 1*1010 gc/ul AAV was

injected at each site through a 33 gauge needle on a 5 ul Hamilton syringe using a

KDS310 Nano Pump (KD Scientific, Holliston, MA) mounted to the stereotaxic frame, at a

rate of 0.1 ul/min. The needle remained in place for 4 additional minutes at each

injection site. The skin was sutured and the animal recovered on a heating pad before

returning to the home cage.

Drugs Desipramine (DMI, Sigma, St. Louis, MO), chlordiazepoxide (CDP, Sigma) and

bromodeoxyuridine (BrdU, Sigma) were dissolved in 0.9% saline. Solutions were

prepared immediately before use and injected intraperitoneally using a volume of 10

ml/kg. For cell proliferation analysis, 200 mg/kg BrdU was administered 24 hours prior

to perfusion. For cell survival analysis, 100 mg/kg BrdU was administered once per day

for 4 days, 4 weeks prior to perfusion. To activate the inducible CreERT2, tamoxifen

(Sigma) was dissolved at a concentration of 20 mg/ml in a mixture of 98% corn oil / 2%

ethanol and was injected intraperitoneally once per day for 4 days.

Quantitative-PCR Cre/Lox recombination was quantified using RT-PCR. Brains were

rapidly removed and hippocampi were dissected out and flash-frozen in liquid nitrogen.

DNA was extracted using Qiagen DNeasy Blood and Tissue Kit (Qiagen, Valencia, CA).

Deletion rate was analyzed by utilizing a Taqman primer/probe set which overlap the 3’

lox sequence and a portion of the excised domain as described (Ruzankina et al., 2009).

Floxed-ATR levels were normalized to the housekeeping gene TBP.

Page 44: Role Of Hippocampal Neurogenesis In The Etiology And

36

Immunohistochemistry Mice were anesthetized with nembutol (10 mg/kg) and

transcardially perfused with cold 0.1 M phosphate-buffered saline (PBS) for 5 minutes,

followed by cold 4% paraformaldehyde (PFA) in PBS for 15 minutes. Brains were

postfixed overnight in PFA at 4◦C and subsequently stored at 4◦C in 30% sucrose. Brains

were frozen on dry ice, sectioned coronally at a thickness of 40 µm, and transferred to

PBS + 0.5% Sodium Azide at 4◦C prior to processing for immunohistochemistry.

For BrdU analysis, sections were wet-mounted on Superfrost/Plus slides (Fisher

Scientific, Pittsburgh, PA) for stereological analysis using a modified version of the

optical fractionator method (West et al., 1991). Mounted sections were incubated in 0.1

M citric acid, pH 6.0, for antigen retrieval. Subsequently, slides were incubated in

trypsin, 2N HCl, then primary antibody (mouse anti-BrdU, 1:200, Becton Dickinson) with

0.5% Tween 20 overnight. On the second day, slides were incubated for 1 hour in

secondary antibody (biotinylated anti-mouse IgG, Vector Laboratories), 1 hour in avidin-

biotin-HRP (1:100, Vector Laboratories), and labeled cells were visualized using

diaminobenzidine (Sigma). PBS-washed slides were then counterstained and cover-

slipped with Permount. BrdU-IR cells in every 9th section of the hippocampus were

counted with a 100x oil immersion lens, omitting cells in the upper-most focal plane.

Cells within clusters were counted by distinguishing nuclear borders while focusing down

through the tissue using an objective with a narrow depth of focus. For proliferation

counts, a cell is included in the subgranular zone (SGZ) of the dentate gyrus if it is

touching, or within two cells of the granule cell layer (GCL).

For doublecortin (DCX) analysis, free-floating sections of a 1/9 series through the

hippocampus were blocked for 1 hour with 3% Normal Horse Serum, 0.5% Tween 20

and 0.2% Triton in PBS (blocking solution). Sections were then incubated for 72 hours

Page 45: Role Of Hippocampal Neurogenesis In The Etiology And

37

at 4◦C in primary antibody (Goat anti-DCX, Santa Cruz #8066) diluted 1:500 in blocking

solution. Sections were washed and incubated for 1 hour in secondary antibody (Horse

anti-goat, Vector Laboratories) diluted 1:200 in blocking solution. Sections were then

washed and treated with 0.75% H2O2 for 20 minutes prior to incubation in avidin-biotin-

HRP (1:200, Vector Laboratories). Labeled cells were visualized using nickel-enhanced

diaminobenzidine. Free-floating sections were mounted onto slides and dried overnight

before dehydrating and cover-slipping with Permount. DCX-IR cells in every 9th section

were counted with a 100x oil immersion lens, omitting cells in the upper-most focal

plane. All cells located within the GCL and SGZ were counted.

Golgi Impregnation Golgi Impregnation was conducted to label neurons and their

processes for analysis of dendritic spine density according to the FD Rapid GolgiStain Kit

(FD NeuroTechnologies, Ellicott City, MD). 100 µm sections were cleared with xylenes

and cover-slipped with Permount mounting medium. Hippocampal CA1 neurons selected

for analysis had the following characteristics: Dark and complete impregnation,

untruncated tertiary dendrites, and isolated cell body with a clear connection to

dendrites that are isolated from neighboring cells to avoid interference. For each brain,

five CA1 neurons were selected for analysis. Within each neuron, six separate 10 µm

segments of tertiary dendrite, located at least 50 µm from the cell body, were selected

for counting. Number of spines per 10 µm was determined manually using a confocal

microscope (Nikon Eclipse E600, Nikon) with a 100x oil immersion lens. For each

treatment group, average spine density of each neuron analyzed was averaged to arrive

at the spine density used for statistical analysis.

Page 46: Role Of Hippocampal Neurogenesis In The Etiology And

38

Elevated Zero Maze The elevated zero maze test was performed as described

previously (Gundersen and Blendy, 2009). The maze (Stoelting, Wood Dale, IL) consists

of two open areas (wall height, 0.5”) and two closed areas (wall height, 12”), and is

elevated 24” from the ground. Lighting in the maze is 15 lux. At the start of testing,

mice were placed into one of the closed areas and were allowed to explore the maze for

300 seconds. The Viewpoint Tracking System (Viewpoint, Champagne au Mont d’or,

France) was used to track time spent in the open areas, number of entries into the open

areas, and distance traveled in each area.

Marble Burying After a 1-hour acclimation period in the testing room, mice were

placed in a standard mouse cage containing 5 cm of bedding and 20 marbles, spaced

equally along the edge of the cage. A wire-mesh lid was placed on the cage. Number of

marbles buried at the end of a 15-minute session was quantified. A marble was

considered buried if it was at least 2/3 beneath the surface of the bedding.

Novelty-Induced Hypophagia (NIH) Test The NIH test was performed as

previously described (Gur et al., 2007). Briefly, mice were pair-housed and trained to

consume peanut butter chips (Nestle) in their home cage. Drug treatment occurred

between the training and testing periods. For chronic AD treatment, mice received 25

days of desipramine (12.5 mg/kg DMI, b.i.d.). This was followed by measuring latency

to consume the palatable food on two consecutive test days. The first test day took

place in the home cage. On the second test day, latency was measured in a novel

environment, which consisted of an empty cage with no bedding, placed in a white box

with a bright light (2150 lux) and a novel smell (dilute pine sol). On home and novel test

Page 47: Role Of Hippocampal Neurogenesis In The Etiology And

39

days, mice received their first daily injection of DMI 1 hour prior to testing latency to

feed, and received their second injection of DMI in the evening, consistent with the

chronic treatment paradigm. For acute CDP treatment, home and novel test days occur

immediately following the training period. CDP (10 mg/kg) is injected 30 minutes prior

to the novel test.

Forced Swim Test Mice were placed in water (15 cm depth, 23 C) in plastic cylinders

(23 cm tall x 14 cm diameter) for 6 minutes. Using Viewpoint automated scoring

(Viewpoint, Champagne au Mont d’Or, France), duration of immobility was measured.

Mice were exposed to the swim tanks twice (pre-swim and swim test), with 24 hours

between exposures. Three injections of DMI were given during the inter-swim interval:

One 24 hours prior to the swim test (10 mg/kg), a second injection 5 hours prior to the

swim test (10 mg/kg), and a third injection 1 hour prior to the swim test (20 mg/kg).

Locomotor Activity Locomotor activity was measured by beam-breaks in a

photobeam frame (Med Associates, St. Albans, VT, USA). During the test, mice were

placed individually into a clean home cage resting within the photobeam frame, and

data were recorded by Med Associates Software. Ambulations and crossings were

measured in 5-min bins for 60 minutes.

Restraint Stress Mice were placed in 50 mL Falcon conical vials for 15 minutes. Holes

were drilled at both ends to allow for breathing and to give access to the tail. Tail blood

(20 uL) was collected at 0, 15, 30, and 90 minutes. Blood was transferred directly into

tubes with chilled EDTA, centrifuged at 5,000 RPM for 10 minutes at 4 C, and serum was

Page 48: Role Of Hippocampal Neurogenesis In The Etiology And

40

used to measure corticosterone levels with a 125I Corticosterone RIA kit (MP Biomedicals,

Orangeburg, NY).

Fear Conditioning Fear conditioning was performed in a rectangular chamber capable

of delivering footshocks (Med Associates, St. Albans, VT). During the training session,

mice were exposed to a 20 second, 2000 Hz tone (conditioned stimulus, CS) at 2 and

3.5 minutes. During the final 2 seconds of each CS, a 0.5 mA scrambled footshock

(unconditioned stimulus, US) was delivered. The training session lasted for a total of 6

minutes, at which time the mice were returned to their home cages. To evaluate

contextual fear learning, 24 hours after training the mice were placed in the chamber for

6 minutes in the absence of the CS or US and their freezing behavior was assessed. To

evaluate cued fear conditioning, 24 hours after the contextual fear test the mice were

placed in an altered chamber and exposed to the CS at 2 minutes. They were removed

from the chamber after a total of 5 minutes and freezing behavior was assessed before

and after the presentation of the CS. To alter the context of the chamber for the cued

fear test, an insert was placed in the chamber to modify its shape and color.

Additionally, a novel scent and increased lighting were present.

Morris Water Maze (MWM) MWM Apparatus: The water maze apparatus consisted

of a white circular swimming pool 150 cm in diameter, located in the center of a square-

shaped room with unique spatial cues on each wall. The water was made opaque by the

addition of a non-toxic white tempera paint and maintained at 23 C. The escape

platform consisted of a translucent square plastic platform 12 cm in length. The water

level was maintained at depth such that the surface of the hidden platform was 1 cm

Page 49: Role Of Hippocampal Neurogenesis In The Etiology And

41

below the water surface. A video camera fixed to the ceiling above the pool recorded

behavior. For swim path analysis the videos were replayed on a computer equipped with

Viewpoint video tracking system. The tracking system allowed for automated

determination of latency to the escape platform, time spent in each quadrant, and total

path length (distance traveled).

MWM Training: Mice were trained during the light cycle with three trials per day and a

10-minute inter-trial interval. Mice that failed to locate the hidden platform after 60

seconds were guided to the platform. Upon reaching the platform, mice remained in

place for 10 seconds prior to being returned to the home cage. For hidden platform

acquisition training the start location varied for each trial and was counterbalanced

across the treatment groups and days. For reversal training the hidden platform was

moved to a novel quadrant and the start location was held constant for each mouse. A

probe test was administered 24 hours following the 9th day of hidden platform

acquisition training. For this probe test the platform was removed and mice were placed

in the maze for 60 seconds and required to locate the platform using spatial extra-maze

cues. Time spent in each quadrant was analyzed. 24 hours after the first probe test,

reversal training commenced. Following 6 days of reversal training, a second probe-test

was administered. In this 60 second reversal training probe test, time to reach the

platform from a novel start location was measured. Analysis of the hidden platform

training and reversal training consisted of averaging the three daily trials and comparing

AAV treatments by RMANOVA.

Statistical Analysis Statistical analyses were performed with GraphPad Prism 5. Data

are reported as mean + SEM. Studies comparing AAV injection were analyzed with

Page 50: Role Of Hippocampal Neurogenesis In The Etiology And

42

Student’s t tests. Studies comparing AAV and environment or treatment were analyzed

by two-way ANOVA followed by pair-wise comparisons with Bonferroni post hoc tests.

Results

Hippocampal ATR deletion reduces neurogenesis

To delete ATR we micro-injected AAV.Cre into both the dorsal and ventral

hippocampus. AAV.Cre and the control virus, AAV.eGFP, are packaged into viral capsids

of the serotype 9. AAV9 was chosen for its ability to spread throughout the

hippocampus, thereby covering the entire rostral-caudal extent of the dentate gyrus

without spreading outside of the hippocampal formation. Maximal deletion of ATR (-

78%, t(16)=4.75, p<0.001) was evident 8 weeks following AAV injection (Fig. 1a).

Photomicrographs of eGFP fluorescence at this time point illustrate spread and

expression of the AAV (Fig. 1b,c)

To determine the effect of hippocampal ATR deletion on neurogenesis, floxed-

ATR mice were injected intraperitoneally (IP) with 200 mg/kg BrdU 8 weeks following

hippocampal AAV microinjection and perfused 24 hours later. Compared to controls,

ATR-deleted mice showed significantly reduced (-45%, t(17)=5.56, p<0.0001) BrdU

incorporation, indicating a robust reduction in basal levels of cell proliferation (Fig.

2a,d,e). To determine effect of ATR deletion on cell survival, BrdU was injected 4 weeks

following AAV microinjection and mice were perfused 4 weeks later. Compared to

controls, ATR-deleted mice showed significantly reduced (-57%, t(22)=5.31, p<0.0001)

BrdU incorporation, indicative of reduced cell survival (Fig. 2b) Additionally, ATR-deleted

mice showed significantly reduced (-54%, t(17)=3.246, p<0.005) expression of DCX,

indicating a reduction in the total number of newly born, immature neurons present in

Page 51: Role Of Hippocampal Neurogenesis In The Etiology And

43

the hippocampus (Fig. 2c,f,g). Taken together, these data demonstrate that

hippocampal ATR deletion results in at least a 50% reduction in neurogenesis. The

requirement of ATR for normal rates of neurogenesis was verified in a second model of

ATR deletion, ATRf/f;CreERT2 mice treated with tamoxifen. 5 weeks after tamoxifen

treatment we already observed a 30% reduction in BrdU incorporation in these mice

(p=0.06, data not shown).

ATR deletion reduces anxiety-like behavior

After establishing that hippocampal ATR deletion results in reduced basal levels

of neurogenesis, we sought to evaluate behavior of the floxed-ATR mice in rodent

models of affective state. Anxiety-like behavior was evaluated using the EZM (Fig. 3a).

ATR-deleted mice spent significantly greater time (t(10)=3.26, p<0.01) in the open arms

of the maze, indicating reduced levels of anxiety compared to controls. This observation

was not due to any overall difference in distance traveled in the maze (t(10)=0.71

p=0.49) (Fig. 3b). Furthermore, deletion of ATR from the hippocampus did not affect

general locomotor activity or corticosterone response to an acute stressor (Fig. S1).

The anxiolytic phenotype observed in the EZM is at odds with work by others

demonstrating an anxiogenic phenotype following reduced neurogenesis (Bergami et al.,

2009; Revest et al., 2009). To determine if reduced anxiety-like behavior was present in

other tests, we examined behavior in the marble burying test in both the AAV and

tamoxifen-inducible models of ATR deletion. As illustrated in Figure 4a, hippocampal

ATR deletion was associated with a significant reduction (t(28)=3.55, p<0.01) in the

number of marbles buried during a 15 minute session. Consistent with the anxiolytic

effect of hippocampal-specific ATR deletion, reduced anxiety-like behavior was also

Page 52: Role Of Hippocampal Neurogenesis In The Etiology And

44

observed in the marble burying test in ATRf/f;CreERT2 mice treated with tamoxifen

(t(10)=5.805, p<0.05) (Fig. 4b).

ATR deletion resulted in reduced hyponeophagia in the NIH test, indicative of a

blunted anxiety-like response to a novel, anxiety-provoking environment. Mice were

trained to consume peanut butter chips in their home cage and subsequently tested for

latency to consume this palatable food in a novel, anxiety provoking environment.

Following hippocampal-specific deletion, ATR-deleted mice showed significantly shorter

latency to feed in the novel environment compared to controls (p<0.001) (Fig. 4c). Two

way ANOVA revealed significant main effects of environment (F(1,28)=57.87, p<0.0001)

and AAV (F(1,28)=15.23, p<0.001), as well as a significant environment x AAV interaction

(F(1,28)=15.65, p<0.001). Consistent with these findings, reduced anxiety was also

observed in the ATRf/f;CreERT2 mice in the NIH test (Fig. 4d). Two way ANOVA revealed

significant main effects of environment (F(1,45)=32.44, p<0.0001) and genotype

(F(1,45)=10.61, p<0.005), as well as a significant environment x genotype interaction

(F(1,45)=10.08; p<0.005). Post hoc analyses confirmed significantly reduced latency to

feed (p<0.001) in the novel environment in ATR-deleted mice compared to controls (Fig.

4d).

Behavioral effects of DMI are attenuated following ATR deletion

As hippocampal neurogenesis has been implicated in the mechanism of action of

ADs, we next sought to determine whether ATR deletion would alter the cellular and

behavioral response to chronic AD treatment. To address this we again utilized the NIH

test but evaluated mice following chronic DMI treatment (12.5 mg/kg i.p., b.i.d.). This

dose effectively reduces feeding latency in mice on a 129SV/C57BL/6 background

Page 53: Role Of Hippocampal Neurogenesis In The Etiology And

45

following chronic, but not acute treatment (Gur et al., 2007; Turner et al., 2010). All

training occurred prior to drug exposure. Home cage testing occurred on the 24th day of

DMI treatment and novel cage testing occurred on the 25th day. Following the novel

cage test, mice were given 200 mg/kg BrdU and perfused 24 hours later. There were no

effects of AAV or AD (AAV main effect: F(1,28)=2.735, p=0.109; AD main effect:

F(1,28)=0.02, p=0.889; AAV x AD interaction: F(1,28)=0.003, p=0.957) on latency to feed

in the home cage test (Fig. 5a). Consistent with our finding that ATR deletion reduced

hyponeophagia in the absence of drug treatment (Fig. 4c), two way ANOVA revealed a

significant main effect of AAV (F(1,25)=23.40, p<0.0001) in the novel environment (Fig.

5a). Additionally, there was a significant main effect of DMI (F(1,25)=4.394, p<0.05) and

a significant AAV x AD interaction (F(1,25)=4.601, p<0.05) in the novel environment (Fig.

5a). Post hoc analyses revealed a significant effect of DMI only in the AAV.eGFP

treatment group (p<0.05), indicating that DMI does not reduce hyponeophagia in ATR-

deleted mice. This is unlikely due to a floor effect of this test, as acute treatment with

CDP (10 mg/kg) 30 minutes prior to the novel cage test resulted in significantly reduced

latency to feed in the novel environment (Fig. 5b) in both control and ATR-deleted mice

(AAV main effect: F(1,24)=4.863, p=<0.05; CDP main effect: F(1,24)=25.45, p<0.0001).

Additionally, ATR-deleted mice and controls respond equally well to DMI in the forced

swim test (Fig. 5c) by demonstrating reduced immobility compared to saline-treated

mice (DMI main effect: F(1,32)=30.44, p<0.0001). Furthermore, the altered behavior

observed in the NIH is not likely due to learning or memory impairment, as ATR deletion

does not induce deficits in associative (Fig. S2) or spatial learning (Fig. S3).

Cellular effects of chronic DMI are altered in ATR-deleted mice

Page 54: Role Of Hippocampal Neurogenesis In The Etiology And

46

Following behavioral testing in the NIH paradigm, the cellular effects of chronic

DMI treatment were evaluated. Two-way ANOVA revealed a significant main effect of

AAV (F(1,29)=31.73; p<0.0001) to reduce cell proliferation in ATR-deleted mice (Fig. 5d),

as observed previously. DMI had no effect on cell proliferation in control or ATR-deleted

mice (AD main effect: F(1,29)=0.173, p=0.68, AAV x AD interaction: F(1,29)=0.877,

p=0.357). In the absence of a neurogenic effect of DMI treatment, we sought to

identify a potential mechanism through which DMI may exert behavioral changes.

Dendritic spine density, a correlate of synaptic transmission strength and neuronal

plasticity (von Bohlen Und Halbach, 2009), can be increased by AD treatment in rodents

(Norrholm and Ouimet, 2001; Hajszan et al., 2005). Reduced spine density has been

observed in post-mortem tissue of depressed individuals (Law et al., 2004). To this end,

we examined whether DMI exerted differential effects on CA1 dendritic spine density in

ATR-deleted mice. Two-way ANOVA revealed significant main effects of AAV

(F(1,145)=25.2, p<0.0001), AD (F(1,145)=159.7, p<0.0001), and a significant AAV x AD

interaction (F(1,145)=81.41, p<0.0001). Post hoc analyses confirmed a robust significant

effect (+34%, p<0.001) of DMI in control mice (Fig 6a,b). This effect was greatly

attenuated (+5%, p<0.05) in ATR-deleted mice (Fig. 6c).

Page 55: Role Of Hippocampal Neurogenesis In The Etiology And

47

Figure 1. Hippocampal microinjection of AAV.Cre deletes ATR.

Figure 1. Hippocampal microinjection of AAV.Cre deletes ATR. A Floxed-ATR mice show

a significant reduction in the floxed allele 8 weeks following hippocampal microinjection

of AAV.Cre (**, p<0.001, n=7-9 per group). B,C Expression pattern of eGFP in the

dorsal (B) and ventral (C) hippocampus 8 weeks following hippocampal microinjection of

AAV.eGFP. Error bars indicate SEM.

ATR Paper - NPP.pzf: Figure 1 Resub - Saturday, October 16, 2010

AAV.eGFP AAV.Cre0.000

0.005

0.010

0.015

**

AT

R/T

BP

(A

rbitra

ry u

nits

)A B C

Page 56: Role Of Hippocampal Neurogenesis In The Etiology And

48

Figure 2. Hippocampal ATR deletion inhibits hippocampal neurogenesis.

ATR Paper - NPP.pzf: Figure 2 Resub - Saturday, October 16, 2010

Cell Proliferation

AAV.eGFP AAV.Cre

0

300

600

900

1200

***

Brd

U-IR

ce

lls

Immature Neurons

AAV.eGFP AAV.Cre

0

1000

2000

3000

4000

**

DC

X-IR

ce

lls

A

B

C

Cell Survival

AAV.eGFP AAV.Cre

0

300

600

900

1200

***

Brd

U-IR

ce

lls

G

F

E

D

AAV.eGFP

AAV.Cre

AAV.eGFP

AAV.Cre

Page 57: Role Of Hippocampal Neurogenesis In The Etiology And

49

Figure 2. Hippocampal ATR deletion inhibits hippocampal neurogenesis. A,B AAV.Cre-

injected mice exhibited significantly fewer BrdU-labeled cells within the subgranular zone

as compared to AAV.eGFP-injected mice, both 24 hours (A, ***, p<0.0001, n=9-10 per

group) and 4 weeks (B, ***, p<0.0001, n=12 per group) after BrdU injection. C Using

sections from the same mice as in Figure 2a, DCX quantification revealed a significant

decrease in immature neurons in the dentate gyrus of AAV.Cre-injected mice as

compared with AAV.eGFP-injected mice (**, p<0.005, n=9-10 per group). D,E

Representative photomicrographs of immunostaining for BrdU. Arrows point to BrdU-IR

cells. F,G Representative photomicrographs of immunostaining for DCX. Error bars

indicate SEM.

Page 58: Role Of Hippocampal Neurogenesis In The Etiology And

50

Figure 3. Hippocampal ATR deletion is associated with reduced anxiety in the

elevated zero maze (EZM).

Figure 3. Hippocampal ATR deletion is associated with reduced anxiety in the elevated

zero maze (EZM). A AAV.Cre-injected mice spent significantly more time in the open

arms of the maze, as compared to AAV.eGFP-injected mice (*, p < 0.01, n=5-7 per

group). B There was no difference between groups in the total distance traveled in the

EZM. Error bars indicate SEM.

ATR Paper - NPP.pzf: Figure 3 - Saturday, October 16, 2010

AAV.eGFP AAV.Cre0

5000

10000

15000

20000

Dis

tan

ce

(a

rbitra

ry u

nits

)

AAV.eGFP AAV.Cre0

50

100

150

200

*

Tim

e in

Op

en

Arm

s (s

ec

)

A B

Page 59: Role Of Hippocampal Neurogenesis In The Etiology And

51

Figure 4. ATR deletion is associated with reduced anxiety.

Figure 4. ATR deletion is associated with reduced anxiety. A,B ATR-deleted mice buried

significantly fewer marbles during a 15-minute marble burying test. This effect was

observed in both hippocampal specific ATR deletion (A, **, p<0.01, n=14-16 per group)

and in the tamoxifen-inducible ubiquitous ATR deletion (B, **, p<0.001, n=6 per

group). C, D ATR-deleted mice exhibited significantly shorter latencies to feed in the

novel environment during the NIH test. This effect was observed both in hippocampal-

specific ATR deletion (C, **, p<0.001, n=8 per group) and in the tamoxifen-inducible

ubiquitous ATR deletion (D, **, p<0.001, n=11-13 per group). Error bars indicate SEM.

ATR Paper - NPP.pzf: Figure 4 Resub - Saturday, October 16, 2010

Home Novel0

100

200

300

400

500AAV.eGFP

AAV.Cre

**

La

ten

cy

to

fe

ed

(s

ec

)A B

C D

Home Novel0

100

200

300

400

500Vehicle

Tamoxifen

**L

ate

nc

y to

fe

ed

(s

ec

)

ATRf/f ATRf/f;CreERT20

5

10

15

20

**Ma

rble

s B

uri

ed

AAV Tamoxifen

Marb

le B

ury

ing

Hyp

on

eo

ph

ag

ia

AAV.eGFP AAV.Cre0

5

10

15

20M

arb

les

Bu

rie

d

**

Page 60: Role Of Hippocampal Neurogenesis In The Etiology And

52

Figure 5. In the NIH paradigm, ATR-deleted mice exhibit altered

antidepressant response.

ATR Paper - Resubmission Figs.pzf: Figure 5 Resub - Sunday, October 24, 2010

Acute Desipramine - FST

eGFP Cre 0

100

200

300

400 Saline

DMI

## ##

Imm

ob

ility

(s

ec

)

Acute Chlordiazepoxide

Home Novel0

200

400

600

800

eGFP-Saline

eGFP-CDP

Cre-Saline

Cre-CDP

#

##

##

La

ten

cy

(s

ec

.)

Chronic Desipramine

Home Novel0

200

400

600

800

eGFP-Saline

eGFP-DMI

Cre-Saline

Cre-DMI

#

*

La

ten

cy

to

fe

ed

(s

ec

)

AAV.eGFP AAV.Cre0

500

1000

1500

2000

Saline

DMI

###

Brd

U-IR

ce

lls

A

C

B

D

Page 61: Role Of Hippocampal Neurogenesis In The Etiology And

53

Figure 5. In the NIH paradigm, ATR-deleted mice exhibit altered antidepressant

response. A In a novel environment, AAV.Cre-injected mice exhibited a reduction in

latency to consume peanut butter chips compared to AAV.eGFP-injected mice (#,

p<0.0001, n=7-8 per group). There was a significant effect of chronic DMI treatment

(12.5 mg/kg b.i.d., 24 days) only in the AAV.eGFP-injected mice (*, p<0.05). B CDP

administered 30 minutes prior to novel testing reduces latency to feed in both control

and ATR-deleted mice (##, p<0.0001 vs. saline-treated, n=8-10 per group). C DMI

significantly reduces immobility in the FST in both control and ATR-deleted mice (##,

p<0.0001 vs. saline-treated, n=8-10 per group). DMI was administered 24 hours (10

mg/kg), 5 hours (10 mg/kg), and 1 hour (20 mg/kg) prior to the 6-minute swim test.

Immobility was measured over the final 4 minutes. D ATR deletion reduces cell

proliferation in the dentate gyrus (###, p<0.001 vs. AAV.eGFP-treated, n=7-8 per

group) Chronic DMI (12.5 mg/kg b.i.d., 24 days) has no effect on cell proliferation. Error

bars indicate SEM.

Page 62: Role Of Hippocampal Neurogenesis In The Etiology And

54

Figure 6. Hippocampal CA1 dendritic spine density following DMI treatment.

Figure 6. Hippocampal CA1 dendritic spine density following DMI treatment. A, B

Representative photomicrographs of Golgi impregnation staining to visualize dendritic

arborization following saline (A) or DMI (B) treatment in AAV.eGFP-injected mice. C

Chronic DMI treatment significantly increased CA1 dendritic spine density in AAV.eGFP-

injected mice (***, p < 0.001, n=30-42 per group). This effect was attenuated in

AAV.Cre-injected mice. (*, p < 0.05). Error bars indicate SEM.

ATR Paper - NPP.pzf: Figure 5 - Saturday, October 16, 2010

eGFP Cre

0

5

10

15

20

Saline

DMI***

*

Sp

ine

s / 1

0 u

mA

B

C

Saline

DMI

Page 63: Role Of Hippocampal Neurogenesis In The Etiology And

55

Figure S1. Hippocampal ATR deletion did not affect general locomotor activity

or HPA axis activation following an acute stress.

Figure S1. Hippocampal ATR deletion did not affect general locomotor activity or HPA

axis activation following an acute stress. A,B Floxed-ATR mice injected with AAV.Cre did

not differ from AAV.eGFP-injected mice in a 60 minute locomotor activity test to

measure ambulation (significant main effect of time, F(11,198)=26.84; p<0.0001; no effect

of AAV, F(1,198)=1.565; p=0.227; no AAV x time interaction, F(11,198)=0.852, p=0.5884,

RMANOVA) and crossings (significant main effect of time, F(11,198)=25.23, p<0.0001; no

effect of AAV, F(1,198)=1.28, p=0.273; no AAV x time interaction, F(11,198)=0.996,

p=0.452, RMANOVA). C ATR deletion did not affect serum corticosterone levels in

response to an acute stressor (significant main effect of time, F(3,39)=140.7, p<0.0001;

no effect of AAV, F(1,39)=0.044, p=0.838; no AAV*time interaction, F(3,39)=1.033;

p=0.3884, RMANOVA).

Page 64: Role Of Hippocampal Neurogenesis In The Etiology And

56

Figure S2. Fear conditioning is not altered by hippocampal ATR deletion.

Figure S2. Fear conditioning is not altered by hippocampal ATR deletion. A Acquisition

of fear conditioning occurred following the first CS-US pairing, as measured by increased

freezing across intervals (significant main effect of time, F(4,88)=38.3, p<0.0001; no

effect of AAV, F(1,88)=1.12, p=0.3013; no AAV x time interaction, F(4,88)=0.159, p=0.958,

RMANOVA). B When tested for contextual memory 24 hours following the training

session, both AAV treatment groups exhibit increased freezing compared to baseline

freezing levels prior to conditioning (significant main effect of time, F(1,22)=93.46,

***p<0.0001; no effect of AAV, F(1,22)=0.115, p=0.738; no AAV x time interaction,

F(1,22)=0.213, p=0.649, RMANOVA) C Both AAV treatment groups respond similarly to a

single CS exposure in a novel context (significant main effect of time, F(1,22)=20.18,

**p<0.001; no effect of AAV, F(1,22)=0.164, p=0.689; no AAV x time interaction,

F(1,22)=0.139, p=0.713, RMANOVA).

Page 65: Role Of Hippocampal Neurogenesis In The Etiology And

57

Figure S3. ATR deletion does not induce impairments in spatial learning and

memory in the Morris water maze.

Figure S3. ATR deletion does not induce impairments in spatial learning and memory in

the Morris water maze. Paradoxically, ATR-deleted mice perform better than controls in

locating the hidden platform. This subtle effect is an interesting area of exploration for

future research, however it is beyond the scope and focus of the present paper. A Both

AAV.eGFP and AAV.Cre-injected mice learned the location of the hidden platform,

measured by reduced latency across 9 days (3 trials/day). RMANOVA revealed

significant main effects of time (F(8.152)=44.04, p<0.0001), and AAV (F(1,152)=5.38,

p<0.05) with no AAV x time interaction (F(8,152) = 0.494, p=0.859). B In the hidden

platform probe test both treatment groups spent significantly greater time than chance

in the target (North) quadrant (***p<0.0001, one sample T-test). C From a constant

starting point, both treatment groups located the platform in a novel location in the

reversal learning task. Additionally, ATR-deleted mice did not exhibit impairments in this

task from a novel start location on the 7th day, suggesting intact spatial relational

memory. RMANOVA revealed a significant main effect of time (F(6,108)=18.11, p<0.0001),

a non-significant trend for an effect of AAV (F(1,108)=4.073, p=0.059), and no AAV x time

interaction (F(6,108)=0.896, p=0.5).

Page 66: Role Of Hippocampal Neurogenesis In The Etiology And

58

Discussion

Since neurogenesis was first proposed to regulate response to antidepressant

treatment (Jacobs et al., 2000), numerous publications have examined hippocampal

neurogenesis in the context of depression. However, its precise role in the etiology and

treatment of mood disorders remains controversial. In this study we showed that ATR is

necessary for maintenance of the basal rate of hippocampal neurogenesis. Deletion of

ATR from the hippocampus resulted in a partial inhibition of neurogenesis and reduced

anxiety-like behavior in multiple mouse models of affective state. Antidepressant

treatment was behaviorally ineffective in the NIH test following ATR deletion. In

addition, the increased dendritic spine density induced by chronic treatment with DMI

was severely attenuated following ATR deletion. These data suggest that capacity for

increased neuronal plasticity in the hippocampus, rather than increased neurogenesis,

may underlie the behavioral efficacy of DMI in the NIH test. A neurogenesis-independent

effect of DMI in the NIH test is additionally supported by the observation that DMI

reduced hyponeophagia in control mice without stimulating cell proliferation.

ATR is important for maintaining genomic integrity during DNA synthesis and

proliferative failure is observed in its absence (Brown and Baltimore, 2000; Brown and

Baltimore, 2003). No obvious behavioral changes are observed when ATR is deleted

from differentiated cells in the forebrain (Ruzankina et al., 2007), suggesting that the

behavioral alterations observed herein can be attributed to deletion of ATR from cells

with proliferative potential following hippocampal AAV microinjection. The partial

reduction in neurogenesis is likely related to the infection rate of the AAV in the neural

stem and progenitor cells. Hypothetically, greater inhibition could be obtained by

utilizing an approach that targets these cells with greater affinity. However, the

Page 67: Role Of Hippocampal Neurogenesis In The Etiology And

59

significant cellular and behavioral effects observed in this model of partially blocked

neurogenesis reinforce the sensitivity of hippocampal functioning to changes in

neurogenesis.

We observed a 45% reduction in hippocampal cell proliferation (Fig. 2a) and a

similar reduction in the total number of 4-week old cells (Fig. 2b), 8 weeks following

ATR deletion. Consistent with this finding, 54% fewer doublecortin-expressing cells were

present in the hippocampus as a result of ATR deletion (Fig. 2c). Doublecortin is

transiently expressed in immature neurons as they mature and migrate (Brown et al.,

2003). These data indicate that ATR is necessary to maintain a normal rate of

hippocampal neurogenesis. In a second model of ATR deletion that does not involve

virus injection, we used a tamoxifen-inducible Cre transgene to delete ATR. In this

model we observed a 30% reduction in cell proliferation as early as 5 weeks after ATR

deletion. While this lesser reduction may be attributed to the abbreviated period

between deletion and BrdU assay, the dose of tamoxifen and level of Cre induction may

also be factors. Regardless, the reduced cell proliferation and reduced anxiety-like

behavior observed in this second model used to delete ATR supports our finding that

ATR plays a critical role in cell proliferation in the hippocampus.

ATR deletion resulted in reduced anxiety in multiple rodent behavioral tests,

including the EZM (Fig. 3), marble burying test (Fig. 4a-b), and NIH test (Fig. 4c-d).

These observations are at odds with studies associating reduced neurogenesis with

increased anxiety in the elevated plus maze (Revest et al., 2009; Conboy et al., 2010)

and light-dark box (Scobie et al., 2009), and finding no baseline change in

hyponeophagia (Santarelli et al., 2003; Revest et al., 2009; Scobie et al., 2009).

Interestingly, recent literature (Fuss et al., 2010a) suggests that intermediate levels of

Page 68: Role Of Hippocampal Neurogenesis In The Etiology And

60

neurogenesis are associated with low levels of anxiety in some paradigms, with exercise

and irradiation, which increase and ablate neurogenesis, respectively, both associated

with elevated anxiety. The anxiolytic phenotype observed in ATR-deleted mice, which

exhibit levels of neurogenesis intermediate between basal levels and a complete

ablation, appear to be in agreement with the aforementioned finding. Furthermore,

though at the opposite end of the spectrum, our data is in line with studies

demonstrating a correlation between increased neurogenesis and increased anxiety

following voluntary exercise (Fuss et al., 2010b). This association between increased

neurogenesis and increased anxiety is also observed in CREB-deficient mice (Gur et al.,

2007).

The NIH test is considered an ethologically relevant test with strong predictive

and construct validity (Dulawa and Hen, 2005). Our lab and others have previously

demonstrated that latency to feed in the novel environment can be reduced by acute

treatment with a benzodiazepine, as well as treatment with chronic, but not acute,

antidepressant compounds (Dulawa and Hen, 2005, for review; Gur et al., 2007;

Gundersen and Blendy, 2009). Our interest in examining the ATR mice in the NIH test

was two-fold. First, baseline behavior in the novel environment provides a measure of

anxiety. Second, this behavior is sensitive to chronic AD treatment that is thought to

depend on increased neurogenesis. However, in studies where x-ray irradiation is used

to ablate neurogenesis, there are discrepancies in the resulting effects on both baseline

and antidepressant-mediated hyponeophagia (Santarelli et al., 2003; Holick et al., 2008;

David et al., 2009). These conflicting findings may stem from variations in mouse strain,

AD treatment, or hyponeophagia protocol, as some investigators utilize the novelty-

suppressed feeding test, a variation of the NIH test. Additionally, reported effects of x-

Page 69: Role Of Hippocampal Neurogenesis In The Etiology And

61

ray irradiation on differentiation (Tada et al., 2000) and inflammatory response

(Mizumatsu et al., 2003) have the potential to interfere with evaluation of hippocampal-

modulated behaviors. Whether or not these factors confound the findings of studies

using x-ray irradiation, confirmation by genetic methods in which neurogenesis is

reduced or ablated will lend confidence to the interpretations and implications of altered

neurogenesis and AD responsivity. To date, no one has reported whether a partial deficit

in neurogenesis generated by genetic manipulation, such as that seen following ATR

deletion, alters AD efficacy in a chronic exposure paradigm.

We observed a significant behavioral effect of DMI treatment in the NIH test only

in our control mice (Fig. 5a). As baseline latencies were lower in the ATR-deleted mice,

we ruled out a floor effect by testing the efficacy of a benzodiazepine, CDP, on latency

in the novel test following ATR deletion. CDP significantly reduced latency to feed

following ATR deletion (Fig. 5b). Both control and ATR-deleted mice exhibited reduced

immobility time in the forced swim test after acute treatment (Fig. 5c), ruling out a more

general deficit in response to DMI following ATR deletion.

Based on the behavioral observations in the NIH test, we hypothesized that DMI

treatment in AAV.eGFP-injected mice would lead to increased neurogenesis, and that

this effect of DMI would be attenuated in ATR-deleted mice. Interestingly, we observed

no effect of DMI treatment on cell proliferation in either the control or ATR-deleted mice

(Fig. 5d). While this was an unexpected observation, previous work has established that

antidepressant effects on neurogenesis can be small in magnitude and depend on

numerous factors including mouse strain and pretreatment (Balu et al., 2009; David et

al., 2009). Furthermore, others have reported no effects of antidepressant treatment on

Page 70: Role Of Hippocampal Neurogenesis In The Etiology And

62

cell proliferation in rodents (Huang et al., 2008; Navailles et al., 2008; Petersen et al.,

2009).

DMI reduces latency to feed without affecting cell proliferation, indicating that

DMI’s effect on latency is through a neurogenesis-independent mechanism. To better

understand this dissociation between the DMI’s effects on neurogenesis and behavior in

the NIH paradigm, we sought to determine if DMI exerts other trophic effects in the

hippocampus, and whether those effects may explain the lack of efficacy of DMI in the

NIH paradigm following ATR deletion. To this end, we observed significantly increased

CA1 dendritic spine density in control mice treated with DMI; this effect was greatly

attenuated following ATR deletion (Fig. 6c). Dendritic spine density is enhanced by

various external stimuli including learning (Moser et al., 1994), enrichment (Zhu et al.,

2009), exercise (Stranahan et al., 2009) and antidepressant treatment; all stimuli which

can have a positive influence on mood and emotional affect in depressed individuals. In

agreement with work by others (Bessa et al., 2009), our results suggest that neuronal

plasticity and/or remodeling, not increased neurogenesis, underlies the behavioral

efficacy of antidepressant treatment.

As ATR is not necessary for the functioning of mature cells, attenuation of DMI-

mediated increases in dendritic spine density in the CA1 region, an area of the

hippocampus composed of mature neurons, hints at broad, circuit-wide changes in

hippocampal function as a result of reduced neurogenesis. Immature neurons in the

adult hippocampus exhibit unique properties including enhanced excitability and a lower

threshold for the induction of long-term potentiation, indicative of enhanced potential for

synaptic plasticity (Schmidt-Hieber et al., 2004). A reduction in the proportion of

hippocampal neurons which exhibit these unique properties, as well a reduction in the

Page 71: Role Of Hippocampal Neurogenesis In The Etiology And

63

number of new neurons available to functionally integrate into the hippocampal circuitry,

may interfere with homeostatic functioning of the hippocampus, indirectly rendering it

less responsive to the changes normally induced by DMI. Consistent with this

hypothesis, ablation of neurogenesis leads to a reduction in evoked perforant path

responses and an increase in the amplitude of spontaneous network activity in the

dentate gyrus in an in vivo model (Lacefield et al., 2010). It is not unreasonable to

predict that changes in network activity in the dentate gyrus due to loss of young,

plastic neurons might extend to interrupting circuitry function within other regions of the

hippocampus. The importance of neurogenesis for the treatment of mood disorders may

thus lie in the role of new neurons in maintaining homeostatic network activity within

the hippocampus, and this concept is supported by evidence that fluoxetine depends on

the plasticity of young neurons for its anxiolytic and antidepressant efficacy (Wang et

al., 2008).

Hippocampal ATR deletion provides a valuable model for studying the behavioral

changes associated with deficits in hippocampal neurogenesis. In future studies this

model will be a useful tool for determining whether different classes of antidepressant

compounds exhibit varying degrees of neurogenesis dependence. In addition, it is of

interest to evaluate susceptibility to depressive-like behaviors in mouse models of

depression such as chronic mild stress and chronic social defeat in the ATR-deleted

mice. Further insight into the pathophysiology and treatment of depression should lead

to the eventual development of more rapid-acting treatments with fewer undesired side

effects.

Page 72: Role Of Hippocampal Neurogenesis In The Etiology And

64

References Balu DT, Hodes GE, Anderson BT, Lucki I (2009). Enhanced sensitivity of the MRL/MpJ

mouse to the neuroplastic and behavioral effects of chronic antidepressant treatments. Neuropsychopharmacology 34(7): 1764-1773.

Balu DT, Lucki I (2009). Adult hippocampal neurogenesis: regulation, functional implications, and contribution to disease pathology. Neurosci Biobehav Rev 33(3): 232-252.

Bergami M, Berninger B, Canossa M (2009). Conditional deletion of TrkB alters adult hippocampal neurogenesis and anxiety-related behavior. Commun Integr Biol 2(1): 14-16.

Bessa JM, Ferreira D, Melo I, Marques F, Cerqueira JJ, Palha JA, Almeida OF, Sousa N (2009). The mood-improving actions of antidepressants do not depend on neurogenesis but are associated with neuronal remodeling. Mol Psychiatry 14(8): 764-773, 739.

Bondolfi L, Ermini F, Long JM, Ingram DK, Jucker M (2004). Impact of age and caloric restriction on neurogenesis in the dentate gyrus of C57BL/6 mice. Neurobiol Aging 25(3): 333-340.

Brown EJ, Baltimore D (2000). ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev 14(4): 397-402.

Brown EJ, Baltimore D (2003). Essential and dispensable roles of ATR in cell cycle arrest and genome maintenance. Genes Dev 17(5): 615-628.

Brown JP, Couillard-Despres S, Cooper-Kuhn CM, Winkler J, Aigner L, Kuhn HG (2003). Transient expression of doublecortin during adult neurogenesis. J Comp Neurol 467(1): 1-10.

Calabrese F, Molteni R, Racagni G, Riva MA (2009). Neuronal plasticity: a link between stress and mood disorders. Psychoneuroendocrinology 34 Suppl 1: S208-216.

Conboy L, Varea E, Castro JE, Sakouhi-Ouertatani H, Calandra T, Lashuel HA, Sandi C (2010). Macrophage migration inhibitory factor is critically involved in basal and fluoxetine-stimulated adult hippocampal cell proliferation and in anxiety, depression, and memory-related behaviors. Mol Psychiatry.

Couillard-Despres S, Wuertinger C, Kandasamy M, Caioni M, Stadler K, Aigner R, Bogdahn U, Aigner L (2009). Ageing abolishes the effects of fluoxetine on neurogenesis. Mol Psychiatry 14(9): 856-864.

David DJ, Klemenhagen KC, Holick KA, Saxe MD, Mendez I, Santarelli L, Craig DA, Zhong H, Swanson CJ, Hegde LG, Ping XI, Dong D, Marzabadi MR, Gerald CP, Hen R (2007). Efficacy of the MCHR1 antagonist N-[3-(1-{[4-(3,4-difluorophenoxy)phenyl]methyl}(4-piperidyl))-4-methylphen yl]-2-

Page 73: Role Of Hippocampal Neurogenesis In The Etiology And

65

methylpropanamide (SNAP 94847) in mouse models of anxiety and depression following acute and chronic administration is independent of hippocampal neurogenesis. J Pharmacol Exp Ther 321(1): 237-248.

David DJ, Samuels BA, Rainer Q, Wang JW, Marsteller D, Mendez I, Drew M, Craig DA, Guiard BP, Guilloux JP, Artymyshyn RP, Gardier AM, Gerald C, Antonijevic IA, Leonardo ED, Hen R (2009). Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron 62(4): 479-493.

Dulawa SC, Hen R (2005). Recent advances in animal models of chronic antidepressant effects: the novelty-induced hypophagia test. Neurosci Biobehav Rev 29(4-5): 771-783.

Duman CH, Schlesinger L, Russell DS, Duman RS (2008). Voluntary exercise produces antidepressant and anxiolytic behavioral effects in mice. Brain Res 1199: 148-158.

Elizalde N, Garcia-Garcia AL, Totterdell S, Gendive N, Venzala E, Ramirez MJ, Del Rio J, Tordera RM (2010). Sustained stress-induced changes in mice as a model for chronic depression. Psychopharmacology (Berl) 210(3): 393-406.

Fuss J, Ben Abdallah NM, Hensley FW, Weber KJ, Hellweg R, Gass P (2010). Deletion of running-induced hippocampal neurogenesis by irradiation prevents development of an anxious phenotype in mice. PLoS One 5(9).

Fuss J, Ben Abdallah NM, Vogt MA, Touma C, Pacifici PG, Palme R, Witzemann V, Hellweg R, Gass P (2010). Voluntary exercise induces anxiety-like behavior in adult C57BL/6J mice correlating with hippocampal neurogenesis. Hippocampus 20(3): 364-376.

Gundersen BB, Blendy JA (2009). Effects of the histone deacetylase inhibitor sodium butyrate in models of depression and anxiety. Neuropharmacology 57(1): 67-74.

Gur TL, Conti AC, Holden J, Bechtholt AJ, Hill TE, Lucki I, Malberg JE, Blendy JA (2007). cAMP response element-binding protein deficiency allows for increased neurogenesis and a rapid onset of antidepressant response. J Neurosci 27(29): 7860-7868.

Hajszan T, MacLusky NJ, Leranth C (2005). Short-term treatment with the antidepressant fluoxetine triggers pyramidal dendritic spine synapse formation in rat hippocampus. Eur J Neurosci 21(5): 1299-1303.

Holick KA, Lee DC, Hen R, Dulawa SC (2008). Behavioral effects of chronic fluoxetine in BALB/cJ mice do not require adult hippocampal neurogenesis or the serotonin 1A receptor. Neuropsychopharmacology 33(2): 406-417.

Huang GJ, Bannerman D, Flint J (2008). Chronic fluoxetine treatment alters behavior, but not adult hippocampal neurogenesis, in BALB/cJ mice. Mol Psychiatry 13(2): 119-121.

Page 74: Role Of Hippocampal Neurogenesis In The Etiology And

66

Jacobs BL, van Praag H, Gage FH (2000). Adult brain neurogenesis and psychiatry: a novel theory of depression. Mol Psychiatry 5(3): 262-269.

Jessberger S, Kempermann G (2003). Adult-born hippocampal neurons mature into activity-dependent responsiveness. Eur J Neurosci 18(10): 2707-2712.

Lacefield CO, Itskov V, Reardon T, Hen R, Gordon JA (2010). Effects of adult-generated granule cells on coordinated network activity in the dentate gyrus. Hippocampus.

Law AJ, Weickert CS, Hyde TM, Kleinman JE, Harrison PJ (2004). Reduced spinophilin but not microtubule-associated protein 2 expression in the hippocampal formation in schizophrenia and mood disorders: molecular evidence for a pathology of dendritic spines. Am J Psychiatry 161(10): 1848-1855.

Lenze EJ, Sheffrin M, Driscoll HC, Mulsant BH, Pollock BG, Dew MA, Lotrich F, Devlin B, Bies R, Reynolds CF, 3rd (2008). Incomplete response in late-life depression: getting to remission. Dialogues Clin Neurosci 10(4): 419-430.

Malberg JE, Duman RS (2003). Cell proliferation in adult hippocampus is decreased by inescapable stress: reversal by fluoxetine treatment. Neuropsychopharmacology 28(9): 1562-1571.

Malberg JE, Eisch AJ, Nestler EJ, Duman RS (2000). Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. J Neurosci 20(24): 9104-9110.

Marchetti C, Tafi E, Middei S, Rubinacci MA, Restivo L, Ammassari-Teule M, Marie H (2010). Synaptic adaptations of CA1 pyramidal neurons induced by a highly effective combinational antidepressant therapy. Biol Psychiatry 67(2): 146-154.

Merikangas KR, Zhang H, Avenevoli S, Acharyya S, Neuenschwander M, Angst J (2003). Longitudinal trajectories of depression and anxiety in a prospective community study: the Zurich Cohort Study. Arch Gen Psychiatry 60(10): 993-1000.

Mizumatsu S, Monje ML, Morhardt DR, Rola R, Palmer TD, Fike JR (2003). Extreme sensitivity of adult neurogenesis to low doses of X-irradiation. Cancer Res 63(14): 4021-4027.

Moser MB, Trommald M, Andersen P (1994). An increase in dendritic spine density on hippocampal CA1 pyramidal cells following spatial learning in adult rats suggests the formation of new synapses. Proc Natl Acad Sci U S A 91(26): 12673-12675.

Murray F, Smith DW, Hutson PH (2008). Chronic low dose corticosterone exposure decreased hippocampal cell proliferation, volume and induced anxiety and depression like behaviours in mice. Eur J Pharmacol 583(1): 115-127.

Navailles S, Hof PR, Schmauss C (2008). Antidepressant drug-induced stimulation of mouse hippocampal neurogenesis is age-dependent and altered by early life stress. J Comp Neurol 509(4): 372-381.

Page 75: Role Of Hippocampal Neurogenesis In The Etiology And

67

Norrholm SD, Ouimet CC (2001). Altered dendritic spine density in animal models of depression and in response to antidepressant treatment. Synapse 42(3): 151-163.

O'Leary OF, Wu X, Castren E (2009). Chronic fluoxetine treatment increases expression of synaptic proteins in the hippocampus of the ovariectomized rat: role of BDNF signalling. Psychoneuroendocrinology 34(3): 367-381.

Papp M, Willner P, Muscat R (1991). An animal model of anhedonia: attenuation of sucrose consumption and place preference conditioning by chronic unpredictable mild stress. Psychopharmacology (Berl) 104(2): 255-259.

Petersen A, Wortwein G, Gruber SH, El-Khoury A, Mathe AA (2009). Nortriptyline mediates behavioral effects without affecting hippocampal cytogenesis in a genetic rat depression model. Neurosci Lett 451(2): 148-151.

Revest JM, Dupret D, Koehl M, Funk-Reiter C, Grosjean N, Piazza PV, Abrous DN (2009). Adult hippocampal neurogenesis is involved in anxiety-related behaviors. Mol Psychiatry 14(10): 959-967.

Ruzankina Y, Pinzon-Guzman C, Asare A, Ong T, Pontano L, Cotsarelis G, Zediak VP, Velez M, Bhandoola A, Brown EJ (2007). Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1(1): 113-126.

Ruzankina Y, Schoppy DW, Asare A, Clark CE, Vonderheide RH, Brown EJ (2009). Tissue regenerative delays and synthetic lethality in adult mice after combined deletion of Atr and Trp53. Nat Genet 41(10): 1144-1149.

Sahay A, Hen R (2007). Adult hippocampal neurogenesis in depression. Nat Neurosci 10(9): 1110-1115.

Salam JN, Fox JH, Detroy EM, Guignon MH, Wohl DF, Falls WA (2009). Voluntary exercise in C57 mice is anxiolytic across several measures of anxiety. Behav Brain Res 197(1): 31-40.

Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen R (2003). Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 301(5634): 805-809.

Saxe MD, Battaglia F, Wang JW, Malleret G, David DJ, Monckton JE, Garcia AD, Sofroniew MV, Kandel ER, Santarelli L, Hen R, Drew MR (2006). Ablation of hippocampal neurogenesis impairs contextual fear conditioning and synaptic plasticity in the dentate gyrus. Proc Natl Acad Sci U S A 103(46): 17501-17506.

Schmidt-Hieber C, Jonas P, Bischofberger J (2004). Enhanced synaptic plasticity in newly generated granule cells of the adult hippocampus. Nature 429(6988): 184-187.

Page 76: Role Of Hippocampal Neurogenesis In The Etiology And

68

Schoevers RA, Van HL, Koppelmans V, Kool S, Dekker JJ (2008). Managing the patient with co-morbid depression and an anxiety disorder. Drugs 68(12): 1621-1634.

Scobie KN, Hall BJ, Wilke SA, Klemenhagen KC, Fujii-Kuriyama Y, Ghosh A, Hen R, Sahay A (2009). Kruppel-like factor 9 is necessary for late-phase neuronal maturation in the developing dentate gyrus and during adult hippocampal neurogenesis. J Neurosci 29(31): 9875-9887.

Stranahan AM, Lee K, Martin B, Maudsley S, Golden E, Cutler RG, Mattson MP (2009). Voluntary exercise and caloric restriction enhance hippocampal dendritic spine density and BDNF levels in diabetic mice. Hippocampus 19(10): 951-961.

Tada E, Parent JM, Lowenstein DH, Fike JR (2000). X-irradiation causes a prolonged reduction in cell proliferation in the dentate gyrus of adult rats. Neuroscience 99(1): 33-41.

Turner JR, Castellano LM, Blendy JA (2010). Nicotinic partial agonists varenicline and sazetidine-A have differential effects on affective behavior. J Pharmacol Exp Ther 334(2): 665-672.

von Bohlen Und Halbach O (2009). Structure and function of dendritic spines within the hippocampus. Ann Anat 191(6): 518-531.

Wang JW, David DJ, Monckton JE, Battaglia F, Hen R (2008). Chronic fluoxetine stimulates maturation and synaptic plasticity of adult-born hippocampal granule cells. J Neurosci 28(6): 1374-1384.

West MJ, Slomianka L, Gundersen HJ (1991). Unbiased stereological estimation of the total number of neurons in thesubdivisions of the rat hippocampus using the optical fractionator. Anat Rec 231(4): 482-497.

Zhu SW, Codita A, Bogdanovic N, Hjerling-Leffler J, Ernfors P, Winblad B, Dickins DW, Mohammed AH (2009). Influence of environmental manipulation on exploratory behaviour in male BDNF knockout mice. Behav Brain Res 197(2): 339-346.

Page 77: Role Of Hippocampal Neurogenesis In The Etiology And

69

CHAPTER III

Low Neurogenesis Does Not Increase Susceptibility To Stress-Induced or Corticosterone-Induced Depressive Behaviors in Mice.

Page 78: Role Of Hippocampal Neurogenesis In The Etiology And

70

Abstract

Depression and mood disorders are associated with deficits in hippocampal size

and function. Additionally, the birth of new neurons, or neurogenesis, in the

hippocampus is thought to underlie the beneficial effects of certain antidepressant

treatments in preclinical models. The relevance of hippocampal neurogenesis in the

etiology of depression is less clear, with numerous studies suggesting reduced

neurogenesis does not precipitate depressive-like phenotypes in rodents. This area of

research is impeded by a poor understanding of the pathophysiology of depressive

disorders in humans. In this study, we examined the effects of a partial blockade of

adult hippocampal neurogenesis on basal and stress-induced affective behavior and

neuroendocrine function. Neurogenesis was inhibited by approximately 50% in adult

mice by hippocampal-specific deletion of ataxia-telangeictasia mutated and Rad-3

related (ATR) protein, a cell cycle checkpoint kinase essential for normal levels of

neurogenesis. In male mice, reduced hippocampal neurogenesis was associated with an

anhedonic phenotype in the sucrose preference test. However, in the learned

helplessness paradigm, reduced neurogenesis did not influence the likelihood of

developing learned helplessness or the extent of the escape deficit observed in

susceptible mice. In addition, ATR deletion did not potentiate the effects of chronic

corticosterone treatment. Consistent with current literature, these findings suggest

reduced neurogenesis is likely not a prominent risk factor for mood disorders.

Page 79: Role Of Hippocampal Neurogenesis In The Etiology And

71

Introduction

Mood disorders, including major depressive disorder, dysthymic disorder and

bipolar disorder, affect approximately 10% of adults in the U.S. each year (Kessler et al.,

2003). Despite this high prevalence, the pathophysiology of mood disorders is poorly

understood. Among individuals suffering from depressive disorders, a high level of

heterogeneity is observed in the manifestation of symptoms as well as in the treatment

efficacy and prognosis. This has led researchers and clinicians to suggest depressive

disorders may be subdivided into a spectrum of subtypes with a diverse, multifactorial

pathophysiology (Akiskal, 1989; Fagiolini and Kupfer, 2003).

The hippocampus has been extensively implicated in the etiology and treatment

of mood disorders. Clinical studies in depressed individuals have demonstrated reduced

hippocampal volume which is reversed by antidepressant treatment (Sheline et al.,

1999). A twin study demonstrated that reduced hippocampal volume may be indicative

of increased vulnerability to psychiatric illness (Gilbertson et al., 2002). In addition, the

hippocampus plays a key regulatory role in the activity of the hypothalamic-pituitary-

adrenal (HPA) axis. In response to stress, a major risk factor for depressive disorders

(Kessler, 1997), HPA axis activation leads to the production of glucocorticoid stress

hormones (Herman et al., 1996): cortisol in humans and corticosterone in rodents.

Stress hormones activate glucocorticoid receptors within the hippocampus, leading to

inhibitory feedback and suppression of subsequent glucocorticoid production (Herman et

al., 1989; Jacobson and Sapolsky, 1991; van Haarst et al., 1997). Approximately 50% of

depressed individuals exhibit HPA axis abnormalities (Gibbons, 1964).

A unique feature of the hippocampus is the continued formation of a substantial

population of new neurons into adulthood, or neurogenesis (Altman and Das, 1965;

Page 80: Role Of Hippocampal Neurogenesis In The Etiology And

72

Ming and Song, 2005, for review). New neurons in the hippocampus have been

extensively implicated in learning and memory processes as well as in mental health

(Eisch et al., 2008, for review; Aimone et al., 2010, for review). Neurogenesis is subject

to regulation by a variety of stimuli; factors such as stress (Gould et al., 1997; Tanapat

et al., 1998) and drugs of abuse (Yamaguchi et al., 2004; Eisch et al., 2008) reduce

hippocampal neurogenesis while learning (Patel et al., 1997; Gould et al., 1999),

environmental enrichment (Kempermann et al., 1997; Kempermann et al., 1998),

exercise (van Praag et al., 1999) and antidepressants (Malberg et al., 2000) can

increase neurogenesis. Newborn neurons mature and functionally integrate into the

hippocampal circuitry in approximately 4 weeks (van Praag et al., 2002), similar to the

time necessary for antidepressants to exert their mood-improving actions in depressed

individuals. A number of studies in which hippocampal neurogenesis is ablated by either

x-ray irradiation or genetic manipulation suggest neurogenesis is necessary for the

efficacy of some antidepressant drugs (Santarelli et al., 2003; Surget et al., 2008; David

et al., 2009; Perera et al., 2011).

On the other hand, there is little direct evidence supporting reduced hippocampal

neurogenesis in the pathophysiology of depression. Although reduced neurogenesis and

depressive-like phenotypes arise in tandem following various chronic stressors including

unpredictable chronic mild stress (Willner et al., 1987; Surget et al., 2008; Bessa et al.,

2009), chronic corticosterone (Gourley et al., 2008; Murray et al., 2008), and social

defeat stress (Berton et al., 2006; Czeh et al., 2007; Van Bokhoven et al., 2011), no one

has demonstrated a requirement for reduced neurogenesis in the development of a

depressive-like phenotype. However, we and others have shown inhibition of

neurogenesis to be associated with altered anxiety-like behaviors (Bergami et al., 2008;

Page 81: Role Of Hippocampal Neurogenesis In The Etiology And

73

Revest et al., 2009; Onksen et al., 2011), which often present in a co-morbid,

overlapping fashion with depressive symptoms.

We previously described the effects of Crelox-conditional deletion of ATR from

the hippocampus of adult mice, specifically a partial reduction of hippocampal

neurogenesis (Onksen et al., 2011). Here, we evaluated the effect of ATR deletion on

rodent phenotypes considered to be representative of symptoms of depressive

disorders, including anhedonia in a sucrose preference test, behavioral despair in a

learned helplessness paradigm, and behavioral and endocrine effects of chronic

corticosterone treatment.

Materials & Methods

Animals Mice homozygous for the Crelox-conditional allele of ATR (ATRf/f) on a

129SV/C57BL/6 background were generated as previously described (Ruzankina et al.,

2007). ATRf/f mice receiving hippocampal microinjection of AAV.Cre and having ATR

deleted throughout the hippocampus are subsequently referred to as ATRΔHipp, while

control mice are injected with AAV.eGFP and retain the ATRf/f designation. Mice (1.5-5

months old, 20-35 gm) were housed in groups of 3-4 following stereotaxic surgery and

during experimental paradigms, with exceptions noted. All behavioral experiments were

conducted at least 5 weeks following stereotaxic surgery or tamoxifen treatment.

Animals were maintained on a 12 hr light/dark cycle with food and water available ad

libitum in accordance with the University of Pennsylvania Institutional Animal Care and

Use Committee.

Page 82: Role Of Hippocampal Neurogenesis In The Etiology And

74

Experimental design

Experiment 1. Sucrose preference test (n=17 male ATRf/f, 21 male ATRΔHipp, 13 female

ATRf/f, 15 female ATRΔHipp). To determine if mice with reduced hippocampal

neurogenesis exhibit anhedonia, we performed a two-bottle choice sucrose preference

test. Five weeks following adeno-associated virus (AAV) injection, mice were single-

housed and given two weeks to acclimate to these new housing conditions. Mice were

then acclimated to the presence of two drinking bottles for one week, and subsequently

given a choice between water and a solution of 1% sucrose dissolved in water.

Preference and total intake were analyzed for a 14 day period. This experiment was run

in two separate cohorts of mice and data were pooled after determining that control

groups did not differ from one another statistically.

Experiment 2. Learned helplessness. (n=31 male ATRf/f, 29 male ATRΔHipp). The

learned helplessness paradigm was used to determine if mice with reduced hippocampal

neurogenesis exhibit heightened susceptibility to the development of behavioral despair.

Only males were used in this experiment, as we observed behavioral effects of ATR

deletion in the sucrose drinking test only in male mice. Additionally, female mice are

resistant to developing learned helplessness (Shors et al., 2007; Dalla et al., 2008). Mice

were assigned to either a no shock control group or an inescapable shock (IS) group.

Mice in the IS group were exposed to two 1-hour sessions of 120 inescapable footshocks

while mice in the control group were placed in the training environment for the same

length of time in the absence of footshocks. Subsequently, latency to escape from a

series of 15 escapable footshocks was measured. The average escape latency during the

final 5 escapable footshock trials was used for data analysis. A subset of mice (n=20

Page 83: Role Of Hippocampal Neurogenesis In The Etiology And

75

ATRf/f, 19 ATRΔHipp) were injected with bromodeoxyuridine (BrdU) 48 hours prior to the

first training session to assess hippocampal cell genesis. These mice were killed 24

hours following the testing session and BrdU-positive cells were quantified by

immunohistochemistry.

Experiment 3. Behavioral and endocrine response to chronic corticosterone. (n=20

ATRf/f, 17 ATRΔHipp). Corticosterone pellets (21 day sustained release) were implanted to

mimic chronic stress and to subsequently determine if reduced neurogenesis potentiates

the effect of chronic stress on behavioral and endocrine endpoints. 26 days following

pellet implantation, the forced swim test (FST) was performed. This consisted of a pre-

swim session and a post-swim session. The antidepressant desipramine was given

intraperitoneally three times in the 24 hour period between swim sessions. Both male

and female mice were used and data was pooled after determining there was no sex

effect in the FST. Three days following the forced swim test, in a subset of mice (n=8

male ATRf/f, 8 male ATRΔHipp), we evaluated endocrine response to an acute stressor.

Only males were used (to avoid sex hormone effects). A 15 minute restraint stress was

performed and tail blood was collected prior to and at three time points following the

restraint to measure serum corticosterone.

Adeno-associated virus Adeno-associated viruses (AAV) expressing Cre recombinase

(AAV2/9.CMV.PI.CRE, titer 2.84*1013 gc/ml) and eGFP (AAV2/9.CMV.eGFP, titer

3.74*1013 gc/ml) were generated by the University of Pennsylvania Vector Core. The

expression cassette consists of the AAV2 terminal repeats flanking the CMV promoter-PI-

Cre recombinase/eGFP sequences packaged into AAV9. The vectors were purified using

Page 84: Role Of Hippocampal Neurogenesis In The Etiology And

76

CsCl sedimentation method. Quantification of vector genome copies (gc) was performed

by Q-PCR. AAVs were diluted in sterile PBS for microinjections.

Hippocampal Injections ATRf/f mice (6-8 weeks) were anesthetized with isoflurane

and secured in a stereotaxic frame (Kopf, Tujuna, CA). Holes were drilled bilaterally in

the skull at sites of injection. Stereotaxic coordinates for the hippocampus are (from

Bregma) anterior-posterior -2.1, lateral +/- 1.4, dorso-ventral -2.0, and anterior-

posterior -2.9, lateral +/- 3.0, dorso-ventral -3.8. 0.5 ul of 1*1010 gc/ul AAV was injected

at each site through a 33 gauge needle on a 5 ul Hamilton syringe using a KDS310 Nano

Pump (KD Scientific, Holliston, MA) mounted to the stereotaxic frame, at a rate of 0.1

ul/min. The needle remained in place for 4 additional minutes at each injection site. The

skin was sutured and the animal recovered on a heating pad before returning to the

home cage.

Drugs BrdU (Sigma) was dissolved in 0.9% saline immediately before use and injected

intraperitoneally using a volume of 10 ml/kg. In the present study, 200 mg/kg BrdU was

administered twice, 2 days prior to inescapable shock exposure in the learned

helplessness paradigm, with injections separated by 8 hours. Corticosterone (5

mg/pellet, 21 day sustained release) or placebo pellets (Innovative Research of America,

Sarasota, FL) were implanted subcutaneously. Each mouse in the corticosterone-

treatment group was implanted with two 5 mg pellets to achieve the equivalent of 20

mg/kg/day. Desipramine (DMI, Sigma) was dissolved in 0.9% saline immediately before

use and injected intraperitoneally using a volume of 10 ml/kg.

Page 85: Role Of Hippocampal Neurogenesis In The Etiology And

77

Sucrose Preference Mice were single-housed 2 weeks prior to beginning the test.

Singly-housed mice were acclimated to the presence of 2 water-containing drinking

bottles in their home cage for 1 week. Subsequently, mice were presented with 1 bottle

containing water and 1 bottle containing a 1% solution of sucrose (Fisher Scientific)

dissolved in water. The position of each bottle within a cage was changed every 24

hours to mitigate bottle location preferences. Bottles were weighed every 24 hours to

track consumption of both water and 1% sucrose. To accurately control for volume, two

bottles were placed in a cage containing no mouse. Volume lost from these bottles

averaged less than 5% of fluid intake and was subtracted from the daily intake prior to

data analysis. Water and sucrose intake over every 2 days was averaged prior to

analysis to account for any potential bottle position bias.

Learned Helplessness Learned helplessness was performed as previously described

(Berton et al., 2008). Training and testing was performed using an active avoidance

system (Med Associates, Georgia, VT) consisting of a shuttle box divided into two

compartments which are separated by an automated sliding door. The grid floor is

capable of delivering scrambled shocks. Photocell detection tracks the location of the

animal in the box. Training consisted of a one hour session in which mice in the

inescapable shock (IS) group were confined to one compartment of the shuttle box and

received 120 randomly distributed foot shocks (0.5 mA, 5 second duration). Training

occurred once per day for two days. Animals in the no-shock control group were placed

in the shuttle box for 1 hour in the absence of foot shocks. The testing session occurred

24 hours following the 2nd training session, and consisted of 15 escape trials spread over

a 1 hour session. During each trial, a shock was delivered continuously and mice were

Page 86: Role Of Hippocampal Neurogenesis In The Etiology And

78

given the opportunity to escape by passing into the adjacent compartment through the

door, which would open at the beginning of each trial. The door closed following

successful escape. If mice failed to escape within 25 seconds, the shock was terminated

and the door was shut.

Forced Swim Test Mice were placed in water (15 cm depth, 23° C) in plastic cylinders

(23 cm tall x 14 cm diameter) for 6 minutes. Using Viewpoint automated scoring

(Viewpoint, Champagne au Mont d’Or, France), duration of immobility was measured.

Mice were exposed to the swim tanks twice (pre-swim and swim test), with 24 hours

between exposures. Three injections of DMI were given during the inter-swim interval:

The first at 24 hours prior to the swim test (10 mg/kg), a second injection 5 hours prior

to the swim test (10 mg/kg), and a third injection 1 hour prior to the swim test (20

mg/kg).

Restraint Stress Mice were placed in 50 mL Falcon conical vials for 15 minutes. Holes

were drilled at both ends to allow for breathing and to give access to the tail. Tail blood

(20 uL) was collected at 0, 15, 30, and 90 minutes. Blood was transferred directly into

tubes with chilled EDTA, centrifuged at 5,000 RPM for 10 minutes at 4 C, and serum was

used to measure corticosterone levels with a 125I Corticosterone RIA kit (MP Biomedicals,

Orangeburg, NY).

Immunohistochemistry Mice were anesthetized with nembutol (10 mg/kg) and

transcardially perfused with cold 0.1 M phosphate-buffered saline (PBS) for 5 minutes,

followed by cold 4% paraformaldehyde (PFA) in PBS for 15 minutes. Brains were

Page 87: Role Of Hippocampal Neurogenesis In The Etiology And

79

postfixed overnight in PFA at 4° C and subsequently stored at 4° C in 30% sucrose.

Brains were frozen on dry ice, sectioned coronally at a thickness of 40 µm, and

transferred to PBS + 0.5% Sodium Azide at 4° C prior to processing for

immunohistochemistry. For BrdU analysis, sections were wet-mounted on

Superfrost/Plus slides (Fisher Scientific, Pittsburgh, PA) for stereological analysis using a

modified version of the optical fractionator method (West et al., 1991). Mounted

sections were incubated in 0.1 M citric acid, pH 6.0, for antigen retrieval. Subsequently,

slides were incubated in trypsin, 2N HCl, then primary antibody (mouse anti-BrdU,

1:200, Becton Dickinson) with 0.5% Tween 20 overnight. On the second day, slides

were incubated for 1 hour in secondary antibody (biotinylated anti-mouse IgG, Vector

Laboratories), 1 hour in avidin-biotin-HRP (1:100, Vector Laboratories), and labeled cells

were visualized using diaminobenzidine (Sigma). PBS-washed slides were then

counterstained and coverslipped with Permount. BrdU-IR cells in every 9th section of the

hippocampus were counted with a 100x oil immersion lens, omitting cells in the upper-

most focal plane. Cells within clusters were counted by distinguishing nuclear borders

while focusing down through the tissue using an objective with a narrow depth of focus.

For proliferation counts, a cell is included in the subgranular zone of the dentate gyrus if

it is touching, or within two cells of the granule cell layer.

Statistical Analysis Sucrose drinking data was analyzed by two-factor ANOVA with

time as a repeated factor. Escape latency and cell proliferation data were analyzed by

two-factor ANOVA. Two-factor ANOVAs were performed using GraphPad Prism 5 and

Bonferroni’s post-tests were applied where appropriate. Stress-induced serum

Page 88: Role Of Hippocampal Neurogenesis In The Etiology And

80

corticosterone and forced swim test immobility were analyzed by three-factor ANOVA

using JMP software. All data are reported as mean + SEM.

Results

Experiment 1: Sucrose preference

Male ATRΔHipp mice exhibit a marked reduction in preference for a 1% sucrose

solution (Fig. 1a) compared to ATRf/f controls (gene main effect, F(1,216)=12.01, p<0.01;

time main effect, F(6,216)=5.92, p<0.0001). This difference is not observed following ATR

deletion in female mice (Fig. 1b; gene main effect, F(1,156)=0.255, p=0.618; time main

effect, F(6,156)=5.5, p<0.0001). ATR deletion does not affect total fluid consumption in

males (Fig. 1c; gene main effect, F(1,216)=2.12, p=0.154; time main effect, F(6,216)=3.35,

p<0.01) or females (Fig. 1d; gene main effect, F(1,156)=1.82, p=0.189; time main effect,

F(6,156)=9.86, p<0.0001).

Experiment 2: Learned helplessness

Learned helplessness. Based on sucrose preference findings, we examined

whether the anhedonic phenotype observed in males would translate to susceptibility in

the learned helplessness paradigm, a rodent model of behavioral despair. Following 2

days of exposure to repeated, inescapable footshocks, latency to escape from escapable

footshocks was measured. Mice were exposed to 15 escapable footshock trials during a

1 hour test session. The average escape latency of the last 5 trials was calculated (Fig.

2a). A significant main effect of inescapable shock was observed (F(1,56)=27.77,

p<0.0001). There was no effect of ATR deletion on average escape latency

Page 89: Role Of Hippocampal Neurogenesis In The Etiology And

81

(F(1,56)=0.037, p=0.848). Variability was observed in the effect of inescapable shock

treatment; some mice were susceptible to developing learned helplessness while others

performed similar to no-shock controls. ATR deletion did not influence the proportion of

mice that were susceptible.

Cell genesis. A subset of mice in the learned helplessness experiment received

two injections of BrdU (200mg/kg, IP) two days prior to inescapable footshock exposure

to evaluate cell genesis in the hippocampus (Fig. 2d). As expected, there was an overall

effect of ATR deletion to reduce BrdU-labeled cells (F(1,32)=35.24, p<0.0001). Exposure

to inescapable footshocks did not have a significant effect on this outcome measure

(F(1,32)=0.296, p=0.59). In addition, no correlation was observed between escape

latency and cell genesis (data not shown).

Experiment 3: Behavioral and endocrine response to chronic corticosterone

Forced swim test. Because stress is a major risk factor in the pathophysiology of

depression, we sought to determine if reduced neurogenesis exacerbates the effects of

chronic stress. To do this, we examined the effects of ATR deletion on behavioral and

endocrine endpoints following chronic corticosterone treatment. 21 day extended-

release pellets were implanted to deliver 20 mg/kg/day corticosterone to mimic the

effects of chronic, mild stress. To assess the behavioral effect of chronic corticosterone,

immobility in the FST was measured 26 days following pellet implantation (Fig. 3a).

Increased immobility in the FST is interpreted as behavioral despair. Three-factor

ANOVA revealed a near-significant effect of corticosterone pellet to increase immobility

(F(1,27)=3.69, p=0.066) and, as a positive control, a significant effect of the

antidepressant desipramine (DMI) to reduce immobility (F(1,27)=9.09, p<0.01). There

Page 90: Role Of Hippocampal Neurogenesis In The Etiology And

82

was not a significant gene effect (F(1,27)=0.035, p=0.85) and no significant interactions

were observed, indicating that ATR deletion does not alter the expected effects of prior

corticosterone experience and of antidepressant treatment on immobility in the FST.

Restraint stress. We previously showed that the partial reduction of neurogenesis

observed following hippocampal ATR deletion is not associated with altered

corticosterone response to an acute restraint stress in naïve mice. Here, we evaluated

the effect of chronic corticosterone on subsequent response to an acute restraint stress.

28 days following corticosterone pellet implantation, mice were subjected to a 15 minute

restraint stress and tail blood was collected at 0, 15, 30 and 90 minutes to measure

serum corticosterone (Fig. 3b). Analysis by three-factor ANOVA revealed a significant

main effect of time (time main effect, F(3,63)=90.37, p<0.0001; gene main effect,

F(1,63)=0.04, p=0.839; pellet main effect, F(1,63)=0.16, p=0.693) and a significant time x

pellet interaction (F(3,63)=9.48, p<0.0001). Bonferroni post-tests to compare

corticosterone treatment at the 4 time points indicated that prior corticosterone pellet

experience blunted the corticosterone response to an acute stressor at the 15 minute

and 30 minute time points. ATR deletion did not influence the effect of chronic

corticosterone exposure on acute stress response.

Page 91: Role Of Hippocampal Neurogenesis In The Etiology And

83

Figure 1. Sucrose preference and fluid intake.

Figure 1. Sucrose preference and total fluid intake. (A,B) Sucrose consumption as a

percent of total fluid intake across 14 days. Males exhibit reduced preference following

ATR deletion (A, *p<0.01). (C,D) ATR deletion does not affect total fluid intake during

the sucrose preference test. Error bars represent SEM.

2 4 6 8 10 12 140

20

40

60

80

100

ATRf/f

ATR!Hipp

Su

cro

se

Pre

fere

nc

e

(Su

cro

se

/ T

ota

l * 1

00

)

2 4 6 8 10 12 140

20

40

60

80

100

ATRf/f

ATR!Hipp

Su

cro

se

Pre

fere

nc

e

(Su

cro

se

/ T

ota

l * 1

00

)

2 4 6 8 10 12 140

2

4

6

ATRf/f

ATR!Hipp

Flu

id C

on

su

mp

tio

n (m

L)

2 4 6 8 10 12 140

2

4

6

ATRf/f

ATR!Hipp

Flu

id C

on

su

mp

tio

n (m

L)

Males Females

A

DC

B

} *

Page 92: Role Of Hippocampal Neurogenesis In The Etiology And

84

Figure 2. Learned helplessness.

Figure 2. Learned helplessness. (A) Mice in the inescapable shock (IS) group exhibit

significantly greater escape latencies during the escape trials compared to the control

no-shock (NS) mice (***p<0.0001 vs. NS group). (B) Mice exposed to inescapable

shock do not differ from no-shock controls in the number of BrdU-positive cells in the

hippocampus. There is a gene main effect such that ATR deletion results in significantly

reduced BrdU-immunoreactive (IR) cells (#p<0.001, two-way ANOVA). Error bars

represent SEM.

Learned Helplessness

NS IS0

5

10

15

20

ATRf/f

ATR!Hipp

***E

sc

ap

e L

ate

nc

y (s

ec

)

A

B Cell Proliferation

NS IS0

500

1000

1500

ATRf/f

ATR!Hipp

Brd

U-IR

ce

lls

#

#

Page 93: Role Of Hippocampal Neurogenesis In The Etiology And

85

Figure 3. Behavioral and endocrine consequences of chronic corticosterone.

Restraint Stress

0 15 30 900

50

100

150

200

250ATRf/f-Placebo

ATRf/f-Cort

ATR!Hipp-Placebo

ATR!Hipp-CortRestraint

*

*

Co

rtic

os

tero

ne

(n

g/u

l)Forced Swim Test

Placebo-Sal Placebo-DMI Cort-Sal Cort-DMI0

50

100

150

200

250 ATRf/f

ATR!Hipp

Imm

ob

ility

(s

ec

on

ds

)A

B

Page 94: Role Of Hippocampal Neurogenesis In The Etiology And

86

Figure 3. Behavioral and endocrine consequences of chronic corticosterone. (A)

Immobility during the final 4 minutes of a 6 minute swim test is graphed. Desipramine

(DMI) reduced immobility and prior corticosterone (Cort) pellet experience increased

immobility. There was no effect of ATR deletion within any of the four

pellet/antidepressant conditions. N = 4-5 per treatment group. (B) Serum cort is plotted

at 4 time points. Restraint stress occurred between the 0 and 15 minute time points.

Prior corticosterone pellet experience resulted in significantly reduced cort at the 15 and

30 minute time points compared to placebo pellet (*, p<0.05). There was no significant

effect of ATR deletion. N = 4 per treatment group. Error bars represent SEM.

Page 95: Role Of Hippocampal Neurogenesis In The Etiology And

87

Discussion

In this study we explored the possibility that a partial reduction in hippocampal

neurogenesis may result in depressive-like phenotypes in mice. We previously showed

an approximately 50% reduction in hippocampal neurogenesis following deletion of ATR

from this brain region and additionally observed alterations in anxiety-like behaviors and

in response to antidepressant treatment. Thus, we sought to determine if these effects

extended to phenotypes representative of the symptoms of depression.

ATR deletion resulted in a significantly reduced preference for a 1% sucrose

solution in male mice (Fig. 1). This effect was not observed in female mice, and was not

due to a change in total fluid intake. Reduced sucrose preference in rodents is an

indicator of anhedonia, a key symptom of depression. Preference for this palatable

solution can be reduced by chronic stress and subsequently brought back to normal

levels by treatment with antidepressants (Katz, 1982; Willner et al., 1987; Papp et al.,

1991; Muscat et al., 1992). Various forms of chronic stress can lead to reduced

neurogenesis as well as reduced sucrose preference in both mice (Aonurm-Helm et al.,

2008; Goshen et al., 2008) and rats (Veena et al., 2009). A survey of the literature to

identify studies in which neurogenesis was knocked down through x-ray irradiation,

cytotoxicity, or genetic manipulation revealed just two studies in rats demonstrating no

effect of reduced neurogenesis on sucrose preference (Bessa et al., 2009; Noonan et al.,

2010), suggesting chronic stress reduces neurogenesis and sucrose preference

independently of each other. We are not aware of studies in mice evaluating sucrose

preference following a non-stress induced reduction of neurogenesis, leaving open the

possibility that reduced neurogenesis blunts hedonic response. Alternatively, it can not

be ruled out that the anhedonic phenotype we observe is not due to an unintended

Page 96: Role Of Hippocampal Neurogenesis In The Etiology And

88

effect of single housing (Hellemans et al., 2004; Wei et al., 2007). Recent work from

Belforte and colleagues found single housing to potentiate anxiogenic and anhedonic

phenotypes in male mutant mice with a conditional knockout of NMDA receptor subunit

NR1 from cortical and hippocampal GABAergic interneurons (Belforte et al., 2010). Mice

in our study were single-housed two weeks prior to the start of the sucrose drinking

task. This mild stressor may have acted synergistically to induce an anhedonic

phenotype in ATR-deleted mice. Greater sensitivity to single-housing among male mice

in this particular measure of anhedonia would explain why we do not see an effect in

females. A possible stress effect of single housing suggests reduced neurogenesis alone

may not be sufficient to induce anhedonia in group-housed animals.

We next evaluated behavioral despair in the learned helplessness paradigm

following ATR deletion. This paradigm, first described by Overmier and Seligman (1967),

models depression through exposure to unpredictable and unavoidable shocks which

lead to deficits in subsequent coping behavior. Due to multiple reports that females are

resistant to the development of behavioral deficits in the learned helplessness paradigm

(Shors et al., 2007; Dalla et al., 2008) only males were used for this portion of the

study. We observed a significant effect of IS on escape latency during the avoidance

test (Fig. 2a), such that IS resulted in a phenotype indicative of behavioral despair, or a

lack of effort to escape from an avoidable footshock. In addition, among IS-treated

mice, some were susceptible to developing learned helplessness while others were

resistant and behaved similar to no shock control mice in the avoidance test. This

phenomenon is responsible for the relatively large standard error observed among IS

treatment groups compared to the NS control groups. This individual susceptibility has

been observed previously in both rats and mice (Vollmayr et al., 2003; Chourbaji et al.,

Page 97: Role Of Hippocampal Neurogenesis In The Etiology And

89

2005). Consistent with other studies investigating the relationship between hippocampal

neurogenesis and learned helplessness (Vollmayr et al., 2003; Reif et al., 2004; but see

Ho and Wang, 2010), we did not observe an effect of ATR deletion on the proportion of

mice susceptible to developing learned helplessness. In addition, escape latency and cell

genesis did not correlate, further supporting the conclusion that rate of neurogenesis

does not directly influence the development of learned helplessness. As expected, ATR

deletion resulted in an overall effect of reduced cell genesis (Fig. 2b).

In the third experiment we examined the effects of reduced hippocampal

neurogenesis on chronic exposure to low dose corticosterone. Exogenous corticosterone

induces behaviors reminiscent of depression and anxiety, similar to those observed

following chronic mild stress (Murray et al., 2008; Zhao et al., 2008; David et al., 2009;

Crupi et al., 2010). The dose used in the present study (20 mg/kg/day, 21 day release)

has been shown to induce increased immobility in the forced swim test (FST) and tail

suspension test without inhibiting cell proliferation (Murray et al., 2008; Zhao et al.,

2008). This dose was selected to determine if corticosterone levels that have a sub-

threshold effect on cell proliferation produce more severe depressive-like symptoms on a

background of low neurogenesis. We found chronic corticosterone exposure to increase

immobility in a subsequent FST (Fig. 3a), indicative of increased behavioral despair.

However, ATR deletion did not alter the corticosterone pellet’s effects on immobility in

the FST. In addition to this behavioral outcome measure, we evaluated endocrine

response to an acute stressor by measuring serum corticosterone during an acute

restraint stress following chronic corticosterone pellet exposure. Prior corticosterone

exposure blunted the subsequent response to an acute stress experience (Fig. 3b).

Similar to the FST, ATR deletion did not increase the severity of the corticosterone

Page 98: Role Of Hippocampal Neurogenesis In The Etiology And

90

pellet’s effects. From these findings we can conclude that the detrimental effects of

chronic exposure to heightened circulating corticosterone are not exacerbated by

reduced basal levels of neurogenesis.

In conclusion, while susceptibility to depression is strongly influenced by

numerous factors such as social stress, environmental stress and genetics, neurogenesis

does not appear to exert a significant influence over the development of depressive-like

behavior in rodents. Mechanistically, new neurons are essential for detecting and

encoding subtle contextual changes, a process also referred to as pattern separation

(Clelland et al., 2009; Sahay et al., 2011). Based on this, Perera and colleagues have

postulated that suppressed neurogenesis, occurring during periods of stress, may result

in uncoupling of affect and external context during negative mood states. Inability to

recognize subtle reductions in stress due to poor pattern separation may then lead to

persistence of a negative mood that is no longer reflective of the external context,

precipitating a depressive disorder (Perera et al., 2008). Due to the diverse

pathophysiology and varied treatment responsiveness across the population of

depressed individuals, it is indeed possible that neurogenesis plays an role in certain

cases of clinical depression. However, current methods of examining depressive-like

behavior in pre-clinical models have not yet revealed reduced neurogenesis to be a

significant risk factor.

Page 99: Role Of Hippocampal Neurogenesis In The Etiology And

91

References Aimone JB, Deng W, Gage FH (2010). Adult neurogenesis: integrating theories and

separating functions. Trends Cogn Sci 14(7): 325-337.

Akiskal HS (1989). New insights into the nature and heterogeneity of mood disorders. J Clin Psychiatry 50 Suppl: 6-10; discussion 11-12.

Altman J, Das GD (1965). Autoradiographic and histological evidence of postnatal hippocampal neurogenesis in rats. J Comp Neurol 124(3): 319-335.

Aonurm-Helm A, Jurgenson M, Zharkovsky T, Sonn K, Berezin V, Bock E, Zharkovsky A (2008). Depression-like behaviour in neural cell adhesion molecule (NCAM)-deficient mice and its reversal by an NCAM-derived peptide, FGL. Eur J Neurosci 28(8): 1618-1628.

Belforte JE, Zsiros V, Sklar ER, Jiang Z, Yu G, Li Y, Quinlan EM, Nakazawa K (2010). Postnatal NMDA receptor ablation in corticolimbic interneurons confers schizophrenia-like phenotypes. Nat Neurosci 13(1): 76-83.

Bergami M, Rimondini R, Santi S, Blum R, Gotz M, Canossa M (2008). Deletion of TrkB in adult progenitors alters newborn neuron integration into hippocampal circuits and increases anxiety-like behavior. Proc Natl Acad Sci U S A 105(40): 15570-15575.

Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, Russo SJ, Graham D, Tsankova NM, Bolanos CA, Rios M, Monteggia LM, Self DW, Nestler EJ (2006). Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 311(5762): 864-868.

Bessa JM, Ferreira D, Melo I, Marques F, Cerqueira JJ, Palha JA, Almeida OF, Sousa N (2009). The mood-improving actions of antidepressants do not depend on neurogenesis but are associated with neuronal remodeling. Mol Psychiatry 14(8): 764-773, 739.

Chourbaji S, Zacher C, Sanchis-Segura C, Dormann C, Vollmayr B, Gass P (2005). Learned helplessness: validity and reliability of depressive-like states in mice. Brain Res Brain Res Protoc 16(1-3): 70-78.

Clelland CD, Choi M, Romberg C, Clemenson GD, Jr., Fragniere A, Tyers P, Jessberger S, Saksida LM, Barker RA, Gage FH, Bussey TJ (2009). A functional role for adult hippocampal neurogenesis in spatial pattern separation. Science 325(5937): 210-213.

Crupi R, Mazzon E, Marino A, La Spada G, Bramanti P, Cuzzocrea S, Spina E (2010). Melatonin treatment mimics the antidepressant action in chronic corticosterone-treated mice. J Pineal Res 49(2): 123-129.

Page 100: Role Of Hippocampal Neurogenesis In The Etiology And

92

Czeh B, Muller-Keuker JI, Rygula R, Abumaria N, Hiemke C, Domenici E, Fuchs E (2007). Chronic social stress inhibits cell proliferation in the adult medial prefrontal cortex: hemispheric asymmetry and reversal by fluoxetine treatment. Neuropsychopharmacology 32(7): 1490-1503.

Dalla C, Edgecomb C, Whetstone AS, Shors TJ (2008). Females do not express learned helplessness like males do. Neuropsychopharmacology 33(7): 1559-1569.

David DJ, Samuels BA, Rainer Q, Wang JW, Marsteller D, Mendez I, Drew M, Craig DA, Guiard BP, Guilloux JP, Artymyshyn RP, Gardier AM, Gerald C, Antonijevic IA, Leonardo ED, Hen R (2009). Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron 62(4): 479-493.

Eisch AJ, Cameron HA, Encinas JM, Meltzer LA, Ming GL, Overstreet-Wadiche LS (2008). Adult neurogenesis, mental health, and mental illness: hope or hype? J Neurosci 28(46): 11785-11791.

Fagiolini A, Kupfer DJ (2003). Is treatment-resistant depression a unique subtype of depression? Biol Psychiatry 53(8): 640-648.

Gibbons JL (1964). Cortisol Secretion Rate in Depressive Illness. Arch Gen Psychiatry 10: 572-575.

Gilbertson MW, Shenton ME, Ciszewski A, Kasai K, Lasko NB, Orr SP, Pitman RK (2002). Smaller hippocampal volume predicts pathologic vulnerability to psychological trauma. Nat Neurosci 5(11): 1242-1247.

Goshen I, Kreisel T, Ben-Menachem-Zidon O, Licht T, Weidenfeld J, Ben-Hur T, Yirmiya R (2008). Brain interleukin-1 mediates chronic stress-induced depression in mice via adrenocortical activation and hippocampal neurogenesis suppression. Mol Psychiatry 13(7): 717-728.

Gould E, Beylin A, Tanapat P, Reeves A, Shors TJ (1999). Learning enhances adult neurogenesis in the hippocampal formation. Nat Neurosci 2(3): 260-265.

Gould E, McEwen BS, Tanapat P, Galea LA, Fuchs E (1997). Neurogenesis in the dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. J Neurosci 17(7): 2492-2498.

Gourley SL, Wu FJ, Taylor JR (2008). Corticosterone regulates pERK1/2 map kinase in a chronic depression model. Ann N Y Acad Sci 1148: 509-514.

Hellemans KG, Benge LC, Olmstead MC (2004). Adolescent enrichment partially reverses the social isolation syndrome. Brain Res Dev Brain Res 150(2): 103-115.

Herman JP, Prewitt CM, Cullinan WE (1996). Neuronal circuit regulation of the hypothalamo-pituitary-adrenocortical stress axis. Crit Rev Neurobiol 10(3-4): 371-394.

Page 101: Role Of Hippocampal Neurogenesis In The Etiology And

93

Herman JP, Schafer MK, Young EA, Thompson R, Douglass J, Akil H, Watson SJ (1989). Evidence for hippocampal regulation of neuroendocrine neurons of the hypothalamo-pituitary-adrenocortical axis. J Neurosci 9(9): 3072-3082.

Ho YC, Wang S (2010). Adult neurogenesis is reduced in the dorsal hippocampus of rats displaying learned helplessness behavior. Neuroscience 171(1): 153-161.

Jacobson L, Sapolsky R (1991). The role of the hippocampus in feedback regulation of the hypothalamic-pituitary-adrenocortical axis. Endocr Rev 12(2): 118-134.

Katz RJ (1982). Animal model of depression: pharmacological sensitivity of a hedonic deficit. Pharmacol Biochem Behav 16(6): 965-968.

Kempermann G, Brandon EP, Gage FH (1998). Environmental stimulation of 129/SvJ mice causes increased cell proliferation and neurogenesis in the adult dentate gyrus. Curr Biol 8(16): 939-942.

Kempermann G, Kuhn HG, Gage FH (1997). More hippocampal neurons in adult mice living in an enriched environment. Nature 386(6624): 493-495.

Kessler RC (1997). The effects of stressful life events on depression. Annu Rev Psychol 48: 191-214.

Kessler RC, Berglund P, Demler O, Jin R, Koretz D, Merikangas KR, Rush AJ, Walters EE, Wang PS (2003). The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R). JAMA 289(23): 3095-3105.

Malberg JE, Eisch AJ, Nestler EJ, Duman RS (2000). Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. J Neurosci 20(24): 9104-9110.

Ming GL, Song H (2005). Adult neurogenesis in the mammalian central nervous system. Annu Rev Neurosci 28: 223-250.

Murray F, Smith DW, Hutson PH (2008). Chronic low dose corticosterone exposure decreased hippocampal cell proliferation, volume and induced anxiety and depression like behaviours in mice. Eur J Pharmacol 583(1): 115-127.

Muscat R, Papp M, Willner P (1992). Reversal of stress-induced anhedonia by the atypical antidepressants, fluoxetine and maprotiline. Psychopharmacology (Berl) 109(4): 433-438.

Noonan MA, Bulin SE, Fuller DC, Eisch AJ (2010). Reduction of adult hippocampal neurogenesis confers vulnerability in an animal model of cocaine addiction. J Neurosci 30(1): 304-315.

Onksen JL, Brown EJ, Blendy JA (2011). Selective deletion of a cell cycle checkpoint kinase (ATR) reduces neurogenesis and alters responses in rodent models of behavioral affect. Neuropsychopharmacology 36(5): 960-969.

Page 102: Role Of Hippocampal Neurogenesis In The Etiology And

94

Overmier JB, Seligman ME (1967). Effects of inescapable shock upon subsequent escape and avoidance responding. J Comp Physiol Psychol 63(1): 28-33.

Papp M, Willner P, Muscat R (1991). An animal model of anhedonia: attenuation of sucrose consumption and place preference conditioning by chronic unpredictable mild stress. Psychopharmacology (Berl) 104(2): 255-259.

Patel SN, Clayton NS, Krebs JR (1997). Spatial learning induces neurogenesis in the avian brain. Behav Brain Res 89(1-2): 115-128.

Perera TD, Dwork AJ, Keegan KA, Thirumangalakudi L, Lipira CM, Joyce N, Lange C, Higley JD, Rosoklija G, Hen R, Sackeim HA, Coplan JD (2011). Necessity of hippocampal neurogenesis for the therapeutic action of antidepressants in adult nonhuman primates. PLoS One 6(4): e17600.

Perera TD, Park S, Nemirovskaya Y (2008). Cognitive role of neurogenesis in depression and antidepressant treatment. Neuroscientist 14(4): 326-338.

Reif A, Schmitt A, Fritzen S, Chourbaji S, Bartsch C, Urani A, Wycislo M, Mossner R, Sommer C, Gass P, Lesch KP (2004). Differential effect of endothelial nitric oxide synthase (NOS-III) on the regulation of adult neurogenesis and behaviour. Eur J Neurosci 20(4): 885-895.

Revest JM, Dupret D, Koehl M, Funk-Reiter C, Grosjean N, Piazza PV, Abrous DN (2009). Adult hippocampal neurogenesis is involved in anxiety-related behaviors. Mol Psychiatry 14(10): 959-967.

Ruzankina Y, Pinzon-Guzman C, Asare A, Ong T, Pontano L, Cotsarelis G, Zediak VP, Velez M, Bhandoola A, Brown EJ (2007). Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1(1): 113-126.

Sahay A, Scobie KN, Hill AS, O'Carroll CM, Kheirbek MA, Burghardt NS, Fenton AA, Dranovsky A, Hen R (2011). Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature 472(7344): 466-470.

Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen R (2003). Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 301(5634): 805-809.

Sheline YI, Sanghavi M, Mintun MA, Gado MH (1999). Depression duration but not age predicts hippocampal volume loss in medically healthy women with recurrent major depression. J Neurosci 19(12): 5034-5043.

Shors TJ, Mathew J, Sisti HM, Edgecomb C, Beckoff S, Dalla C (2007). Neurogenesis and helplessness are mediated by controllability in males but not in females. Biol Psychiatry 62(5): 487-495.

Page 103: Role Of Hippocampal Neurogenesis In The Etiology And

95

Surget A, Saxe M, Leman S, Ibarguen-Vargas Y, Chalon S, Griebel G, Hen R, Belzung C (2008). Drug-dependent requirement of hippocampal neurogenesis in a model of depression and of antidepressant reversal. Biol Psychiatry 64(4): 293-301.

Tanapat P, Galea LA, Gould E (1998). Stress inhibits the proliferation of granule cell precursors in the developing dentate gyrus. Int J Dev Neurosci 16(3-4): 235-239.

Van Bokhoven P, Oomen CA, Hoogendijk WJ, Smit AB, Lucassen PJ, Spijker S (2011). Reduction in hippocampal neurogenesis after social defeat is long-lasting and responsive to late antidepressant treatment. Eur J Neurosci.

van Haarst AD, Oitzl MS, de Kloet ER (1997). Facilitation of feedback inhibition through blockade of glucocorticoid receptors in the hippocampus. Neurochem Res 22(11): 1323-1328.

van Praag H, Kempermann G, Gage FH (1999). Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci 2(3): 266-270.

van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD, Gage FH (2002). Functional neurogenesis in the adult hippocampus. Nature 415(6875): 1030-1034.

Veena J, Srikumar BN, Raju TR, Shankaranarayana Rao BS (2009). Exposure to enriched environment restores the survival and differentiation of new born cells in the hippocampus and ameliorates depressive symptoms in chronically stressed rats. Neurosci Lett 455(3): 178-182.

Vollmayr B, Simonis C, Weber S, Gass P, Henn F (2003). Reduced cell proliferation in the dentate gyrus is not correlated with the development of learned helplessness. Biol Psychiatry 54(10): 1035-1040.

Wei XY, Yang JY, Dong YX, Wu CF (2007). Anxiolytic-like effects of oleamide in group-housed and socially isolated mice. Prog Neuropsychopharmacol Biol Psychiatry 31(6): 1189-1195.

West MJ, Slomianka L, Gundersen HJ (1991). Unbiased stereological estimation of the total number of neurons in thesubdivisions of the rat hippocampus using the optical fractionator. Anat Rec 231(4): 482-497.

Willner P, Towell A, Sampson D, Sophokleous S, Muscat R (1987). Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology (Berl) 93(3): 358-364.

Yamaguchi M, Suzuki T, Seki T, Namba T, Juan R, Arai H, Hori T, Asada T (2004). Repetitive cocaine administration decreases neurogenesis in adult rat hippocampus. Ann N Y Acad Sci 1025: 351-362.

Zhao Y, Ma R, Shen J, Su H, Xing D, Du L (2008). A mouse model of depression induced by repeated corticosterone injections. Eur J Pharmacol 581(1-2): 113-120.

Page 104: Role Of Hippocampal Neurogenesis In The Etiology And

96

CHAPTER IV

Mice Deficient in Hippocampal Neurogenesis Do Not Develop an Anxiety

Phenotype Following Wheel Running

This chapter has been submitted for publication:

Onksen JL, Galante RJ, Pack AI, Blendy JA. Mice deficient in hippocampal neurogenesis do not develop an anxiety phenotype following wheel running. Submitted to Genes, Brain & Behavior (05/31/2011).

Page 105: Role Of Hippocampal Neurogenesis In The Etiology And

97

Abstract

Exercise, specifically voluntary wheel running, is a potent stimulator of hippocampal

neurogenesis in adult mice. In addition, exercise induces behavioral changes in

numerous measures of anxiety in rodents. However, the physiological underpinnings of

these changes are poorly understood. To investigate the role of neurogenesis in

exercise-mediated anxiety, we examined the cellular and behavioral effects of voluntary

wheel running in mice with a reduction in hippocampal neurogenesis, achieved through

conditional deletion of ataxia telangeictasia-mutated and rad-3 related protein (ATR), a

cell cycle checkpoint kinase necessary for normal levels of neurogenesis. Following

hippocampal microinjection of an adeno-associated virus expressing Cre recombinase to

delete ATR, mice were exposed to four weeks of voluntary wheel running and

subsequently evaluated for anxiety-like behavior. Wheel running resulted in increased

cell proliferation and neurogenesis, as measured by bromodeoxyuridine and

doublecortin, respectively. Wheel running also resulted in heightened anxiety in the

novelty-induced hypophagia, open field, and light-dark box tests. However, both the

neurogenic and anxiogenic effects of wheel running were attenuated following

hippocampal ATR deletion, suggesting increased neurogenesis is an important mediator

of exercise-induced anxiety.

Page 106: Role Of Hippocampal Neurogenesis In The Etiology And

98

Introduction

Exercise is associated with a wide array of health benefits (van Praag, 2008, for

review). In addition to the physical benefits, exercise can improve cognition (van Praag

et al., 1999; Van der Borght et al., 2007; Winter et al., 2007; Nichol et al., 2009),

alleviate symptoms of neurodegenerative diseases (Tillerson et al., 2003; Nichol et al.,

2009), as well as aid in recovery from mood disorders (Blumenthal et al., 1999; Babyak

et al., 2000; Duman et al., 2008).

Rodents exhibit a strong penchant for voluntary wheel running, which activates

brain reward pathways (Brene et al., 2007) and is used as a model to investigate the

mechanisms underlying the benefits of exercise in humans. Voluntary running results in

a robust increase in hippocampal neurogenesis in adult mice. Indeed, running has been

shown to increase proliferation of neural progenitor cells (Kronenberg et al., 2003), and

increase both the number and percentage of newborn cells which become mature

neurons (van Praag et al., 1999). Running and neurogenesis have been directly

correlated, both within and across mouse strains, further highlighting the potent

influence of exercise on hippocampal neurogenesis (Allen et al., 2001; Rhodes et al.,

2003; Clark et al., 2011). A multitude of changes are implicated in the therapeutic

effects of exercise, including heightened synaptic plasticity (van Praag et al., 1999;

Farmer et al., 2004), angiogenesis (Van der Borght et al., 2009) and growth factor

expression (Neeper et al., 1996; Kitamura et al., 2003). As evidenced by the importance

of newborn hippocampal neurons in cognition and mood regulation (Sahay et al., 2011;

Surget et al., 2011), neurogenesis is an additional mechanism which may contribute to

the therapeutic effects of exercise.

Page 107: Role Of Hippocampal Neurogenesis In The Etiology And

99

In light of clinical evidence for the mood-improving effects of exercise

(Deslandes et al., 2009, for review), we set out to investigate the underlying

mechanisms associated with these beneficial effects. A number of studies have

investigated the effects of voluntary wheel running on mood and anxiety-related

behaviors in rodents, the results of which are inconsistent. While some studies find

decreased anxiety (Dishman et al., 1996; Salam et al., 2009), others demonstrate

increased anxiety (Burghardt et al., 2004; Fuss et al., 2010). Yet others demonstrate

either increased or decreased anxiety depending on the specific anxiety measure used

and the timing of the measurement in relation to time after removal from running

wheels (Binder et al., 2004; Duman et al., 2008). Reports of heightened anxiety

following wheel running appear counterintuitive to the beneficial effects of exercise on

mood in humans. However, a recent study demonstrating that running-induced anxiety

was prevented by x-ray irradiation, a technique to ablate neurogenesis (Fuss et al.,

2010), supports a role for neurogenesis in the development of exercise induced-anxiety.

We previously showed partial suppression of hippocampal neurogenesis following

hippocampal deletion of ATR, a cell cycle checkpoint kinase (Onksen et al., 2011). Here,

we explored the possibility that deletion of ATR from the hippocampus would result in

resistance to the neurogenic effects of voluntary exercise and examined the behavioral

implications of that resistance.

Materials & Methods

Animals Mice homozygous for the Cre/lox-conditional allele of ATR (ATRf/f) on a

129SV/C57BL/6 background were generated as previously described (Ruzankina et al.,

Page 108: Role Of Hippocampal Neurogenesis In The Etiology And

100

2007). ATRf/f mice receiving hippocampal microinjection of AAV.Cre and having ATR

deleted throughout the hippocampus are subsequently referred to as ATRΔHipp, while

control mice are injected with AAV.eGFP and retain the ATRf/f designation.

Mice (1.5-4 months old, 20-35 gm) were housed in groups of 3-4 following

stereotaxic surgery and subsequently single-housed at least one week prior to being

placed into cages containing running wheels. Cages with running wheels (Mini Mitter,

Bend OR) measured 20x36 cm with an 11.5 cm diameter wheel mounted to the cage

top. Wheel rotations were monitored continuously via VitalView (Mini Mitter). Sedentary

control animals were housed in identical cages with immobilized wheels.

All behavioral experiments were conducted on adult males and females at least 6

weeks following stereotaxic surgery. Animals were maintained on a 12 hr light/dark

cycle with food and water available ad libitum in accordance with the University of

Pennsylvania Institutional Animal Care and Use Committee.

Experimental Design Two cohorts of ATRf/f mice were used in this study. Mice in

both cohorts received hippocampal micro-injections at 8 weeks of age. For a schematic

of the experimental design see Figure 1a. Mice in cohort #1 were used for the novelty-

induced hypophagia (NIH) test and were therefore single housed and trained to

consume peanut butter chips in their home cages prior to being placed in cages

containing running wheels. Cohort #2 was used to evaluate behavior in the open field,

marble-burying and light-dark box tests (separated by 3 days). All behavioral testing

was performed during the 4th week of wheel running. All mice were injected with 200

mg/kg bromodeoxyuridine (BrdU, Sigma) 24 hours following the last behavioral test and

subsequently perfused 24 hours following BrdU injection. Mice had ad libitum access to

Page 109: Role Of Hippocampal Neurogenesis In The Etiology And

101

running wheels until they were perfused (except when removed from the cages for

behavioral testing). Data from both cohorts was pooled to examine effects of AAV

injection and voluntary running on neurogenesis.

Adeno-associated virus Adeno-associated viruses (AAV) expressing Cre recombinase

(AAV2/9.CMV.PI.CRE, titer 2.84*1013 gc/ml) and eGFP (AAV2/9.CMV.eGFP, titer

3.74*1013 gc/ml) were generated by the University of Pennsylvania Vector Core. The

expression cassette consists of the AAV2 terminal repeats flanking the CMV promoter-PI-

Cre recombinase/eGFP sequences packaged into AAV9. The vectors were purified using

CsCl sedimentation method. Quantification of vector genome copies (gc) was performed

by Q-PCR. AAVs were diluted in sterile PBS for microinjections.

Hippocampal Injections ATRf/f mice (6-8 weeks) were anesthetized with isoflurane

and secured in a stereotaxic frame (Kopf, Tujuna, CA). Holes were drilled bilaterally in

the skull at sites of injection. Stereotaxic coordinates for the hippocampus are (from

Bregma) anterior-posterior -2.1, lateral +/- 1.4, dorso-ventral -2.0, and anterior-

posterior -2.9, lateral +/- 3.0, dorso-ventral -3.8. 0.5 ul of 1*1010 gc/ul AAV was injected

at each site through a 33 gauge needle on a 5 ul Hamilton syringe using a KDS310 Nano

Pump (KD Scientific, Holliston, MA) mounted to the stereotaxic frame, at a rate of 0.1

ul/min. The needle remained in place for 4 additional minutes at each injection site. The

skin was sutured and the animal recovered on a heating pad before returning to the

home cage.

Page 110: Role Of Hippocampal Neurogenesis In The Etiology And

102

Immunohistochemistry Mice were anesthetized with nembutol (10 mg/kg) and

transcardially perfused with cold 0.1 M phosphate-buffered saline (PBS) for 5 minutes,

followed by cold 4% paraformaldehyde (PFA) in PBS for 10 minutes. Brains were

postfixed overnight in PFA at 4◦C and subsequently stored at 4◦C in 30% sucrose. Brains

were frozen on dry ice, sectioned coronally at a thickness of 40 µm, and transferred to

PBS + 0.5% Sodium Azide at 4◦C prior to processing for immunohistochemistry.

For BrdU analysis, sections were wet-mounted on Superfrost/Plus slides (Fisher

Scientific, Pittsburgh, PA) for stereological analysis using a modified version of the

optical fractionator method (West et al., 1991). Mounted sections were incubated in 0.1

M citric acid, pH 6.0, for antigen retrieval. Subsequently, slides were incubated in

trypsin, 2N HCl, then primary antibody (mouse anti-BrdU, 1:200, Becton Dickinson) with

0.5% Tween 20 overnight. On the second day, slides were incubated for 1 hour in

secondary antibody (biotinylated anti-mouse IgG, Vector Laboratories), 1 hour in avidin-

biotin-HRP (1:100, Vector Laboratories), and labeled cells were visualized using

diaminobenzidine (Sigma). PBS-washed slides were then counterstained and

coverslipped with Permount. BrdU-IR cells in every 9th section of the hippocampus were

counted with a 100x oil immersion lens, omitting cells in the upper-most focal plane.

Cells within clusters were counted by distinguishing nuclear borders while focusing down

through the tissue using an objective with a narrow depth of focus. For proliferation

counts, a cell is included in the subgranular zone (SGZ) of the dentate gyrus if it is

touching, or within two cells of the granule cell layer (GCL).

For doublecortin (DCX) analysis, free-floating sections of a 1/9 series through the

hippocampus were blocked for 1 hour with 3% Normal Horse Serum, 0.5% Tween 20

and 0.2% Triton in PBS (blocking solution). Sections were then incubated for 72 hours

Page 111: Role Of Hippocampal Neurogenesis In The Etiology And

103

at 4◦C in primary antibody (Goat anti-DCX, Santa Cruz #8066) diluted 1:500 in blocking

solution. Sections were washed and incubated for 1 hour in secondary antibody (Horse

anti-goat, Vector Laboratories) diluted 1:200 in blocking solution. Sections were then

washed and treated with 0.75% H2O2 for 20 minutes prior to incubation in avidin-biotin-

HRP (1:200, Vector Laboratories). Labeled cells were visualized using nickel-enhanced

diaminobenzidine. Free-floating sections were mounted onto slides and dried overnight

before dehydrating and coverslipping with Permount. DCX-IR cells in every 9th section

were counted with a 100x oil immersion lens, omitting cells in the upper-most focal

plane. All cells located within the GCL and SGZ were counted.

Novelty-induced Hypophagia For 1 week prior to the training period, and through

the experiment, mice were single housed. On 11 consecutive training days, mice were

exposed to highly palatable food (peanut butter chips, Nestle, Glendale CA) in a plastic

dish in their home cage. On each training day mice were acclimated to the presence of a

plastic cage divider for 1 hour, consistent with previous NIH studies (Gur et al., 2007;

Onksen et al., 2011). Peanut butter chips were then placed in the cage for 15 minutes

and latency to initiate feeding was measured. Subsequent to training, mice were placed

in cages containing running wheels and left for 28 days. On days 25 and 26, a home

cage test was performed in which mice were removed from wheel-containing cages and

placed into a standard mouse cage, allowed to acclimate to the cage and divider for 1

hour, and latency to feed was subsequently measured. On day 27, latency to feed in a

novel environment was measured. Mice were removed from wheel-containing cages,

placed for 1 hour in the same standard mouse cage with divider that was used for home

cage tests, and subsequently placed in a novel, anxiety provoking environment where

Page 112: Role Of Hippocampal Neurogenesis In The Etiology And

104

latency to feed was measured. The novel environment consisted of an empty cage with

no bedding, set inside a white box with a bright light and a novel odor (pine sol).

Following each test session, mice were returned to their running wheel-containing

cages.

Open Field Open field behavior was monitored in a white box measuring 41x35 cm in

dim light conditions. Activity was recorded by an overhead camera and analyzed with

TopScanLite 2.0 (Clever Sys, Inc., Reston, VA) Parameters assessed were time spent

and distance traveled in the center and outer zones. The center zone was defined as

25% of the total area.

Light-Dark Box The light-dark box consisted of two adjacent chambers, connected by

a 5x5 cm opening. Each chamber measured 17x20 cm. The dark chamber was black and

covered. The light chamber was white and illuminated with bright light. Each test

session lasted 5 minutes and was initiated by placing the mouse in the corner of the

light chamber. Behavior was recorded by an overhead camera and analyzed with

TopScanLite 2.0. Parameters assessed were transitions between the two chambers, time

spent in each chamber, and distance traveled in the light chamber.

Statistical analysis Statistical analyses were performed with GraphPad Prism 5 and

JMP software. Data are reported as mean + SEM. Studies involving two variables were

analyzed by two-way ANOVA, with time as a repeated measure where appropriate.

Bonferroni post hoc tests were performed to compare individual treatment groups. Data

Page 113: Role Of Hippocampal Neurogenesis In The Etiology And

105

from the NIH test was analyzed by three-factor ANOVA to account for gene, exercise,

and test environment.

Results

ATR deletion attenuates the neurogenic effect of voluntary wheel running

To determine if ATR deletion confers resistance to the neurogenic effects of

wheel running, we microinjected AAV.Cre or AAV.eGFP into the hippocampus of ATRf/f

mice and assigned them to either the running or the sedentary condition. Mice were

single housed in running wheel-containing cages 6 weeks following AAV injection.

Running distance plateaued in the 2nd week in both ATRf/f and ATRΔHipp mice (Fig. 1b),

which is consistent with other studies involving wheel running (Clark et al., 2009). No

differences were observed in wheel running distance between treatment groups (ATRf/f,

7.73+2.49 km/day; ATRΔHipp, 8.31+2.14 km/day; gene main effect, F(1,72)=0.404,

p=0.53; time main effect, F(1,72)=16.57, p<0.001).

Following 28 days of running, immunohistochemical analysis was performed to

measure cell proliferation and number of immature neurons. BrdU (200 mg/kg) was

injected 24 hours prior to perfusing the animals. Two-way ANOVA of BrdU-positive cells

in the hippocampus (Fig. 2a) revealed main effects of gene (F(1,50)=28.89, p<0.0001)

and exercise (F(1,50)=23.33, p<0.0001), as well as a significant interaction (F(1,50)=5.26,

p<0.05). Post tests indicate that wheel running resulted in a significant increase in cell

proliferation in ATRf/f runners compared to all other treatment groups (+89% vs. ATRf/f

sedentary, +206% vs. ATRΔHipp sedentary, +102% vs. ATRΔHipp runners, each p<0.001).

Unlike ATRf/f runners, ATRΔHipp runners did not exhibit significantly increased cell

Page 114: Role Of Hippocampal Neurogenesis In The Etiology And

106

proliferation compared to their sedentary counterparts. In addition, as previously

observed, ATR deletion resulted in significantly reduced cell proliferation among

sedentary mice (-38%, p<0.05). Two-way ANOVA of doublecortin (DCX)-positive cells in

the hippocampus (Fig. 2b) revealed significant main effects of gene (F(1,51)=32.65,

p<0.0001) and exercise (F(1,51)=39.73, p<0.0001), as well as a significant interaction

(F(1,51)=10.08, p<0.01). Similar to our BrdU observations, post tests indicated DCX

expression was significantly increased in ATRf/f runners compared to all other groups

(+172% vs. ATRf/f sedentary, +406% vs. ATRΔHipp sedentary, +146% vs. ATRΔHipp

runners, each p<0.001). ATRΔHipp runners did not exhibit significantly increased DCX-

expressing cells compared to their sedentary counterparts. Post tests did not show a

significant effect of ATR deletion among sedentary mice. However, we previously

demonstrated an effect of ATR deletion to reduce DCX-expressing cells, so we therefore

performed a Student’s t-test comparing sedentary ATRf/f and ATRΔHipp mice to verify this

effect had again been achieved, and indeed it had (-46%, t(30)=4.00, p<0.001). Taken

together, BrdU and DCX data indicate that mice lacking hippocampal ATR exhibit a

reduced neurogenic response to wheel running.

Running-induced anxiety in the NIH test is attenuated in ATRΔHipp mice

We recently observed reduced feeding latency in a novel environment in mice

with reduced levels of neurogenesis (Onksen et al., 2011). The NIH paradigm consists of

training mice to consume a palatable food in their home cage, and subsequently

measuring their latency to consume this same food in a novel, anxiety provoking

environment. Thus, reduced neurogenesis may contribute to reduced anxiety in this

particular paradigm. As others have demonstrated heightened anxiety as a result of

Page 115: Role Of Hippocampal Neurogenesis In The Etiology And

107

voluntary exercise, we sought to determine whether our voluntary wheel running

paradigm would result in increased anxiety in the NIH test, and whether this anxiogenic

phenotype would be altered by reduced neurogenesis.

Six weeks following hippocampal microinjection of AAV to delete ATR, mice were

single housed and subsequently trained to consume peanut butter chips in their home

cage. No differences were observed in training performance between ATRf/f and ATRΔHipp

mice (Fig. 3a), as measured by two-way ANOVA of feeding latency on days 6-11, with

day as a repeated measure (day main effect, F(5,150)=15.91, p<0.0001; gene main

effect, F(1,150)=0.339, p=0.565). Training was followed by 28 days of housing in running-

wheel-containing cages.

On the 25th and 26th days of running, mice were given access to peanut butter

chips in a home cage environment. Latency was measured on the 2nd home environment

exposure. Our unpublished observations indicated exercise to be anxiogenic when mice

are initially removed from running wheels and tested for feeding latency in a home cage.

Thus, we measured feeding latency during the second day of home cage feeding, during

which all treatment groups performed similarly (Fig. 3b). Normalization of home cage

behavior allows for clear interpretation of any anxiety phenotypes observed in the novel

test. On the 27th day of running, feeding latency in a novel, anxiety-provoking

environment was measured.

Three-factor ANOVA of feeding latency revealed significant main effects of gene

(F(1,51)=7.64, p<0.01), exercise (F(1,51)=7.94, p<0.01) and environment (F(1,51)=23.31,

p<0.001), in addition to a significant gene x exercise x environment interaction

(F(1,51)=24.29, p<0.001). Post test comparisons indicated significantly greater latency in

the ATRf/f runners compared to all other treatment groups in the novel environment (Fig.

Page 116: Role Of Hippocampal Neurogenesis In The Etiology And

108

3b, p<0.05). Among ATRΔHipp mice, there was no difference in latency between runners

and their sedentary counterparts in the novel environment (Fig. 3b), indicating ATR

deletion blocked the anxiogenic effects of running on feeding latency.

Running-induced anxiety in an open field is attenuated in ATRΔHipp mice

Voluntary wheel running results in increased anxiety in an open field test, as

measured by reduced time spent in the center zone. This anxiogenic phenotype was

attenuated in ATRΔHipp mice. Two-way ANOVA of time spent in the center zone (Fig. 4a)

revealed a near-significant effect of exercise (F(1,24)=3.67, p=0.067) and a near-

significant exercise x gene interaction (F(1,25)=2.5, p=0.12). Bonferroni post tests to

compare individual treatment groups indicated significantly reduced time spent in the

center zone in ATRf/f runners compared to their sedentary counterparts (-67%,

t(13)=2.445, p<0.05). ATRΔHipp runners did not differ from either sedentary control

group. While the reduced distance traveled in the center zone in ATRf/f runners appears

to be blunted in ATRΔHipp mice, two-way ANOVA of distance traveled in the center zone

(Fig. 4b) revealed significant main effects of exercise (F(1,24)=5.646, p<0.05) and gene

(F(1,24)=5.232, p<0.05), but no exercise x gene interaction (F(1,24)=0.58, p=0.45). Similar

results were obtained when measuring total distance traveled in the open field (Fig. 4c;

exercise main effect F(1,24)=5.75, p<0.05; gene main effect F(1,24)=9.77, p<0.01, exercise

x gene interaction F(1,24)=0.06, p=0.81), suggesting exercise exerts an overall anxiogenic

effect on exploratory behavior while ATR deletion exerts an overall anxiolytic effect on

exploratory behavior in the open field.

Page 117: Role Of Hippocampal Neurogenesis In The Etiology And

109

Running-induced anxiety in the light-dark box test is attenuated in ATRΔHipp

mice

As an additional measure of anxiety, we evaluated behavior in the light-dark box

following voluntary wheel running. Two-way ANOVA revealed a significant main effect of

exercise (F(1,25)=12.82, p<0.01) on light-to-dark transitions (Fig. 5a). There was no

effect of gene (F(1,25)=0.004, p<0.95) and no exercise x gene interaction (F(1,25)=0.19,

p<0.67) on this outcome measure, suggesting the effect of exercise on transitions is not

dependent on a robust stimulation of neurogenesis. Analysis of distance traveled in the

light compartment revealed a trend towards an anxiogenic effect of running which was

blunted in ATRΔHipp mice (Fig. 5b). Two-way ANOVA revealed a near-significant main

effect of exercise (F(1,25)=3.97, p=0.057) and a near-significant exercise x gene

interaction (F(1,25)=3.105, p=0.09). Post tests revealed a significant decrease in distance

traveled in the light compartment in ATRf/f runners compared to their sedentary

counterparts (p<0.05). This effect was not apparent in ATRΔHipp mice. No effects of

running or ATR deletion were observed on time spent in the light compartment (exercise

main effect F(1,25)=0.01, p<0.92; gene main effect F(1,25)=1.14, p=0.30; exercise x gene

interaction F(1,25)=0.001, p=0.97). Reduced distance traveled in the light compartment in

the absence of any changes in total time spent in the compartment may be indicative of

reduced exploration.

Page 118: Role Of Hippocampal Neurogenesis In The Etiology And

110

Figure 1. Experimental design

Figure 1. Experimental design. A Two cohorts of mice were used in this study, the first

for novelty-induced hypophagia and the second for marble burying, open field and light-

dark box tests. Both cohorts were injected with AAV, single-housed approximately 5

weeks later, and then placed in running wheels for 4 weeks. Behavioral testing occurred

in the final week of running. BrdU (200 mg/kg, IP) was injected 24 hours following the

last behavioral test and mice were perfused 24 hours following BrdU injections. B No

significant differences were observed in running distance between ATRf/f and ATRΔHipp

mice. Error bars represent SEM.

Running Distance

1 2 3 40

5

10

15

ATRf/f

ATR!Hipp

Week

km

/da

y

6 weeks 4 weeks

anxiety tests

NIH test

Cohort #1:

Cohort #2:

NIH training

AAV BrdUSingle-housed

text in boxes/large arrows indicate manipulations to both cohorts

Into wheels

Page 119: Role Of Hippocampal Neurogenesis In The Etiology And

111

Figure 2. ATRΔHipp mice exhibit deficits in exercise-induced neurogenesis.

Figure 2. ATRΔHipp mice exhibit deficits in exercise-induced neurogenesis. A Voluntary

exercise resulted in a significant increase in BrdU-expressing cells in ATRf/f mice (***,

p<0.001 vs. ATRf/f-S, ATRΔHipp-S and ATRΔHipp-R groups, n=11-16 per treatment group).

Among sedentary controls, ATR deletion results in significantly reduced BrdU-expressing

cells (#, p<0.05 vs. ATRf/f-S group). B Voluntary exercise resulted in a significant

increase in DCX-expressing cells in ATRf/f mice (***, p<0.001 vs. ATRf/f-S, ATRΔHipp-S

and ATRΔHipp-R groups, n=11-16 per treatment group). In addition, among sedentary

controls, ATR deletion results in reduced DCX-expressing cells (γ, p<0.001 vs. ATRf/f-S

group by Student’s t-test). Error bars represent SEM. Abbreviations: S - sedentary; R -

runner.

Cell Proliferation

0

1000

2000

3000***

#

ATRf/f-S ATRf/f-R ATR!Hipp-S ATR!Hipp-R

Brd

U-IR

ce

lls

A B Immature Neurons

0

10000

20000

30000

***

"

ATRf/f-S ATRf/f-R ATR!Hipp-S ATR!Hipp-R

DC

X-IR

ce

lls

Page 120: Role Of Hippocampal Neurogenesis In The Etiology And

112

Figure 3. Running-induced anxiety in the NIH test is attenuated in ATRΔHipp

mice

Figure 3. Running-induced anxiety in the NIH test is attenuated in ATRΔHipp mice. A No

differences were observed between AAV treatment groups in home cage NIH training

prior to placement in running wheels. B In the home test all mice exhibited comparable

feeding latencies. On the novel test day ATRf/f runners exhibited significantly greater

latency compared to each of the other treatment groups (***, p<0.001 vs. ATRf/f-S,

ATRΔHipp-S and ATRΔHipp-R, n=4-8 per group). Error bars represent SEM. Abbreviations:

S - sedentary; R - runner.

Training

6 7 8 10 11 120

5

10

15

20

ATRf/f

ATR!Hipp

Day of Training

La

ten

cy

(s

ec

.)

Novelty-Induced Hypophagia

Home Novel0

200

400

600

800ATRf/f-S

ATRf/f-R

ATR!Hipp-S

ATR!Hipp-R

***

La

ten

cy

(s

ec

.)

A B

Page 121: Role Of Hippocampal Neurogenesis In The Etiology And

113

Figure 4. Running-induced anxiety in an open field is attenuated in ATRΔHipp

mice.

Time in Center

0

50

100

150

*

ATRf/f-S ATRf/f-R ATR!Hipp-S ATR!Hipp-R

Tim

e (s

ec

.)

Distance Traveled in Center

0

500

1000

1500

2000

2500

ATRf/f-S ATRf/f-R ATR!Hipp-S ATR!Hipp-R

Dis

tan

ce

(m

m)

A

B

C Total Distance Traveled

0

5000

10000

15000

ATRf/f-S ATRf/f-R ATR!Hipp-S ATR!Hipp-R

Dis

tan

ce

(m

m)

Page 122: Role Of Hippocampal Neurogenesis In The Etiology And

114

Figure 4. Running-induced anxiety in an open field is attenuated in ATRΔHipp mice. A

ATRf/f runners spent less time in the center zone compared to their sedentary

counterparts (*, p<0.05 vs. ATRf/f-S). B,C Running exerts a main effect of reduced

distance traveled in the center and reduced total distance traveled compared to

sedentary mice. ATR deletion exerts a main effect of increased distance traveled in the

center zone and increased total distance traveled compared to control ATRf/f mice. There

are no significant differences between individual groups. N=6-8 per group. Error bars

represent SEM. Abbreviations: S - sedentary; R - runner.

Page 123: Role Of Hippocampal Neurogenesis In The Etiology And

115

Figure 5. Running-induced anxiety in the light-dark box is attenuated in

ATRΔHipp mice

Transitions

0

5

10

15

ATRf/f-S ATRf/f-R ATR!Hipp-S ATR!Hipp-R

Tra

ns

itio

ns

Distance in Light Compartment

0

500

1000

1500

2000

**

ATRf/f-S ATRf/f-R ATR!Hipp-S ATR!Hipp-R

Dis

tan

ce

(m

m)

Time in Light Compartment

0

20

40

60

80

100

ATRf/f-S ATRf/f-R ATR!Hipp-S ATR!Hipp-R

Tim

e (s

ec

.)

A

B

C

Page 124: Role Of Hippocampal Neurogenesis In The Etiology And

116

Figure 5. Running-induced anxiety in the light-dark box is attenuated in ATRΔHipp mice.

A Running exerts a main effect of reduced transitions compared to sedentary mice. B

ATRf/f runners explored the light compartment less than their sedentary counterparts

(**, p<0.01 vs. ATRf/f-S), as measured by distance traveled in the light compartment.

This effect is not observed in ATRΔHipp mice. C No differences were observed in total

time spent in the light compartment. N=6-8 per group. Error bars represent SEM.

Abbreviations: S - sedentary; R - runner.

Page 125: Role Of Hippocampal Neurogenesis In The Etiology And

117

Discussion

Exercise is associated with many health benefits in humans, ranging from the

cognitive to the physiological (van Praag, 2008). Preclinical research has established

exercise as a potent enhancer of hippocampal neurogenesis (van Praag et al., 1999),

which may underlie its health benefits. Interestingly, exercise leads to changes in

anxiety-like behavior in rodents. The potential role of neurogenesis in modulating

anxiety-related behaviors following exercise is only now being explored. In this study,

we utilized a transgenic mouse in which Cre recombinase-inducible deletion of ATR from

the hippocampus of adult mice leads to reduced levels of hippocampal neurogenesis.

Our previous characterization of hippocampal ATR deletion examined effects on basal

neurogenesis only. Here, we found that ATR deletion attenuated the neurogenic effect

of wheel running. Additionally, wheel running resulted in heightened anxiety in the

novelty-induced hypophagia, open field, and light-dark box tests. Many of the anxiety

phenotypes induced by wheel running were absent following ATR deletion. While our

findings are in agreement with work by others (Fuss et al., 2010), there are conflicting

reports in the literature regarding effects of exercise on anxiety (Dishman et al., 1996;

Binder et al., 2004; Burghardt et al., 2004; Duman et al., 2008; Salam et al., 2009).

These discrepancies may arise from a multitude of factors including variation in housing

conditions and exercise parameters. However, the most relevant factor is likely

differences in running distance and neurogenic response to running across mouse

strains (Clark et al., 2011), which will subsequently influence behavioral outcome

measures.

We utilized a 4 week, ad libitum access wheel running paradigm, as we

previously observed increased cell proliferation and BDNF mRNA in the hippocampus in

Page 126: Role Of Hippocampal Neurogenesis In The Etiology And

118

naïve ATRf/f mice using this paradigm (J.L.O., unpublished observations). ATR deletion

did not alter running behavior, as measured by running distance across 4 weeks (Fig.

1b). Wheel running exerted a potent stimulatory effect on neurogenesis in ATRf/f mice,

as indicated by increased BrdU and DCX immunostaining to measure cell proliferation

and immature neurons, respectively (Fig. 2). This effect of wheel running was

attenuated in ATRΔHipp mice; levels of neurogenesis in ATRΔHipp runners were

comparable to those of sedentary ATRf/f controls. Our neurogenesis data suggests

ATRΔHipp mice retain some neurogenic capacity in response to a strong stimuli. However,

the overall magnitude of the increase remains well below that which is observed in the

control group, allowing for investigation of the causal role of excessive neurogenesis in

the behavioral changes observed following exercise.

To identify behavioral implications of a blunted neurogenic response to exercise,

we examined the anxiety state of the animals. We first examined behavior in the NIH

paradigm following exercise. Both ATRf/f and ATRΔHipp mice exhibited similar feeding

latencies during the training period, prior to exercise (Fig 3a). In the novel environment,

we observed a large increase in latency to feed in ATRf/f runners compared to sedentary

ATRf/f controls (Fig. 3b). This increase was not apparent in the ATRΔHipp runners,

suggesting heightened neurogenesis may underlie the effect observed in ATRf/f mice.

Because ATR deletion does not influence feeding latency in the home cage test, it is

possible that the role of neurogenesis in exercise-induced anxiety is context-dependent

and is most relevant in novel environments. Previously, we observed a statistically

significant reduction in latency to feed in a novel environment following ATR deletion

(Onksen et al., 2011), whereas here we observe only a trend in the NIH test (Fig. 3b)

and ATR deletion alone does not reduce all measures of anxiety examined in the present

Page 127: Role Of Hippocampal Neurogenesis In The Etiology And

119

study. We hypothesize that the single-housing conditions necessary for the wheel

running studies result in alterations to anxiety state (Kwak et al., 2009) such that certain

differences are less likely to be observed.

In addition to the NIH paradigm, we examined anxiety-like behavior in the open

field and light-dark box tests. In the open field test (Fig. 4), ATRf/f runners spent less

time in the center zone compared to their sedentary counterparts, indicative of

increased anxiety. This effect was not observed in ATRΔHipp runners. When distance

traveled in the open field was analyzed, we found exercise to reduce distance traveled,

indicative of increased anxiety or reduced exploratory behavior. Additionally, ATR

deletion increased distance traveled, indicative of reduced anxiety or increased

exploratory behavior, consistent with our previous work showing reduced anxiety

following ATR deletion (Onksen et al., 2011). In the light-dark box test (Fig. 5), running

resulted in fewer transitions between the compartments. This effect was present in both

ATRf/f and ATRΔHipp runners. Wheel running resulted in reduced distance traveled in the

light compartment in ATRf/f mice. This effect was completely blocked in ATRΔHipp mice,

and was not an effect of total time spent in the light, which was similar across groups.

Among previous reports using the light-dark or dark-light tests, some report changes in

both transitions and time spent in the light compartment as measures of anxiety

(Chaouloff et al., 1997; Frye et al., 2008; Varadarajulu et al., 2011), while others report

one or the other (Binder et al., 2004; Correa et al., 2008; Fuss et al., 2010; Pilhatsch et

al., 2010). Variability may be dependent on differences in mouse strain or parameters of

the testing environment, including size and lighting conditions. Thus, we analyzed

transitions, distance traveled in the light compartment, and time in the light

compartment in our mice.

Page 128: Role Of Hippocampal Neurogenesis In The Etiology And

120

Taken together, this data from multiple anxiety tests suggests mice with

increased neurogenesis exhibit heightened anxiety. This phenotype may also be

interpreted as reduced exploration or slower habituation to a unfamiliar environment.

This alternative explanation is supported by observations of reduced distance traveled in

the open field (Fig. 4) and in the light compartment of the light-dark box (Fig. 5)

following wheel running in ATRf/f mice. Because increased neurogenesis is associated

with heightened cognition in rodent models (Sahay et al., 2011), the phenotype

exhibited by ATRf/f runners may be due to increased awareness of novel surroundings

and subsequent caution in exploring the environment.

In conclusion, we have shown here that hippocampal neurogenesis is an

important determinant of some anxiety-like behaviors in mice. Findings of heightened

anxiety would seem at odds with clinical evidence for the beneficial effects of moderate

exercise. However, the data presented herein, in addition to recent literature

highlighting the prevention of running-induced anxiety by x-ray irradiation (Fuss et al.,

2010), strengthens the hypothesis that neurogenesis directly influences the anxiety-like

behavior induced by running. As further explanation of exercise effects which seem

counterintuitive to the beneficial effects observed in humans, running distances achieved

by rodents may not be representative of the exercise behavior that is beneficial to the

treatment of mood and anxiety disorders in humans and may indicate that too much

neurogenesis can be detrimental to certain behaviors (Saxe et al., 2007). Indeed,

moderate exercise is recommended as therapy for depressed mood in humans (Greer

and Trivedi, 2009), often in conjunction with pharmacotherapy, and excessive exercise

can have negative consequences (Peluso and Guerra de Andrade, 2005; Purvis et al.,

2010; Czepluch et al., 2011). Thus, while rodent models of exercise are a valuable tool

Page 129: Role Of Hippocampal Neurogenesis In The Etiology And

121

for studying neurogenesis, caution must be exercised in applying the results of unlimited

voluntary wheel running to the human population. Future work should focus on

developing rodent models of exercise which accurately mirror moderate exercise in

humans, and on understanding the mechanisms through which newborn neurons in the

hippocampus influence anxiety-like behaviors. This is especially pertinent in the context

of efforts to develop therapeutic compounds that stimulate hippocampal neurogenesis

for the treatment of mood disorders.

Page 130: Role Of Hippocampal Neurogenesis In The Etiology And

122

References

Allen DM, van Praag H, Ray J, Weaver Z, Winrow CJ, Carter TA, Braquet R, Harrington E, Ried T, Brown KD, Gage FH, Barlow C (2001). Ataxia telangiectasia mutated is essential during adult neurogenesis. Genes Dev 15(5): 554-566.

Babyak M, Blumenthal JA, Herman S, Khatri P, Doraiswamy M, Moore K, Craighead WE, Baldewicz TT, Krishnan KR (2000). Exercise treatment for major depression: maintenance of therapeutic benefit at 10 months. Psychosom Med 62(5): 633-638.

Binder E, Droste SK, Ohl F, Reul JM (2004). Regular voluntary exercise reduces anxiety-related behaviour and impulsiveness in mice. Behav Brain Res 155(2): 197-206.

Blumenthal JA, Babyak MA, Moore KA, Craighead WE, Herman S, Khatri P, Waugh R, Napolitano MA, Forman LM, Appelbaum M, Doraiswamy PM, Krishnan KR (1999). Effects of exercise training on older patients with major depression. Arch Intern Med 159(19): 2349-2356.

Brene S, Bjornebekk A, Aberg E, Mathe AA, Olson L, Werme M (2007). Running is rewarding and antidepressive. Physiol Behav 92(1-2): 136-140.

Burghardt PR, Fulk LJ, Hand GA, Wilson MA (2004). The effects of chronic treadmill and wheel running on behavior in rats. Brain Res 1019(1-2): 84-96.

Chaouloff F, Durand M, Mormede P (1997). Anxiety- and activity-related effects of diazepam and chlordiazepoxide in the rat light/dark and dark/light tests. Behav Brain Res 85(1): 27-35.

Clark PJ, Bhattacharya TK, Miller DS, Rhodes JS (2011). Induction of c-Fos, Zif268, and Arc from acute bouts of voluntary wheel running in new and pre-existing adult mouse hippocampal granule neurons. Neuroscience.

Clark PJ, Brzezinska WJ, Puchalski EK, Krone DA, Rhodes JS (2009). Functional analysis of neurovascular adaptations to exercise in the dentate gyrus of young adult mice associated with cognitive gain. Hippocampus 19(10): 937-950.

Clark PJ, Kohman RA, Miller DS, Bhattacharya TK, Brzezinska WJ, Rhodes JS (2011). Genetic influences on exercise-induced adult hippocampal neurogenesis across 12 divergent mouse strains. Genes Brain Behav 10(3): 345-353.

Correa M, Manrique HM, Font L, Escrig MA, Aragon CM (2008). Reduction in the anxiolytic effects of ethanol by centrally formed acetaldehyde: the role of catalase inhibitors and acetaldehyde-sequestering agents. Psychopharmacology (Berl) 200(4): 455-464.

Czepluch FS, Barres R, Caidahl K, Olieslagers S, Krook A, Rickenlund A, Zierath JR, Waltenberger J (2011). Strenuous physical exercise adversely affects monocyte chemotaxis. Thromb Haemost 105(1): 122-130.

Page 131: Role Of Hippocampal Neurogenesis In The Etiology And

123

Deslandes A, Moraes H, Ferreira C, Veiga H, Silveira H, Mouta R, Pompeu FA, Coutinho ES, Laks J (2009). Exercise and mental health: many reasons to move. Neuropsychobiology 59(4): 191-198.

Dishman RK, Dunn AL, Youngstedt SD, Davis JM, Burgess ML, Wilson SP, Wilson MA (1996). Increased open field locomotion and decreased striatal GABAA binding after activity wheel running. Physiol Behav 60(3): 699-705.

Duman CH, Schlesinger L, Russell DS, Duman RS (2008). Voluntary exercise produces antidepressant and anxiolytic behavioral effects in mice. Brain Res 1199: 148-158.

Farmer J, Zhao X, van Praag H, Wodtke K, Gage FH, Christie BR (2004). Effects of voluntary exercise on synaptic plasticity and gene expression in the dentate gyrus of adult male Sprague-Dawley rats in vivo. Neuroscience 124(1): 71-79.

Frye CA, Edinger K, Sumida K (2008). Androgen administration to aged male mice increases anti-anxiety behavior and enhances cognitive performance. Neuropsychopharmacology 33(5): 1049-1061.

Fuss J, Ben Abdallah NM, Hensley FW, Weber KJ, Hellweg R, Gass P (2010). Deletion of running-induced hippocampal neurogenesis by irradiation prevents development of an anxious phenotype in mice. PLoS One 5(9).

Fuss J, Ben Abdallah NM, Vogt MA, Touma C, Pacifici PG, Palme R, Witzemann V, Hellweg R, Gass P (2010). Voluntary exercise induces anxiety-like behavior in adult C57BL/6J mice correlating with hippocampal neurogenesis. Hippocampus 20(3): 364-376.

Greer TL, Trivedi MH (2009). Exercise in the treatment of depression. Curr Psychiatry Rep 11(6): 466-472.

Gur TL, Conti AC, Holden J, Bechtholt AJ, Hill TE, Lucki I, Malberg JE, Blendy JA (2007). cAMP response element-binding protein deficiency allows for increased neurogenesis and a rapid onset of antidepressant response. J Neurosci 27(29): 7860-7868.

Kitamura T, Mishina M, Sugiyama H (2003). Enhancement of neurogenesis by running wheel exercises is suppressed in mice lacking NMDA receptor epsilon 1 subunit. Neurosci Res 47(1): 55-63.

Kronenberg G, Reuter K, Steiner B, Brandt MD, Jessberger S, Yamaguchi M, Kempermann G (2003). Subpopulations of proliferating cells of the adult hippocampus respond differently to physiologic neurogenic stimuli. J Comp Neurol 467(4): 455-463.

Kwak C, Lee SH, Kaang BK (2009). Social Isolation Selectively Increases Anxiety in Mice without Affecting Depression-like Behavior. Korean J Physiol Pharmacol 13(5): 357-360.

Page 132: Role Of Hippocampal Neurogenesis In The Etiology And

124

Neeper SA, Gomez-Pinilla F, Choi J, Cotman CW (1996). Physical activity increases mRNA for brain-derived neurotrophic factor and nerve growth factor in rat brain. Brain Res 726(1-2): 49-56.

Nichol K, Deeny SP, Seif J, Camaclang K, Cotman CW (2009). Exercise improves cognition and hippocampal plasticity in APOE epsilon4 mice. Alzheimers Dement 5(4): 287-294.

Onksen JL, Brown EJ, Blendy JA (2011). Selective Deletion of a Cell Cycle Checkpoint Kinase (ATR) Reduces Neurogenesis and Alters Responses in Rodent Models of Behavioral Affect. Neuropsychopharmacology 36(5): 960-969.

Peluso MA, Guerra de Andrade LH (2005). Physical activity and mental health: the association between exercise and mood. Clinics (Sao Paulo) 60(1): 61-70.

Pilhatsch M, Winter C, Nordstrom K, Vennstrom B, Bauer M, Juckel G (2010). Increased depressive behaviour in mice harboring the mutant thyroid hormone receptor alpha 1. Behav Brain Res 214(2): 187-192.

Purvis D, Gonsalves S, Deuster PA (2010). Physiological and psychological fatigue in extreme conditions: overtraining and elite athletes. PM R 2(5): 442-450.

Rhodes JS, van Praag H, Jeffrey S, Girard I, Mitchell GS, Garland T, Jr., Gage FH (2003). Exercise increases hippocampal neurogenesis to high levels but does not improve spatial learning in mice bred for increased voluntary wheel running. Behav Neurosci 117(5): 1006-1016.

Ruzankina Y, Pinzon-Guzman C, Asare A, Ong T, Pontano L, Cotsarelis G, Zediak VP, Velez M, Bhandoola A, Brown EJ (2007). Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1(1): 113-126.

Sahay A, Scobie KN, Hill AS, O'Carroll CM, Kheirbek MA, Burghardt NS, Fenton AA, Dranovsky A, Hen R (2011). Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature 472(7344): 466-470.

Sahay A, Scobie KN, Hill AS, O'Carroll CM, Kheirbek MA, Burghardt NS, Fenton AA, Dranovsky A, Hen R (2011). Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature.

Salam JN, Fox JH, Detroy EM, Guignon MH, Wohl DF, Falls WA (2009). Voluntary exercise in C57 mice is anxiolytic across several measures of anxiety. Behav Brain Res 197(1): 31-40.

Saxe MD, Malleret G, Vronskaya S, Mendez I, Garcia AD, Sofroniew MV, Kandel ER, Hen R (2007). Paradoxical influence of hippocampal neurogenesis on working memory. Proc Natl Acad Sci U S A 104(11): 4642-4646.

Page 133: Role Of Hippocampal Neurogenesis In The Etiology And

125

Surget A, Tanti A, Leonardo ED, Laugeray A, Rainer Q, Touma C, Palme R, Griebel G, Ibarguen-Vargas Y, Hen R, Belzung C (2011). Antidepressants recruit new neurons to improve stress response regulation. Mol Psychiatry.

Tillerson JL, Caudle WM, Reveron ME, Miller GW (2003). Exercise induces behavioral recovery and attenuates neurochemical deficits in rodent models of Parkinson's disease. Neuroscience 119(3): 899-911.

Van der Borght K, Havekes R, Bos T, Eggen BJ, Van der Zee EA (2007). Exercise improves memory acquisition and retrieval in the Y-maze task: relationship with hippocampal neurogenesis. Behav Neurosci 121(2): 324-334.

Van der Borght K, Kobor-Nyakas DE, Klauke K, Eggen BJ, Nyakas C, Van der Zee EA, Meerlo P (2009). Physical exercise leads to rapid adaptations in hippocampal vasculature: temporal dynamics and relationship to cell proliferation and neurogenesis. Hippocampus 19(10): 928-936.

van Praag H (2008). Neurogenesis and exercise: past and future directions. Neuromolecular Med 10(2): 128-140.

van Praag H, Christie BR, Sejnowski TJ, Gage FH (1999). Running enhances neurogenesis, learning, and long-term potentiation in mice. Proc Natl Acad Sci U S A 96(23): 13427-13431.

van Praag H, Kempermann G, Gage FH (1999). Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci 2(3): 266-270.

Varadarajulu J, Lebar M, Krishnamoorthy G, Habelt S, Lu J, Bernard Weinstein I, Li H, Holsboer F, Turck CW, Touma C (2011). Increased anxiety-related behaviour in Hint1 knockout mice. Behav Brain Res 220(2): 305-311.

West MJ, Slomianka L, Gundersen HJ (1991). Unbiased stereological estimation of the total number of neurons in thesubdivisions of the rat hippocampus using the optical fractionator. Anat Rec 231(4): 482-497.

Winter B, Breitenstein C, Mooren FC, Voelker K, Fobker M, Lechtermann A, Krueger K, Fromme A, Korsukewitz C, Floel A, Knecht S (2007). High impact running improves learning. Neurobiol Learn Mem 87(4): 597-609.

Page 134: Role Of Hippocampal Neurogenesis In The Etiology And

126

CHAPTER V

CONCLUSIONS AND FUTURE DIRECTIONS

The overall aim of this dissertation was to examine the role of hippocampal

neurogenesis in the etiology and treatment of mood and anxiety disorders. Post-mortem

and fMRI studies of depressed patients’ brains suggest the hippocampus is relevant to

these mental illnesses (Webster et al., 2002; Gaughran et al., 2006; Lorenzetti et al.,

2009) and the process of neurogenesis within the dentate gyrus of the hippocampus is

of particular interest. However, current methods utilized to study hippocampal

neurogenesis in rodents are invasive and cannot be employed in depressed patients.

This limitation has led the field to rely heavily on rodent models. Our examination of the

influence of neurogenesis in mood disorders was accomplished through a transgenic

mouse model of suppressed hippocampal neurogenesis. ATR deletion from the adult

mouse was previously shown to induce aging-like phenotypes due to a depletion of stem

and progenitor cells (Ruzankina et al., 2007); however a specific effect of ATR deletion

on neurogenesis in the dentate gyrus of the hippocampus was not previously evaluated.

We demonstrated a partial suppression of hippocampal neurogenesis following ATR

deletion from the hippocampus of the adult mouse. Spatial and temporal regulation of

ATR deletion was accomplished through hippocampal microinjection of an AAV

expressing Cre-recombinase.

Other approaches to inhibiting hippocampal neurogenesis, including x-ray

irradiation, produce a near-complete ablation of new neurons, and are thus valuable for

evaluating the function of new neurons in certain cases. While the dynamics of

hippocampal neurogenesis are poorly understood in humans, the partial suppression

Page 135: Role Of Hippocampal Neurogenesis In The Etiology And

127

observed following ATR deletion may be more representative of a pathological state

than a complete ablation would be. Using ATR deletion to suppress hippocampal

neurogenesis, we found changes in behaviors representative of mood and anxiety

disorders and alterations in response to antidepressant treatment. These insights are a

valuable contribution to the field of hippocampal neurogenesis and psychiatric illness.

The following paragraphs contain a more extensive discussion of the findings and

implications of this dissertation, and highlight areas of interest for future investigation.

In characterizing the cellular effects of hippocampal ATR deletion, we observed

an approximately 50% reduction in cell proliferation and in the number of immature

neurons present 8 weeks following AAV injection. This partial suppression is in contrast

to the near-complete ablation that can be achieved with other methods. The partial

effect observed here may be due to the affinity of the AAV to neural stem/progenitor

cells. A future study using either an alternative viral approach or a transgenic approach,

such as Cre recombinase expression through the Nestin promoter, would shed light on

whether ATR deletion is a viable approach for obtaining an even greater suppression of

neurogenesis. Subsequently, one could compare the behavioral effects of partial and

complete suppression within the same transgenic line.

ATR deletion resulted in reduced anxiety in the novelty-induced hypophagia,

marble burying, and zero maze tests. These observations are at odds with some studies

reporting increased anxiety in mice with reduced neurogenesis (Revest et al., 2009;

Scobie et al., 2009; Conboy et al., 2010). However, our observation of reduced anxiety

makes sense in the context of models in which heightened anxiety is associated with

heightened neurogenesis (Gur et al., 2007; Fuss et al., 2010), including our own work

highlighting the anxiogenic effect of exercise and blockade of this effect in ATR-deleted

Page 136: Role Of Hippocampal Neurogenesis In The Etiology And

128

mice. In addition, hippocampal lesion studies have reported reduced anxiety in social,

exploratory, and novelty tasks (Bannerman et al., 2002). As anxiety can be parsed into

these various aforementioned categories, a more extensive analysis of which types of

anxiety measures are affected by ATR deletion may improve understanding of how

neurogenesis modulates anxiety.

We observed a lack of antidepressant response in the NIH paradigm following

ATR deletion, similar to work by others suggesting antidepressants work though

stimulating neurogenesis (Santarelli et al., 2003). However, in our hands we did not

observe an effect of DMI on cell proliferation in control animals and did find the effects

of DMI on CA1 dendritic spine density to be attenuated in ATR-deleted mice, indicative

of a blunted capacity for synaptic plasticity. Work by others has indicated that neuronal

remodeling is the relevant underlying factor in antidepressant efficacy, rather than

neurogenesis (Bessa et al., 2009). It is possible that these two phenomena,

neurogenesis and neuronal plasticity/remodeling, are intimately connected. Future

studies to examine broad hippocampal circuitry function in ATR-deleted mice, using

methods such as voltage-sensitive dye imaging and electrophysiology, may shed light on

whether a partial suppression of neurogenesis can alter homeostatic functioning of the

hippocampus, as was demonstrated following x-ray irradiation to ablate neurogenesis

(Lacefield et al., 2010). In addition, we examined only one antidepressant, DMI, a

tricyclic which mainly inhibits norepinephrine reuptake and, to a lesser extent, serotonin

reuptake. Antidepressants with more potent effects on serotonergic signaling, as well as

antidepressants with non-monoaminergic mechanisms, may exhibit a differential

requirement for hippocampal neurogenesis in modulating hyponeophagia behavior.

Page 137: Role Of Hippocampal Neurogenesis In The Etiology And

129

While many reports argue that reduced hippocampal neurogenesis is not

sufficient to induce depressive-like behaviors, we were compelled to consider the

potential for depressive-like phenotypes, as ATR deletion is a novel approach to

inhibiting neurogenesis and results in a unique, partial suppression. We observed

anhedonia in male ATR-deleted mice, an effect that was absent in females. Additional

research should exclude possible metabolic differences after ATR deletion, as well as

rule out the possibility that single-housing played a role in the appearance of the

anhedonic phenotype. Future studies to delve further into the sex-specific effect might

include ovariectomizing females to determine the contribution of sex hormones to the

females’ resiliency to anhedonia in the sucrose drinking task. Subsequent to observing

an anhedonic phenotype in males, we explored the possibility that they would be more

susceptible to helplessness in the learned helplessness paradigm. The proportion of

ATR-deleted mice which developed helplessness did not differ from that of control mice.

In addition, the average escape latency in avoidance trials following inescapable shock

was not affected by ATR deletion. Finally, in the learned helplessness studies, we did not

observe any correlation between escape latency and cell genesis, indicating

neurogenesis does not influence the extent of learned helplessness or the likelihood that

a particular mouse will be susceptible.

As stress is a major risk factor for depression, we examined whether ATR

deletion would potentiate the effects of chronic corticosterone treatment. The

consequences of this treatment, thought to mirror some of the effects of chronic stress,

were not exacerbated by ATR deletion. Low dose corticosterone administered through a

21-day sustained-release pellet (20 mg/kg/day), to mimic chronic mild stress, resulted in

increased immobility in the forced swim test and reduced corticosterone response to an

Page 138: Role Of Hippocampal Neurogenesis In The Etiology And

130

acute stressor. No interaction between the corticosterone treatment and ATR deletion

was observed. The restraint stress study was performed in males only. As sex

differences were observed in the sucrose drinking task, it is possible that males and

females are differentially susceptible to certain depressive-like phenotypes. Implanting

corticosterone pellets in females and subsequently measuring response to an acute

stressor might thus yield a potentiating effect of ATR deletion in females, even though it

is not observed in the males.

In addition to examining baseline behaviors following ATR deletion, we examined

a stimuli which normally exerts a potent neurogenic effect in rodents, voluntary wheel

running. Our interest in wheel running stems from two sources. First, wheel running

exerts a potent influence on neurogenesis and alters anxiety state. Second, in humans

exercise is recommended as a treatment for mood disorders, often in conjunction with

pharmacotherapy. These two points present an interesting contrast; a stimulus which

improves mood clinically can increase anxiety in pre-clinical models, an effect

contradictory to the treatment of psychiatric disease. Further, in the NIH test, chronic

wheel running in place of chronic antidepressant treatment does not result in the

antidepressant-like effect of reduced hyponeophagia. One significant difference between

voluntary wheel running in rodents and voluntary exercise in humans is the extent of the

exercise. Mice can run more than 10 km/day when given unlimited wheel access;

accounting for the difference in size between a mouse and human, this is significantly

farther than a human runs in a given day when partaking in moderate, voluntary

exercise. Thus, the rodent model of ad libitum access to voluntary wheel running may

not be the ideal model for understanding precisely the mechanisms through which

Page 139: Role Of Hippocampal Neurogenesis In The Etiology And

131

exercise improves mood in humans. Nonetheless, wheel running is a valuable tool for

understanding how neurogenesis influences anxiety.

The potent neurogenic effect of wheel running is attenuated in ATR-deleted

mice, such that ATR-deleted runners exhibit levels of cell proliferation and neurogenesis

similar to a sedentary control animal. Additionally, wheel running resulted in anxiety in

the NIH paradigm, marble burying test, open field, and light-dark box, in agreement

with work by others (Burghardt et al., 2004; Fuss et al., 2010b; Fuss et al., 2010). In

some of our outcome measures, ATR deletion blocked the anxiogenic effect of running.

In addition to increased anxiety, the behavior observed following exercise is reminiscent

of reduced exploration and slowed habituation to novel situations. As increased

neurogenesis is often associated with heightened cognition, the anxiety phenotype

observed in control runners and blocked in ATR-deleted runners may be due to

increased alertness and awareness in a novel environment. Additional studies should be

designed to parse these potentially independent phenomena, pure anxiety versus

heightened alertness and caution in response to novel surroundings in mice with

elevated hippocampal neurogenesis.

In conclusion, our studies to examine mood and anxiety disorders in the ATR

transgenic mouse support a complex and essential role for hippocampal neurogenesis in

these psychiatric diseases. The high prevalence of these disorders and lack of optimal

treatment options support the continued exploration and characterization of novel drug

development targets, such as hippocampal neurogenesis, in hope of identifying

compounds which improve symptoms with greater efficacy and fewer unwanted side

effects.

Page 140: Role Of Hippocampal Neurogenesis In The Etiology And

132

References Bannerman DM, Deacon RM, Offen S, Friswell J, Grubb M, Rawlins JN (2002). Double

dissociation of function within the hippocampus: spatial memory and hyponeophagia. Behav Neurosci 116(5): 884-901.

Bessa JM, Ferreira D, Melo I, Marques F, Cerqueira JJ, Palha JA, Almeida OF, Sousa N (2009). The mood-improving actions of antidepressants do not depend on neurogenesis but are associated with neuronal remodeling. Mol Psychiatry 14(8): 764-773, 739.

Burghardt PR, Fulk LJ, Hand GA, Wilson MA (2004). The effects of chronic treadmill and wheel running on behavior in rats. Brain Res 1019(1-2): 84-96.

Conboy L, Varea E, Castro JE, Sakouhi-Ouertatani H, Calandra T, Lashuel HA, Sandi C (2010). Macrophage migration inhibitory factor is critically involved in basal and fluoxetine-stimulated adult hippocampal cell proliferation and in anxiety, depression, and memory-related behaviors. Mol Psychiatry.

Fuss J, Ben Abdallah NM, Hensley FW, Weber KJ, Hellweg R, Gass P (2010). Deletion of running-induced hippocampal neurogenesis by irradiation prevents development of an anxious phenotype in mice. PLoS One 5(9).

Fuss J, Ben Abdallah NM, Vogt MA, Touma C, Pacifici PG, Palme R, Witzemann V, Hellweg R, Gass P (2010). Voluntary exercise induces anxiety-like behavior in adult C57BL/6J mice correlating with hippocampal neurogenesis. Hippocampus 20(3): 364-376.

Gaughran F, Payne J, Sedgwick PM, Cotter D, Berry M (2006). Hippocampal FGF-2 and FGFR1 mRNA expression in major depression, schizophrenia and bipolar disorder. Brain Res Bull 70(3): 221-227.

Gur TL, Conti AC, Holden J, Bechtholt AJ, Hill TE, Lucki I, Malberg JE, Blendy JA (2007). cAMP response element-binding protein deficiency allows for increased neurogenesis and a rapid onset of antidepressant response. J Neurosci 27(29): 7860-7868.

Lacefield CO, Itskov V, Reardon T, Hen R, Gordon JA (2010). Effects of adult-generated granule cells on coordinated network activity in the dentate gyrus. Hippocampus.

Lorenzetti V, Allen NB, Fornito A, Yucel M (2009). Structural brain abnormalities in major depressive disorder: a selective review of recent MRI studies. J Affect Disord 117(1-2): 1-17.

Revest JM, Dupret D, Koehl M, Funk-Reiter C, Grosjean N, Piazza PV, Abrous DN (2009). Adult hippocampal neurogenesis is involved in anxiety-related behaviors. Mol Psychiatry 14(10): 959-967.

Page 141: Role Of Hippocampal Neurogenesis In The Etiology And

133

Ruzankina Y, Pinzon-Guzman C, Asare A, Ong T, Pontano L, Cotsarelis G, Zediak VP, Velez M, Bhandoola A, Brown EJ (2007). Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1(1): 113-126.

Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen R (2003). Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 301(5634): 805-809.

Scobie KN, Hall BJ, Wilke SA, Klemenhagen KC, Fujii-Kuriyama Y, Ghosh A, Hen R, Sahay A (2009). Kruppel-like factor 9 is necessary for late-phase neuronal maturation in the developing dentate gyrus and during adult hippocampal neurogenesis. J Neurosci 29(31): 9875-9887.

Webster MJ, Knable MB, O'Grady J, Orthmann J, Weickert CS (2002). Regional specificity of brain glucocorticoid receptor mRNA alterations in subjects with schizophrenia and mood disorders. Mol Psychiatry 7(9): 985-994, 924.