+ All Categories
Home > Documents > The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune...

The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune...

Date post: 23-Jun-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
107
The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders by Cong Yang (Ingrid) Xuan A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Pharmaceutical Sciences University of Toronto © Copyright by Cong Yang (Ingrid) Xuan, 2013
Transcript
Page 1: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders

by

Cong Yang (Ingrid) Xuan

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

Graduate Department of Pharmaceutical Sciences University of Toronto

© Copyright by Cong Yang (Ingrid) Xuan, 2013

Page 2: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

ii

The Maternal Immune Activation Mouse Model of Autism

Spectrum Disorders

Cong Yang (Ingrid) Xuan

Master of Science

Department of Pharmaceutical Sciences

University of Toronto

2013

Abstract

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments

in social interaction and communication as well as ritualistic repetitive behaviors.

Epidemiological studies suggest that maternal immune activation (MIA) during pregnancy may

be a risk factor for ASD. To study MIA in a laboratory setting, we injected mouse dams

(C57BL/6) with lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (Poly IC) during

mid-gestation to mimic a bacterial or viral infection, respectively. We also performed the same

Poly IC treatment on a mouse model of Fragile X syndrome (i.e. Fmr1 knockout), a genetic

disease with high incidences of ASD. We found modest female-specific impairments in social

interaction and striking male-specific increases in repetitive behavior in adult MIA offspring.

Moreover, prenatal Poly IC treatment caused genotype-specific deficits in sociability in addition

to reduced body weight and rearing in Fmr1 knockout mice only. Therefore, ASD-related

behaviors caused by MIA may be sex, treatment, and/or genotype-dependent.

Page 3: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

iii

Acknowledgments

I would like to express my deepest appreciation for my supervisor Dr. David Hampson for his

helpful guidance and support during this project as well as all the current and past lab members.

I would like to thank my committee members, Dr. Peter Wells and Dr. Paul Frankland for

sharing their expertise during my committee meetings. Lastly, I am extremely grateful to Dr.

Ramsey and her lab members for their generosity with the behavior equipment.

Page 4: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

iv

Table of Contents

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

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

List of Figures ................................................................................................................................ vi

List of Appendices ........................................................................................................................ vii

List of Abbreviations ................................................................................................................... viii

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

1.1 Autism Spectrum Disorder in Human ................................................................................. 1

1.2 Etiology of ASD: Genetic Makeup ..................................................................................... 6

1.2.1 Fragile X Syndrome ................................................................................................ 7

1.3 Etiology of ASD: Immune Activation ................................................................................ 7

1.3.1 Evidence Based on Epidemiological Studies .......................................................... 7

1.3.2 Evidence Based on Human Pathological and Biochemical Studies ....................... 8

1.4 Maternal Immune Activation: Possible Mechanism in ASD Patients .............................. 10

1.5 Modeling ASD in Rodents Based on the MIA Hypothesis .............................................. 11

2 Rational, Hypotheses and Objectives ....................................................................................... 14

2.1 Rational for MIA Mouse Model ....................................................................................... 14

2.2 Hypotheses and Objectives ............................................................................................... 14

3 Methods .................................................................................................................................... 17

3.1 Animals ............................................................................................................................. 17

3.2 Motor Activity .................................................................................................................. 20

3.3 Social Interaction and Social Preference Test .................................................................. 20

3.4 Marble Burying Test ......................................................................................................... 22

3.5 Grooming .......................................................................................................................... 22

Page 5: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

v

3.6 Western Blot ..................................................................................................................... 23

3.7 Statistical Analysis ............................................................................................................ 24

4 Results ...................................................................................................................................... 25

4.1 The Effect of MIA on Body Weight ................................................................................. 25

4.2 The effect of MIA on Motor Activity ............................................................................... 25

4.3 The Effect of MIA on Social Behavior ............................................................................. 28

4.4 The Effect of MIA on Repetitive Behavior Evaluated Using Marble Burying Test ........ 36

4.5 The Effect of MIA on Repetitive Behavior Evaluated by Grooming ............................... 38

4.6 The Effect of MIA on the Adult Cerebellum Evaluated by Western Blot ........................ 38

5 Discussion ................................................................................................................................ 41

5.1 The Effect of MIA on Body Weight ................................................................................. 41

5.2 The Effect of MIA on Motor activity ............................................................................... 42

5.3 The Effect of MIA on Social behavior ............................................................................. 43

5.4 The Effect of MIA on Ritualistic and Repetitive Behavior .............................................. 46

5.5 The Effect of MIA on the Adult Cerebellum .................................................................... 49

5.6 Potential Biological Mechanisms for Sex-Dependent Behavioral Changes ..................... 50

6 Conclusions .............................................................................................................................. 52

7 Future Directions ...................................................................................................................... 53

References ..................................................................................................................................... 55

Appendices .................................................................................................................................... 82

Page 6: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

vi

List of Figures

Fig. 1 Injection schedule of MIA dams.. ...................................................................................... 18

Fig. 2 Translating human ASD behaviors to mouse model of ASD.. ........................................... 19

Fig. 3 The schedule of behavioral and biochemical analysis of MIA offspring. .......................... 20

Fig. 4 Body weight of wild-type and Fmr1 knockout MIA offspring on PND46 ....................... 26

Fig. 5 Motor activity of LPS wild-type offspring.. ....................................................................... 27

Fig. 6 Motor activity of Poly IC wild-type offspring .................................................................... 29

Fig. 7 Motor activity of Poly IC Fmr1 knockout offspring.. ........................................................ 30

Fig. 8 Social behavior of LPS treated wild-type MIA offspring evaluated by the modified 3-

chamber paradigm. ........................................................................................................................ 32

Fig. 9 Social behavior of Poly IC treated wild-type MIA offspring evaluated by the modified 3-

chamber paradigm.. ....................................................................................................................... 33

Fig. 10 Social behavior of Poly IC treated Fmr1 knockout MIA offspring evaluated by the

modified 3-chamber paradigm.. .................................................................................................... 35

Fig. 11 Repetitive behavior of wild-type and Fmr1 knockout MIA offspring evaluated by the

marble burying test. ...................................................................................................................... 37

Figure 12 Repetitive behavior of MIA wild-type and Fmr1 knockout MIA offspring evaluated by

grooming behavior ........................................................................................................................ 39

Fig. 13 Summary of the expression of different glial markers in the adult cerebellum of LPS

offspring quantified by western blot ............................................................................................. 40

Page 7: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

vii

List of Appendices

Appendix 1 Comparison between computer and manual scoring of social interaction of LPS

treated wild-type MIA female offspring evaluated by the modified 3-chamber paradigm... ……82

Appendix 2 Summary of behavioral findings in wild-type MIA offspring and Fmr1 knockout

MIA offspring.. ............................................................................................................................. 83

Appendix 3 Summary of literature research regarding Poly IC MIA rodent model……………..85

Appendix 4 Summary of literature research regarding LPS MIA rodent model………………...92

Page 8: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

viii

List of Abbreviations

1X: immunologically “naïve” dams that were only injected once with either Poly IC or LPS

2X: dams that have been injected with the same agent prior to second injection of Poly IC or LPS

ADHD: attention deficit hyperactivity disorder

ADI-R: Autistic Diagnostic Interview, Revised

Akt (or PKB): protein kinase B

ASD: autism spectrum disorder

ASSQ: autism spectrum screening questionnaire

CDC: Center for Disease Control

CNS: central nervous system

DSM: Diagnostic and Statistical Manual of Mental Disorder

Fmr1: Fragile X mental retardation 1 gene

FMRP: Fragile X mental retardation protein

FXS: Fragile X syndrome

GABRB3: gamma-aminobutyric acid receptor subunit beta-3

GAD: glutamate decarboxylase

GAPDH: anti-glyceraldehyde 3-phosphate-dehydrogenase

GFAP: Glial fibrillary acidic protein

Iba-1: ionized calcium-binding adapter molecule 1

IL: interleukin

INF-γ: interferon- γ IP-10: Interferon gamma-induced protein 10

KO: knockout

LPS: lipopolysaccharide

mTOR: mammalian target of rapamycin

MAPK: mitogen-activated protein kinase

MBP: myelin basic protein

MCP-1: monocyte chemoattractant protein-1

MeCP2: methyl CpG binding protein 2

MHC1: major histocompatibility complex 1

MIA: maternal immune activation

Page 9: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

ix

MRI: magnetic resonance imaging

mTORC1: mammalian target of rapamycin complex 1

NO: nitric oxide

PCs: Purkinje cells

PND: post-natal day

Poly IC or Poly: Polyinosinic-polycytidylic acid

PPI: prepulse inhibition

RANTES: Regulated on Activation, Normal T cell Expressed and Secreted

RELN: reelin

SH3: SRC homology 3

SHANK1: multiple ankyrin repeat domains

STAT: screening tool for autism in two-year-olds

Tsc: tuberous sclerosis complex

TGF-β: transforming growth factor-β

TLR: toll-like receptor

TNF-α: tumour necrosis factor-α

WT: wild-type

Page 10: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

1

1 Introduction

1.1 Autism Spectrum Disorder in Human

Autism spectrum disorder (ASD) is a behaviorally defined neurodevelopmental disorder that is

estimated to affect 1 in 88 children in the United States (Center for Disease Control, 2012). The

5th

edition of Diagnostic and Statistical Manual of Mental Disorder (DSM), released in 2013,

requires four criteria to be met for an ASD diagnosis, they are as follows: 1) early childhood

presentation of the symptoms; 2) impaired social communication and interaction; 3) ritualistic

repetitive behavior; 4) symptoms limit everyday functioning.

With regards to the first criterion, ASD is generally diagnosed before the age of two via two

stages (CDC, 2012). The first stage involves screening for developmental problems around 18

months. This is usually conducted by pediatricians in the form of questionnaires such as

checklist of autism in toddlers, screening tool for autism in two-year-olds, and autism spectrum

screening questionnaire. If problems arise, the child would undergo more comprehensive

evaluations to identify the possible origins of the symptoms. These evaluations include genetic

testing, hearing testing, and toxin screening (CDC, 2010).

Autism is diagnosed four times more frequently in males than in females (National Institute of

Health, 2013). The male bias is even stronger in Asperger syndrome, a form of ASD, where the

ratio of male : female is between 9 : 1 and 11 : 1 (Gillberg et al., 2006). Several theories have

been hypothesized with regards to this phenomenon (Baron-Cohen et al., 2011). The first theory

is the natural sexual dimorphism in the human brain during early development. Infant females

tend to have smaller total brain volume and amygdala size than average corresponding males

(Gilmore et al., 2007, Good et al., 2001). This increase in size is further exacerbated in ASD

patients (Schumann et al., 2004, Schumann et al., 2010). The second theory suggests that the

gender bias is due to genetic predisposition. A small percentage of ASDs is caused by X-linked

mutations. This means females will be naturally protected if the activated X-chromosome does

not have the mutation(s). The third theory suggests that the high prevalence of males in ASD

could be caused by elevated fetal testosterone levels. In typically developing children, autistic

traits measured by Childhood Autism Spectrum Test, Child Autism Spectrum Quotient, and

Quantitative Checklist for Autism in Toddlers were positively associated with fetal testosterone

Page 11: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

2

levels (Auyeung et al., 2009, Auyeung et al., 2010). In congenital adrenal hyperplasia, female

patients are exposed to high levels of androgens such as testosterone during most of gestation.

Knickmeyer et al. 2006 found that female patients with congenital adrenal hyperplasia had

higher Autism Spectrum Quotient than controls suggesting that the high fetal testosterone

exposure may be responsible for this outcome. Regardless of the gender-bias in ASD, the core

symptoms are the same for both male and female patients.

One of the core symptoms of ASD is deficits in social-emotional reciprocity, impaired

verbal/nonverbal communication during social interaction, and difficulty developing and

maintaining relationships appropriate for their developmental level. Specific behaviors include

lack of reciprocal interaction, gaze aversion and inability to initiate conversation in social

settings. With regards to repetitive behavior or stereotypies, ASD patients must demonstrate at

least two of the following behaviors: repetitive speech or motor movements, insistence on

sameness, fixation of a particular interest, and hyper- or hypo-reactivity to sensory input

(Tchaconas and Adesman, 2013).

Apart from the core symptoms, there are several additional associated conditions of ASD that are

correlated with each other and vary greatly in their severity. For example, some ASD patients

experience heightened or reduced sensitivity to sensory stimuli such as sound and temperature.

Interestingly, behavioral responses to sensory stimuli, such as electrodermal reactivity to tone

(78 decibels), were able to accurately predict sleeping dysfunction in ASD patients (Reynolds et

al., 2012). Furthermore, Baranek et al., 2006 reported that sensory symptoms were inversely

related to the mental age of ASD patients. The level of impairment for specific symptoms also

varies depending on the individual. For instance, in a recent cohort study conducted with 156

ASD patients between ages of 10 and 14, 55% had intellectual disability (IQ < 70), 17% had

below average intelligence (IQ 70-84), 25% had average intelligence (IQ 85-114), and 3% had

above average intelligence (IQ > 115) (Charman, 2011). Other associated symptoms include

seizure, motor impairment, and increased anxiety (Matson et al., 2007, Maski et al., 2011).

Overall, the behavioral impairments of ASD patients are highly variable and difficult to

delineate. This propelled DSM-V to merge the distinct disorders of ASD (with the exception of

Rett syndrome) and reclassify this disorder based on level of severity.

Page 12: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

3

There is some evidence suggesting that the severity of the impairments may be higher in second

born siblings compare to first siblings. Martin et al. 2012 showed in a large study (n > 300) that

second siblings demonstrated a decline in nonverbal and verbal IQ scores and increase in autism

severity via social responsiveness scale. This result was reinforced in a separate study where

later-born siblings of children with ASD were at elevated risk of social impairments (Yoder et

al., 2009). In contrast, the repetitive behavior, measured by Autism Diagnostic Interview-

Revised (ADI-R), was more severe in first born siblings compared to second born siblings

(Reichenberg et al., 2007).

To better understand the origin of behavioral abnormalities, scientists investigated the brains of

ASD patients using magnetic resonance imaging (MRI) and biochemical analysis of post-mortem

brain tissues. The cerebellum in particular, has been consistently found to be implicated in the

neuropathology of ASD. Although this brain region is generally associated with motor

coordination, it is also involved in cognitive functions due to its interconnections with other brain

regions such as the cerebral cortex (Fatemi et al., 2012; Broussard, 2013). In a study with 20

cases where the disease was confined to the cerebellum, patients demonstrated impaired

executive functions, personality changes, language deficits, and difficulties with spatial cognition

(Schmahmann and Sherman, 1998). Similarly, in a study consisting of 27 patients with

cerebellar malformations, 74% of patients presented with some degree of mental retardation.

Specifically, patients with cerebellar vermal agenesis or diffuse cerebellar hypoplasia presented

core ASD symptoms including language deficits, social interaction impairments, and some

repetitive and stereotyped behavior. Interestingly, motor deficits in these patients were less

severe (Tavano et al., 2007). These studies highlight the multitude of brain functions that the

cerebellum is involved in.

Structural MRI analyses of autistic children between ages of two and four revealed significant

increases in whole cerebellum volume and cerebellar white matter volume but no change in gray

matter volume (Courchesne et al., 2001, Sparks et al., 2002). However, this overgrowth subsided

by the time the children reached adolescence (Courchesne et al., 2004). The abnormal brain

growth may be partially responsible for the behavioral impairments observed in ASD patients

since the period of overgrowth coincided with the onset of symptoms. In addition, several

groups demonstrated decreases in the size of vermis lobules VI and VII (Akshoomoff et al.,

2004, Kaufmann et al., 2003, Stanfield et al., 2008). This region is involved in oculomotor

Page 13: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

4

functions which may be impaired in autism (Takarae et al., 2004). As well, hypoplasia in these

lobules was found to be correlated with decreased exploration and higher rate of stereotyped

behaviors in ASD patients (Pierce and Courchesne, 2001). Interestingly, diffusion tensor

imaging showed increases in axonal integrity in cerebellar white matter regions between the

fastigium of the fourth ventricle and parts of vermis lobules V and VIII which are in close

proximity to lobule VI and VII respectively. Noriuchi et al. 2010 suggested that this may be a

compensatory mechanism for the functional deficits linked with lobule VI and VII.

To understand the neurobiological basis behind these changes in the cerebellar anatomy, analyses

were conducted on post-mortem brain tissues of ASD patients. One of the most reproducible

findings was significant decreases in the number and/or size of Purkinje cells (PCs) (Ritvo et al.,

1986, Kemper and Bauman, 1993, Bailey et al., 1998, Fatemi et al., 2002). It should be noted

that abnormalities in PCs were not observed in all ASD patients, rather, a subpopulation. Using

stereological method, Whitney et al. 2008 showed that only 3 out of 6 ASD patients presented

lower density of PCs compared to controls. The disparity may be due to the heterogeneity of

patient history, small sample size, and method of tissue staining. In addition, the lack of change

in PC density does not exclude the possible changes in its physiology. In fact, Yip et al., 2007

found a 40% reduction in glutamate decarboxylase (GAD) 67 mRNA in PCs based on 8 adult

autistic cases with matched controls.

PCs are the major GABAergic neurons that send inhibitory projections to the deep cerebellar

nuclei. Physical or functional vulnerability of these cells may result in behavioral abnormalities

associated with the cerebellum. Patients with tuberous sclerosis, a genetic disorder that has high

comorbidity with ASD, demonstrated cerebellar pathology that was positively correlated with the

severity of ASD symptoms (Ertan et al., 2010, Weber et al., 2000, Eluvathingal et al., 2006). In

rodents, loss of tuberous sclerosis complex (Tsc) 1 or Tsc2 in PCs both resulted in PC

degeneration that caused ASD-like behaviors including social impairment, increased repetitive

behavior and abnormal vocalization (Tsai et al., 2012, Reith et al., 2013). Similarly, lurcher

mice, which exhibit depletion of PCs during development, displayed ASD characteristics

including increases in repetitive behavior and deficits during reversal learning tasks (Martin et

al., 2010, Dickson et al., 2010). Therefore, abnormalities in PCs may contribute to ASD

symptomology.

Page 14: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

5

In addition to the cerebellum, pathologies in other brain regions may also contribute to the

development of ASD. One such region is the amygdala which is part of the limbic system

responsible for emotion and memory (Phelps, 2006). Similar to the growth progression of the

cerebellum, the amygdala was also found to be enlarged during early childhood compared to

controls but the growth subsided during adolescence and thereafter (Sparks et al., 2002,

Schumann et al., 2004). More importantly, in ASD children between age 3 and 7, this

enlargement was associated with deficits in social and communication skills (Munson et al.,

2006, Kim et al., 2010). In a small study of 9 cases, autistic patients had significantly fewer

neurons in the amygdala than respective controls (Schumann and Amaral, 2006). Moreover,

quantitative MRI revealed that the grey matter volume was significantly increased in the anterior

temporal and dorsolateral prefrontal region and reduced in the occipital and medial parietal

regions of ASD patients compared to controls. These changes were significantly correlated with

the severity of ASD symptoms measured by ADI-R in the social, communication, and/or

repetitive behavior domain (Ecker et al., 2012). To summarize, several regions of the brain are

involved in the pathology of ASD, particularly the cerebellum.

Despite major scientific advances in the field of autism, no cure has currently been identified.

The effectiveness of treatment can be improved with early diagnosis and intervention (Reichow

et al., 2009, Rogers and Vismara, 2008). The primary treatment for ASD is through educational

therapy. These therapies focus on relationship building as well as teaching social and

communication skills. The most widely accepted therapy is the applied behavior analysis (ABA)

which uses small groups to focus on shaping and reinforcing social communication by prompting

and encouraging play time (Couper et al., 2003). Medications used to treat ASD-related

conditions have been found to be effective for some patients. Risperidone and aripiprazole,

which are antipsychotic drugs, were shown to be effective for treating repetitive behaviors in

children with ASDs but also caused significant adverse effects (McPheeters et al., 2011). Nasal

oxytocin spray is currently investigated as an ASD drug to potentially improve social

impairments. Thus far, a small clinical trial of 7 patients showed promising results using

functional MRI studies to evaluate brain patterns associated with social functions (Eisenstein,

2012). Methylphenidate (Ritalin), a psychostimulant that is commonly prescribed for attention

deficit hyperactivity disorder (ADHD), were administered to alleviate hyperactivity in ASD

patients but not all responded to the treatment (Posey et al., 2007). Furthermore, some

Page 15: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

6

complementary and alternative medicine therapies have been reported to be beneficial in certain

cases of ASD. Examples include gluten-free diet, omega-3 fatty acid supplements, hyperbaric

oxygen therapy, and chemical chelation. However, the validity of these treatments remains to be

investigated since they were mostly anecdotal (Tchaconas and Adesman, 2013).

Finding a cure is difficult because we still do not know the etiology of this disorder. Due to the

heterogeneous nature of ASD, there is almost certainly more than one possible cause. The most

established theories by the scientific community are genetic makeup and immune

activation/dysregulation (Onore et al., 2012). Below, I will discuss the evidence supporting these

two theories based on human and rodent studies.

1.2 Etiology of ASD: Genetic Makeup

Autism spectrum disorder can be classified as either idiopathic or syndromic. Idiopathic ASD

represents cases where ASD is the primary diagnosis and the cause is unknown. Twin studies in

ASD suggest that the genetic makeup of the individual could provide one explanation for its

etiology. The concordance rate for dizygotic twins was found to range from 0% - 30% whereas

for monozygotic twins, it is increased to 60% - 90% (Steffenburg et al., 1989, Bailey et al., 1995,

Rutter et al., 1999, Taniai et al., 2008, Rosenberg et al., 2009, Lichtenstein et al., 2010, Ronald et

al., 2011). This encouraged more in-depth genetic analysis which revealed several ASD risk

genes including reelin (RELN), SRC homology 3 and multiple ankyrin repeat domains

(SHANK1), and gamma-aminobutyric acid receptor subunit beta-3 (GABRB3) (Buxbaum et al.,

2002, Skaar et al., 2005, Sato et al., 2012). More convincingly, knockout (KO) mice or mice

with mutation(s) in these genes demonstrated ASD-related behaviors and pathologies (DeLorey

et al., 2008, Silverman et al., 2010, Won et al., 2012). Syndromic ASDs are genetic disorders

where ASD is a comorbid condition in addition to a pre-existing genetic mutation(s). Examples

include Rett syndrome, Fragile X syndrome (FXS), and tuberous sclerosis which are caused by

single gene mutations in the methyl CpG binding protein 2 (MeCP2), Fragile X mental

retardation 1 (Fmr1), and tuberous sclerosis complex (Tsc1 or Tsc2) gene, respectively. The

corresponding incidences of ASD in these diseases were estimated to be 25 - 40%, 21 - 50%, and

24 - 60% (Moss et al., 2009). Nevertheless, only 10 - 20% of the ASD population can be

attributed to genetic mutations and risk factors (Abrahams and Geschwind, 2008).

Page 16: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

7

1.2.1 Fragile X Syndrome

Fragile X syndrome (FXS) is genetic disorder caused by a single gene mutation in the Fmr1 gene

on the X chromosome. This results in an expansion of the number of CGG trinucleotide repeats

(>200) in the 5’ untranslated region which silences the FMR1 gene via hypermethylation. This

prevents the production of Fragile X mental retardation protein (FMRP) which is an mRNA

binding protein, primarily found in the brain, testes, and ovaries. It is responsible for shuttling

target mRNA to dendritic spines and inhibiting its translation by associating with specific

ribosomes. The lack of this protein leads to perturbations in synaptic plasticity and protein

translation (Bassell and Warren, 2008). FXS patients have characteristic physical phenotypes

including large, protruding ears, long face, and macroorchidism. Behavioral consequences of

this disease heavily overlap with ASD which include mental retardation, stereotypic movements

and abnormal social development (Jin and Warren, 2003). In fact, approximately 2 – 6% of

children with autism have FXS (Estecio et al., 2002, Hagerman and Hagerman, 2002; Harris et

al., 2008; Reddy, 2005, Wassink et al., 2001).

1.3 Etiology of ASD: Immune Activation

1.3.1 Evidence Based on Epidemiological Studies

Compared to twins, the incidence of ASD in siblings was estimated to be only 3 – 8% (Smalley

et al., 1991, Muhle et al., 2004, Lauritsen et al., 2005, Sumi et al., 2006). A recent twin study

found that 58% of heritability in ASD can be explained by environmental factors (Hallmayer et

al., 2011). This suggests that environmental factors such as the maternal environment during

fetal development could contribute to ASD since fraternal twins share the same placenta.

One of the most established environmental factors linked to increasing risk of ASD is infection

during pregnancy. Studies published as early as the 1970s found that the rubella epidemic in

1964 was associated with significant increased incidences of ASD in children (Chess, 1971,

Swisher and Swisher, 1975, Chess et al., 1978). As well, congenital infection of

cytomegalovirus was reported in ASD patients (Sweeten et al., 2004). Perhaps the most

convincing evidence was presented recently by a Danish cohort study of more than 20,000 ASD

patients spanning from 1980 to 2005. In this study, mothers that gave birth to ASD children had

higher incidences of autoimmune disease, such as rheumatoid arthritis, compared to control.

Page 17: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

8

More importantly, the risk of autism was increased by 3-fold with maternal viral infection and by

2-fold with maternal bacterial infection (specifically respiratory infection) in the first trimester

(Atladóttir et al., 2010). In a more recent study, the same group reported that out of 976 children

diagnosed with ASD, mothers who had fever lasting more than 7 days during first and second

trimester increased the risk of their baby developing infantile autism by 2.9 and 4.2 fold,

respectively (Atladóttir et al., 2012). These studies support the theory that environmental insults,

such as an infection during pregnancy, could increase the risk of the fetus developing ASD.

1.3.2 Evidence Based on Human Pathological and Biochemical Studies

Within the central nervous system, microglia and astrocytes are activated as part of the innate

immune response during neuroinflammation. Microglia is able to migrate to the site of insult

and clear debris by increasing the rate of phagocytosis and exerting cytotoxicity (Napoli and

Neumann, 2009). In addition, both astrocytes and microglia can activate the adaptive immune

system by triggering cytokine production and recruiting T-cells by expressing major

histocompatibility complex (Kreutzberg, 1996, Hamo et al., 2007). In ASD patients between the

age of 5 and 44, increased activation in microglia and astrocytes was observed in the cerebellum,

middle frontal gyrus, and anterior cingulate gyrus of post-mortem tissues (Vargas et al., 2005).

This result was replicated in a recent study of 20 male ASD patients (age 18 – 31) using positron

emission tomography where they found activation of microglia in brain regions including the

cerebellum, midbrain, pons, fusiform gyri and the anterior cingulate and orbitofrontal cortices

with the cerebellum showing the most prominent increases (Suzuki et al., 2013). In a separate

study of ASD patients of similar age range, microglial somal volume and cell density were

increased in the white matter and gray matter of the dorsolateral prefrontal cortex of ASD

patients (Morgan et al., 2010). The large age range of the population sample suggests that

immune activation in ASD may persist into adulthood.

Cytokines, produced mainly by immune cells, are the pleiotropic mediators between the immune

system and the CNS. They participate in a variety of neurological processes in the CNS such as

progenitor cell differentiation/migration, synaptic network modification and pruning, and cell

signaling cascade induction (Deverman and Patterson, 2009). The cytokines that were most

consistently found to be implicated in ASD pathogenesis are: interleukins-1β (IL-1β),

interleukin-6 (IL-6), interleukin-4 (IL-4), interferon- γ (INF-γ) and transforming growth factor-β

Page 18: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

9

(TGF-β) (Goines and Ashwood, 2012). IL-1β and IL-6 are proinflammatory cytokines that were

found to be both elevated in the plasma of ASD children and adults (Ashwood et al., 2011,

Emanuele et al., 2010, Suzuki et al., 2011). In contrast, IL-4, INF-γ and TGF-β serve more

neuroprotective roles in the CNS. IL-4 level was reported to be increased in the serum and

amniotic fluid of mothers with ASD children (Goin et al., 2011, Abdallah et al., 2011). INF-γ

and TGF-β levels were elevated and decreased in the plasma of ASD patients, respectively

(Singh, 1996, Croonenberghs et al., 2002, Vargas et al., 2005, Okada et al., 2007, Ashwood et

al., 2008, Li et al., 2009).

These cytokines activate similar pathways such as the mitogen-activated protein kinase (MAPK)

signalling pathway (Nelms et al., 1999, O’Neill, 2000, Shi and Massague, 2003, Platanias, 2005,

Hsiao et al., 2011). Therefore, it is not surprising that they can elicit similar functions in the

CNS. IL-1β was found to be expressed immediately upon insult and was able to inhibit neural

progenitor cell proliferation and promote gliogenesis (Crampton et al., 2012, Peng et al., 2008).

Furthermore, intracerebral injection of IL-1β was able to decrease the expression of myelin basic

protein (MBP, a marker for mature oligodendrocytes) in neonatal rats (Cai et al., 2004). On the

contrary, microglia activated by IL-4 was able to promote oligodendrogenesis (Butovsky et al.,

2006). Therefore, both cytokines could be implicated in the region-specific growth

abnormalities and white matter alternations found in ASD brains (de la Mano et al., 2007).

Synapses are also influenced by dysregulation of these cytokines. For instance, chronic IL-6

expression was able to shift the ratio of excitatory to inhibitory synapses in cerebellar granular

cell cultures (Wei et al., 2011). As well, INF-γ was able to induce synaptic alternations by

increasing the expression of major histocompatibility complex 1 (MHC1), triggering T cells and

natural killer cells (Shatz, 2009). Furthermore, lack of TGF-β in mice showed deficits in

glutamatergic and GABAergic synapses (Vashlishan et al., 2008). Changes at these synapses

may be involved in the behavioral symptoms found in ASD patients.

Cytokines are multifunctional and work within a network of signaling molecules. They are able

to exert both neuroprotective and neurotoxic responses in the brain. Too much or too little

expression can both be detrimental to proper brain functioning. For instance, both

overexpression and under expression of IL-1β in the hippocampus were associated with impaired

memory and learning (Goshen et al., 2007, Barrientos et al, 2009, Labrousse et al., 2009). The

time that these cytokines are activated is also important. The overexpression of TGF-β1 in the

Page 19: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

10

dentate gyrus during development led to deficits in social interaction and increased repetitive

behavior. However, these behavioral characteristics were the opposite when the overexpression

was introduced during adulthood (Depino et al., 2011). In addition, these cytokines are able to

regulate each other. They induce immune responses by interacting closely with glial cells. For

example, IL-1β was found to stimulate astrocyte growth as well as induce the activation of TGF-

β (Ransohoff and Benveniste, 1996). Other cytokines such as tumour necrosis factor-α (TNF-α),

interleukin-10 (IL-10), and monocyte chemoattractant protein-1(MCP-1) were also found to be

involved in ASD (Goines and Ashwood, 2012).

Overall, there is strong evidence that the level of cytokines is altered in maternal serum as well

as in the serum and brain tissue of ASD patients. Changes in these cytokine levels could

contribute to the pathological and behavioral deficits in ASDs. The exact mechanism is still

unclear because responses elicited by cytokines vary depending on the time of activation and the

environmental conditions. Nevertheless, it is clear that the balance of these cytokines is vital for

normal brain functioning.

1.4 Maternal Immune Activation: Possible Mechanism in ASD Patients

The MIA hypothesis is supported by epidemiological studies and the presence of immune

dysregulation observed in ASD patients as described above. The hypothesis suggests that an

infection to the mother during pregnancy can cause the production of various cytokines. The

imbalance of these signaling molecules may trigger the fetal immune system and perturb proper

brain maturation resulting in brain pathologies and ASD-associated behaviors.

The mechanism of how the activation of the maternal immune system causes changes in the fetal

brain is still unclear. As discussed in the previous section, cytokines are the main mediators

upon immune activation. However, most of the cytokines were found to be unable to cross the

placenta, with the exception of IL-6 (Zaretsky et al., 2004, Aaltonen et al., 2005). In fact, in fetal

inflammatory response syndrome, the level of IL-6 was found to be increased in the amniotic

fluid as well as maternal, fetal and umbilical cord serum (Gomez et al., 1998, Madsen-Bouterse

et al., 2010). Furthermore, incubation of human placenta with lipopolysaccharide (LPS), which

stimulated a bacterial infection, produced increased level of IL-6 (Holmlund et al., 2002).

Therefore, it is likely that IL-6 plays a major role in translating the maternal immune activation

Page 20: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

11

to the fetus. Smith et al., 2007 explored this hypothesis in rodents and found that injection of

recombinant IL-6 to wild-type mouse dams during mid-gestation resulted in ASD-like behaviors

such as social deficits and decreased prepulse inhibition in the litters. However, this was not

observed in litters from IL-6 knockout dams. More importantly, the behavioral deficits can be

reversed with injection of IL-6 antibody. Therefore, IL-6 signaling could be one of the key

participants in the MIA mechanistic pathway.

1.5 Modeling ASD in Rodents Based on the MIA Hypothesis

Rodent models can be used to further investigate the potential effects of maternal immune

activation on the fetal brain. In this model, pregnant dams were injected with an infectious agent

during pregnancy to activate the maternal immune system. The agents that were most commonly

used are polyinosinic:polycytidylic acid (Poly IC) and LPS which simulate viral and bacterial

infections, respectively. Poly IC is a synthetic double stranded RNA that mimics a viral

infection by binding to toll-like receptor (TLR) 3 while LPS is an endotoxin that mimics a

bacterial infection by binding to TLR4 (Muzio et al., 2000, Alexopoulou et al., 2001, Visintin et

al., 2001, Netea et al., 2002). In humans, both types of TLRs are highly expressed in microglia.

However, only TLR3 is also expressed in astrocytes and oligodendrocytes (Bsibsi et al, 2002;

Farina et al, 2005). The activation of these TLRs on different types of cells could potentiate

different downstream effects. The offspring from these immune activated dams were subjected

to behavioral tests linked to symptoms of ASD. As well, their brain and serum were analyzed to

identify the presence of immune activation and ASD associated neuropathologies.

Thus far, the behavioral and pathological phenotypes of the MIA model, summarized in the

Appendix 3 and 4, look promising. However, it is difficult directly compare these models

because they differ in the specific agent used, route of administration, dose, and injection regime.

The most common route of administration was through intraperitoneal (i.p.) injection. However,

subcutaneous, intravenous and tail vein injections were also performed. The dose is also

important because higher doses may be lethal to the litter or the dam. This was the case with

LPS (serotype: O111:B4) where i.p. injection of 100 μg/kg on E10 and E11 resulted in 0% litter

delivery whereas injection of 50 μg/kg on the same dates resulted in 100% litter delivery (Fortier

et al., 2007). In addition, the serotype of LPS is an important factor because certain serotypes

may be more potent than others. Compare to injection of LPS with serotype of O111:B4 (Fortier

Page 21: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

12

et al., 2007), a much higher dose (1 mg/kg) of LPS with serotype of O26:B6 was injected using

the same method and at the same time, but 100% of the litter survived (Ling et al., 2009). The

gestation day(s) and frequency of the injection(s) should be considered as well. LPS or Poly IC

injection(s) during early pregnancy may cause embryonic resorption associated with oxidative

stress. Oxidative stress occurs as a consequence of the body’s inability to properly detoxify

reactive oxygen species such as peroxides and free radicals. Nitric oxide (NO) is a free radical

that can exert numerous toxic effects including inhibition of mitochondrial respiratory chain,

DNA break, apoptosis, and necrosis (Karima et al., 1999). Ogando et al., 2003 found that high

doses of LPS injection (500 μg/kg) during early pregnancy (E7) increased the decidual and

uterine NO production due to increased mRNA expression of inducible and neuronal nitric oxide

synthase. This resulted in 100% embryonic resorption 24 hours post injection. Additional

injections of aminoguanidine, an inducible nitric oxide synthase inhibitor, were able to rescue

this outcome. Similarly, prenatal Poly IC injection on the same gestation day (E7) also increased

embryonic resorption rate to 54.8%. As well, the mRNA expression of iNOS was also increased

in the embryonic unit which consisted of the embryo, placenta and uterine muscle (Shimada et

al., 2003).

The fetal immune system can be induced by activating the maternal immune system. In the LPS

model, the cytokine levels (i.e. IL-1β, IL-6, and/or TNF-α) were elevated in the placenta,

amniotic fluid, fetal serum and fetal brain tissue for multiple serotypes of LPS (Urakubo et al.,

2001, Ashdown et al., 2006, Liverman et al., 2006, Salminen et al., 2008, Ning et al., 2008,

Romero et al., 2010, Girard et al., 2010). For Poly IC model, the increase in IL-6 and MCP-1

was observed in the amniotic fluid and dam serum, respectively (Forrest et al., 2012, Mandal et

al., 2010, 2011). Garay et al. 2012 reported that changes in the levels of different cytokines in

the Poly IC model were dependent on the post-natal age and specific brain regions. For example,

they found that the level of IL-1β in the frontal cortex was significantly elevated at PND0 but

decreased at PND14 and PND30. In comparison, level of IL-1β was significantly reduced in the

hippocampus at PND0 and not changed at later time points. Remarkably, the cytokines that were

changed in the MIA model were also found to be altered in ASD patients. One such example is

IL-1β, which was found to be significantly elevated in the sera of both young mice prenatally

injected with Poly IC and ASD children.

Page 22: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

13

In the LPS model, injection(s) during mid- and late-gestation yielded increased in the expression

of astrocyte (Cai et al., 2000, Paintlia et al., 2004, Paintlia et al., 2008) and microglia marker(s)

(Larouche et al., 2005, Ling et al., 2006, Ling et al., 2009, Roumier et al., 2008, Girard et al.,

2010) as well as decreased in the expression of mature oligodendrocyte marker(s) (Cai et al.,

2000, Kumral et al., 2007, Yesilirmak et al, 2007, Paintlia et al., 2004, 2008, Rousset et al., 2006,

2008). For Poly IC, Juckel et al., 2011 and Makinodan et al., 2008 showed mid-gestation

injection (E9 - 9.5) increased microglia activation in the hippocampus and striatum as well as

decreased myelin in the hippocampus. These data mirror the pathologies observed in ASD

patients and support the idea that maternal immune activation is able to elicit brain perturbations

in the fetus.

Ideally, the MIA model for ASD should demonstrate the core symptoms of this disorder.

Malkova et al, 2012 accomplished this using Poly IC mouse model where they showed that male

offspring displayed deficits in social interaction, impaired communication and increased

repetitive behavior. Furthermore, the deficits in social interaction in the Poly IC model were

confirmed by Soumiya et al., 2011. Similarly, several groups have also reported impairments in

social interaction in the LPS model (Golan et al., 2006, Kirsten et al., 2010, 2012, Oskvig et al.,

2012). In addition, both Poly IC and LPS model demonstrated several ASD-associated behaviors

such as increased anxiety in novel environment (Bakos et al., 2004, Smith et al., 2007, de

Miranda et al., 2010, Wang et al., 2010, Soumiya et al., 2011, Hsiao et al, 2011) and deficits in

sensory motor gating (Borrell et al., 2002, Shi et al., 2003, Fortier et al., 2007, Romero et al.,

2007, 2010, Makinodan et al., 2008, de Miranda et al., 2010, Hsiao and Patterson, 2011).

Page 23: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

14

2 Rational, Hypotheses and Objectives

2.1 Rational for MIA Mouse Model

We decided to inject wild-type C57/B6 mice with Poly IC (20 mg/kg, i.p.) on E12.5 or LPS

(serotype O127:B8, 75 μg/kg, i.p.) on E11.5 and E12.5. We also performed the same Poly IC

injection on Fmr1 KO C57/B6N mice. The Poly IC model was established based on published

ASD-related behavioral and neuropathological findings using the same injection regime (Smith

et al., 2007, Shi et al., 2009, Mandal et al., 2010, 2011, Hsiao and Patterson, 2011).

Furthermore, the gestation day(s) of the injections coincided with the peak of PC generation in

the cerebellum and amygdala neurogenesis which occurs at 10 – 13 days and 11 – 14 days

respectively (Workman et al., 2013). As previously discussed, these two brain regions were

heavily implicated in ASD. LPS was injected around the same time because we wanted to

directly compare the differences between viral and bacterial infection. The LPS serotype

O127:B8 was used because it was the most common serotype used in mice by other groups.

Using this particular serotype, these groups demonstrated immune activation in amniotic fluid

and/or fetal brain (Liverman et al., 2006, Ning et al., 2008) as well as some behavioral changes

including increased anxiety (Wang et al., 2010) and hyperactivity after acute amphetamine

challenge (Zager et al., 2012).

2.2 Hypotheses and Objectives

We hypothesized that maternal immune activation in mice using Poly IC and LPS during mid-

gestation will elicit ASD-like behaviors and cerebellar pathologies in the offspring that may be

dependent on sex of the offspring and type of prenatal infection (i.e. LPS vs. Poly IC).

Furthermore, the effect of MIA may be exacerbated in offspring from dams with immunological

memory (i.e. Poly IC 2X and LPS 2X) and/or genetic mutation (i.e. Fmr1 KO).

Objective 1: To compare the effects of bacterial (LPS) and viral (Poly IC) prenatal infection in

MIA offspring

Rational: LPS and Poly IC mimic different types of infection in the mother by acting on different

toll like receptors. Thus far, only Harvey and Boksa, 2012 has directly compared these two

models and found prenatal treatment-dependent changes in neuronal density (at PND14) and

Page 24: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

15

reelin-positive cells (at PND28) in the stratum oriens of CA1. No other groups have investigated

the behavioral differences between these two models.

Objective 2: To investigate whether male vs. female MIA mice show differential effects in ASD-

related behaviors and cerebellar pathologies

Rational: Because of the strong male-bias in ASD, it would be interesting to investigate if this

phenomenon is also present in the MIA mouse model. Most of the studies published to date

were conducted on male mice only, or a mixture of both sexes. By combining them during the

analysis, it could mask the significance that may be present for only one sex. For example,

Wang et al., 2010 showed that female offspring (PND200) from LPS treated dams had impaired

exploration in open field test whereas male offspring of the same age did not showed such

difference.

Objective 3: To investigate cerebellar pathology in adult MIA offspring

Rational: Human pathological studies suggest that the cerebellum is perturbed in ASD.

However, most groups that investigated neuroinflammation focused on the forebrain during early

development. Our study will focus on specifically investigating the pathological changes in the

adult cerebellum as it pertains to ASD.

Objective 4: To investigate the effects of “immunological memory” of dams on MIA offspring

Rational: All of the studies published to date administered LPS or Poly IC injections on

immunologically “naïve” mice. In contrast to rodents, most pregnant woman would have been

exposed to different pathogens prior to their pregnancy. Therefore, it would be biologically

relevant for human to investigate the MIA model using previously immunized dams. So far,

only Mandal et al., 2011 has investigated the difference in immune response between litters from

naïve and immunized dams injected with Poly IC. They found that although both naïve and

previously immunized dams had comparable serum and amniotic fluid levels of IL-6 before and

after Poly IC injection, only offspring from immunized dams demonstrated differentiation of T-

helper 17 (Th17) cells. These cells are a subset of T helper cells that produce IL-17 which can

contribute to neuroinflammation by recruiting monocytes and neutrophils to the site of

inflammation (Korn et al., 2009). Our goal was to compare the severity of neuropathological

Page 25: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

16

and/or behavioral outcomes between litters from immunologically “naïve” dams (i.e. Poly 1X or

LPS 1X) and litters from dams re-injected with Poly IC or LPS (i.e. Poly 2X or LPS 2X).

Objective 5: To investigate the effects of MIA on a genetic model of ASD (i.e. Fmr1 KO mouse)

Rational: There is some evidence of immune dysregulation in Fragile X syndrome. Ashwood et

al., 2010 reported that the serum level of several cytokines (IL-1α, RANTES, and IP-10) in

Fragile X patients with or without ASD was significantly different from typically developing

individuals. In addition, increased GFAP expression was observed in several brain regions

including the cerebellum of adult Fmr1 knockout mice, a possible sign of neuroinflammation

(Yuskaitis et al., 2010, L. K.K. Pacey, I. Xuan, D. R. Hampson, unpublished data). We injected

Poly IC into Fmr1 knockout dams to determine whether the effects of prenatal viral insult could

be exacerbated in offspring with pre-existing genetic vulnerability.

Page 26: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

17

3 Methods

3.1 Animals

All animal experiments were carried out in accordance with the guidelines set out by the

Canadian Council on Animal Care and were approved by the University of Toronto Animal Care

Committee. FMR1 knockout mice (backcrossed > 10 generations on the C57BL/6 background)

were generously provided by Dr. William Greenough, University of Illinois, and bred at the

University of Toronto. Only homozygous female Fmr1 knockout mice were used for breeding

therefore genotyping was not performed. Mice were mated overnight and vaginal plug was

checked at 9 am and 4 pm each day. The day that the vaginal plug was observed was marked as

embryonic day 0.5. One group of dams were given intraperitoneal (i.p.) injections (5 μl/g) of 75

μg/kg of LPS (lipopolysaccharide O127:B8; L3129; Sigma) on both E11.5 and E12.5. A

separate group of dams were injected (i.p. 5 μl/g) with 20 mg/kg polyinosinic:polycytidylic acid

(Poly IC) potassium salt (P9582; Sigma) once on E12.5. Poly IC was dissolved in saline at 4

mg/mL (based on the weight of Poly IC itself, which was 10% of total weight of the salt). Saline

injections were made on either E11.5 and E12.5 or E12.5 only, and were used as control. A

schematic of the injection schedule is shown in Fig. 1.

All pups were housed with their mother until post-natal day (PND) 21 when they were weaned

and ear tagged. After weaning, two to four pups of the same sex and treatment were housed

together per cage. In case where only one single female or male was born in one litter, it was

housed with other mice of same sex and similar age (may be of different treatment group). All

animals were left undisturbed except for biweekly cage changes and behavior tests. All

behavioral testing with the exception of social test was conducted between 9 am and 2 pm.

Social testing was performed between 9 am and 5 pm. Fig. 2 describes the core and associated-

symptoms of ASD in human and how it can be evaluated in rodent ASD models. A schematic

diagram of the schedule of behavior and biochemical analysis for this project is shown in Fig. 3.

Page 27: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

18

Fig. 1 Injection schedule of MIA dams. The day that the plug was observed is considered E0.5.

The litters are kept with their mother until weaning at PND21. Male and female offspring were

housed separately (2 to 4 mice per cage). Behavioral analysis commenced at 6 weeks.

Page 28: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

19

Fig. 2 Translating human ASD behaviors to mouse model of ASD. The table lists some of the

mouse behavior tests that can be used for certain core and associated – ASD behaviors

manifested in humans (Crawley, 2004, Roullet and Crawley, 2011).

Deficit in social interaction and communication

Ritualistic Repetitive Behaviour

Human ASD behaviors Mouse Behavioral Tests

Gaze aversion eye blink test

Deficit in social -emotional reciprocity 3-chamber paradigm Deficit in developing and maintaining relationships tube test

Lack of initiation during social interaction

Language delay ultrasonic vocalization

Motor Stereotypies grooming marble burying test

Insistence on sameness reversal of a position habi t in an appetitive T -maze task reversal of a position habit in the Morris water maze

Hyperactivity motor activity test

Intellectual disability contextual and cued fear conditioning Operant learning tasks Acquisition of T -maze tasks

Seizures audiogenic seizure test

Sensory hyper/hyposensitivity prepulse inhibition Tactile startle

Sleep dysfunction observation of home cage sleep patterns

Increased anxiety elevated plus maze light-dark exploration open field test

Co

re B

eh

avio

rs o

f A

SD

Asso

cia

ted

Beh

avio

rs o

f A

SD

Page 29: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

20

Fig. 3 The schedule of behavioral and biochemical analysis of MIA offspring.

3.2 Motor Activity

Motor activity was measured using the automated VersaMax animal activity monitoring system

(AccuScan Instruments, Inc., US) as previously described (Pacey et al., 2011). The animals were

tested at 6 weeks of age. The detection system consisted of a 42 x 42 x 30 cm Plexiglas box

placed inside the activity monitor that emitted 16 x 16 x 16 laser beams, each separated by 2.5

cm. The system measured the animal’s activity by tracking the number of beams that was

broken. Two smaller Plexiglas boxes (21 x 21 x 21 cm) were placed at diagonal corners inside

the larger Plexiglas box for activity measurement of each individual mouse. All animals were

habituated under dim lighting (the room was evenly lit by three 14 W incandescent light fixtures)

for at least one hour prior to testing. Before each test, the smaller Plexiglas box was wiped

thoroughly with Virox (0.4% hydrogen peroxide), followed by water. Thereafter, the animal was

gently placed in the center of the smaller Plexiglas box and a lid was immediately placed on top

to prevent the animal from escaping. The test was performed under dim lighting and the data

generated by the monitor was tabulated every 5 minutes for an hour. The following parameters

were measured: distance travelled, horizontal movement time, vertical activity, and vertical

movement time.

3.3 Social Interaction and Social Preference Test

The modified three chamber paradigm protocol was based on Ramsey et al., 2011 with some

modifications. The entire test was conducted under dim lighting (approximately 40 lux). A

Page 30: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

21

digital camera was mounted on the ceiling approximately one meter above the social apparatus to

track the locomotion of the animal. Rather than physically separating the chambers, we virtually

defined two identical circular zones (diameter: 19.8 cm) in a white Plexiglas box (61.7 x 40.8 x

23 cm) using Viewer2 software (BIOBSERVE GmbH, NJ). These two zones were parallel from

each other and equidistant from the edge of the box (7.1 cm lengthwise, 10.5 cm widthwise). An

empty white wire cage (top diameter 7.5 cm, bottom diameter 10 cm, height 10 cm) was placed

at the center of each zone, covered by a white circular disk (diameter: 10 cm) and held down by a

standard 400 mL glass beaker. The beaker prevented the test animals from climbing on top of

the wire cages. Two removable, white cardboard dividers (40 x 23 cm) were inserted into the

middle of the box, shielding both zones. The activity of the animal in each zone was recorded

every 2 minutes for 10 minutes.

The stimulus animals (wild-type C57BL/6, age and sex matched with test animal) were always

kept at the opposite corner from the test animal to ensure its novelty. Stimulus animals were

habituated for at least 30 minutes in their respective home cage under dim lighting as previously

described. Each animal was then trained inside a clean cylindrical wire cage placed inside the

white Plexiglas box. A maximum of four stimulus animals were trained at the same time for two

15-minute sessions. The stimulus animals were used for subsequent social testing if no persistent

aggressive behavior was observed (e.g. climbing and/or biting of the cage).

All social testing was conducted at 7 to 8 weeks of age. Test animals were habituated for at least

one hour in their home cages under dim lighting as previously described. Before each test, all

apparatus were wiped vigorously with Virox, followed by water. To begin, the test animal was

gently placed between the dividers. Habituation commenced when the dividers were

simultaneously lifted, exposing the animal to the two zones, each with an empty wire cage held

down by a beaker as previously described. After the animal was allowed to habituate for 10 min,

it was then isolated between the two dividers again, shielding it from the two zones. If side

preference was present (i.e. the difference in time spent between the two zones was more than 30

seconds), the animal was returned to home cage and tested the next day. Otherwise, one stimulus

animal (of same sex and similar age) was placed in one of the wire cages while a novel object

was placed in the other cage. The designation of the zones was determined at random. The

novel object used was a green Biobag tied into a knot placed on top of a roll of green tape

(diameter: 6 cm). The dividers were then simultaneously lifted and the animal was allowed to

Page 31: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

22

explore for 10 minutes. Social preference index for the social interaction test was calculated by

the difference in time spent between the two zones (social zone minus non-social zone) divided

by the sum of time spent in both zones. At the end of the social interaction test, the animal was

again isolated between the dividers to shield it from both zones. Another novel stimulus mouse

(of same sex and similar age) was placed in the non-social zone, replacing the novel object. This

now becomes the non-familiar zone. The dividers were then lifted simultaneously and the

animal was allowed to explore for 10 minutes. Social preference index for social preference test

was calculated by the difference of time spent between the two zones (non-familiar zone minus

familiar zone) divided by the sum of time spent in both zones. At the end of the experiment, the

test and stimulus animals were returned to their respective cages. A T-test was performed

between the social zone and non-social zone or familiar zone and non-familiar zone for each

treatment group separately to determine if there was a significant preference for one zone over

the other (Ehninger et al., 2012, Lipina et al., 2013).

3.4 Marble Burying Test

Marble burying test is a test for repetitive/compulsive behavior and was conducted at 10 weeks

of age as previously described (Thomas et al., 2009). Clean cages (29.5 x 17.5 x 12.5 cm) were

filled up to 5 cm with corn cob bedding. Twenty identical navy marbles (diameter: 1.2 cm) were

placed in a 4 x 5 rectangular matrix occupying 2/3 of the cage. The animals were first habituated

in the biosafety cabinet class A/B for at least 30 minutes with the blower on as background noise.

After habituation, the test mouse was gently placed into 1/3 of the cage without of the marbles

and a lid was immediately placed over the cage. Individual test animals were allowed to explore

the cage for 30 minutes after which they were returned to the home cages. The marbles were

cleaned with Virox and rinsed with water between each test. A marble was considered buried

when more than 50% of its surface area was covered by the bedding.

3.5 Grooming

Self-grooming behavior entails licking and scratching of any body parts of the mouse (Kalueff

and Tuohimaa, 2005). The total time spent grooming (in seconds) was recorded manually based

on previously recorded videos of the social preference test (first 5 minutes of exploration period).

The experimenter was blinded to the treatment group of the test animal.

Page 32: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

23

3.6 Western Blot

Each cerebellum was dissected, frozen on dry ice, and stored at -80°C. Once one set of

cerebellar samples (i.e. saline, LPS1X, LPS 2X or saline, Poly IC 1X, Poly IC 2X; 2-3 sample

for each treatment group) have been accumulated, the tissues were homogenized in tandem on

the same day in lysis buffer (50 mM Tris-HCl, 1% SDS, 1X protease inhibitor cocktail (PIC,

Sigma-Aldrich, St. Louis, MO) pH 7.4). Protein concentrations were measured using BCA assay

kit (Sigma-Aldrich).

Equal amounts of total protein were loaded in each lane and electrophoretically separated on

12% polyacrylamide gels (one sample set per gel). The amount of total protein loaded (4 – 40

μg) depended on the abundance of the target protein in the cerebellum. The protein was then

transferred to a nitrocellulose membrane using the Bio-Rad Trans-Blot semi-dry transfer system

and blocked in wash buffer (0.01 M Tris, 0.015 M NaCl, 0.05% Tween-20, pH 7.6) with 5%

skim milk at room temperature for 30 minutes. The membrane was then washed 3 x 15 minutes

with wash buffer and incubated with primary antibodies (made in wash buffer) overnight at 4°C.

The primary antibodies used were as follows: anti-GFAP (1: 500, clone N206A/8, NeuroMab),

rabbit anti-Iba-1 (1: 250, Wako), rat anti-MBP (1:3000, Millipore), and anti-glyceraldehyde 3-

phosphate-dehydrogenase (GAPDH) (1:20 000 to 1: 100 000 depending on the amount of protein

loaded, Sigma-Aldrich). The next day, the membranes were washed 3 x 15 minutes with wash

buffer and incubated for 2 hours at room temperature in secondary antibodies (made in wash

buffer with 3% skim milk). The horseradish peroxidase-conjugated secondary antibodies used

were as follows: goat anti-rabbit (1:2500, Jackson ImmunoResearch), goat anti-mouse (1:5000,

Jackson ImmunoResearch), and donkey anti-rat (1:2500, Jackson ImmunoResearch). This was

continued by 3 x 15-minute washes with wash buffer. Immunoreactivity was detected using

SuperSignal West Pico Chemiluminescence substrate (Fisher Scientific, Pittsburgh, PA) and

imaged using Alpha Innotech FluorChem® Chemiluminescent Imaging System. The integrated

density value per area of the target bands were quantified using AlphaEaseFC image analysis

software normalized to the loading control (i.e. GAPDH). Each LPS sample set consisted of

saline (n = 3), LPS 1X (n = 3) and LPS 2X (n = 2 or 3). The protein expression of LPS treated

groups were expressed as a percentage of saline treated groups. The values from different

sample sets were then averaged between different sample sets.

Page 33: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

24

3.7 Statistical Analysis

Statistical analysis was conducted only for groups of mice with n ≥ 8 and analyzed separately for

each treatment (i.e. Poly IC or LPS), genotype (i.e. wild-type or Fmr1 knockout) and sex.

Statistical analysis was performed using GraphPad Prism 5. For studies with two sample groups,

a student’s t-test was performed with 95% confidence interval. For studies with three sample

groups, one way ANOVA was performed followed by Bonferroni post hoc test. Grubbs’ test

was used to eliminate outliers.

Page 34: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

25

4 Results

4.1 The Effect of MIA on Body Weight

The weight of MIA offspring was measured on PND46 as an indicator of their overall health.

Prenatal LPS treatment did not affect the weight of wild-type female (F(2, 40) = 0.2763, p =

0.7600) or male mice (F(2, 36) = 2.72, p = 0.0793) (Fig. 4A). Similarly, prenatal Poly IC

treatment did not alter the weight of wild-type female (F(2, 32) = 1.31, p = 0.2850) or male

offspring (F(2, 23) = 0.80, p = 0.4627) (Fig. 4B).

Saline Fmr1 KO mice were heavier in comparison to WT mice of the same treatment. This was

significant for male (t(24) = 2.87, p < 0.01) but not female mice (t(32) = 0.80, p = 0.4314).

Treatment with Poly IC decreased the weight of female (F(2, 38) = 3.81, p < 0.05; Bonferroni

post hoc test Poly 1X p > 0.05, Poly 2X p > 0.05) and male Fmr1 KO mice (F(2, 29) = 8.29, p <

0.01; Bonferroni post hoc test Poly 1X p < 0.01, Poly 2X p < 0.05) (Fig. 4C).

4.2 The effect of MIA on Motor Activity

Locomotor and exploratory behavior of the MIA offspring was investigated by measuring

horizontal and vertical activity in an automated activity box. It should be noted that for all

treatment groups, male mice demonstrated higher vertical activity compared to female mice.

LPS treatment (i.e. both LPS 1X and 2X) did not affect the horizontal activity in the offspring in

terms of total distance travelled (female: F(2, 54) = 0.09, p = 0.9181; male: F(2, 59) = 1.14, p =

0.3268) and total horizontal movement time (female: F(2, 54) = 0.01, p = 0.9908; male: F(2, 59)

= 1.24, p = 0.2958) (Fig. 5A, B). In addition, it did not affect the total vertical activity (female:

F(2, 54) = 2.75, p = 0.0728; male: F(2, 59) = 0.44, p = 0.6467) or total vertical movement time

(female: F(2, 54) = 3.05, p = 0.06, male: F(2, 59) = 0.70, p = 0.5020) (Fig. 5C, D).

Single Poly IC prenatal injection significantly decreased the total distance travelled (F(2, 52) =

7.17, p < 0.01, Bonferroni post hoc test p < 0.01) and total horizontal movement time (F(2, 52) =

6.28, p < 0.01, Bonferroni post hoc test p < 0.01) in male but did not affect female offspring.

Page 35: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

26

Fig. 4 Body weight of wild-type and Fmr1 knockout MIA offspring on PND46. LPS (A)

and Poly IC (B) treatment did not affect the weight of wild-type mice. Prenatal Poly IC injection

significantly decreased the body weight of male Fmr1 KO mice (C). Each column represents the

average ± S.E.M. One-way ANOVA was performed for each genotype and sex separately,

followed by Bonferroni’s post hoc analysis. Student’s t-test was performed for saline WT vs.

saline KO. Significant outliers were removed using the Grubbs’ test. Statistical significance is

not shown for groups with n < 8. * p < 0.05, ** p < 0.01.

Page 36: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

27

Fig. 5 Motor activity of LPS wild-type offspring. LPS treatment did not affect the total

distance travelled (A), total horizontal movement time (B), total vertical activity (C) and total

vertical movement time (D) of the offspring. Each column represents the average ± S.E.M.

One-way ANOVA was performed for each sex separately, followed by Bonferroni’s post hoc

analysis.

In contrast, double Poly IC prenatal injections significantly increased the horizontal activity of

female offspring with regards to total distance travelled (F(2, 61) = 4.24, p < 0.05; Bonferroni

post hoc test p < 0.05) and total horizontal movement time (F(2, 61) = 3.91, p < 0.05; Bonferroni

post hoc test p < 0.05) but did not affect the male offspring (Fig. 6A, B). Both Poly IC 1X and

2X treatment did not affect the total vertical activity (female: F(2, 61) = 0.40, p = 0.6699; male:

F(2, 52) = 0.57, p = 0.5685) and total vertical movement time (female: F(2, 61) = 1.26, p =

0.2911, male: F(2, 52) = 0.40, p = 0.6735) of MIA offspring (Fig. 6C, D).

Saline Fmr1 KO mice were hyperactive compare to saline wild-type mice with regards to total

distance travelled (female: t(40) = 8.66, p < 0.0001; male: t(32) = 6.28, p < 0.0001), horizontal

movement time (female: t(40) = 7.49, p < 0.0001; male: t(32) = 5.39, p < 0.0001), vertical

Page 37: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

28

activity (female: t(40) = 4.51, p < 0.0001; male: t(32) = 4.79, p < 0.0001), and vertical movement

time (female: t(40) = 5.10, p < 0.0001; male: t(32) = 5.00, p < 0.0001) (Fig. 7A – D).

In Fmr1 KO mice, Poly IC treatment (1X and 2X) did not further increase the total distance

travelled (female: F(2, 45) = 2.07, p = 0.1378; male: F(2, 35) = 0.18, p = 0.8371) or horizontal

movement time (female: F(2, 45) = 2.57, p = 0.0876; male: F(2, 35) = 0.47, p = 0.6318) (Fig. 7A,

B). In contrast, Poly 1X and 2X female Fmr1 KO mice showed significantly decreased total

vertical activity (F(2, 45) = 6.86, p < 0.01, Bonferroni post hoc test p < 0.05 for Poly 1X, p <

0.01 for Poly 2X) and vertical movement time (F (2, 45) = 8.61, p < 0.001, Bonferroni post hoc

test p < 0.01). A similar trend was observed for Poly 1X male but not Poly 2X male Fmr1 KO

mice for total vertical activity (F (2, 35) = 3.09, p = 0.0582) and vertical movement time (F (2,

35) = 4.46, p < 0.05, Bonferroni post hoc test p < 0.05 for Poly 1X, p > 0.05 for Poly 2X).

To summarize the motor activity results, while prenatal LPS injection into wild-type mice did not

affect the motor activity of the offspring, prenatal Poly IC injection caused sex and treatment-

dependent changes in the horizontal activity of wild-type offspring and also significantly

decreased the vertical activity of Fmr1 knockout mice.

4.3 The Effect of MIA on Social Behavior

Social impairment is one of the core symptoms of ASD and is often linked with deficits in

communication. In rodents, this behavior can be assessed using the 3-chamber paradigm

developed by Crawley’s group (Silverman et al., 2010). This test was divided into two stages.

In the social interaction test, sociability was measured by comparing the time the animal spent in

the zone with novel mouse vs. novel object. In the social preference test, the second stage,

sociability was measured by comparing the time the animal spent in the zone with the familiar

mouse vs. non-familiar mouse. In both tests, normal social behavior entailed preference for the

social and non-familiar zone over the non-social and familiar zone, respectively.

Page 38: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

29

Fig. 6 Motor activity of Poly IC wild-type offspring. For female offspring, double Poly IC

injection significantly increased the total distance travelled (A) and total horizontal movement

time (B). For male offspring, single Poly IC injection significantly decreased these two

parameters. Poly IC treatment did not affect the total vertical activity (C) and total vertical

movement time (D) of MIA offspring. Each column represents the average ± S.E.M. One-way

ANOVA was performed for each sex separately, followed by Bonferroni’s post hoc analysis.

* p < 0.05. *** p < 0.001.

Page 39: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

30

Fig. 7 Motor activity of Poly IC Fmr1 knockout offspring. Compared to saline WT animals,

saline Fmr1 KO mice were significantly more hyperactive with respect to all four parameters (A

– D). Poly IC treatment did not affect the total distance travelled (A) and total horizontal

movement time (B) of Fmr1 KO offspring. Poly IC treatment decreased the total vertical activity

(C) and total vertical movement (D) of Fmr1 KO offspring with the exception of Poly 2X male

mice. Each column represents the average ± S.E.M. One-way ANOVA was performed for each

sex separately, followed by Bonferroni’s post hoc analysis. Student’s t-test was performed for

saline wild-type vs. saline knockout for each sex separately. * p < 0.05. ** p < 0.01. **** p <

0.0001.

Page 40: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

31

LPS treatment caused decreased sociability in female mice only (Fig. 8). In the social interaction

test, LPS 1X female offspring showed no statistically significant preference towards the social

zone (saline: t(32) = 4.00, p < 0.001; LPS 1X: t(20) = 1.77, p = 0.0914; LPS 2X: t(14) = 3.01, p

< 0.01). As well, female LPS mice showed a trend towards lower social interaction index

suggesting their preference for the social zone was not as strong as the respective control groups

(F(2,33) = 1.942, p = 0.1595). In contrast, LPS male offspring displayed normal social behavior

by preferring the social zone over the non-social zone (saline: t(30) = 3.86, p < 0.01; LPS 1X:

t(20) = 6.30, p < 0.0001; LPS 2X: t(24) = 5.70, p < 0.0001) (Fig. 8A). The social interaction

index for male LPS offspring was comparable to that of the controls (F(2, 37) = 1.40, p = 0.2586)

(Fig. 8C). For social preference test, LPS offspring displayed decreased sociability by showing

no preference for the non-familiar zone (saline: t(28) = 2.93, p < 0.01; LPS 1X: t(20) = 0.080, p

= 0.9369; LPS 2X: t(14) = 0.83, 0.4223) (Fig. 8B). LPS female offspring also showed a trend

towards decreased social preference index (F(2, 33) = 2.57, p = 0.0916) (Fig. 8D). Male saline

and LPS offspring showed no preference for the non-familiar zone (saline: t(28) = 1.34, p =

0.1900; LPS 1X: t(20) = 1.31, p = 0.2002; LPS 2X: t(24) = 1.89, p = 0.0709) (Fig. 8B). Male

LPS offspring also had comparable social preference index with saline control animals (F(2, 31)

= 0.0082, p = 0.8591) (Fig. 8D).

Poly IC wild-type offspring showed no deficits in sociability. For female offspring, both Poly

1X and Poly 2X offspring displayed normal sociability by spending more time in the social zone

(Poly 1X: t(18) = 1.89, p = 0.0745), Poly 2X: t(14) = 2.96, p < 0.05) (Fig. 9A). Their respective

social interaction index did not significantly differ from controls (F(2, 32) = 1.39, p = 0.2643)

(Fig. 9C). Although both Poly 1X and 2X female offspring did not show a significant preference

for the non-familiar zone (Poly 1X: t(18) = 0.83, p = 0.4198, Poly 2X: t(14) = 1.07, p = 0.3050),

their social preference index was comparable to the saline group (F(2,32) = 0.83, p = 0.4466). In

comparison, Poly 1X male offspring demonstrated normal social behavior by spending more

time in the social zone (t(14) = 3.26, p < 0.01) and non-familiar zone (t(10) = 2.53, p < 0.05).

They also did not significantly differ from controls with respect to social interaction index (F(2,

28) = 0.49, p = 0.6151) and social preference index (F(2, 26) = 1.41, p = 0.2615) (Fig. 9C, D).

Statistical tests were not performed for Poly 2X male wild-type offspring because the n-value

was less than 8.

Page 41: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

32

Fig. 8 Social behavior of LPS treated wild-type MIA offspring evaluated by the modified 3-

chamber paradigm. For social interaction test, only LPS 1X female mice displayed deficits in

sociability by showing no preference for the social zone (A). This deficit was reflected by the

decreased social interaction index for this group, although not statistically significant (C). For

social preference test, deficits in sociability were observed for both LPS 1X and LPS 2X female

mice where they showed no preference for the non-familiar zone (B). This lack of preference

was observed for all male groups, including control. The social preference index was decreased

for female but not male mice, although not statistically significant (D). Time spent in each zone

was measured in seconds. Each column represents the average ± S.E.M. One-way ANOVA was

performed for each sex separately, followed by Bonferroni’s post hoc analysis. Using the

Grubbs’ test, one value that was furthest from the mean was omitted for each group, except for

those with n ≤ 8. * p < 0.05. ** p < 0.01. *** p < 0.001.

Page 42: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

33

Fig. 9 Social behavior of Poly IC treated wild-type MIA offspring evaluated by the

modified 3-chamber paradigm. For social interaction test, only Poly 1X female mice displayed

deficits in sociability by showing no preference for the social zone (A). This deficit was not

reflected by the social interaction index (C). For social preference test, deficits in sociability

were observed for all groups except for female saline and male Poly 1X offspring where they

showed a significant preference for the non-familiar zone (B). The Poly IC treated groups did

not differ in social preference index (D). Time spent in each zone was measured in seconds.

Each column represents the average ± S.E.M. One-way ANOVA was performed for each sex

separately, followed by Bonferroni’s post hoc analysis. Using the Grubbs’ test, one value that

was furthest from the mean was omitted for each group, except for those with n ≤ 8. * p < 0.05.

** p < 0.01. *** p < 0.001.

Page 43: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

34

Similar to wild-type saline treated mice, both female and male saline treated Fmr1 KO mice

demonstrated normal social behavior during social interaction test by spending significantly

more time in the social zone (female: t(16) = 4.33, p < 0.001; male: t(16) = 3.56, p < 0.01) (Fig.

10A). However, in the social preference test, both female and male saline treated Fmr1 KO mice

showed no significant preference for either the non-familiar zone or the familiar zone (female:

t(16) = 1.54, p = 0.1355; male: t(16) = 0.29, p = 0.7745) (Fig. 10B).

With regards to the effect of Poly IC on Fmr1 KO mice, decreases in sociability was only

observed in female Poly 1X group where it did not show a preference for the social zone during

social interaction test (t(26) = 1.85, p = 0.07449) (Fig. 10A). This result was also reflected by a

significantly lower social interaction index in Poly 1X female mice compared to control (F(2, 32)

= 3.78, p < 0.05, Bonferroni post hoc test p < 0.05) (Fig. 10C). In contrast, Poly IC treatment did

not impair the sociability of other Fmr1 KO groups during social interaction test (Poly 2X

female: t(22) = 2.98, p < 0.01; Poly 1X male: t(16) = 3.56, p < 0.01; Poly 2X male: t(22) = 5.99,

p < 0.001). Comparison of social interaction index revealed the same findings for these groups

with the exception of Poly 2X male offspring which showed a trend towards decreased index

(female: F(2, 32) = 3.78, p < 0.05, Bonferroni post hoc test p > 0.05 for Poly 2X; male: F(2, 26)

= 4.79, p < 0.05, Bonferroni post hoc test p > 0.05 for Poly 1X and Poly 2X). For social

preference test, none of the groups demonstrated a preference for either zone with the exception

of female Poly 2X and male Poly 1X Fmr1 KO offspring that showed a preference for the non-

familiar zone (female Poly 1X: t(26) = 1.62, p = 0.1167; female Poly 2X: t(22) = 3.93, p < 0.001;

male Poly IX: t(22) = 2.47, p < 0.05; male Poly 2X: t(14) = 1.64, p = 0.1121) (Fig. 10B). Similar

results were observed with respect to the social preference index where a trend towards more

preference for the non-familiar zone was found for Poly 2X Fmr1 KO female (F(2, 32) = 2.54, p

= 0.0950) and Poly 1X Fmr1 KO male mice (F(2, 26) = 1.72, p = 0.1168) (Fig. 10D).

Page 44: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

35

Fig. 10 Social behavior of Poly IC treated Fmr1 knockout MIA offspring evaluated by the

modified 3-chamber paradigm. For social interaction test, all groups demonstrated normal

social behavior except for female Poly 1X mice where it showed no significant preference for the

social zone (A). The impairment in this group was reflected by the significant decrease in social

interaction index (C). For social preference test, only female Poly 2X and male Poly 1X mice

showed normal social behavior by preferring the non-familiar zone (B). This result was partially

reflected in the social preference index where Poly 2X female showed increased score, although

not statistically significant (D). Wild-type saline groups (grey) were included as a comparison.

Time spent in each zone was measured in seconds. Each column represents the average ±

S.E.M. One-way ANOVA was performed for each sex separately, followed by Bonferroni’s post

hoc analysis. Using the Grubbs’ test, one value that was furthest from the mean was omitted for

each group. * p < 0.05. ** p < 0.01. *** p < 0.001.

Page 45: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

36

It should be noted that the average time spent in all four zones did not significantly differ

between controls and MIA offspring. Modest deficits were only evident through social

interaction and/or social preference index. Despite this, the deficits observed in social behavior

were limited to female MIA offspring only. Single prenatal LPS treatment caused a strong trend

towards deficits in both social interaction and preference test. This impairment was maintained

in the offspring from double injected dams (i.e. LPS 2X), particularly in the last 5 minutes of the

exploration period (see Appendix 1). In contrast, only female offspring from Poly 1X FXS dams

showed deficits in social interaction and the deficits were not preserved in female Poly 2X

offspring. Therefore, social impairments in MIA offspring were dependent on sex and genotype

of the offspring as well as the immunological memory of the dam (for Poly IC only).

4.4 The Effect of MIA on Repetitive Behavior Evaluated Using Marble Burying Test

Ritualistic and repetitive behavior is the other core symptom of ASD. This behavior can be

assessed in rodents using the marble burying test where increased repetitive behavior is

correlated with more marbles buried (Thomas et al., 2009). The response of MIA offspring was

sex-dependent for this test. For female wild-type mice, MIA offspring showed no difference

with regards to the number of marbles buried (LPS: F(2, 46) = 1.98, p = 0.1237; Poly IC: F(2,

49) = 2.18, p = 0.1237). In contrast, both LPS and Poly IC male wild-type offspring buried

significantly more marbles compared to controls (LPS: F(2, 44) = 8.66, p < 0.001; Bonferroni

post hoc test LPS 1X: p < 0.05, LPS 2X: p < 0.001; Poly IC: F (2, 37) = 5.55, p < 0.01;

Bonferroni post hoc test p < 0.05) (Fig. 11A, B). Saline treated male but not female Fmr1 KO

mice showed a trend towards increased repetitive behavior compared to saline WT mice (female:

t(38) = 0.6257, p = 0.5352; male: t(28) = 1.903, p = 0.0673). Poly IC treatment did not

exacerbate the repetitive behavior of Fmr1 KO mice (female: F (2, 45) = 2.95, p = 0.0628, male:

F (2, 30) = 0.13, p = 0.8746) (Fig. 11C).

To summarize, MIA wild-type offspring demonstrated increased repetitive behavior that was

found in male but not female mice. For Fmr1 knockout mice, marble burying was not affected

by prenatal Poly IC treatment.

Page 46: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

37

Fig. 11 Repetitive behavior of wild-type and Fmr1 knockout MIA offspring evaluated by

the marble burying test. Male wild-type LPS (A) and Poly IC (B) treated MIA offspring

displayed increased repetitive behavior by burying significantly more marbles compared to

control. For female wild-type MIA mice, the number of marbles buried was not different in 1X

offspring and decreased in 2X offspring, although not statistically significant. Compared to

saline wild-type mice, male saline Fmr1 knockout mice showed a trend towards more marble

burying. This increase was not exacerbated by Poly IC treatment in Fmr1 knockout mice (C).

Each column represents the average ± S.E.M. One-way ANOVA was performed for each sex

separately, followed by Bonferroni’s post hoc analysis. Using the Grubbs’ test, one value that

was furthest from the mean was omitted for each group, except for those with n ≤ 8. * p < 0.05.

** p < 0.01. *** p < 0.001.

Page 47: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

38

4.5 The Effect of MIA on Repetitive Behavior Evaluated by Grooming

Repetitive behavior was also evaluated by scoring the time the animal spent grooming during the

first five minutes of social preference test. Similar to marble burying test, LPS female offspring

showed a trend towards decreased repetitive behavior by spending less time grooming (F(2, 33)

= 2.47, p = 0.1005). No significant differences in grooming was found in LPS male offspring

(F(2, 30) = 0.50, p = 0.6142) (Fig. 12A). Interestingly, the lack of change in repetitive behavior

(via marble burying test) in female Poly IC offspring was mirrored in grooming results of wild-

type (F(2, 32) = 0.05, p = 0.9547) and FXS (F(2, 31) = 0.06, p = 0.9463) animals. A strong trend

towards increased repetitive behavior was observed in both Poly IC 1X wild-type (F(2, 25) =

3.23, p = 0.06) and FXS (F(2, 25) = 3.09, p = 0.06) offspring but not Poly IC 2X offspring (Fig.

12B, C).

4.6 The Effect of MIA on the Adult Cerebellum Evaluated by Western Blot

Quantitative western blotting was performed to investigate the expression of markers for

microglia (Iba-1), astrocytes (GFAP) and mature oligodendrocytes (MBP) in the adult

cerebellum. A representative blot for each marker is shown in Fig. 13A – C and the summary of

data and statistics is shown in Fig. 13D. Interestingly, both female and male LPS1X offspring

had approximately the same magnitude of changes in the expression of Iba-1 (~16% increase),

GFAP (~20% increase) and MBP (~10% decrease) compared to control, although not

statistically significant. For MBP, the 17 and 18.5 kDa bands were analyzed together because

they were not well separated on the gel.

Page 48: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

39

Figure 12 Repetitive behavior of MIA wild-type and Fmr1 knockout MIA offspring

evaluated by grooming behavior. This parameter was recorded during the first 5 minutes of the

social preference test. For female, only LPS 2X offspring (A) showed a trend towards decreased

grooming time while other groups did not differ from control. For male, single Poly IC prenatal

injection increased grooming time in both wild-type (B) and Fmr1 knockout (C) offspring,

although not statistically significant. Double Poly IC prenatal treatment did not affect grooming

time in male offspring compared to control (B, C). Each column represents the average ± S.E.M.

One-way ANOVA was performed for each sex separately, followed by Bonferroni’s post hoc

analysis. Using the Grubbs’ test, one value that was furthest from the mean was omitted for each

group, except for those with n ≤ 8.

Page 49: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

40

Fig. 13 Summary of the expression of different glial markers in the adult cerebellum of

LPS offspring quantified by western blot. Representative blots are shown in A – C. Ionized

calcium binding adaptor molecule 1 (Iba-1) (A), glial fibrillary acidic protein (GFAP) (B), and

myelin basic protein (MBP) (C) were used as markers for microglia, astrocytes, and

oligodendrocytes, respectively. Numerical values and statistics are tabulated in D. Each value

represents the average (expressed as a percentage of the respective saline controls) ± S.E.M.

One-way ANOVA was performed for each sex separately. Cerebellar tissue samples were

collected between PND70-90.

Page 50: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

41

5 Discussion

5.1 The Effect of MIA on Body Weight

Sickness behaviors such as weight loss have been reported in animals injected with LPS

(Cloutier et al., 2012) or Poly IC (Cunningham et al., 2012). We postulated that the immune

response from dams injected with these agents will perturb the development of the fetal brain.

However, it is important that the immune responses were not severe enough to become

detrimental to the well-being of the offspring since autistic patients are typically healthy and

have normal body weight (Bolte et al., 2001, Curtin et al., 2005). Therefore, we decided to

assess the general health of the offspring by measuring their weight on PND46.

We found that prenatal LPS injections did not affect the weight of the wild-type offspring (Fig.

4A). Similar results were observed by other groups where they found normal body weight in

offspring from dams that received an acute LPS injection during early or late gestation (Golan et

al., 2005, Ling et al., 2009). It should be noted that chronic LPS injections to the dam decreased

the body weight of male offspring (PND21 to adulthood) (Bakos et al., 2004). In contrast, a high

dose (790 μg/kg) of LPS injection during early pregnancy (compared to 75 μg/kg used in this our

animal model) increased the body weight of adult offspring (Nilsson et al., 2001, Wei et al.,

2007). Therefore, the effects of LPS on offspring weight gain may be dependent on the

treatment regime of the dams. Our model used a low and acute dose of LPS which could explain

the lack of effect on the body weight of the litters.

Saline treated male Fmr1 KO offspring were significantly heavier compared to wild-type mice

(Fig. 4C). This sex difference in body weight was also observed at PND30 by Pacey et al. 2011.

In addition, Bhattacharya et al. 2012 reported that the weight gain was significantly increased

after 6 weeks in male mice which was approximately the age that we weighed our animals. This

weight gain mirrored the accelerated prepubescent growth in human Fragile X patients (Loesch

et al., 1995). Moreover, we found that prenatal Poly IC injection significantly decreased the

weight of male but not female Fragile X offspring (Fig. 4C). This decrease was absent in Poly

IC wild-type animals (Fig. 4B). Several groups with different treatment regimens reported that

Poly IC injection during pregnancy did not affect the weight of wild-type offspring from birth to

early adulthood (Yee et al., 2011, Malkova et al., 2012, Vorhees et al., 2012). However,

offspring from dams that demonstrated a weight loss following Poly IC injection showed reduced

Page 51: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

42

weight at approximately three months of age (Bronson et al., 2011, Vorhee et al., 2012). It is

possible that because body weight was assessed at a single time point, we were unable to capture

the weight change later on in life. It is also possible that the genetic predisposition of Fmr1

knockout mice contributed to an earlier onset of weight loss caused by prenatal Poly IC

treatment. A longitudinal analysis of body weight would help us determine if this was the case.

5.2 The Effect of MIA on Motor activity

Motor activity was tested to evaluate locomotion and exploratory behavior in a novel

environment. Similar to published findings (Golan et al., 2005, Girard et al., 2009, Zager et al.,

2012), acute prenatal LPS treatment did not affect the horizontal or vertical motor activity of the

offspring (Fig. 5). The lack of change in locomotor activity could be due to the serotype of LPS

as well as the frequency and dose of LPS given prenatally. For example, Ling et al., 2009

showed that prenatal injection of a much higher dose of LPS (1 mg/kg) during early gestation

increased the locomotor activity of the offspring at PND90.

Several groups reported that offspring from Poly IC injected dams showed decreased total

distance travelled during open field test (Meyer et al., 2006, Smith et al. 2007, de Miranda et al.,

2010, Malkova, et al, 2012). Some groups also reported thigomotaxis behavior where the Poly

IC offspring spent less time in the center of the open field box (Ozawa et al., 2006, Smith et al.,

2007, Soumiya et al., 2011). Similar to these previous reports, we found that male Poly IC 1X

offspring travelled less distance compare to respective controls (Fig. 6). However, this was not

observed in the female Poly IC 1X offspring or male Poly IC 2X offspring. Rather, female Poly

2X offspring showed significantly increased horizontal activity compare to controls. Most

groups analyzed either only male offspring or a combination of male and female offspring. To

our knowledge, no published reports have studied female Poly IC offspring specifically. It is

possible that the effect of Poly IC on motor activity could be sex-dependent. For example, in the

valporic acid model of ASD, Kim et al., 2013 reported that locomotor activity was only affected

in male and not female offspring.

With regards to thigomotaxis behavior, we did not find that Poly IC animals preferred to occupy

the margin zone more than the center zone (data not shown). This could be because we tested

the animals in smaller activity boxes under dim lighting whereas other groups tested the animals

Page 52: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

43

in larger activity boxes under bright lighting. The latter test conditions may have created a more

stressful environment for the animals, hence inducing thigomotaxis behavior.

In agreement with published literature, we found that both male and female Fmr1 KO mice

exhibited hyperactivity (Spencer et al, 2005, Pacey et al, 2011). Although prenatal Poly IC

treatment did not affect the horizontal activity of Fragile X mice, it did significantly decrease the

vertical movement of both female and male offspring with the exception of male Poly 2X FXS

mice (Fig. 7). Vertical activity can be considered as a form of exploratory behavior that is

associated with the animals’ arousal state and attention in a novel environment (Sadile et al.,

1995, Aspide et al., 1998, 2000). In fact, decreased vertical activity (i.e. rearing) has been

observed in other ASD models including Gabrb3 knockout mice (DeLorey et al., 2008) and wild-

type LPS MIA mice (Liu et al., 2004). Interestingly, Walker et al., 2007 reported significant

decreases in exploratory behavior in adult male rats that underwent developmental cerebellar

suction lesions or intracerebroventricular saporin injections specifically targeting the Purkinje

cells. The association between cerebellar pathologies and lack of exploratory behavior was

reflected in a human study as well. Pierce and Courchesne, 2001 showed that decreased

exploration in autistic patients, measured by the time spent investigating different novel objects

in a large room, was correlated with the extent of cerebellar hypoplasia of vermal lobules VI-VII.

Therefore, decreases in both horizontal and vertical activity could be considered as ASD-

associated phenotypes in mice, and the cerebellum might be a key brain region linked to these

behaviors.

5.3 The Effect of MIA on Social behavior

Impairments in social interaction and communication are one of the core symptoms of ASD.

Since the symptoms persist throughout life, we decided to investigate the social behavior of the

adult MIA offspring. First, prenatal infections caused moderate deficits in female offspring only

(Fig. 8, Fig. 10). All male MIA groups displayed normal social behavior, evident by

demonstrating preference of the social and non-familiar zone. As well, the male social

interaction and social preference index did not significantly differ from the respective controls.

Second, tandem injections of LPS (i.e. LPS 2X) maintained the deficits in social behavior

observed in the LPS 1X female offspring (Fig. 8). Lastly, whereas Poly IC had no effects in any

of the wild-type injected mice, Poly IC 1X treatment did induce deficits in female Fragile X

Page 53: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

44

knockout animals, suggesting a possible genotype effect (Fig. 10). Unlike the LPS 2X female

offspring, tandem injections of Poly IC in FXS animals did not preserve the social deficits

observed in Poly 1X female offspring (Fig. 10).

Overall, the deficits observed in social behavior were modest since the average time the MIA

offspring spent interacting with the social or non-familiar zone did not significantly differ from

the respective saline controls. Deficits were only apparent through the social interaction and/or

preference index. Previous published findings have reported social impairments in both the LPS

(Golan et al., 2006, Kirsten et al., 2010, 2012) and Poly IC model (Shi et al., 2003, Smith et al.,

2007, Soumiya et al., 2011, Malkova et al., 2012). Possible reasons why we did not see such

strong impairments are discussed below.

Kirsten et al. 2010 investigated sociability in rats prenatally treated with LPS by subjecting the

offspring to novel free moving stimulus animal in a cage and manually scoring their play

behavior such as sniffing and mounting. They found that they were only able to detect deficits in

social interaction in adult rats when they isolated the rats (i.e. one rat per cage) for a week before

testing. This was done to increase the rats’ motivation to engage in social interaction with the

novel stimulus rat. We housed our animals in a group setting without social isolation prior to

testing. Thus lower social motivation in the test animals may explain why we were unable to

detect stronger deficits in the MIA model.

Contrary to our findings, Kirsten et al, 2010, 2012 found male-specific deficits in social

interaction in the rat LPS model. They found significant reductions in the male offspring’s

frequency to crawl under/over and mount on the stimulus rat. These two parameters are more

aggressive behaviors that are usually more common in male rodents. Female mice are generally

more docile. For example, male rats had a mounting frequency of 4 (Kirsten et al., 2010) during

the 30 minutes of exploration whereas female only had a frequency of 2.5 (Kirsten et al., 2012).

Since the frequency was already reduced in female control rats, deficits may be harder to detect

in the LPS offspring. Furthermore, they found no difference in less aggressive social behaviors

(i.e. sniffing and following stimulus rat) in either male or female adult LPS rats compared to

controls. Our test condition was less aggressive in comparison because the stimulus mice were

constrained in wire cups and were not in direct contact with the test mouse. Therefore, the lack

Page 54: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

45

of social interaction in female rats observed by Kirsten et al. 2012 does not conflict with the

social deficits we observed using the modified 3-chamber paradigm.

In the Poly IC model, deficits in sociability were previously reported in three different strains

including C57/B6 (Smith et al., 2007, Malkova et al., 2012), ddY (Soumiya et al., 2011), and

BALB (Shi et al. 2003) mice using the 3-chamber paradigm assay. It should be noted that all

findings were reported by Patterson’s group with the exception of Soumiya et al., 2011. The

impairments were present in either only male mice or a combination of both female and male

mice. To our knowledge, no sex differences in social interaction analyses have been specifically

investigated. It should be emphasized that we used a modified version of the 3-chamber

paradigm. Rather than chambers, we confined the area of social interaction to zones which was

much smaller in size. Therefore, interaction with either the stimulus mouse or novel object was

only recorded when the test mouse was in close proximity. This may be a more stringent

measure because in the typical 3-chamber paradigm, time is recorded after the animal enters the

chamber, even though they may not be actually interacting with the stimulus.

Although we were unable to detect a difference in sociability in the Poly IC wild-type animals,

we did find significant decreases in social interaction index in the FXS Poly IC 1X female

offspring (Fig. 10). This suggested that the lack of FMRP in the FXS animals may have

rendered them more vulnerable in the presence of maternal Poly IC injection. Ehninger et al.,

2012 performed a similar study using Tsc2 halpodeficient mice, a genetic model of tuberous

sclerosis which also has high incidences of ASD (Moss et al., 2009). Similar to our results, they

found that deficits in social interaction were only observed in Tsc2 halpodeficient mice (not

wild-type mice) with mid-gestational viral infection using Poly IC. Tsc2 is able to from a

complex with Tsc1 to negatively regulate mammalian target of rapamycin complex 1

(mTORC1). Interestingly, cytokines such as TNF-α are able to inhibit the formation of Tsc1 and

Tsc2 complex via activation of Akt (also known as protein kinase B) (Lee et al, 2007).

Therefore, changes in levels of cytokines plus dampened function of Tsc2 may elevate mTORC1

signaling. The mTORC1 signaling pathway plays an important role in the regulation of the

innate and adaptive immune system (Weichhart et al., 2008, Thomson et al. 2009). For example,

mTORC1 is able to activate signal transducer and activator of transcription 3 (STAT3) which is

essential for the differentiation of Th17 cells (Stepkowski et al., 2008). Interestingly, Sharma et

al., 2010 found that mTOR phosphorylation and activity were elevated in the hippocampus of

Page 55: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

46

juvenile Fmr1 knockout mice. This suggests that deficits observed in Poly IC 1X FXS offspring

may also be linked to disruptions in the mTOR pathway.

In summary, deficits in social interaction were limited to female offspring only. Prenatal

bacterial (LPS) not viral infection (Poly IC) to naïve dams caused modest social impairments in

the female offspring. Furthermore, this deficit was preserved in offspring from LPS dams with

immunological memory (i.e. LPS 2X wild-type). Lastly, prenatal viral infection in the Fmr1 KO

mice caused deficits in social behavior that was not observed in Poly IC treated wild-type

animals, suggestive of possible gene x environment interaction.

5.4 The Effect of MIA on Ritualistic and Repetitive Behavior

Ritualistic and repetitive behavior is the other core symptom of ASD. According to DSM-V, it

can be manifested through stereotyped motor movements, ritualistic behavior (e.g. insistence on

sameness), highly restricted interests, and hyper or hypo-reactivity to sensory input. These

behaviors can be evaluated in animals via different assays. The most established tests to assess

stereotyped motor movements are the marble burying assay and self-grooming behavior (Roullet

and Crawley, 2011). Rodents that exhibit an autistic-like phenotype will bury more marbles and

spend more time grooming.

Our results from the marble burying tests and grooming analyses showed a striking sex

difference in the MIA mice. In female offspring, LPS treatment caused a trend towards

decreased marble burying and grooming (Fig. 11A, 12A) whereas Poly IC treatment caused no

changes in either parameter (Fig. 11B, 12B). In contrast, both male LPS and Poly IC offspring

showed increased repetitive behavior by burying significantly more marbles compared to

controls (Fig. 11A, B). The male-specific increases in repetitive behavior were also observed in

male Poly 1X wild-type offspring where they spent more time grooming (Fig. 12A).

Other groups have reported increased repetitive behavior using the MIA model. Malkova et al.,

2012 found significant increases in marble burying and grooming in male Poly IC offspring. As

well, Kirsten et al., 2012 found that male LPS offspring with an additional LPS exposure had

increased reluctance to switch arms in the T-maze spontaneous alternation test. Although this

test was used to evaluate learning and spatial memory, they suggested that it may also reflect

increased insistence on sameness, which is a form of repetitive behavior. Increased time spent

Page 56: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

47

grooming has been observed in several different ASD mouse models. These include MIA Poly

IC model (Malkova et al., 2012) as well as genetic models of ASD including BTBR T+tf/J mice

(McFarlane et al., 2008, Pearson et al., 2011) and Shank 3 mutant mice (Peca et al., 2011).

However, all of the groups mentioned only used male mice to assess the repetitive behavior.

We saw a male-specific trend towards increased marble burying in saline FXS mice compared to

saline wild-type mice. Similarly, Bhattacharya et al., 2012 reported significant increases in the

number of marbles buried in FXS animals compared to WT animals. However, it should be

noted that three papers published by Paylor’s group found no significant difference between

wild-type and FXS animals (Veeraragavan et al., 2011, Henderson et al., 2012, Thomas et al.,

2012). Both groups used mice with the same genetic background as our animals (C57/B6). One

explanation for this disparity could be that the bedding Paylor’s group used (i.e. SANI-CHIP)

was much easier to bury with than the corn cob bedding used in our study. In fact, the wild-type

animals in their study buried almost 3 times more marbles than in our study. Prenatal Poly IC

treatment to Fmr1 knockout mice caused a trend towards increased grooming time (similar to

WT animals) but did not exacerbate the number of marbles buried. The lack of change in marble

burying is likely due to the ceiling effect where the increases in control Fmr1 KO mice narrowed

the window that Poly IC treatment could exert. In other words, the Fmr1 KO mice may have

already reached the maximum number of marbles that they are able to bury.

In addition to repetitive behavior, increases in self-grooming and marble burying may be

associated with increased anxiety, although there are some caveats. Increases in self-grooming

occur in both low-stress and high-stress situations. In low-stress environments, mice tend to

groom for longer period of time and follow normal progression of grooming activity. In contrast,

during high-stress conditions, mice generally have more bursts of short grooming activity with

abnormal progression, in addition to numerous incomplete and interrupted bouts (Kalueff et al.,

2004). Therefore, more comprehensive analyses of grooming behavior should to be conducted to

identify whether increases in grooming was related to increased anxiety. In terms of marble

burying test, several groups have reported that administration of various anxiolytic (alprazolam,

diazepam, chlordiazepoxide) or antidepressant (paroxetine, fluoxetine, citalopram) drugs

significantly decreased the number of marbles buried (Njung’e and Handley, 1991, Nicolas et al.,

2006, Gomez et al., 2011). However, administration of anxiogenic agents (pramipexole, methyl-

β-carboline-3-carboxylate, yohimbine, meta-chlorophenyl) not only did not increase the number

Page 57: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

48

of marbles buried but significantly decreased burying behavior (Njung’e and Handley, 1991,

Nicolas et al., 2006, Gomez et al., 2011). Furthermore, Thomas et al., 2009 found that marble

burying behavior did not correlate with anxiety in open field test and light-dark exploration.

This suggests that marble burying may be more suitable as a predicative test to screen the effects

of anxiolytic drugs rather than a direct test for anxiety. Therefore, self-grooming and marble

burying test may, at best, only partially reflect anxiety in rodents. More specific tests such as

elevated plus maze and open field test may be more suitable for evaluating anxious behavior.

Our study was the first to demonstrate the sex difference in stereotyped repetitive behavior in

MIA animal models. The strong sex-bias observed in marble burying assay and grooming is

reflected in human ASD patients. In fact, several studies demonstrated that males have higher

repetitive and/or restricted interests compared to females (Hattier, 2011, Sipes et al., 2011,

Mandy et al., 2012, Werling et al, 2013). However, I should note that there are also published

studies that found no gender differences in young ASD patients in terms of repetitive behavior

(Worley et al., 2011 (children and adolescent), Andersson et al., 2013 (pre-school children with

suspected of ASD)). In addition, the lack of change in motor stereotypies in females does not

exclude the fact that they may still demonstrate other forms of repetitive behavior such as

restricted interest and ritualistic behavior.

Both animal and human studies suggest that increased repetitive behavior may be linked to

cerebellar pathologies. In rodents, increases in repetitive behavior were reported in mice with

mutations restricted to Purkinje cell degeneration (i.e. Tsc1 mutant mice, Tsai et al., 2012) or

depletion of Purkinje cells during development (i.e. lurcher mice, Martin et al, 2010, Dickson et

al., 2010). In humans, Rojas et al., 2006 reported significant negative partial correlations

between grey matter volume in specific regions of the cerebellum (i.e. left cerebellar lobule VI

and right cerebellar Crus 1) and severity of repetitive and stereotype behaviors in 24 male ASD

patients. Similarly, in a study with 14 ASD patients, Pierce and Courchesne, 2001 found that the

area of cerebellar vermis lobules VI – VII was significantly negatively correlated with the rates

of stereotyped behavior. These studies suggest that increased repetitive behavior may be linked

to cerebellar pathologies.

A summary chart of all behavioral findings for wild-type and Fmr1 knockout MIA offspring can

be found in Appendix 2.

Page 58: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

49

5.5 The Effect of MIA on the Adult Cerebellum

We wanted to investigate the pathology of the cerebellum because this brain structure has been

found to be consistently implicated in autism spectrum disorder. Our goal was to investigate

whether the pathologies observed in human ASD patients are present in our MIA mouse model.

First, we wanted to investigate if the presence of neuroinflammation persisted until adulthood.

We expected to see increases in microglia and/or astrocyte expression because this was observed

in human post-mortem tissues of ASD patients (Vargas et al, 2005). As well, several groups

have shown increases in the expression of microglia in certain brain regions of the LPS model

(Larouche et al, 2005, Roumier et al., 2008, Girard et al, 2010) and Poly IC model (Jackel et al.,

2011). Ling et al., 2009 reported that a single LPS injection of during early gestation increased

activated microglia in substantia nigra of adult offspring at 4, 14, and 17 months. Our results

thus far also show possible increases in microglia in the adult cerebellum (Fig. 13). However, it

was difficult to detect microglia marker (Iba-1) on western blots because the expression was very

low in whole cerebellar samples and thus required large amount of protein to be loaded.

Therefore, for future experiments, immunohistochemistry may be a better method for evaluating

the density of microglia in this particular brain structure.

Increases in the expression of astrocyte marker (GFAP) were reported in several brain regions of

the LPS model (Cai et al., 2000, Paintlia et al., 2004, 2008). Our results thus far showed

approximately a 20% increase in GFAP expression in the cerebellum of both male and female

LPS 1X groups, although not statistically significant. One possible reason for this could be

because we used tissue homogenates of the entire cerebellum. If GFAP expression was

increased in only certain regions of the cerebellum, analysis with the entire tissue may mask the

changes in a specific region(s).

Apart from the changes in microglia and astrocytes, several groups also reported decreases in

expression of MBP, a marker for mature oligodendrocytes. This was reported for various ages in

several brain regions including periventricular white matter at PND7 (Kumral et al., 2007,

Yesilirmak et al., 2007), corpus callosum at PND9 – 30 (Paintlia et al., 2004, 2008), and external

and internal capsule at PND7 (Rousset et al., 2006, 2008). Decreases in MBP were also found in

the hippocampal CA1 and CA3 region of Poly IC treated mice at PND14 (Makinodan et al.,

2008). Thus far, no group has reported MBP changes in the cerebellum. Our results showed

Page 59: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

50

decreases in MBP in both LPS 1X and LPS 2X female and male offspring, although not

statistically significant. It may be beneficial to confirm this with another myelin marker such as

2',3'-Cyclic-nucleotide 3'-phosphodiesterase (CNPase) or proteolipid protein (PLP) to determine

if the decrease is real and reproducible.

To summarize, western blotting results were suggestive of potential signs of neuroinflammation

and white matter changes in LPS offspring. However, the n-values for these experiments were

too low to reach a firm conclusion.

5.6 Potential Biological Mechanisms for Sex-Dependent Behavioral Changes

In the marble burying test, we found that prenatal immune activation caused significant male-

specific increases in repetitive behavior. This suggests that the immune response maybe more

severe in male rather than female mice. Some explanations for this sex difference in immune

response are as follows. First of all, the placenta, derived from maternal and fetal origin may

contribute to the sex differences during prenatal programming (Rossant and Cross, 2001). In the

placenta of mothers with mild asthma, the mRNA expression of various cytokines (TNFα, IL-1β,

IL-6, IL-8, and IL5) was increased for female but not male fetuses (Scott et al., 2009). The

production of these cytokines can affect the hypothalamus-pituitary-adrenal axis which is

involved in the stress pathway activation and immune system. Cytokines such as IL-1β, which is

up-regulated after LPS injection, can promote the release of corticotropin releasing hormone and

adrenocorticotropic hormone from the hypothalamus and pituitary, respectively (Irwin and

Miller, 2007). In turn, glucocorticoids production, triggered by adrenocorticotropic hormone,

can readily pass through the placenta and participate in fetal brain development. In fact, Cui et

al., 2011 found LPS injection during mid or late-gestation increased the plasma

adrenocorticotropic hormone and corticosterone (a main glucocorticoid) levels in the dam. One

of the most important functions of glucocorticoids is its regulation of placental glucose

transporters. Exogenous glucocorticoid administration led to the reduction of glucose transporter

1 placental expression. However, this reduction was maintained in males but not in females later

in gestation (O’Connell et al., 2011).

Another explanation for the sex differences could be contributed to genetic factors. In contrast to

males who have only one X chromosome, females have two X chromosomes where the alleles

Page 60: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

51

on one or the other are inactive. However, approximately 15% of X-linked genes escape this

inactivation to some degree (Carrel et al., 2005). There are many immune related genes on the

X-chromosome. These include immune-associated receptors (e.g. IL receptors), immune-

response related proteins (e.g. ELK1 – involved in B-cell development), and transcriptional and

translational control effectors (e.g. NF-kB repressing factor) (Fish, 2008). This could explain the

differences in female and male fetuses’ response to immune activation. In fact, 65 genes were

found to be changed in the placentae of mothers with chronic asthma. Interestingly, only 6 genes

were altered in male placentae compared to the 59 genes that were altered in female placentae.

The genes investigated are involved in a variety of functions including immune response, cellular

growth, cell-cell signaling, and gene expression (Osei-Kumah et al., 2011). Depending on the

function of the gene, dramatic changes in gene expression could be a protective mechanism for

female fetuses that is lacking in the male fetuses.

Page 61: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

52

6 Conclusions

The main goal of this thesis was to evaluate the validity of the MIA mouse model for autism

spectrum disorder by investigating the core and associated-ASD behaviors and adult cerebellar

pathologies. We found that prenatal bacterial infection via LPS to naïve dams induced both core

symptoms of ASD in wild-type offspring (i.e. LPS 1X). In addition, the deficits in social

interaction and increases in repetitive behavior were preserved in LPS 2X offspring. In

comparison, prenatal viral infection (Poly IC) in wild-type offspring was only able to increase

the repetitive behavior.

Interestingly, although social interaction was not impaired in Poly 1X and 2X wild-type mice,

prenatal Poly IC injection to naïve dams impaired the social behavior in Fmr1 knockout mice. In

fact, prenatal Poly IC treatment caused other phenotypes in the Fmr1 knockout mice that were

absent in the Poly IC wild-type mice. These included significant reductions in body weight and

rearing. Genotype-specific impairments in Poly IC Fmr1 knockout mice suggested that the lack

of FMRP protein may have rendered the mice more vulnerable to prenatal viral infection.

Lastly, prenatal bacterial and viral infection caused striking sex-dependent phenotypes that were

absent in saline control mice. Specifically, we found that deficits in social interaction were

female-specific whereas increases in repetitive behavior were male-specific. In addition, western

blots performed on the adult cerebellum were suggestive of possible increases and decreases in

the expression of astrocytes and mature oligodendrocytes, respectively. To summarize, we found

that the ASD-related behavioral phenotypes observed in the MIA model were dependent on the

sex and genotype of the offspring, type of prenatal infection, and/or immunological memory of

the dam.

Page 62: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

53

7 Future Directions

Although we investigated the core symptoms of ASD in this study, it would be beneficial to see

if these core phenotypes can be replicated using other behavioral assays. One of the most

interesting effects observed based on the MIA models was the strong male-specific increases in

motor stereotypies. It would be interesting to investigate if the sex difference is preserved in

other forms of repetitive behavior. For example, insistence on sameness can be evaluated using

the reversal learning tasks to assess the likelihood of the animal to change their established

habits. Restricted interest is another form of repetitive behavior that can be measured by

counting the frequency of nose-pokes into holes baited with a stimulus. We also found female-

specific modest deficits in social interaction. This may be improved by subjecting the animals to

isolation prior to testing to increase their motivation to engage in social interaction. As well, we

can look at the progression of the social behavior across different time intervals. Other assays

can be used to assess other forms of social behavior such as social dominance using the tube test.

We did not specifically investigate communication in the MIA model, although it is linked with

social behavior. This can be evaluated using ultrasonic vocalization and measuring pup retrieval

time by the dams. Associated-ASD behaviors such as anxiety should also be investigated.

Grooming is a type of repetitive behavior that may be linked with increased anxiety (Kalueff et

al., 2004, Thomas et al., 2009). Grooming in high stress situations involves “frequent bursts of

rapid short grooming activity with abnormal progression and frequent incomplete and interrupted

bouts” (Fentress, 1977). It may be worthwhile to perform more in-depth analyses of grooming

behavior to identify if excessive grooming observed in MIA offspring is caused by the high

stress social environment during the social preference test. If grooming was increased due to

stress, it may be of interest to use more specific assays that are catered to anxiety such as open

field test, light/dark box, and elevated plus maze test. Animals that are more anxious will spend

less time in the open arm, brightly lit area of the box, and center of the open field.

Molecularly, we can further our investigation on the presence of neuroinflammation in the adult

cerebellum of the MIA model. Our results thus far with the LSP model were suggestive of

possible changes in the expression of microglia, astrocyte and oligodendrocyte markers.

Additional analyses using immunohistochemistry would be helpful to identify the particular

region that may be affected. Furthermore, the number and size of Purkinje cells could be

Page 63: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

54

examined to determine whether the reductions observed in ASD patients are present in the MIA

model. Lastly, since Fmr1 knockout mice showed more severe phenotypes after prenatal Poly IC

treatment compared to control, it would be interesting to elucidate the possible pathway(s)

involved. Further investigation of the mTOR pathway may be a fruitful endeavor since it is

misregulated (over-active) in Fragile X syndrome and is involved in the modulation of both the

innate and adaptive immune responses.

Page 64: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

55

References

Aaltonen, R., Heikkinen, T., Hakala, K., Laine, K., & Alanen, A. (2005). Transfer of

proinflammatory cytokines across term placenta. Obstetrics and Gynecology, 106(4), 802-807.

Abdallah, M. W., Larsen, N., Mortensen, E. L., Atladóttir, H. Ó., Nørgaard-Pedersen, B.,

Bonefeld-Jørgensen, E. C., . . . Hougaard, D. M. (2012). Neonatal levels of cytokines and risk of

autism spectrum disorders: An exploratory register-based historic birth cohort study utilizing the

danish newborn screening biobank. Journal of Neuroimmunology, 252(1-2), 75-82.

Abrahams, B. S., & Geschwind, D. H. (2008). Advances in autism genetics: On the threshold of

a new neurobiology. Nature Reviews Genetics, 9(5), 341-355.

Akshoomoff, N., Lord, C., Lincoln, A. J., Courchesne, R. Y., Carper, R. A., Townsend, J., &

Courchesne, E. (2004). Outcome classification of preschool children with autism spectrum

disorders using MRI brain measures.Journal of the American Academy of Child and Adolescent

Psychiatry, 43(3), 349-357.

Alexopoulou, L., Holt, A. C., Medzhitov, R., & Flavell, R. A. (2001). Recognition of double-

stranded RNA and activation of NF-κB by toll-like receptor 3. Nature, 413(6857), 732-738.

Andersson, G. W., Gillberg, C., & Miniscalco, C. (2013). Pre-school children with suspected

autism spectrum disorders: Do girls and boys have the same profiles? Research in

Developmental Disabilities, 34(1), 413-422.

Ashdown, H., Dumont, Y., Ng, M., Poole, S., Boksa, P., & Luheshi, G. N. (2006). The role of

cytokines in mediating effects of prenatal infection on the fetus: Implications for schizophrenia.

Molecular Psychiatry, 11(1), 47-55.

Ashwood, P., Enstrom, A., Krakowiak, P., Hertz-Picciotto, I., Hansen, R. L., Croen, L. A., . . .

Van de Water, J. (2008). Decreased transforming growth factor beta1 in autism: A potential link

between immune dysregulation and impairment in clinical behavioral outcomes. Journal of

Neuroimmunology, 204(1-2), 149-153.

Page 65: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

56

Ashwood, P., Krakowiak, P., Hertz-Picciotto, I., Hansen, R., Pessah, I., & Van de Water, J.

(2011). Elevated plasma cytokines in autism spectrum disorders provide evidence of immune

dysfunction and are associated with impaired behavioral outcome. Brain, Behavior, and

Immunity, 25(1), 40-45.

Aspide, R., Fresiello, A., De Filippis, G., Gironi Carnevale, U. A., & Sadile, A. G. (2000). Non-

selective attention in a rat model of hyperactivity and attention deficit: Subchronic

methylphenydate and nitric oxide synthesis inhibitor treatment. Neuroscience and Biobehavioral

Reviews, 24(1), 59-71.

Aspide, R., Gironi Carnevale, U. A., Sergeant, J. A., & Sadile, A. G. (1998). Non-selective

attention and nitric oxide in putative animal models of attention-deficit hyperactivity disorder.

Behavioral Brain Research, 95(1), 123-133.

Atladóttir, H. Ó, Thorsen, P., Østergaard, L., Schendel, D. E., Lemcke, S., Abdallah, M., et al.

(2010). Maternal infection requiring hospitalization during pregnancy and autism spectrum

disorders. Journal of Autism and Developmental Disorders, 40(12), 1423-1430.

Atladóttir, H. Ó., Henriksen, T. B., Schendel, D. E., & Parner, E. T. (2012). Autism after

infection, febrile episodes, and antibiotic use during pregnancy: An exploratory study. Pediatrics,

130(6), e1447-e1454.

Auyeung, B., Baron-Cohen, S., Ashwin, E., Knickmeyer, R., Taylor, K., & Hackett, G. (2009).

Fetal testosterone and autistic traits. British Journal of Psychology, 100(1), 1-22.

Auyeung, B., Taylor, K., Hackett, G., & Baron-Cohen, S. (2010). Foetal testosterone and autistic

traits in 18 to 24-month-old children. Molecular Autism, 1(1)

Bailey, A., Le Couteur, A., Gottesman, I., Bolton, P., Simonoff, E., Yuzda, E., & Rutter, M.

(1995). Autism as a strongly genetic disorder: Evidence from a british twin study. Psychological

Medicine, 25(1), 63-77.

Bailey, A., Luthert, P., Dean, A., Harding, B., Janota, I., Montgomery, M., . . . Lantos, P. (1998).

A clinicopathological study of autism. Brain, 121(5), 889-905.

Page 66: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

57

Bakos, J., Duncko, R., Makatsori, A., Pirnik, Z., Kiss, A., & Jezova, D. (2004). Prenatal immune

challenge affects growth, behavior, and brain dopamine in offspring

Bassell, G. J., & Warren, S. T. (2008). Fragile X syndrome: Loss of local mRNA regulation

alters synaptic development and function. Neuron, 60(2), 201-214.

Baranek, G. T., David, F. J., Poe, M. D., Stone, W. L., & Watson, L. R. (2006). Sensory

experiences questionnaire: Discriminating sensory features in young children with autism,

developmental delays, and typical development. Journal of Child Psychology and Psychiatry and

Allied Disciplines, 47(6), 591-601.

Baron-Cohen, S., Lombardo, M. V., Auyeung, B., Ashwin, E., Chakrabarti, B., & Knickmeyer,

R. (2011). Why are autism spectrum conditions more prevalent in males? PLoS Biology, 9(6),

e1001081.

Barrientos, R. M., Frank, M. G., Hein, A. M., Higgins, E. A., Watkins, L. R., Rudy, J. W., &

Maier, S. F. (2009). Time course of hippocampal IL-1 β and memory consolidation impairments

in aging rats following peripheral infection. Brain, Behavior, and Immunity, 23(1), 46-54.

Bhattacharya, A., Kaphzan, H., Alvarez-Dieppa, A. C., Murphy, J. P., Pierre, P., & Klann, E.

(2012). Genetic removal of p70 S6 kinase 1 corrects molecular, synaptic, and behavioral

phenotypes in fragile X syndrome mice. Neuron, 76(2), 325-337.

Bolte, S., Ozkara, N., & Poustka, F. (2002). Autism spectrum disorders and low body weight: Is

there really a systematic association? International Journal of Eating Disorders, 31(3), 349-351.

Borrell, J., et al. "Prenatal Immune Challenge Disrupts Sensorimotor Gating in Adult Rats:

Implications for the Etiopathogenesis of Schizophrenia." Neuropsychopharmacology 26.2

(2002): 204-15.

Bronson, S. L., Ahlbrand, R., Horn, P. S., Kern, J. R., & Richtand, N. M. (2011). Individual

differences in maternal response to immune challenge predict offspring behavior: Contribution of

environmental factors. Behavioral Brain Research, 220(1), 55-64.

Broussard, Dianne M. The Cerebellum: Learning, Language, Movement, and Social Skills. 1st

ed. N.p.: J. Wiley, 2013. Print.

Page 67: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

58

Bsibsi, M., Ravid, R., Gveric, D., & Van Noort, J. M. (2002). Broad expression of toll-like

receptors in the human central nervous system. Journal of Neuropathology and Experimental

Neurology, 61(11), 1013-1021.

Butovsky, O., Ziv, Y., Schwartz, A., Landa, G., Talpalar, A. E., Pluchino, S., . . . Schwartz, M.

(2006). Microglia activated by IL-4 or IFN-γ differentially induce neurogenesis and

oligodendrogenesis from adult stem/progenitor cells. Molecular and Cellular Neuroscience,

31(1), 149-160.

Buxbaum, J. D., Silverman, J. M., Smith, C. J., Greenberg, D. A., Kilifarski, M., Reichert, J., . . .

Vitale, R. (2002). Association between a GABRB3 polymorphism and autism. Molecular

Psychiatry, 7(3), 311-316.

Cai, Z., Pan, Z. -., Pang, Y., Evans, O. B., & Rhodes, P. G. (2000). Cytokine induction in fetal rat

brains and brain injury in neonatal rats after maternal lipopolysaccharide administration.

Pediatric Research, 47(1), 64-72.

Cai, Z., Lin, S., Pang, Y., & Rhodes, P. G. (2004). Brain injury induced by intracerebral injection

of interleukin-1beta and tumor necrosis factor-alpha in the neonatal rat. Pediatric Research,

56(3), 377-384.

Carrel, L., & Willard, H. F. (2005). X-inactivation profile reveals extensive variability in X-

linked gene expression in females. Nature, 434(7031), 400-404.

Center for Health and Disease. (2010, May). Autism spectrum disorder (ASD) screening and

diagnosis. Retrieved from http://www.cdc.gov/ncbddd/autism/screening.html

Center for Health and Disease. (2012, March). Autism spectrum disorder (ASD) facts about

ASD. Retrieved from http://www.cdc.gov/ncbddd/autism/facts.html

Charman, T., Pickles, A., Simonoff, E., Chandler, S., Loucas, T., & Baird, G. (2011). IQ in

children with autism spectrum disorders: Data from the special needs and autism project

(SNAP). Psychological Medicine, 41(3), 619-627.

Chess, S. (1971). Autism in children with congenital rubella. Journal of Autism and Childhood

Schizophrenia, 1(1), 33-47.

Page 68: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

59

Chess, S., Fernandez, P., & Korn, S. (1978). Behavioral consequences of congenital rubella.

Journal of Pediatrics, 93(4), 699-703.

Cloutier, C. J., Rodowa, M. -., Cross-Mellor, S. K., Chan, M. Y. T., Kavaliers, M., &

Ossenkopp, K. -. (2012). Inhibition of LiCl-induced conditioning of anticipatory nausea in rats

following immune system stimulation: Comparing the immunogens lipopolysaccharide, muramyl

dipeptide, and polyinosinic: Polycytidylic acid. Physiology and Behavior, 106(2), 243-251..

Couper, J. J., & Sampson, A. J. (2003). Children with autism deserve evidence-based

intervention. Medical Journal of Australia, 178(9), 424-425.

Courchesne, E., Karns, C. M., Davis, H. R., Ziccardi, R., Carper, R. A., Tigue, Z. D., . . .

Courchesne, R. Y. (2001). Unusual brain growth patterns in early life in patients with autistic

disorder: An MRI study. Neurology,57(2), 245-254.

Courchesne, E., Redcay, E., & Kennedy, D. P. (2004). The autistic brain: Birth through

adulthood. Current Opinion in Neurology, 17(4), 489-496.

Crampton, S. J., Collins, L. M., Toulouse, A., Nolan, Y. M., & O'Keeffe, G. W. (2012).

Exposure of foetal neural progenitor cells to IL-1β impairs their proliferation and alters their

differentiation - A role for maternal inflammation? Journal of Neurochemistry, 120(6), 964-973.

Croonenberghs, J., Bosmans, E., Deboutte, D., Kenis, G., & Maes, M. (2002). Activation of the

inflammatory response system in autism. Neuropsychobiology, 45(1), 1-6.

Cui, K., Luheshi, G. N., & Boksa, P. (2011). Effects of endogenous glucocorticoid secretion on

the interleukin-6 response to bacterial endotoxin in pregnant and non-pregnant rats. Journal of

Endocrinology, 209(1), 95-103.

Cunningham, C., Campion, S., Teeling, J., Felton, L., & Perry, V. H. (2007). The sickness

behavior and CNS inflammatory mediator profile induced by systemic challenge of mice with

synthetic double-stranded RNA (poly I:C). Brain, Behavior, and Immunity, 21(4), 490-502.

Curtin, C., Bandini, L. G., Perrin, E. C., Tybor, D. J., & Must, A. (2005). Prevalence of

overweight in children and adolescents with attention deficit hyperactivity disorder and autism

spectrum disorders: A chart review. BMC Pediatrics, 5.

Page 69: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

60

de la Mano, A., Gato, A., Alonso, M. I., Carnicero, E., Martín, C., & Moro, J. A. (2007). Role of

interleukin-1β in the control of neuroepithelial proliferation and differentiation of the spinal cord

during development.Cytokine, 37(2), 128-137.

de Miranda, J., Yaddanapudi, K., Hornig, M., Villar, G., Serge, R., & Ian Lipkin, W. (2010).

Induction of toll-like receptor 3-mediated immunity during gestation inhibits cortical

neurogenesis and causes behavioral disturbances. MBio, 1(4)

DeLorey, T. M., Sahbaie, P., Hashemi, E., Homanics, G. E., & Clark, J. D. (2008). Gabrb3 gene

deficient mice exhibit impaired social and exploratory behaviors, deficits in non-selective

attention and hypoplasia of cerebellar vermal lobules: A potential model of autism spectrum

disorder. Behavioral Brain Research, 187(2), 207-220.

Depino, A. M., Lucchina, L., & Pitossi, F. (2011). Early and adult hippocampal TGF-β1

overexpression have opposite effects on behavior. Brain, Behavior, and Immunity, 25(8), 1582-

1591.

Deverman, B. E., & Patterson, P. H. (2009). Cytokines and CNS development. Neuron, 64(1),

61-78.

Di Castro, M. A., Chuquet, J., Liaudet, N., Bhaukaurally, K., Santello, M., Bouvier, D., . . .

Volterra, A. (2011). Local ca 2+ detection and modulation of synaptic release by astrocytes.

Nature Neuroscience, 14(10), 1276-1284.

Dickson, P. E., Rogers, T. D., Mar, N. D., Martin, L. A., Heck, D., Blaha, C. D., . . . Mittleman,

G. (2010). Behavioral flexibility in a mouse model of developmental cerebellar purkinje cell

loss. Neurobiology of Learning and Memory, 94(2), 220-228.

Ecker, C., Suckling, J., Deoni, S. C., Lombardo, M. V., Bullmore, E. T., Baron-Cohen, S., . . .

Murphy, D. G. M. (2012). Brain anatomy and its relationship to behavior in adults with autism

spectrum disorder: A multicenter magnetic resonance imaging study. Archives of General

Psychiatry, 69(2), 195-209.

Page 70: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

61

Ehninger, D., Sano, Y., De Vries, P. J., Dies, K., Franz, D., Geschwind, D. H., . . . Silva, A. J.

(2012). Gestational immune activation and Tsc2 haploinsufficiency cooperate to disrupt fetal

survival and may perturb social behavior in adult mice. Molecular Psychiatry, 17(1), 62-70.

Eisenstein, M. (2012). Treatments: In the waiting room. Nature, 491(7422 SUPPL.), S14-S16.

Eluvathingal, T. J., Behen, M. E., Chugani, H. T., Janisse, J., Bernardi, B., Chakraborty, P., . . .

Chugani, D. C. (2006). Cerebellar lesions in tuberous sclerosis complex: Neurobehavioral and

neuroimaging correlates.Journal of Child Neurology, 21(10), 846-851.

Emanuele, E., Boso, M., Brondino, N., Pietra, S., Barale, F., Ucelli di Nemi, S., & Politi, P.

(2010). Increased serum levels of high mobility group box 1 protein in patients with autistic

disorder. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 34(4), 681-683.

Ertan, G., Arulrajah, S., Tekes, A., Jordan, L., & Huisman, T. A. G. M. (2010). Cerebellar

abnormality in children and young adults with tuberous sclerosis complex: MR and diffusion

weighted imaging findings. [Anomalie cérébelleuse chez des enfants et adultes jeunes présentant

une sclérose tubéreuse complexe: aspects en IRM cérébrale et imagerie de diffusion] Journal of

Neuroradiology, 37(4), 231-238.

Estécio, M. R. H., Fett-Conte, A. C., Varella-Garcia, M., Fridman, C., & Silva, A. E. (2002).

Molecular and cytogenetic analyses on brazilian youths with pervasive developmental disorders.

Journal of Autism and Developmental Disorders, 32(1), 35-41.

Farina, C., Krumbholz, M., Giese, T., Hartmann, G., Aloisi, F., & Meinl, E. (2005). Preferential

expression and function of toll-like receptor 3 in human astrocytes. Journal of

Neuroimmunology, 159(1-2), 12-19.

Fatemi, S. H., Aldinger, K. A., Ashwood, P., Bauman, M. L., Blaha, C. D., Blatt, G. J., . . .

Welsh, J. P. (2012). Consensus paper: Pathological role of the cerebellum in autism. Cerebellum,

11(3), 777-807.

Fatemi, S. H., Halt, A. R., Realmuto, G., Earle, J., Kist, D. A., Thuras, P., & Metz, A. (2002).

Purkinje cell size is reduced in cerebellum of patients with autism. Cellular and Molecular

Neurobiology, 22(2), 171-175.

Page 71: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

62

Fish, E. N. (2008). The X-files in immunity: Sex-based differences predispose immune

responses.Nature Reviews Immunology, 8(9), 737-744.

Forrest, C. M., Khalil, O. S., Pisar, M., Smith, R. A., Darlington, L. G., & Stone, T. W. (2012).

Prenatal activation of toll-like receptors-3 by administration of the viral mimetic poly(I:C)

changes synaptic proteins, N-methyl-D-aspartate receptors and neurogenesis markers in

offspring. Molecular Brain,5(1)

Fortier, M. -., Luheshi, G. N., & Boksa, P. (2007). Effects of prenatal infection on prepulse

inhibition in the rat depend on the nature of the infectious agent and the stage of pregnancy.

Behavioral Brain Research,181(2), 270-277.

Garay, P. A., Hsiao, E. Y., Patterson, P. H., & McAllister, A. K. (2012). Maternal immune

activation causes age- and region-specific changes in brain cytokines in offspring throughout

development. Brain, Behavior, and Immunity,

Gillberg, C., Cederlund, M., Lamberg, K., & Zeijlon, L. (2006). Brief report: "the autism

epidemic". the registered prevalence of autism in a swedish urban area. Journal of Autism and

Developmental Disorders, 36(3), 429-435.

Gilmore, J. H., Lin, W., Prastawa, M. W., Looney, C. B., Vetsa, Y. S. K., Knickmeyer, R. C., . . .

Gerig, G. (2007). Regional gray matter growth, sexual dimorphism, and cerebral asymmetry in

the neonatal brain. Journal of Neuroscience, 27(6), 1255-1260.

Girard, S., Kadhim, H., Beaudet, N., Sarret, P., & Sébire, G. (2009). Developmental motor

deficits induced by combined fetal exposure to lipopolysaccharide and early neonatal

hypoxia/ischemia: A novel animal model for cerebral palsy in very premature infants.

Neuroscience, 158(2), 673-682.

Girard, S., et al. "IL-1 Receptor Antagonist Protects Against Placental and Neurodevelopmental

Defects Induced by Maternal Inflammation." J.Immunol. (2010).

Goines, P. E., & Ashwood, P. (2012). Cytokine dysregulation in autism spectrum disorders

(ASD): Possible role of the environment. Neurotoxicology and Teratology.

Page 72: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

63

Goines, P. E., Croen, L. A., Braunschweig, D., Yoshida, C. K., Grether, J., Hansen, R., . . . Van

De Water, J. (2011). Increased midgestational IFN-γ, IL-4 and IL-5 in women bearing a child

with autism: A case-control study. Molecular Autism, 2(1).

Golan, H. M., Lev, V., Hallak, M., Sorokin, Y., & Huleihel, M. (2005). Specific

neurodevelopmental damage in mice offspring following maternal inflammation during

pregnancy. Neuropharmacology, 48(6), 903-917.

Gomez, R., Romero, R., Ghezzi, F., Bo Hyun Yoon, Mazor, M., & Berry, S. M. (1998). The fetal

inflammatory response syndrome. American Journal of Obstetrics and Gynecology, 179(1), 194-

202.

Good, C. D., Johnsrude, I., Ashburner, J., Henson, R. N. A., Friston, K. J., & Frackowiak, R. S.

J. (2001). Cerebral asymmetry and the effects of sex and handedness on brain structure: A voxel-

based morphometric analysis of 465 normal adult human brains. NeuroImage, 14(3), 685-700.

Goshen, I., Kreisel, T., Ounallah-Saad, H., Renbaum, P., Zalzstein, Y., Ben-Hur, T., . . . Yirmiya,

R. (2007). A dual role for interleukin-1 in hippocampal-dependent memory processes.

Psychoneuroendocrinology, 32(8-10), 1106-1115.

Hagerman, R. J., & Hagerman, P. J. (2002). The fragile X premutation: Into the phenotypic fold.

Current Opinion in Genetics and Development, 12(3), 278-283.

Hallmayer, J., Cleveland, S., Torres, A., Phillips, J., Cohen, B., Torigoe, T., . . . Risch, N. (2011).

Genetic heritability and shared environmental factors among twin pairs with autism. Archives of

General Psychiatry,68(11), 1095-1102.

Hamo, L., Stohlman, S. A., Otto-Duessel, M., & Bergmann, C. C. (2007). Distinct regulation of

MHC molecule expression on astrocytes and microglia during viral encephalomyelitis. Glia,

55(11), 1169-1177.

Hattier, M. A., Matson, J. L., Tureck, K., & Horovitz, M. (2011). The effects of gender and age

on repetitive and/or restricted behaviors and interests in adults with autism spectrum disorders

and intellectual disability. Research in Developmental Disabilities, 32(6), 2346-2351.

Page 73: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

64

Harris, S. W., Hessl, D., Goodlin-Jones, B., Ferranti, J., Bacalman, S., Barbato, I., . . . Hagerman,

R. J. (2008). Autism profiles of males with fragile X syndrome. American Journal on Mental

Retardation, 113(6), 427-438.

Henderson, C., Wijetunge, L., Kinoshita, M. N., Shumway, M., Hammond, R. S., Postma, F. R.,

. . . Healy, A. M. (2012). Reversal of disease-related pathologies in the fragile X mouse model by

selective activation of GABA B receptors with arbaclofen. Science Translational Medicine,

4(152)

Holmlund, U., Cebers, G., Dahlfors, A. R., Sandstedt, B., Bremme, K., Ekström, E. S., &

Scheynius, A. (2002). Expression and regulation of the pattern recognition receptors toll-like

receptor-2 and toll-like receptor-4 in the human placenta. Immunology, 107(1), 145-151.

Hsiao, E. Y., & Patterson, P. H. (2011). Activation of the maternal immune system induces

Irwin, M. R., & Miller, A. H. (2007). Depressive disorders and immunity: 20 years of progress

and discovery. Brain, Behavior, and Immunity, 21(4), 374-383.

Jimenez-Gomez, C., Osentoski, A., & Woods, J. H. (2011). Pharmacological evaluation of the

adequacy of marble burying as an animal model of compulsion and/or anxiety. Behavioral

Pharmacology,22(7), 711-713.

Jin, P., & Warren, S. T. (2003). New insights into fragile X syndrome: From molecules to

neurobehaviors. Trends in Biochemical Sciences, 28(3), 152-158.

Juckel, G., Manitz, M. P., Brüne, M., Friebe, A., Heneka, M. T., & Wolf, R. J. (2011). Microglial

activation in a neuroinflammational animal model of schizophrenia - a pilot study. Schizophrenia

Research,131(1-3), 96-100.

Kalueff, A. V., & Tuohimaa, P. (2004). Grooming analysis algorithm for neurobehavioral stress

research. Brain Research Protocols, 13(3), 151-158.

Page 74: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

65

Kalueff, A. V., & Tuohimaa, P. (2005). The grooming analysis algorithm discriminates between

different levels of anxiety in rats: Potential utility for neurobehavioral stress research. Journal of

Neuroscience Methods, 143(2), 169-177.

Karima, R., Matsumoto, S., Higashi, H., & Matsushima, K. (1999). The molecular pathogenesis

of endotoxic shock and organ failure. Molecular Medicine Today, 5(3), 123-132.

Kaufmann, W. E., Cooper, K. L., Mostofsky, S. H., Capone, G. T., Kates, W. R., Newschaffer,

C. J., . . . Lanham, D. C. (2003). Specificity of cerebellar vermian abnormalities in autism: A

quantitative magnetic resonance imaging study. Journal of Child Neurology, 18(7), 463-470.

Kemper, T. L., & Bauman, M. L. (1993). The contribution of neuropathologic studies to the

understanding of autism. Neurologic Clinics, 11(1), 175-187.

Kim, J. E., Lyoo, I. K., Estes, A. M., Renshaw, P. F., Shaw, D. W., Friedman, S. D., . . . Dager,

S. R. (2010). Laterobasal amygdalar enlargement in 6- to 7-year-old children with autism

spectrum disorder. Archives of General Psychiatry, 67(11), 1187-1197.

Kirsten, T. B., Chaves-Kirsten, G. P., Chaible, L. M., Silva, A. C., Martins, D. O., Britto, L. R., .

. . Bernardi, M. M. (2012). Hypoactivity of the central dopaminergic system and autistic-like

behavior induced by a single early prenatal exposure to lipopolysaccharide. Journal of

Neuroscience Research,90(10), 1903-1912.

Kirsten, T. B., Taricano, M., Flório, J. C., Palermo-Neto, J., & Bernardi, M. M. (2010). Prenatal

lipopolysaccharide reduces motor activity after an immune challenge in adult male offspring.

Behavioral Brain Research,211(1), 77-82.

Knickmeyer, R., Baron-Cohen, S., Fane, B. A., Wheelwright, S., Mathews, G. A., Conway, G.

S., . . . Hines, M. (2006). Androgens and autistic traits: A study of individuals with congenital

adrenal hyperplasia. Hormones and Behavior, 50(1), 148-153.

Korn, T., Bettelli, E., Oukka, M., & Kuchroo, V. K. (2009). IL-17 and Th17 cells

Kreutzberg, G. W. (1996). Microglia: A sensor for pathological events in the CNS. Trends in

Neurosciences, 19(8), 312-318.

Page 75: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

66

Kumral, A., Baskin, H., Yesilirmak, D. C., Ergur, B. U., Aykan, S., Genc, S., . . . Ozkan, H.

(2007). Erythropoietin attenuates lipopolysaccharide-induced white matter injury in the neonatal

rat brain. Neonatology, 92(4), 269-278.

Labrousse, V. F., Costes, L., Aubert, A., Darnaudéry, M., Ferreira, G., Amédée, T., & Layé, S.

(2009). Impaired interleukin-1β and c-fos expression in the hippocampus is associated with a

spatial memory deficit in P2X7 receptor-deficient mice. PLoS ONE, 4(6)

Larouche, A., Roy, M., Kadhim, H., Tsanaclis, A. M., Fortin, D., & Sébire, G. (2005). Neuronal

injuries induced by perinatal hypoxic-ischemic insults are potentiated by prenatal exposure to

lipopolysaccharide: Animal model for perinatally acquired encephalopathy. Developmental

Neuroscience, 27(2-4), 134-142.

Lauritsen, M. B., Pedersen, C. B., & Mortensen, P. B. (2005). Effects of familial risk factors and

place of birth on the risk of autism: A nationwide register-based study. Journal of Child

Psychology and Psychiatry and Allied Disciplines, 46(9), 963-971.

Lee, D. -., Kuo, H. -., Chen, C. -., Hsu, J. -., Chou, C. -., Wei, Y., . . . Hung, M. -. (2007). IKKβ

suppression of TSC1 links inflammation and tumor angiogenesis via the mTOR pathway. Cell,

130(3), 440-455.

Li, X., Chauhan, A., Sheikh, A. M., Patil, S., Chauhan, V., Li, X. -., . . . Malik, M. (2009).

Elevated immune response in the brain of autistic patients. Journal of Neuroimmunology, 207(1-

2), 111-116.

Lichtenstein, P., Carlström, E., Råstam, M., Gillberg, C., & Anckarsäter, H. (2010). The genetics

of autism spectrum disorders and related neuropsychiatric disorders in childhood. American

Journal of Psychiatry,167(11), 1357-1363.

Ling, Z., Zhu, Y., Tong, C. w., Snyder, J. A., Lipton, J. W., & Carvey, P. M. (2006). Progressive

dopamine neuron loss following supra-nigral lipopolysaccharide (LPS) infusion into rats exposed

to LPS prenatally. Experimental Neurology, 199(2), 499-512.

Page 76: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

67

Ling, Z., Zhu, Y., Tong, C. W., Snyder, J. A., Lipton, J. W., & Carvey, P. M. (2009). Prenatal

lipopolysaccharide does not accelerate progressive dopamine neuron loss in the rat as a result of

normal aging. Experimental Neurology, 216(2), 312-320.

Lipina, T. V., Zai, C., Hlousek, D., Roder, J. C., & Wong, A. H. C. (2013). Maternal immune

activation during gestation interacts with Disc1 point mutation to exacerbate schizophrenia-

related behaviors in mice. Journal of Neuroscience, 33(18), 7654-7666.

Liu, X., Lee, J. G., Yee, S. K., Bresee, C. J., Poland, R. E., & Pechnick, R. N. (2004). Endotoxin

exposure in utero increases ethanol consumption in adult male offspring. Neuroreport, 15(1),

203-206.

Liverman, C. S., Kaftan, H. A., Cui, L., Hersperger, S. G., Taboada, E., Klein, R. M., & Berman,

N. E. J. (2006). Altered expression of pro-inflammatory and developmental genes in the fetal

brain in a mouse model of maternal infection. Neuroscience Letters, 399(3), 220-225.

Loesch, D. Z., Huggins, R. M., & Hoang, N. H. (1995). Growth in stature in fragile X families:

A mixed longitudinal study. American Journal of Medical Genetics, 58(3), 249-256.

Madsen-Bouterse, S. A., Romero, R., Tarca, A. L., Kusanovic, J. P., Espinoza, J., Kim, C. J., . . .

Draghici, S. (2010). The transcriptome of the fetal inflammatory response syndrome. American

Journal of Reproductive Immunology, 63(1), 73-92.

Makinodan, M., Tatsumi, K., Manabe, T., Yamauchi, T., Makinodan, E., Matsuyoshi, H., et al.

(2008). Maternal immune activation in mice delays myelination and axonal development in the

hippocampus of the offspring.Journal of Neuroscience Research, 86(10), 2190-2200.

Malkova, N. V., Yu, C. Z., Hsiao, E. Y., Moore, M. J., & Patterson, P. H. (2012). Maternal

immune activation yields offspring displaying mouse versions of the three core symptoms of

autism. Brain, Behavior, and Immunity, 26(4), 607-616.

Page 77: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

68

Mandal, M., Marzouk, A. C., Donnelly, R., & Ponzio, N. M. (2011). Maternal immune

stimulation during pregnancy affects adaptive immunity in offspring to promote development of

TH17 cells. Brain, Behavior, and Immunity, 25(5), 863-871.

Mandal, M., Marzouk, A. C., Donnelly, R., & Ponzio, N. M. (2010). Preferential development of

Th17 cells in offspring of immunostimulated pregnant mice. Journal of Reproductive

Immunology, 87(1-2), 97-100.

Mandy, W., Chilvers, R., Chowdhury, U., Salter, G., Seigal, A., & Skuse, D. (2012). Sex

differences in autism spectrum disorder: Evidence from a large sample of children and

adolescents. Journal of Autism and Developmental Disorders, 42(7), 1304-1313.

Marín-Teva, J. L., Dusart, I., Colin, C., Gervais, A., Van Rooijen, N., & Mallat, M. (2004).

Microglia promote the death of developing purkinje cells. Neuron, 41(4), 535-547.

Martin, L. A., & Horriat, N. L. (2012). The effects of birth order and birth interval on the

phenotypic expression of autism spectrum disorder. PLoS ONE, 7(11)

Martin, L. A., Goldowitz, D., & Mittleman, G. (2010). Repetitive behavior and increased activity

in mice with purkinje cell loss: A model for understanding the role of cerebellar pathology in

autism. European Journal of Neuroscience, 31(3), 544-555.

Maski, K. P., Jeste, S. S., & Spence, S. J. (2011). Common neurological co-morbidities in autism

spectrum disorders. Current Opinion in Pediatrics, 23(6), 609-615.

Matson, J. L., & Nebel-Schwalm, M. S. (2007). Comorbid psychopathology with autism

spectrum disorder in children: An overview. Research in Developmental Disabilities, 28(4), 341-

352.

McFarlane, H. G., Kusek, G. K., Yang, M., Phoenix, J. L., Bolivar, V. J., & Crawley, J. N.

(2008). Autism-like behavioral phenotypes in BTBR T+tf/J mice. Genes, Brain and Behavior,

7(2), 152-163.

Page 78: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

69

McPheeters, M. L., Warren, Z., Sathe, N., Bruzek, J. L., Krishnaswami, S., Jerome, R. N., &

Veenstra-VanderWeele, J. (2011). A systematic review of medical treatments for children with

autism spectrum disorders.Pediatrics, 127(5), e1312-e1321.

Meyer, U., Nyffeler, M., Engler, A., Urwyler, A., Schedlowski, M., Knuesel, I., et al. (2006).

The time of prenatal immune challenge determines the specificity of inflammation-mediated

brain and behavioral pathology.Journal of Neuroscience, 26(18), 4752-4762.

Morgan, J. T., Chana, G., Pardo, C. A., Achim, C., Semendeferi, K., Buckwalter, J., . . . Everall,

I. P. (2010). Microglial activation and increased microglial density observed in the dorsolateral

prefrontal cortex in autism.Biological Psychiatry, 68(4), 368-376.

Moss, J., & Howlin, P. (2009). Autism spectrum disorders in genetic syndromes: Implications for

diagnosis, intervention and understanding the wider autism spectrum disorder population.

Journal of Intellectual Disability Research, 53(10), 852-873.

Muhle, R., Trentacoste, S. V., & Rapin, I. (2004). The genetics of autism. Pediatrics, 113(5),

e472-486.

Munson, J., Dawson, G., Abbott, R., Faja, S., Webb, S. J., Friedman, S. D., . . . Dager, S. R.

(2006). Amygdalar volume and behavioral development in autism. Archives of General

Psychiatry, 63(6), 686-693.

Muzio, M., Bosisio, D., Polentarutti, N., D'amico, G., Stoppacciaro, A., Mancinelli, R., et al.

(2000). Differential expression

Napoli, I., & Neumann, H. (2009). Microglial clearance function in health and disease.

Neuroscience, 158(3), 1030-1038.

Nelms, K., Keegan, A. D., Zamorano, J., Ryan, J. J., & Paul, W. E. (1999). The IL-4 receptor:

Signaling mechanisms and biologic functions

Page 79: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

70

Netea, M. G., Van Deuren, M., Kullberg, B. J., Cavaillon, J. -., & Van Der Meer, J. W. M.

(2002). Does the shape of lipid A determine the interaction of LPS with toll-like receptors?

Trends in Immunology, 23(3), 135-139.

Nicolas, L. B., Kolb, Y., & Prinssen, E. P. M. (2006). A combined marble burying-locomotor

activity test in mice: A practical screening test with sensitivity to different classes of anxiolytics

and antidepressants. European Journal of Pharmacology, 547(1-3), 106-115.

Nilsson, C., Larsson, B. -., Jennische, E., Eriksson, E., Björntorp, P., York, D. A., & Holmäng,

A. (2001). Maternal endotoxemia results in obesity and insulin resistance in adult male offspring.

Endocrinology,142(6), 2622-2630.

Ning, H., Wang, H., Zhao, L., Zhang, C., Li, X. -., Chen, Y. -., & Xu, D. -. (2008). Maternally-

administered lipopolysaccharide (LPS) increases tumor necrosis factor alpha in fetal liver and

fetal brain: Its suppression by low-dose LPS pretreatment. Toxicology Letters, 176(1), 13-19.

Njung'E, K., & Handley, S. L. (1991). Evaluation of marble-burying behavior as a model of

anxiety.Pharmacology Biochemistry and Behavior, 38(1), 63-67.

Noriuchi, M., Kikuchi, Y., Yoshiura, T., Kira, R., Shigeto, H., Hara, T., et al. (2010). Altered

white matter fractional anisotropy and social impairment in children with autism spectrum

disorder. Brain Research, 1362, 141-149.

O'Connell, B. A., Moritz, K. M., Roberts, C. T., Walker, D. W., & Dickinson, H. (2011). The

placental response to excess maternal glucocorticoid exposure differs between the male and

female conceptus in spiny mice. Biology of Reproduction, 85(5), 1040-1047.

Ogando, D. G., Paz, D., Cella, M., & Franchi, A. M. (2003). The fundamental role of increased

production of nitric oxide in lipopolysaccharide-induced embryonic resorption in mice.

Reproduction, 125(1), 95-110.

Okada, K., Hashimoto, K., Iwata, Y., Nakamura, K., Tsujii, M., Tsuchiya, K. J., . . . Mori, N.

(2007). Decreased serum levels of transforming growth factor-β1 in patients with autism.

Progress in Neuro-Psychopharmacology and Biological Psychiatry, 31(1), 187-190.

Page 80: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

71

O'Neill, L. (2000). The Toll/interleukin-1 receptor domain: A molecular switch for inflammation

and host defence. Biochemical Society Transactions, 28(5), 557-563.

Onore, C., Careaga, M., & Ashwood, P. (2012). The role of immune dysfunction in the

pathophysiology of autism. Brain, Behavior, and Immunity, 26(3), 383-392.

Osei-Kumah, A., Smith, R., Jurisica, I., Caniggia, I., & Clifton, V. L. (2011). Sex-specific

differences in placental global gene expression in pregnancies complicated by asthma. Placenta,

32(8), 570-578.

Oskvig, D. B., Elkahloun, A. G., Johnson, K. R., Phillips, T. M., & Herkenham, M. (2012).

Maternal immune activation by LPS selectively alters specific gene expression profiles of

interneuron migration and oxidative stress in the fetus without triggering a fetal immune

response. Brain, Behavior, and Immunity, 26(4), 623-634.

Ozawa, K., Hashimoto, K., Kishimoto, T., Shimizu, E., Ishikura, H., & Iyo, M. (2006). Immune

activation during pregnancy in mice leads to dopaminergic hyperfunction and cognitive

impairment in the offspring: A neurodevelopmental animal model of schizophrenia. Biological

Psychiatry, 59(6), 546-554.

Pacey, L. K. K., Doss, L., Cifelli, C., der Kooy, D. V., Heximer, S. P., & Hampson, D. R. (2011).

Genetic deletion of regulator of G-protein signaling 4 (RGS4) rescues a subset of fragile X

related phenotypes in the FMR1 knockout mouse. Molecular and Cellular Neuroscience, 46(3),

563-572.

Paintlia, M. K., Paintlia, A. S., Barbosa, E., Singh, I., & Singh, A. K. (2004). N-acetylcysteine

prevents endotoxin-induced degeneration of oligodendrocyte progenitors and hypomyelination in

developing rat brain. Journal of Neuroscience Research, 78(3), 347-361.

Paintlia, M. K., Paintlia, A. S., Contreras, M. A., Singh, I., & Singh, A. K. (2008).

Lipopolysaccharide-induced peroxisomal dysfunction exacerbates cerebral white matter injury:

Attenuation by N-acetyl cysteine.Experimental Neurology, 210(2), 560-576.

Page 81: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

72

Paolicelli, R. C., Bolasco, G., Pagani, F., Maggi, L., Scianni, M., Panzanelli, P., . . . Gross, C. T.

(2011). Synaptic pruning by microglia is necessary for normal brain development. Science,

333(6048), 1456-1458.

Pearson, B. L., Pobbe, R. L. H., Defensor, E. B., Oasay, L., Bolivar, V. J., Blanchard, D. C., &

Blanchard, R. J. (2011). Motor and cognitive stereotypies in the BTBR T+tf/J mouse model of

autism. Genes, Brain and Behavior,10(2), 228-235.

Peça, J., Feliciano, C., Ting, J. T., Wang, W., Wells, M. F., Venkatraman, T. N., . . . Feng, G.

(2011). Shank3 mutant mice display autistic-like behaviors and striatal dysfunction. Nature,

472(7344), 437-442.

Pellock, J. M. (2004). Understanding co-morbidities affecting children with epilepsy. Neurology,

62(5 SUPPL. 2), S17-S23.

Peng, H., Whitney, N., Wu, Y., Tian, C., Dou, H., Zhou, Y., & Zheng, J. (2008). HIV-1-infected

and/or immune-activated macrophage-secreted TNF-α affects human fetal cortical neural

progenitor cell proliferation and differentiation. Glia, 56(8), 903-916.

Phelps, E. A. (2006). Emotion and cognition: Insights from studies of the human amygdala.

Annual Review of Psychology, 57, 27-53.

Phillips, M. L., Drevets, W. C., Rauch, S. L., & Lane, R. (2003). Neurobiology of emotion

perception I: The neural basis of normal emotion perception. Biological Psychiatry, 54(5), 504-

514.

Pierce, K., & Courchesne, E. (2001). Evidence for a cerebellar role in reduced exploration and

stereotyped behavior in autism. Biological Psychiatry, 49(8), 655-664.

Platanias, L. C. (2005). Mechanisms of type-I- and type-II-interferon-mediated signalling. Nature

Reviews Immunology, 5(5), 375-386.

Posey, D. J., Aman, M. G., McCracken, J. T., Scahill, L., Tierney, E., Arnold, L. E., . . .

McDougle, C. J. (2007). Positive effects of methylphenidate on inattention and hyperactivity in

pervasive developmental disorders: An analysis of secondary measures. Biological Psychiatry,

61(4), 538-544.

Page 82: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

73

Ramsey, A. J., Milenkovic, M., Oliveira, A. F., Escobedo-Lozoya, Y., Seshadri, S., Salahpour,

A., . . . Caron, M. G. (2011). Impaired NMDA receptor transmission alters striatal synapses and

DISC1 protein in an age-dependent manner. Proceedings of the National Academy of Sciences

of the United States of America, 108(14), 5795-5800.

Ransohoff, R.M., & Benveniste, E. N. (1996). Cytokines and the CNS. Boca Raton: CRC Press.

References

Reddy, K. S. (2005). Cytogenetic abnormalities and fragile-x syndrome in autism spectrum

disorder. BMC Medical Genetics, 6

Reichenberg, A., Smith, C., Schmeidler, J., & Silverman, J. M. (2007). Birth order effects on

autism symptom domains. Psychiatry Research, 150(2), 199-204.

Reichow, B., & Wolery, M. (2009). Comprehensive synthesis of early intensive behavioral

interventions for young children with autism based on the UCLA young autism project model.

Journal of Autism and Developmental Disorders, 39(1), 23-41.

Reith, R. M., McKenna, J., Wu, H., Hashmi, S. S., Cho, S. -., Dash, P. K., & Gambello, M. J.

(2013). Loss of Tsc2 in purkinje cells is associated with autistic-like behavior in a mouse model

of tuberous sclerosis complex.Neurobiology of Disease, 51, 93-103.

Reul, J. M. H. M., Stec, I., Wiegers, G. J., Labeur, M. S., Linthorst, A. C. E., Arzt, E., &

Holsboer, F. (1994). Prenatal immune challenge alters the hypothalamic-pituitary- adrenocortical

axis in adult rats.Journal of Clinical Investigation, 93(6), 2600-2607.

Reynolds, S., Lane, S. J., & Thacker, L. (2012). Sensory processing, physiological stress, and

sleep behaviors in children with and without autism spectrum disorders. OTJR Occupation,

Participation and Health, 32(1), 246-257.

Ritvo, E. R., Freeman, B. J., Scheibel, A. B., Duong, T., Robinson, H., & Guthrie, D. (1986).

Lower purkinje cell counts in the cerebella of four autistic subjects: Initial findings of the UCLA-

NSAC autopsy research report.American Journal of Psychiatry, 143(7), 862-866.

Page 83: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

74

Rogers, S. J., & Vismara, L. A. (2008). Evidence-based comprehensive treatments for early

autism. Journal of Clinical Child and Adolescent Psychology, 37(1), 8-38.

Rojas, D. C., Peterson, E., Winterrowd, E., Reite, M. L., Rogers, S. J., & Tregellas, J. R. (2006).

Regional gray matter volumetric changes in autism associated with social and repetitive behavior

symptoms. BMC Psychiatry, 6

Romero, E., et al. "Neurobehavioral and Immunological Consequences of Prenatal Immune

Activation in Rats. Influence of Antipsychotics." Neuropsychopharmacology 32.8 (2007): 1791-

804.

Romero, E., Guaza, C., Castellano, B., & Borrell, J. (2010). Ontogeny of sensorimotor gating

and immune impairment induced by prenatal immune challenge in rats: Implications for the

etiopathology of schizophrenia. Molecular Psychiatry, 15(4), 372-383.

Ronald, A., Larsson, H., Anckarsäter, H., & Lichtenstein, P. (2011). A twin study of autism

symptoms in sweden. Molecular Psychiatry, 16(10), 1039-1047.

Rosenberg, R. E., Law, J. K., Yenokyan, G., McGready, J., Kaufmann, W. E., & Law, P. A.

(2009). Characteristics and concordance of autism spectrum disorders among 277 twin pairs.

Archives of Pediatrics and Adolescent Medicine, 163(10), 907-914.

Rossant, J., & Cross, J. C. (2001). Placental development: Lessons from mouse mutants. Nature

Reviews Genetics, 2(7), 538-548.

Rousset, C. I., Chalon, S., Cantagrel, S., Bodard, S., Andres, C., Gressens, P., & Saliba, E.

(2006). Maternal exposure to LPS induces hypomyelination in the internal capsule and

programmed cell death in the deep gray matter in newborn rats. Pediatric Research, 59(3), 428-

433.

Rousset, C. I., Kassem, J., Olivier, P., Chalon, S., Gressens, P., & Saliba, E. (2008). Antenatal

bacterial endotoxin sensitizes the immature rat brain to postnatal excitotoxic injury. Journal of

Neuropathology and Experimental Neurology, 67(10), 994-1000.

Page 84: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

75

Roumier, A., Pascual, O., Béchade, C., Wakselman, S., Poncer, J. -., Réal, E., . . . Bessis, A.

(2008). Prenatal activation of microglia induces delayed impairment of glutamatergic synaptic

function. PLoS ONE, 3(7)

Rutter, M., Silberg, J., O'Connor, T., & Simonoff, E. (1999). Genetics and child psychiatry: II.

empirical research findings. Journal of Child Psychology and Psychiatry and Allied Disciplines,

40(1), 19-55.

Sadile AG. Long-term habituation of theta-related activity components of albino rats in the Lat-

maze. In: Sanberg P, Ossenkopp K, Kavaliers M,editors. Motor activity andmovement disorders:

measurement and analysis.New York: Humana; 1995. p. 1–54.

Salminen, A., Paananen, R., Vuolteenaho, R., Metsola, J., Ojaniemi, M., Autio-Harmainen, H.,

& Hallman, M. (2008). Maternal endotoxin-induced preterm birth in mice: Fetal responses in

toll-like receptors, collectins, and cytokines. Pediatric Research, 63(3), 280-286.

Sato, D., Lionel, A. C., Leblond, C. S., Prasad, A., Pinto, D., Walker, S., . . . Scherer, S. W.

(2012). SHANK1 deletions in males with autism spectrum disorder. American Journal of Human

Genetics, 90(5), 879-887.

Schmahmann, J. D., & Sherman, J. C. (1998). The cerebellar cognitive affective syndrome.

Brain, 121(4), 561-579.

Schumann, C. M., & Amaral, D. G. (2006). Stereological analysis of amygdala neuron number in

autism. Journal of Neuroscience, 26(29), 7674-7679.

Schumann, C. M., Bloss, C. S., Barnes, C. C., Wideman, G. M., Carper, R. A., Akshoomoff, N., .

. . Courchesne, E. (2010). Longitudinal magnetic resonance imaging study of cortical

development through early childhood in autism. Journal of Neuroscience, 30(12), 4419-4427.

Schumann, C. M., Hamstra, J., Goodlin-Jones, B. L., Lotspeich, L. J., Kwon, H., Buonocore, M.

H., . . . Amaral, D. G. (2004). The amygdala is enlarged in children but not adolescents with

autism; the hippocampus is enlarged at all ages. Journal of Neuroscience, 24(28), 6392-6401.

Page 85: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

76

Scott, N. M., Hodyl, N. A., Murphy, V. E., Osei-Kumah, A., Wyper, H., Hodgson, D. M., . . .

Clifton, V. L. (2009). Placental cytokine expression covaries with maternal asthma severity and

fetal sex.Journal of Immunology, 182(3), 1411-1420.

Sharma, A., Hoeffer, C. A., Takayasu, Y., Miyawaki, T., McBride, S. M., Klann, E., & Suzanne

Zukin, R. (2010). Dysregulation of mTOR signaling in fragile X syndrome. Journal of

Neuroscience, 30(2), 694-702.

Shatz, C. J. (2009). MHC class I: An unexpected role in neuronal plasticity. Neuron, 64(1), 40-

45.

Shi, L., et al. "Maternal Influenza Infection Causes Marked Behavioral and Pharmacological

Changes in the Offspring." Journal of Neuroscience 23.1 (2003): 297-302.

Shi, Y., & Massagué, J. (2003). Mechanisms of TGF-β signaling from cell membrane to the

nucleus. Cell, 113(6), 685-700.

Shimada, S., Iwabuchi, K., Watano, K., Shimizu, H., Yamada, H., Minakami, H., & Onoé, K.

(2003). Expression of allograft inflammatory factor-1 in mouse uterus and poly(I:C)-induced

fetal resorption. American Journal of Reproductive Immunology, 50(1), 104-112.

Silverman, J. L., Yang, M., Lord, C., & Crawley, J. N. (2010). Behavioral phenotyping assays

for mouse models of autism. Nature Reviews Neuroscience, 11(7), 490-502.

Singh, V. K. (1996). Plasma increase of interleukin-12 and interferon-gamma pathological

significance in autism. Journal of Neuroimmunology, 66(1-2), 143-145.

Sipes, M., Matson, J. L., Worley, J. A., & Kozlowski, A. M. (2011). Gender differences in

symptoms of autism spectrum disorders in toddlers. Research in Autism Spectrum Disorders,

5(4), 1465-1470.

Skaar, D. A., Shao, Y., Haines, J. L., Stenger, J. E., Jaworski, J., Martin, E. R., . . . Pericak-

Vance, M. A. (2005). Analysis of the RELN gene as a genetic risk factor for autism. Molecular

Psychiatry, 10(6), 563-571.

Page 86: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

77

Smalley, S. L. (1991). Genetic influences in autism. Psychiatric Clinics of North America, 14(1),

125-139.

Smith, S. E. P., et al. "Maternal Immune Activation Alters Fetal Brain Development through

Interleukin-6." Journal of Neuroscience 27.40 (2007): 10695-702.

Soumiya, H., Fukumitsu, H., & Furukawa, S. (2011). Prenatal immune challenge compromises

development of upper-layer but not deeper-layer neurons of the mouse cerebral cortex. Journal of

Neuroscience Research,89(9), 1342-1350.

Sparks, B. F., Friedman, S. D., Shaw, D. W., Aylward, E. H., Echelard, D., Artru, A. A., . . .

Dager, S. R. (2002). Brain structural abnormalities in young children with autism spectrum

disorder. Neurology, 59(2), 184-192.

Spencer, C. M., Alekseyenko, O., Serysheva, E., Yuva-Paylor, L. A., & Paylor, R. (2005).

Altered anxiety-related and social behaviors in the Fmr1 knockout mouse model of fragile X

syndrome. Genes, Brain and Behavior, 4(7), 420-430.

Stanfield, A. C., McIntosh, A. M., Spencer, M. D., Philip, R., Gaur, S., & Lawrie, S. M. (2008).

Towards a neuroanatomy of autism: A systematic review and meta-analysis of structural

magnetic resonance imaging studies. European Psychiatry, 23(4), 289-299.

Steffenburg, S., Gillberg, C., Hellgren, L., Andersson, L., Gillberg, I. C., Jakobsson, G., &

Bohman, M. (1989). A twin study of autism in denmark, finland, iceland, norway and sweden.

Journal of Child Psychology and Psychiatry and Allied Disciplines, 30(3), 405-416.

Stepkowski, S. M., Chen, W., Ross, J. A., Nagy, Z. S., & Kirken, R. A. (2008). STAT3: An

important regulator of multiple cytokine functions. Transplantation, 85(10), 1372-1377.

Sumi, S., Taniai, H., Miyachi, T., & Tanemura, M. (2006). Sibling risk of pervasive

developmental disorder estimated by means of an epidemiologic survey in nagoya, japan. Journal

of Human Genetics, 51(6), 518-522.

Suzuki, K., Matsuzaki, H., Iwata, K., Kameno, Y., Shimmura, C., Kawai, S., . . . Mori, N.

(2011). Plasma cytokine profiles in subjects with high-functioning autism spectrum disorders.

PLoS ONE, 6(5)

Page 87: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

78

Suzuki, S., Kawamata, J., Matsushita, T., Matsumura, A., Hisahara, S., Takata, K., . . .

Shimohama, S. (2013). 3-[(2,4-dimethoxy)benzylidene]-anabaseine dihydrochloride protects

against 6-hydroxydopamine-induced parkinsonian neurodegeneration through α7 nicotinic

acetylcholine receptor stimulation in rats. Journal of Neuroscience Research, 91(3), 462-471.

Sweeten, T. L., Posey, D. J., & McDougle, C. J. (2004). Brief report: Autistic disorder in three

children with cytomegalovirus infection. Journal of Autism and Developmental Disorders, 34(5),

583-586.

Swisher, C. N., & Swisher, L. (1975). Congenital rubella and autistic behavior. New England

Journal of Medicine, 293(4), 198.

Tavano, A., Grasso, R., Gagliardi, C., Triulzi, F., Bresolin, N., Fabbro, F., & Borgatti, R. (2007).

Disorders of cognitive and affective development in cerebellar malformations. Brain, 130(10),

2646-2660.

Takarae, Y., Minshew, N. J., Luna, B., & Sweeney, J. A. (2004). Oculomotor abnormalities

parallel cerebellar histopathology in autism. Journal of Neurology, Neurosurgery and Psychiatry,

75(9), 1359-1361.

Taniai, H., Nishiyama, T., Miyachi, T., Imaeda, M., & Sumi, S. (2008). Genetic influences on

the broad spectrum of autism: Study of proband-ascertained twins. American Journal of Medical

Genetics, Part B: Neuropsychiatric Genetics, 147(6), 844-849.

Tchaconas, A., & Adesman, A. (2013). Autism spectrum disorders: A pediatric overview and

update. Current Opinion in Pediatrics, 25(1), 130-144.

Thomas, A. M., Bui, N., Perkins, J. R., Yuva-Paylor, L. A., & Paylor, R. (2012). Group i

metabotropic glutamate receptor antagonists alter select behaviors in a mouse model for fragile X

syndrome.Psychopharmacology, 219(1), 47-58.

Thomson, A. W., Turnquist, H. R., & Raimondi, G. (2009). Immunoregulatory functions of

mTOR inhibition. Nature Reviews Immunology, 9(5), 324-337.

Page 88: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

79

Tsai, P. T., Hull, C., Chu, Y., Greene-Colozzi, E., Sadowski, A. R., Leech, J. M., . . . Sahin, M.

(2012). Autistic-like behavior and cerebellar dysfunction in purkinje cell Tsc1 mutant mice.

Nature, 488(7413), 647-651.

Urakubo, A., Jarskog, L. F., Lieberman, J. A., & Gilmore, J. H. (2001). Prenatal exposure to

maternal infection alters cytokine expression in the placenta, amniotic fluid, and fetal brain.

Schizophrenia Research, 47(1), 27-36.

Vargas, D. L., Nascimbene, C., Krishnan, C., Zimmerman, A. W., & Pardo, C. A. (2005).

Neuroglial activation and neuroinflammation in the brain of patients with autism. Annals of

Neurology, 57(1), 67-81.

Vashlishan, A. B., Madison, J. M., Dybbs, M., Bai, J., Sieburth, D., Ch'ng, Q., . . . Kaplan, J. M.

(2008). An RNAi screen identifies genes that regulate GABA synapses. Neuron, 58(3), 346-361.

Veeraragavan, S., Bui, N., Perkins, J. R., Yuva-Paylor, L. A., & Paylor, R. (2011). The

modulation of fragile X behaviors by the muscarinic M4 antagonist, tropicamide. Behavioral

Neuroscience, 125(5), 783-790.

Veeraragavan, S., Bui, N., Perkins, J. R., Yuva-Paylor, L. A., Carpenter, R. L., & Paylor, R.

(2011). Modulation of behavioral phenotypes by a muscarinic M1 antagonist in a mouse model

of fragile X syndrome.Psychopharmacology, 217(1), 143-151.

Visintin, A., Mazzoni, A., Spitzer, J. H., Wyllie, D. H., Dower, S. K., & Segal, D. M. (2001).

Regulation of toll-like receptors in human monocytes and dendritic cells. Journal of

Immunology, 166(1), 249-255.

Vorhees, C. V., Graham, D. L., Braun, A. A., Schaefer, T. L., Skelton, M. R., Richtand, N. M., &

Williams, M. T. (2012). Prenatal immune challenge in rats: Altered responses to dopaminergic

and glutamatergic agents, prepulse inhibition of acoustic startle, and reduced route-based

learning as a function of maternal body weight gain after prenatal exposure to poly IC. Synapse,

66(8), 725-737.

Page 89: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

80

Walker, B. R., Diefenbach, K. S., & Parikh, T. N. (2007). Inhibition within the nucleus tractus

solitarius (NTS) ameliorates environmental exploration deficits due to cerebellum lesions in an

animal model for autism. Behavioral Brain Research, 176(1), 109-120.

Wang, H., Meng, X. -., Ning, H., Zhao, X. -., Wang, Q., Liu, P., . . . Xu, D. -. (2010). Age- and

gender-dependent impairments of neurobehaviors in mice whose mothers were exposed to

lipopolysaccharide during pregnancy. Toxicology Letters, 192(2), 245-251.

Wassink, T. H., Piven, J., & Patil, S. R. (2001). Chromosomal abnormalities in a clinic sample of

individuals with autistic disorder. Psychiatric Genetics, 11(2), 57-63.

Weber, A. M., Egelhoff, J. C., Mckellop, J. M., & Franz, D. N. (2000). Autism and the

cerebellum: Evidence from tuberous sclerosis. Journal of Autism and Developmental Disorders,

30(6), 511-517.

Wei, H., Zou, H., Sheikh, A. M., Malik, M., Dobkin, C., Brown, W. T., & Li, X. (2011). IL-6 is

increased in the cerebellum of autistic brain and alters neural cell adhesion, migration and

synaptic formation. Journal of Neuroinflammation, 8

Wei, Y. -., Li, X. -., & Zhou, J. -. (2007). Prenatal exposure to lipopolysaccharide results in

increases in blood pressure and body weight in rats. Acta Pharmacologica Sinica, 28(5), 651-656.

Weichhart, T., Costantino, G., Poglitsch, M., Rosner, M., Zeyda, M., Stuhlmeier, K. M., . . .

Säemann, M. D. (2008). The TSC-mTOR signaling pathway regulates the innate inflammatory

response.Immunity, 29(4), 565-577.

Werling, D. M., & Geschwind, D. H. (2013). Sex differences in autism spectrum disorders.

Current Opinion in Neurology, 26(2), 146-153.

Whitney, E. R., Kemper, T. L., Bauman, M. L., Rosene, D. L., & Blatt, G. J. (2008). Cerebellar

purkinje cells are reduced in a subpopulation of autistic brains: A stereological experiment using

calbindin-D28k.Cerebellum, 7(3), 406-416.

Won, S. J., Kim, S. H., Xie, L., Wang, Y., Mao, X. O., Jin, K., & Greenberg, D. A. (2006).

Reelin-deficient mice show impaired neurogenesis and increased stroke size. Experimental

Neurology, 198(1), 250-259.

Page 90: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

81

Workman, A. D., Charvet, C. J., Clancy, B., Darlington, R. B., & Finlay, B. L. (2013). Modeling

transformations of neurodevelopmental sequences across mammalian species. Journal of

Neuroscience, 33(17), 7368-7383.

Worley, J. A., & Matson, J. L. (2011). Psychiatric symptoms in children diagnosed with an

autism spectrum disorder: An examination of gender differences. Research in Autism Spectrum

Disorders,5(3), 1086-1091.

Yamasaki, H., LaBar, K. S., & McCarthy, G. (2002). Dissociable prefrontal brain systems for

attention and emotion. Proceedings of the National Academy of Sciences of the United States of

America, 99(17), 11447-11451.

Yee, N., Ribic, A., de Roo, C. C., & Fuchs, E. (2011). Differential effects of maternal immune

activation and juvenile stress on anxiety-like behavior and physiology in adult rats: No evidence

for the "double-hit hypothesis". Behavioral Brain Research, 224(1), 180-188.

Yesilirmak, D. C., Kumral, A., Baskin, H., Ergur, B. U., Aykan, S., Genc, S., . . . Ozkan, H.

(2007). Activated protein C reduces endotoxin-induced white matter injury in the developing rat

brain. Brain Research, 1164(1), 14-23.

Yip, J., Soghomonian, J. -., & Blatt, G. J. (2007). Decreased GAD67 mRNA levels in cerebellar

purkinje cells in autism: Pathophysiological implications. Acta Neuropathologica, 113(5), 559-

568.

Yoder, P., Stone, W. L., Walden, T., & Malesa, E. (2009). Predicting social impairment and

ASD diagnosis in younger siblings of children with autism spectrum disorder. Journal of Autism

and Developmental Disorders,39(10), 1381-1391.

Zager, A., Mennecier, G., & Palermo-Neto, J. (2012). Maternal immune activation in late

gestation enhances locomotor response to acute but not chronic amphetamine treatment in male

mice offspring: Role of the D1 receptor. Behavioral Brain Research, 232(1), 30-36.

Zaretsky, M. V., Alexander, J. M., Byrd, W., & Bawdon, R. E. (2004). Transfer of inflammatory

cytokines across the placenta. Obstetrics and Gynecology, 103(3), 546-550.

Page 91: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

82

Appendices

Appendix 1 Comparison between computer and manual scoring of social interaction of

LPS treated wild-type MIA female offspring evaluated by the modified 3-chamber

paradigm. The computer (A) and manual (B) analysis of social interaction test results were

comparable. The similarity between the two methods was also reflected by the social interaction

index (D, E). When the analysis was performed separately for each half of the exploration

period, both LPS 1X and LPS 2X offspring showed deficits in sociability in the last 5 minutes

(C). This result was reflected by the social interaction index (F). Each column represents the

average ± S.E.M. One-way ANOVA was performed, followed by Bonferroni’s post hoc

analysis. Using the Grubbs’ test, one value that was furthest from the mean was omitted for each

group, except for those with n ≤ 8. * p < 0.05. ** p < 0.01. *** p < 0.001. **** p < 0.0001.

Page 92: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

83

Appendix 2 Summary of behavioral findings in wild-type MIA offspring and Fmr1

knockout MIA offspring. The solid red line represents the behavioral outcome of control saline

WT (A) and saline FXS (B) mice. The light grey areas represent a trend and the dark grey areas

represent statistical significance of at least p < 0.05.

Page 93: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 3. Summary of literature research regarding Poly IC MIA rodent model. All findings are of offspring from Poly IC injected dams in comparison to its respective control. N/A: not applicable. 84

Author (Year) Target Disease Species (Strain)

Dose (route)

Time of Injection

Behavioral Findings Cytokine Findings Neuroinflammation-related Findings

Arrode-Bruses et al. (2012)

not specified mouse (C57/B6)

20 mg/kg (i.p.)

E16 N/A pro-inflammatory cytokines (IL-1β, IL-13),

chemokines (MCP-1, MIP-1α, IP-10, MIG) and CSF VEGF in fetal brain (6 or

24 hr after injection)

N/A

Bronson et al. (2011)

Schizophrenia rat (Sprague-Dawley)

8 mg/kg (i.p.)

E 14 offspring showed bimodal weight change; male offspring from Poly IC dams with weight loss had decreased MK801-stimulated (P56) and

amphetamine-stimulated (P90) locomotion

compared to offspring from Poly IC dams with weight gain and vehicle dams; weight loss of the

Poly IC dam increased with increasing maternal age and more frequent

social isolation

N/A N/A

Dickerson et al. (2012)

schizophrenia rat (Sprague-Dawley)

4.0 mg/kg (iv)

E15 theta frequency coherence between the

prefrontal cortex and hippocampus was

significantly reduced in the MIA group during baseline resting-state

recordings (adult)

N/A N/A

Page 94: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 3. Summary of literature research regarding Poly IC MIA rodent model. All findings are of offspring from Poly IC injected dams in comparison to its respective control. N/A: not applicable. 85

Author (Year) Target Disease Species (Strain)

Dose (route)

Time of Injection

Behavioral Findings Cytokine Findings Neuroinflammation-related Findings

Forrest et al. (2012)

not specified rat (Wistar)

10 mg/kg (i.p.)

E14, E16, E18

N/A ↑ MCP-1 in serum of Poly IC dams (5 hr after

injection)

N/A

Gilmore et al. (2005)

not specified rat (Sprague-Dawley)

10 or 20 mg/kg (i.p.)

E16 N/A 20 mg/kg: TNFα (plasma and placenta, no

change in fetal brain) 10 mg/kg: ↓ TNFα whole

brain (P1)

N/A

Han et al. (2011)

not specified rat (Sprague-Dawley)

0.5 mg/kg (i.p.)

daily from E15 to E18

no difference in spatial learning (Morris water maze); poly IC offspring

showed impaired reversal learning; Poly IC offspring had higher TNFα in serum

(> p35)

N/A N/A

Hsiao and Patterson

(2011)

neurodegenerative disease

mouse (C57/B6)

20 mg/kg (i.p.)

E12.5 ↓ in PPI, ↑ % freezing, ↓ entries into the center of

the open field (P42)

N/A N/A

Juckel et al. (2011)

Schizophrenia mice (BALB/c)

20 mg/kg (i.p.)

E9 N/A N/A in Iba1-positive cells in hippocampus and

striatum, not frontal and occipital cortex

Koga et al. (2009)

not specified mouse (C57/B6)

4.5 mg/kg (i.p.)

E16.5 N/A IL6, MCP-1, IL-12p40, RANTES, KC, MIP-1β in the placenta (2 hr, 4 hr

after injection)

↑ NF-ĸB activation in the placenta (2hr after

injection)

Li et al. (2009) Schizophrenia mouse (C57/B6)

5 mg/kg (iv)

E9 or E17 E9: ↓ in PPI (P91); E17: no change in PPI (P91)

N/A N/A

Page 95: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 3. Summary of literature research regarding Poly IC MIA rodent model. All findings are of offspring from Poly IC injected dams in comparison to its respective control. N/A: not applicable. 86

Author (Year) Target Disease Species (Strain)

Dose (route)

Time of Injection

Behavioral Findings Cytokine Findings Neuroinflammation-related Findings

Makinodan et al. (2008)

Schizophrenia mouse (C57/B6)

60 mg/kg (i.p.)

E9.5 ↓ PPI (P63) N/A ↓ mRNA for MBP in hippocampus, ↓ myelin thickness in hippocampal

CA1; ↓ MBP immunostaining in

hippocampal CA1 and CA3 (P14)

Malkova et al. (2012)

Autism mouse (C57/B6,

male)

5 mg/kg (i.p.)

E10.5, E12.5, E14.5

↓ number of USV to social stimuli (P10, 8-9

weeks); ↓ PPI (6 weeks); ↓ total distance travelled in open field (7 weeks); ↓

social interaction (10 weeks); deficit in female

urine-induced scent marking (11 weeks); ↑

marble burying (12 weeks); self-grooming (13 weeks); no change in olfactory sensitivity (13--

14 weeks)

N/A N/A

Mandal et al. (2010, 2011)

neurodegenerative disease

mouse (C57/B6)

20 mg/kg (i.p.)

E12 N/A increase in IL6 in the amniotic fluid is partly

contributed by the fetus

Lymphocytes from offspring of Poly (I:C)

injected immune dams showed preferential

ability to develop into Th 17 cells compared to PBS-injected control and Poly

(I:C) injected immunologically naïve

dams

Page 96: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 3. Summary of literature research regarding Poly IC MIA rodent model. All findings are of offspring from Poly IC injected dams in comparison to its respective control. N/A: not applicable. 87

Author (Year) Target Disease Species (Strain)

Dose (route)

Time of Injection

Behavioral Findings Cytokine Findings Neuroinflammation-related Findings

Meyer et al. (2006)

Schizophrenia mouse (C57/B6)

5 mg/kg (iv)

E9 no change in latent inhibition (P35-45), disruption in latent inhibition (P95-135)

N/A N/A

Meyer et al. (2006)

Schizophrenia, neurodegenerative

diseases

mouse (C57/B6)

5 mg/kg (iv)

E9 or E17 Injection at E9: ↓ PPI (P98-P112); ↓ in open-field exploration (P98-112); Injection at E17:

preservative behaviour (reversal learning of left-

right discrimination) (male only P98-112); ↓

working memory in water maze (P98-P112)

E9 and E17: IL-1β, IL6, IL-10, TNFα in brain (3 hr after injection); ↑ IL-6, IL10 in brain (6 hr after

injection)

N/A

Meyer et al. (2008)

Schizophrenia mouse (C57/B6)

5 mg/kg (iv)

E9 ↑ AMPH- and/or MK801-induced locomotion (P35,

P100-110)

N/A N/A

Meyer et al. (2008)

Schizophrenia mouse (C57/B6)

5 mg/kg (iv)

E9 ↓ PPI (P90-P120); deficit in PPI was improved by

periadolescent treatment of clozapine and

fluoxetine

N/A N/A

Miranda et al. (2010)

neurodegenerative disease

mouse (C57/B6)

5 mg/kg (i.p.)

E16 or E15-17

E16: ↓ in open-field locomotor activity (P8);

↓ in PPI at 4 dB and 8 dB (P8)

N/A N/A

Ozawa et al. (2006)

Schizophrenia mouse (Balb/c)

5 mg/kg (i.p.)

E12-E17 ↓ PPI, ↑ thigmotaxis, ↓ novel object recognition;

↑ MAP-induced locomotion (P63-P70)

N/A N/A

Page 97: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 3. Summary of literature research regarding Poly IC MIA rodent model. All findings are of offspring from Poly IC injected dams in comparison to its respective control. N/A: not applicable. 88

Author (Year) Target Disease Species (Strain)

Dose (route)

Time of Injection

Behavioral Findings Cytokine Findings Neuroinflammation-related Findings

Shi et al. (2003)

neurodegenerative disease

mouse (Balb/c)

20 mg/kg (i.p.)

E9.5 ↓ PPI (P42-P56) N/A N/A

Shi et al. (2009)

neurodegenerative disease

mouse (C57/B6)

20 mg/kg (i.p.)

E12.5 N/A N/A ↓ cerebellar Purkinje cells (P11)

Smith et al. (2007)

neurodegenerative disease

mouse (C57/B6)

20 mg/kg (i.p.)

E12.5 ↓ PPI, ↓ LI, ↓ exploration in open field,

↓ social interaction (adult, age unspecified)

N/A N/A

Soumiya et al. (2011)

neurodegenerative disease

mouse (ddY)

20 mg/kg (i.p.)

E9.5 ↑ anxiety (i.e. reluctance to enter the center

portion of the well-lit open field) and ↓ in

social interaction (i.e. showed no preference for

social chamber)

N/A N/A

Page 98: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 3. Summary of literature research regarding Poly IC MIA rodent model. All findings are of offspring from Poly IC injected dams in comparison to its respective control. N/A: not applicable. 89

Author (Year) Target Disease Species (Strain)

Dose (route)

Time of Injection

Behavioral Findings Cytokine Findings Neuroinflammation-related Findings

Vorhees et al. (2012)

Schizophrenia rat (Harlan

Sprague-Dawley)

8 mg/kg (i.p.)

E14 ↓ in the rate of route-based learning; ↓

prepulse inhibition of acoustic startle in females (not males), exaggerated hyperactivity in response to (+)-amphetamine, and attenuated hyperactivity in response to MK-801; no change in elevated zero maze, open-field

activity, object burying, light-dark test, straight

channel swimming, Morris water maze spatial

acquisition, reversal, or shift navigation or spatial

working or reference memory, or conditioned

contextual or cued fear or latent inhibition (P65-87)

N/A N/A

Wolff and Bilkey (2008)

Schizophrenia rat (Sprague-Dawley)

4 mg/kg (iv)

E15 ↓ PPI (P56) N/A N/A

Wolff et al. (2011)

schizophrenia rat (Sprague-Dawley)

4.0 mg/kg (iv)

E15 ↓ time spent exploring novel object; ↑ reversal

learning; more rapid within trial habituation in

the novel context recognition (adult)

N/A N/A

Page 99: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 3. Summary of literature research regarding Poly IC MIA rodent model. All findings are of offspring from Poly IC injected dams in comparison to its respective control. N/A: not applicable. 90

Author (Year) Target Disease Species (Strain)

Dose (route)

Time of Injection

Behavioral Findings Cytokine Findings Neuroinflammation-related Findings

Yee et al. (2011)

not specified rat (Sprague-Dawley)

4.0 mg/kg (tail vein)

E15 ↓ prepulse inhibition; ↓ litter size; ↑ pup weight

(PND2),↑ anxiety in elevated maze

no difference in IL6 serum level (P69), TNFα and IL-1β was under the limit of

detection

N/A

Yee et al. (2012)

not specified rat (Sprague-Dawley)

4 mg/kg (tail vein)

E15 ↑ total number of 22-kHz ultrasonic vocalization

during fear conditioning compared to control

(>P71); No difference in habituation, fear

conditioning, or fear testing in terms of rearing, grooming,

defecation or US induced jumps;

N/A N/A

Zuckerman et al. (2003,

2005)

Schizophrenia rat (Wistar)

4 mg/kg (iv)

E15 or E17 ↓ LI, ↑ reversal learning of left-right

discrimination, ↑ AMPH- and MK801-induced

locomotion (P90)

N/A N/A

Page 100: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 4. Summary of literature research regarding LPS MIA rodent model. All findings are of offspring from LPS injected dams in comparison to its respective control. N/A: not applicable. 91

Author (Year)

Target Disease Species (Strain)

Serotype of LPS

Dose (route)

Time of injection

Behavioural findings Cytokine Findings Neuroinflammation-related Findings

Ashdown et al.

(2006)

Schizophrenia rat 0111:B4 50 μg/kg (i.p.)

E18 N/A ↑ IL-1β, IL-6, TNFα in maternal plasma (2–4 hr after injection); ↑

IL-1β, IL-6, TNFα in placenta (2–8 hr after injection); ↑ IL-1β in

fetal plasma (4 hr after injection)

N/A

Bakos et al. (2004)

chronic inflammation

rat (Wistar)

0111:B4 (L3012)

20, 20, 40, 40, 80 μg/kg

(N/A)

E15-19 ↓ locomotor activity in elevated plus maze,

↑ slips in the beam walking test (not

significant) (female offspring only)

N/A N/A

Borrell et al. (2002)

Schizophrenia rat (Wistar)

026:B6 (L-3755)

1 mg/kg (sc) alternate days for whole

pregnancy

↓ PPI (P70, P100, P200)

N/A N/A

Cai et al. (2000)

PVL, associated with cerebral palsy

rat (Sprague-Dawley)

055:B5 500 μg/kg (i.p.)

E18-19 N/A N/A ↑ GFAP in hippocampus,

thalamus, hypothalamus, frontal

cortex but not cerebellum (P8), ↓

MBP (P8), no change microglia (OX-42 and

tomato lectin)

Page 101: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 4. Summary of literature research regarding LPS MIA rodent model. All findings are of offspring from LPS injected dams in comparison to its respective control. N/A: not applicable. 92

Author (Year)

Target Disease Species (Strain)

Serotype of LPS

Dose (route)

Time of injection

Behavioural findings Cytokine Findings Neuroinflammation-related Findings

Chang et al. (2011)

not specified mouse (CD1)

N/A 100 μg/kg (intrauterine)

E15/20 N/A ↑ IL6 expression in myometrium and

placenta which was attenuated by N-acetylcysteine in

myometrium; in fetal brain, ↑ IL6, TNFα,

and IL-1β

N/A

Coyle et al. (2009)

hypozincaemia mouse (C57/B6)

0111:B4 300 μg/kg (sc)

E8 ↑ exploration of familiar vs. novel

object (reversed by administering

zinc)(P84); no change spatial learning or

memory (cross-maze escape task) (P65,

P79)

N/A N/A

Dowling et al. (2012)

not specified rat (Spraque-Dawley)

055:B5 1 mg/kg (i.p.) E 19 in the placenta: CCL2, IL6, and TNFα,

this was reversed with the administration of mangesium sulfate

N/A N/A

Fortier et al. (2004)

Schizophrenia rat (Sprague-

Dawley Male)

0128:B12 (L-2755)

50 μg/kg (i.p.)

E18-19 ↓ PPI, ↑ AMPH-induced locomotion, enhanced acoustic

startle responses (P70)

N/A N/A

Fortier et al. (2007)

Schizophrenia rat (Sprague-

Dawley Male)

0111:B4 (L-2630)

50 or 100 or 50 μg/kg

(i.p.)

E10-11 or E15-16 or

E18-19

↓ PPI E15-16 and E18-19 (P70), no change in

PPI E10-11 (P70)

N/A N/A

Page 102: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 4. Summary of literature research regarding LPS MIA rodent model. All findings are of offspring from LPS injected dams in comparison to its respective control. N/A: not applicable. 93

Author (Year)

Target Disease Species (Strain)

Serotype of LPS

Dose (route)

Time of injection

Behavioural findings Cytokine Findings Neuroinflammation-related Findings

Gayle et al. (2004)

not specified rat 100 μg/kg (i.p.)

E18 N/A mRNA for IL-1β, IL6, TNFα in placenta

(1–24 hr after injection); ↑ IL-1β,

IL6, TNFα in amniotic fluid (1–12 hr after

injection); no change in these cytokine

levels was observed in the fetal brain (1-24 hr

after injection)

N/A

Gilmore et al. (2005)

Neurodevelopmental disorders

rat (Sprague-Dawley)

055:B5 100 μg/kg (i.p.)

E14-16 N/A ↓ TNFα in frontal cortex (P6)

N/A

Girard et al. (2009)

cerebral palsy rat (Lewis)

0127:B8 200 μg/kg (i.p.)

twice daily from

E17 to birth

↓ in weight before P3, no change in total

distance travelled or number of crossing

events in motor activity test; ↓ latency to fall off rotarod (P30,

P35, P40)

N/A N/A

Girard et al. (2010)

Neurodevelopmental disorders

rat (Lewis)

0127:B8 200 μg/kg (i.p.)

twice daily from

E18-20

motor coordination deficit by rotarod

(P30, P35, P40)

↑ IL-1β, TNFα, IL6, IL-1Ra in placenta

↑ CD68 (labyrinth) and proliferating

microglia (brain white matter) (P9)

Page 103: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 4. Summary of literature research regarding LPS MIA rodent model. All findings are of offspring from LPS injected dams in comparison to its respective control. N/A: not applicable. 94

Author (Year)

Target Disease Species (Strain)

Serotype of LPS

Dose (route)

Time of injection

Behavioural findings Cytokine Findings Neuroinflammation-related Findings

Golan et al. (2005)

Neurodevelopmental disorders

mouse (C57/B6)

N/A 120 μg/kg (i.p.)

E17 no change in exploratory behaviour,

sensory gain, and motor function;↓

associative learning in cued version of water maze, ↑ novel object

recognition,

↑ IL6 in fetal brain (3 hr after injection)

N/A

Golan et al. (2006)

Neurodevelopmental disorders

mouse (C57/B6)

N/A 120 μg/kg (i.p.)

E17 ↑ exploration in open field (P240, P600); ↑ anxiety (elevated plus

maze), ↓ social activity, ↓

aggressiveness towards cagemates (P240); age is a risk

factor for later neurodegenerative

diseases, not systemic maternal

inflammation

N/A N/A

Haesaert and Ornoy

(1986)

Neurodevelopmental disorders

mouse (-) 026:B6 (Difco Co.)

2.5 μg/kg (-) E10 , E12 ↑ audiogenic seizures (P28)

N/A N/A

Kumral et al. (2007); Yesilirmak

et al. (2007)

PVL, associated with cerebral palsy

rat (Sprague-Dawley)

055:B5 500 μg/kg (i.p.)

E18-19 N/A ↑ IL-1β, IL-6, TNFα in whole brain (P7)

↓ MBP immunostaining in

periventricular white matter (P7)

Page 104: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 4. Summary of literature research regarding LPS MIA rodent model. All findings are of offspring from LPS injected dams in comparison to its respective control. N/A: not applicable. 95

Author (Year)

Target Disease Species (Strain)

Serotype of LPS

Dose (route)

Time of injection

Behavioural findings Cytokine Findings Neuroinflammation-related Findings

Lante et al (2007, 2008)

Neurodevelopmental disorders

rat (Sprague-Dawley)

055:B5 500 μg/kg (i.p.)

E19 ↓ spatial learning in water maze (P28)

N/A N/A

Larouche et al.

(2005); Girard et al. (2010)

Hypoxia/Ischemia rat (Lewis)

0127:B8 200 μg/kg (i.p.)

twice daily from

E17 to birth

N/A N/A ↑ proliferating and total microglia in forebrain white

matter (P9)

Ling et al. (2002, 2004,

2006); Zhu et al.

(2007)

Parkinson's disease rat (Sprague-Dawley)

026:B6 (L8274)

1 mg/kg (i.p.) E10.5 N/A ↑ TNFα in striatum (P21, P120, P210,

P510) and SN (P210)

N/A

Ling et al. (2006, 2009)

Parkinson's disease rat (Sprague-Dawley)

026:B6 (L8274)

1 mg/kg (i.p.) E10.5 N/A N/A ↑ activated microglia in SN basally (P120,

P210, P420, P510) and in response to

postnatal intranigral LPS (P210-P294)

Ling et al. (2009)

Parkinson's disease rat (Sprague-Dawley)

026:B6 (L8274)

1 mg/kg (i.p.) E10.5 to E11

↑ locomotor activity (P90); ↓ locomotor

activity (P480)

N/A N/A

Liu et al. (2004)

Ethanol consumption

rat (Sprague-Dawley)

026:B6 (L3773)

1 mg/kg (sc) alternate days for whole

pregnancy

↑ ethanol intake and preference, ↓ rearing

(P100-P130)

N/A N/A

Page 105: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 4. Summary of literature research regarding LPS MIA rodent model. All findings are of offspring from LPS injected dams in comparison to its respective control. N/A: not applicable. 96

Author (Year)

Target Disease Species (Strain)

Serotype of LPS

Dose (route)

Time of injection

Behavioural findings Cytokine Findings Neuroinflammation-related Findings

Liverman et al.

(2006)

cerebral palsy mouse (C57/B6)

0127:B8 50 μg (i.p.) E18 N/A N/A ↑ expresion of MCP-1, IL6, IL-1β, VEGF; YB-

1, needin; ↓ expression of

semaphorin 5b and groucho (RT-PCR was performed on fetal

brain removed 0.5-6 hr after injection)

Ning et al. (2008)

not specified mouse 0127:B8 500 μg/kg (i.p.)

E17 N/A ↑ TNFα in fetal brain, liver and

amniotic fluid (1.5 hr after injection); no

change in mRNA for TNFα in fetal brain

(1.5 hr after injection)

N/A

Paintlia et al. (2004,

2008)

PVL, associated with cerebral palsy

rat (Sprague-Dawley)

055:B5 700 or 1000 μg/kg (i.p.)

E18 N/A ↑ mRNA for IL-1β (1-48 hr after injection), TNFα (1-24 hr after

injection) in fetal brain

↓ OPCs and immature oligodendrocytes in corpus callosum and

lateral ventricles with ↓ mRNA for MBP and PLP in brain; ↓ MBP,

↑ GFAP immunostaining in

corpus callosum and/or cingulum (P9,

P16, P23, P30)

Romero et al. (2007,

2010)

Neurodevelopmental disorders

rat (Wistar)

026:B6 (L-3755)

2 mg/kg (sc) daily for whole

pregnancy

↓ PPI (P35, P70, P170, P180, P400)

↑ in IL2, IL6, TNFα in adult rat serum

N/A

Page 106: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 4. Summary of literature research regarding LPS MIA rodent model. All findings are of offspring from LPS injected dams in comparison to its respective control. N/A: not applicable. 97

Author (Year)

Target Disease Species (Strain)

Serotype of LPS

Dose (route)

Time of injection

Behavioural findings Cytokine Findings Neuroinflammation-related Findings

Roumier et al.

(2008)

Neurodevelopmental disorders

mouse (C57/B6)

055:B5 120 μg/kg (i.p.)

E15 N/A N/A ↑ microglial density in hippocampus (P0)

Rousset et al. (2006,

2008)

PVL, associated with cerebral palsy

rat (Wistar)

055:B5 300 or 400 μg/kg (i.p.)

E19-20 N/A ↑ mRNA for IL-1β (P1), ↓ mRNA for

TNFα (P7) in whole brain

↓ MBP immunostaining in

external and internal capsule (P7); ↑

ibotenate-induced microglial activation

and astrogliosis in various regions (P9)

Salminen et al.

(2008)

not specified mouse (C57/B6)

0111:B4 1 mg/kg (i.p.) E16 or 17 N/A ↑ TNFα, MCP-1, and IL6 in the maternal serum and amniotic fluid; ↑ TNFα and

MCP-1 in fetal serum (1, 3, or 8 hr after

injection)

N/A

Urakubo et al.

(2001)

Schizophrenia rat (Sprague-Dawley)

055:B5 0.5 mg/kg (i.p.)

E16 N/A 0.5 mg/kg: in IL-1β, IL6, TNFα in placenta

and in IL6 in amniotic fluid (2 or 8 hr after injection) 2.5 mg/kg: in IL6, TNFα

in placenta and TNFα in amniotic fluid, ↓TNFα in fetal brain (2 hr after injection)

N/A

Page 107: The Maternal Immune Activation Mouse Model of Autism … · 2013-12-11 · ii The Maternal Immune Activation Mouse Model of Autism Spectrum Disorders Cong Yang (Ingrid) Xuan Master

Appendix 4. Summary of literature research regarding LPS MIA rodent model. All findings are of offspring from LPS injected dams in comparison to its respective control. N/A: not applicable. 98

Author (Year)

Target Disease Species (Strain)

Serotype of LPS

Dose (route)

Time of injection

Behavioural findings Cytokine Findings Neuroinflammation-related Findings

Wang et al. (2010)

Neurodevelopmental disorders

mouse (CD1)

0127:B8 8 μg/kg (i.p.) daily from E8-15

↓ learning and retention in radical

arm maze (P200 female, P400 male); ↑ anxiety in open field,

impaired beam walking (P200 female);

↑ weight burrowed (P70 male, P600

female); ↓ weight hoarded (P200

female)

N/A N/A

Wijkstra et al. (1991)

Sexual behaviour rat (Wistar)

N/A 0.2 or 2 μg/kg (iv)

E18 ↑ latency to initiate sexual behaviour, ↓

intromission by males (P91)

N/A N/A

Zager et al. (2012)

not specified mouse (Swiss)

0127:B8 120 μg/kg (i.p.)

E17 no change in total locomotion and mean velocity. After acute

amphetamine treatment, total

locomotion and mean velocity compare to control and baseline

(P70-90)

N/A N/A


Recommended