145
Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome by Shervin Gholizadeh Moghaddam A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Pharmaceutical Sciences University of Toronto © Copyright by Shervin Gholizadeh Moghaddam, 2016

Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the

Treatment of Fragile X Syndrome

by

Shervin Gholizadeh Moghaddam

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Department of Pharmaceutical Sciences University of Toronto

© Copyright by Shervin Gholizadeh Moghaddam, 2016

Page 2: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

ii

Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the

Treatment of Fragile X Syndrome

Shervin Gholizadeh Moghaddam

Doctor of Philosophy

Department of Pharmaceutical Sciences

University of Toronto

2016

Abstract

Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a trinucleotide

repeat expansion in the FMR1 gene that codes for fragile X mental retardation protein (FMRP).

The goal of the present study was to determine whether restoring FMRP expression in the brains

of the Fmr1 knockout (KO) mouse model of FXS could correct the disorder. Initially, we

investigated major factors affecting tropism, expression level, and cell-type specificity of adeno-

associated viral vectors (AAV), including encapsidation of different AAV serotypes, promoter

selection, and the timing of vector administration, using intra-cerebroventricular injections of

AAV vectors expressing enhanced green fluorescent protein. Our results favored the use of

AAV serotype 9 (AAV9) vectors containing the neuron-selective synapsin-1 promoter, injected

into the lateral ventricles of neonatal mice. To determine if FMRP expression in the central

nervous system could reverse phenotypic deficits in the Fmr1 KO mouse model of FXS, we

used a single-stranded AAV9 that contained a major isoform of FMRP. The vector was

delivered to the brain via a single bilateral intra-cerebroventricular injection into neonatal Fmr1

KO mice on postnatal day 5 (in the first phase of the study) or 0-1 (in the second phase).

Transgene expression and behavioral assessments were conducted either 22-26 or 50-56 days

post-injection. Western blotting and immunocytochemical analyses of AAV-FMRP-injected

Page 3: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

iii

mice revealed neuron-specific FMRP expression in the striatum, hippocampus, retrosplenial

cortex, and cingulate cortex. AAV-FMRP injections reversed the pathologically elevated

repetitive behavior and the deficit in social dominance behavior seen in Fmr1 KO mice in phase

1, as well as the elevated startle response and lower anxiety in phase 2. These results provide

the first proof-of-principle that gene therapy can correct specific behavioral abnormalities in the

mouse model of FXS.

Page 4: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

vi

Table of Contents

Chapter 1. Introduction .................................................................................................................. 1

1.1. Fragile X syndrome ............................................................................................................. 1

1.2. FMRP expression in the brain of the wild-type mouse ....................................................... 3

1.3. FMRP: Structure and molecular functions ......................................................................... 4

1.4. The mGluR theory of fragile X syndrome .......................................................................... 7

1.5. Prospects of gene therapy application for FXS ................................................................... 9

1.6. Viral vectors for gene delivery into the CNS .................................................................... 10

1.7. Adeno-associated viral vectors ......................................................................................... 13

1.8. Subsequent steps in AAV cell entry and implications for gene delivery into the CNS .... 15

1.9. Routes of AAV administration into the CNS ................................................................... 15

1.9.1. Intra-cranial Administration ....................................................................................... 17

1.9.2. Intra-vascular Administration..................................................................................... 19

1.9.3. Intra-CSF .................................................................................................................... 21

1.9.4. Retrograde transport delivery ..................................................................................... 22

1.10. Timing of AVV administration ....................................................................................... 22

1.11. Promoter selection .......................................................................................................... 24

1.12. AAV transport within the CNS ....................................................................................... 26

1.13. Hypotheses and objectives .............................................................................................. 26

Chapter 2. Materials and Methods .............................................................................................. 28

2.1. Animals ............................................................................................................................. 28

2.2. AAV vectors ..................................................................................................................... 28

2.3. Vector injections ............................................................................................................... 29

2.3.1. i.c.v. injections in neonatal mice ................................................................................ 29

2.3.2. Stereotaxic injections in juvenile mice ....................................................................... 30

2.4. Behavioral analysis ........................................................................................................... 30

2.4.1. Locomotor activity measurements ............................................................................. 30

2.4.2. Marble Burying for stereotypic behavior ................................................................... 31

2.4.3. Ultrasonic vocalizations ............................................................................................. 31

2.4.4. Tube test of social dominance .................................................................................... 31

2.4.5. Elevated plus maze test of anxiety ............................................................................. 32

2.4.6. Pre-pulse inhibition for sensorimotor gating .............................................................. 32

Page 5: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

vi

2.4.7. Audiogenic Seizures ................................................................................................... 33

2.5. Tissue preparation, immunocytochemistry and confocal microscopy .............................. 34

2.5.1. Semi-quantitative and quantitative analysis of transgene expression in the brain ..... 35

2.6. Quantitative western blotting ............................................................................................ 36

2.7. Statistical analyses ............................................................................................................ 37

Chapter 3. Expression of FMRP protein in neurons and glia of the developing and adult mouse

brain ............................................................................................................................................. 38

3.1. Specific hypotheses, objectives and rationale ................................................................... 38

3.2. Gradual reduction in the number of FMRP-positive cells from the early postnatal period

to adulthood ............................................................................................................................. 38

3.3. Predominant neuronal expression of FMRP throughout development ............................. 39

3.4. Developmental expression of FMRP in astrocytes, microglia and oligodendrocyte

precursor cells .......................................................................................................................... 41

Chapter 4. Transduction efficiency of adeno-associated viral vectors expressing eGFP after

intra-cerebroventricular administration in neonatal and juvenile mice ....................................... 47

4.1. Specific hypotheses, objectives and rationale ................................................................... 47

4.2. AAV9-CMV-eGFP targets astrocytes in neonatal mice but neurons in juvenile mice .... 48

4.3. Use of the synapsin-1 promoter to achieve neuron-specific transduction ........................ 50

4.4. Low transduction efficiency of AAV5-GFP vectors ........................................................ 54

Chapter 5. FXS gene therapy-Phase 1: Reduced phenotypic severity following adeno-associated

virus mediated Fmr1 gene delivery in postnatal day 5 Fmr1 mice.............................................. 57

5.1. Specific hypotheses, objectives and rationale ................................................................... 57

5.2. Quantification of FMRP expression levels following neonatal i.c.v. injection of AAV-

FMRP in postnatal day 5 mice ................................................................................................. 59

5.3. Distribution and cellular selectivity of transgene expression ........................................... 60

5.4. Phase 1 - Behavioral analyses ........................................................................................... 63

Chapter 6. FXS gene therapy-Phase 2: ........................................................................................ 71

Fmr1 transgene delivery in postnatal day 0 Fmr1 mice .............................................................. 71

6.1. Specific hypotheses, objectives and rationale ................................................................... 71

6.2. Quantification of FMRP expression levels following neonatal i.c.v. injection of AAV-

FMRP in postnatal day 0 mice ................................................................................................. 73

6.2.1. Levels of FMRP substrates in AAV-FMRP-treated mice .......................................... 77

6.3. Distribution and cellular selectivity of transgene expression ........................................... 79

6.4. Phase 2 - Behavioral Analyses .......................................................................................... 86

Page 6: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

vi

Chapter 7. Discussion ................................................................................................................. 93

7.1. FMRP expression in the developing and adult brain ........................................................ 93

7.2. Gene therapy as a potential therapeutic avenue for the treatment of Fragile X syndrome 96

7.3. Major factors affecting cellular tropism, efficiency and distribution of AAV-induced

transduction .............................................................................................................................. 97

7.4. FMRP transgene expression in the brains of Fmr1 KO mice ........................................... 99

7.5. Behavioral effects of FMRP restoration in the brains of Fmr1 KO mice ....................... 101

7.6. Possible contributions of aberrant MeCP2 expression to pathological abnormalities in

FXS ........................................................................................................................................ 104

7.7. Concluding remarks and future directions ...................................................................... 107

References .............................................................................................................................. 111

Page 7: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

vii

List of Publications

1. Hampson DR, Gholizadeh S, Pacey LK. Pathways to drug development for autism

spectrum disorders. Clin Pharmacol Ther. 2012 Feb;91(2):189-200. Review.

2. Gholizadeh S, Tharmalingam S, Macaldaz ME, Hampson DR. Transduction of the

central nervous system after intra-cerebroventricular injection of adeno-associated viral

vectors in neonatal and juvenile mice. Hum Gene Ther Methods. 2013 Aug;24(4):205-

13.

3. Gholizadeh S, Arsenault J, Xuan IC, Pacey LK, Hampson DR. Reduced Phenotypic

Severity Following Adeno-Associated Virus-Mediated Fmr1 Gene Delivery in Fragile X

Mice. Neuropsychopharmacology. 2014 Jul 7.

4. Gholizadeh S, Halder SK, Hampson DR. Expression of fragile X mental retardation

protein in neurons and glia of the developing and adult mouse brain. Brain Research.

2015 Jan 30;1596:22-30.

5. Arsenault J, Gholizadeh S, Pacey LK, Halder SK, Koxhioni E, Hampson DR. Viral

vector-mediated FMRP transgene expression: from phenotypic rescue in Fragile X mice

to pathological over-expression (Manuscript submitted, 2016)

Page 8: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

viii

List of Tables

Table 1. Comparison between fragile X patients and Fmr1 KO mice .......................................... 2

Table 2. Clinical trials for gene therapy using AAV vectors in neurological disorders. ............. 11

Table 3. Comparison of AAV viral vectors against the ideal vector for gene delivery to the CNS.

..................................................................................................................................................... 11

Table 4. Primary receptors and gene delivery tropism of different AAV serotypes in the

mammalian CNS .......................................................................................................................... 16

Table 5. Promoters commonly used for gene delivery into the CNS. ......................................... 25

Table 6. Quantitative analysis of cell type-specific expression of FMRP in the cingulate cortex

and corpus callosum. ................................................................................................................... 39

Table 7. Semi-quantitative analysis of cell type-specific FMRP expression in the developing and

adult mouse brain. ........................................................................................................................ 40

Table 8. Analysis of AAV-eGFP vector transduction patterns in discrete areas of the brain after

i.c.v. delivery in PND 5 or PND 21 mice. ................................................................................... 56

Table 9. Summary of the main differences in the experimental design between Phase 1 and

Phase 2 ......................................................................................................................................... 73

Table 10. Quantitative analysis of AAV-FMRP transduction in cingulate cortex. ..................... 82

Page 9: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

xi

List of Figures

Fig. 1. FMR1 gene expansion and FMRP protein structure .......................................................... 5

Fig. 2. FMRP shuttling between the nucleus and cytoplasm ......................................................... 7

Fig. 3. Major factors affecting tropism, transduction and transgene expression properties of

AAV ............................................................................................................................................. 17

Fig. 4. The brain barriers in the developing and adult brain........................................................ 20

Fig. 5. AAV9 tropism in the CNS at different developmental stages ......................................... 24

Fig. 6. Expression of FMRP in neurons of the WT mouse brain. ............................................... 41

Fig. 7. Expression of FMRP in the astrocytes of mouse brain. ................................................... 44

Fig. 8. Expression of FMRP in oligodendrocyte precursor cells in the mouse brain. ................. 45

Fig. 9. Expression of FMRP in microglia. ................................................................................... 46

Fig. 10. Schematic diagrams depicting (A) the AAV-eGFP vector constructs used and (B)

bilateral intra-cerebroventricular injections on postnatal days 5, and 21. ................................... 48

Fig. 11. AAV-eGFP transgene expression in the mouse striatum. .............................................. 51

Fig. 12. AAV-eGFP transgene expression in the retrosplenial cortex. ....................................... 52

Fig. 13. AAV-eGFP transgene expression in hippocampus. ....................................................... 53

Fig. 14. AAV-eGFP transgene expression in the cerebellum. ..................................................... 55

Fig. 15. Fmr1 gene delivery (Phase 1): Overview of viral vector construction, experimental plan

for injections, and behavioral analyses. ....................................................................................... 58

Fig. 16. Western blots of FMRP expression in control and AAV-FMRP-treated mice. ............. 60

Fig. 17. Immunocytochemical analysis of AAV-FMRP transduction in discrete brain regions . 62

Fig. 18. FMRP transgene expression in the striatum, hippocampus, cingulate cortex, and

retrosplenial cortex 61 days after i.c.v. administration of AAV-FMRP on PND 5. .................... 65

Fig. 19. Quantitative analysis of AAV-FMRP transduction in cingulate cortex. ........................ 66

Fig. 20. Summary of behavioral analyses of the short arm of the study. .................................... 67

Fig. 21. Summary of the behavioral results from the long arm of the study. .............................. 68

Fig. 22. Correlation between the number of marbles buried and FMRP transgene expression

levels in the cerebral cortex. ........................................................................................................ 70

Fig. 23. Fmr1 gene delivery (Phase 2): Overview of experimental plan for injections, and

behavioral analyses ...................................................................................................................... 73

Fig. 24. Analysis of GFAP following AAV-FMRP injections in WT animals. .......................... 74

Fig. 25. Western blots of FMRP expression in AAV-FMRP-treated KO and WT mice. ........... 76

Page 10: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

xi

Fig. 26. Characterization of MeCP2 as an mRNA substrate for FMRP and quantification of

MeCP2 levels in AAV-treated WT and KO mice. ...................................................................... 79

Fig. 27. Immunocytochemical analysis of AAV-FMRP transduction in discrete brain regions of

AAV-treated KO mice. ................................................................................................................ 80

Fig. 28. Immunocytochemical analysis of FMRP over-expression in different brain regions of

AAV-SYN-Fmr1-treated WT mice. ............................................................................................ 84

Fig. 29. Quantitative analysis of FMRP over-expression in AAV-SYN- Fmr1- treated WT mice.

..................................................................................................................................................... 85

Fig. 30. Analysis of locomotor activity. ...................................................................................... 88

Fig. 31. Analysis of sensorimotor gating. .................................................................................... 90

Fig. 32. Analysis of anxiety, stereotypical behavior, and courtship behavior. ............................ 92

Fig. 33. Overlapping characteristics of FXS, Rett syndrome, FMR1 duplication syndrome, and

MeCP2 duplication syndrome. .................................................................................................. 106

Page 11: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

xi

Abbreviations

AAV Adeno-associated virus

BBB blood brain barrier

CBA chicken β-actin

CMV cytomegalovirus

CNS central nervous system

CSF cerebrospinal fluid

eGFP enhanced green fluorescent protein

FMRP fragile X mental retardation protein

FXS fragile X syndrome

GFAP glial fibrillary acidic protein

ITR inverted terminal repeat

KO knockout

LTD long-term depression

mGluR metabotropic glutamate receptors

PBS phosphate-buffered saline

PND postnatal day

SYN synapsin

WPRE woodchuck hepatitis post-transcriptional regulatory element

WT wild-type

Page 12: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

1

Chapter 1. Introduction

1.1. Fragile X syndrome

Fragile X syndrome (FXS) is a genetic disorder and a leading cause of cognitive impairment

and autism (Bagni et al., 2012). The disorder is caused by a pathological CGG trinucleotide

triplet repeat expansion in the 5’ untranslated region of the FMR1 gene, located on the X

chromosome. The CGG repeat range in unaffected individuals is 5-55, while expansions of 200

or more result in hyper-methylation of the FMR1 gene and FXS. The highly expanded CGG

repeat also causes the transcribed mRNA to form RNA-DNA heteroduplexes (Colak et al.,

2014), which together with gene hyper-methylation severely reduces or abrogates the expression

of the encoded protein, fragile X mental retardation protein (FMRP). Trinucleotide expansion

in the intermediate range of 55 – 199 repeats results in the formation of toxic intra-nuclear

inclusions, a mild reduction of FMRP, and is associated with the neurodegenerative disorder

Fragile X-associated Tremor and Ataxia Syndrome (Bagni et al., 2012; LaFauci et al., 2013;

Ludwig et al., 2014). Reduced FMRP expression has also been reported in other mental

disorders including autism, schizophrenia, bipolar disorder, and major depression (Fatemi and

Folsom, 2011).

Patients with FXS experience a wide range of symptoms including cognitive deficits, social

anxiety, attention deficit and hyperactivity disorder, repetitive stereotyped behaviors, seizures,

and sensory hypersensitivity (Wijetunge et al., 2013). Parallel to human FXS, Fmr1 knockout

(KO) mice also display deficits consistent with an “autistic phenotype”. Fmr1 KO mice display

hyperactivity, reduced ultrasonic vocalizations during courtship, increased repetitive/stereotypic

behaviors, increased susceptibility to audiogenic seizures, and reduced social interactions (Pacey

et al., 2009) (Table 1). Furthermore, numerous studies have demonstrated altered synaptic

plasticity in animal models of the disorder. Studies in mice lacking FMRP revealed disrupted

Page 13: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

2

ocular dominance plasticity in visual cortex (Dolen et al., 2007) and impaired synaptic plasticity

in barrel cortex (Harlow et al., 2010), and auditory cortex (Kim et al., 2013a). Hypersensitivity

to auditory stimuli in humans with FXS and mice lacking FMRP suggests that the synaptic

plasticity in the auditory system may also be perturbed by the loss of FMRP (Chen and Toth,

2001; Kim et al., 2013a; Rotschafer and Razak, 2014). Fmr1 KO mice also show deficient fear

memory (Zhao et al., 2005; Neuwirth et al., 2015), impaired cerebellar eyeblink

conditioning (Koekkoek et al., 2005; Tobia and Woodruff-Pak, 2009; Reeb-Sutherland and Fox,

2015), inhibitory avoidance learning (Dolen et al., 2007; Yuskaitis et al., 2010b; Michalon et al.,

2014). Drosophila and zebrafish mutants lacking FMRP also have an impairment in long-term

memory (Bolduc et al., 2008; Bolduc et al., 2010; Kanellopoulos et al., 2012) and avoidance

learning (Ng et al., 2013), respectively.

Table 1. Comparison between fragile X patients and Fmr1 KO mice Fragile X patients Fmr1 Knockout mice

Mutation Expansion CGG repeat in 5′

untranslated region of exon 1

Insertion neomycin cassette in

exon 5

FMRP expression Very low or absent Absent

Physical features Elongated face with

prominent ears

Craniofacial abnormalities

(Heulens et al., 2013)

Macro-orchidism Macro-orchidism

Cognitive and

behavioural features

Intellectual disability

(IQ < 70)

Mild learning and memory

deficits

Autistic features Repetitive, perseverative

digging; Impaired social

behaviour

Hyperactivity Increased locomotor activity

Anxiety Decreased non-social anxiety

(e.g. in elevated plus maze) and

increased social anxiety

Epileptic seizures Increased susceptibility to

audiogenic seizures

Page 14: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

3

Deficit in sensorimotor gating Altered sensorimotor gating

(acoustic startle response and

pre-pulse inhibition)

Neuroanatomical

features

Increased density and

immature morphology of

dendritic spines

Age- and brain-region-

dependent spine abnormalities

Comparison of the physical, neuro-anatomical and behavioral features in fragile X patients

and Fmr1 KO mice.

1.2. FMRP expression in the brain of the wild-type mouse

FMRP is ubiquitously expressed, most abundantly in brain, testes, and ovaries (Devys et al.,

1993; Hinds et al., 1993; Ascano et al., 2012). In the adult mouse brain FMRP is abundantly

expressed in most neurons throughout the CNS. In neurons, the protein is primarily located in

the cytoplasm, including in the cell body, dendrites and axons and in synaptic spines where it

plays a role in spine maturation (Cruz-Martín et al., 2010). A portion of total cellular FMRP is

also present in the nucleus, where its role is less well-characterized. FMRP is highly abundant

in “fragile X granules” in neuronal axons and pre-synaptic terminals where it apparently

regulates recurrent neuronal activity (Akins et al., 2012). FMRP is also present in neural stem

cells (Luo et al., 2010) where it has been shown to control hippocampal-dependent neurogenesis

and learning in the mature brain (Guo et al., 2011).

In contrast to the expression of FMRP in neurons of the adult brain, relatively little is known

about the types of glial cells that express FMRP during CNS development. FMRP was reported

in primary cultures of rodent astrocytes (Yuskaitis et al., 2010a), and in the mouse hippocampus,

FMRP is expressed in astrocytes within the first week of birth and then its expression declines to

low or undetectable levels (Pacey and Doering, 2007). These findings highlight the important

role for FMRP expression in astrocytes during early postnatal weeks, which coincide with the

peak of synaptogenesis. It is also present in oligodendroglia (Wang et al., 2004) and

Page 15: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

4

oligodendrocyte precursor cells in the immature cerebellum where it appears to be a factor in the

proper progression of myelination (Pacey et al., 2013).

1.3. FMRP: Structure and molecular functions

The primary transcript of the mouse Fragile X Mental Retardation-1 gene (Fmr1) spans 39

kb, and is composed of 17 exons (Eichler et al., 1993). Similar to many transcripts in the CNS,

it undergoes alternative splicing and possesses alternative transcription start sites such that at

least 12 isoforms are generated (Tassone et al., 2011; Brackett et al., 2013). Isoform 1 is the

longest form and codes for a protein of 632 amino acids and a molecular weight of 71

kilodaltons. Isoform 1 is also known to account for about 40% of total FMRP in the human

brain (Huang et al., 1996). FMRP isoform 1 is the full-length protein which contains both a

nuclear localization signal (NLS) and a nuclear export signal (NES), and sites for post-

translational modification through phosphorylation and methylation (Sittler et al., 1996; Ceman

et al., 2003). Furthermore, nuclear localization of FMRP is isoform dependent (Dury et al.,

2013). Lack of the NES signal, in FMRP isoforms 4,6,10 and 12, results in predominant nuclear

localization of these FMRP isoforms, indicating its functional significance (Sittler et al., 1996;

Dury et al., 2013). Interestingly, isoforms 6 and 12 are also associated with Cajal bodies in the

nucleus (Dury et al., 2013). In contrast, the most common FMRP isoforms (isoforms 1 and 7)

which are associated with the translation machinery (Ascano et al., 2012), are mainly

cytoplasmic (Dury et al., 2013). These observations suggest that the nuclear FMRP isoforms

might have independent functions from the dominant cytoplasmic FMRP isoforms.

Page 16: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

5

Fig. 1. FMR1 gene expansion and FMRP protein structure

Expansion of CGG repeats in the 5' UTR region of the fragile X mental retardation 1 (FMR1)

gene that encodes FMRP underlies FXS. Repeats that contain >200 copies (full mutation) lead

to loss of FMRP expression.

FMRP is an mRNA binding protein that controls the expression of hundreds of genes in the

CNS (Darnell et al., 2011). FMRP has three RNA-binding domains, including two K homology

domains (KH1 and KH2) and an arginine-glycine-glycine (RGG) box, and binds a subset of

neuronal mRNAs (Darnell et al., 2005) (Fig. 1). KH domains bind tertiary motifs in mRNAs

which are generally known as the “kissing-complexes” (Darnell et al., 2005). Furthermore, the

RGG boxes recognize stem-G-quartet loops, potentially in a methylation-dependent mechanism

(Blackwell et al., 2010). The presence of nuclear localization and export signals suggests that

FMRP is a nucleo-cytoplasmic shuttling protein involved in transport of a subset of RNAs from

the nucleus to ribosomes (Eberhart et al., 1996; Fridell et al., 1996; Feng et al., 1997), however

it has been shown that FMRP binds its cargo mRNAs in the nucleus (Kim et al., 2009).

FMRP binds to more than 800 different identified gene transcripts in mouse brain tissue and

in human cell lines (Darnell et al., 2011; Ascano et al., 2012). This is equivalent to

Page 17: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

6

approximately 4% of total mRNAs that are expressed in human brain (Darnell et al., 2011). For

many of these substrates, FMRP has been shown to act as a translational suppressor (Wang et

al., 2012). However, FMRP has been shown to also act as a positive modular of protein

translation for some of its mRNA substrates (Bechara et al., 2009), possibly by enhancing

mRNA stability (Zalfa et al., 2007) or through potentiating the actions of the translational

activating gene Sod1 (Bechara et al., 2009). Interestingly, many of the mRNA targets of FMRP

encode pre- and post-synaptic proteins, including several proteins that are implicated in other

autism spectrum disorders, indicating molecular communalities between FXS and other

neurodevelopmental disorders (Darnell et al., 2011; Ouwenga and Dougherty, 2015).

Despite FMRP is commonly acknowledged as a regulator of mRNA translation, the precise

mechanism by which FMRP influences the translational machinery remains to be further

identified. Considering the fact that FMRP co-sediments with polyribosomes, it was initially

believed that FMRP might inhibit translation by blocking elongation (Tamanini et al., 1996;

Ceman et al., 2003). This hypothesis received strong support in a more recent study by the

Darnell group (Darnell et al., 2011). It was further shown that FMRP binds the vast majority of

its 842 mRNA substrates within the coding sequence, instead of the 5′ or 3′ untranslated regions.

Ribosomal run-off assays confirmed that FMRP forms a complex with its mRNA targets and

stalled ribosomes (Darnell et al., 2011). Furthermore, mounting evidence suggests that FMRP

stalls ribosomes and represses translation via interaction with the RNA-induced silencing

complex including several micro-RNAs (Kelley et al., 2012).

Page 18: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

7

Fig. 2. FMRP shuttling between the nucleus and cytoplasm

FMRP enters the nucleus by its nuclear localization signal and forms a messenger

ribonucleoprotein (mRNP) complex together with its target mRNAs. FMRP also interacts with

members of the RNA-induced silencing complex, including micro-RNAs and this messenger

ribonucleoprotein complex is transported to the synapses transporting dendritically localized

mRNAs. After synaptic stimulation, regulated by mGluR signalling pathway, the transported

mRNA targets get detached from FMRP and are locally translated in dendrites. FMRP interacts

with its cargo mRNAs either directly or through non-coding RNAs (ncRNA), and micro-RNAs

(miRNA) (Fig. adapted from Bagni and Greenough, 2005).

1.4. The mGluR theory of fragile X syndrome

The mGluR theory is one of the most prominent theories of the underlying cause for fragile

X symptoms. This theory postulates that increased activation of group 1 metabotropic

glutamate receptors (mGluRs) in FXS, and its aberrant downstream molecular consequences

contribute to many molecular and symptomatic abnormalities of FXS. Local dendritic protein

synthesis is essential for many forms of long-term changes in synaptic strength or synaptic

plasticity, mechanisms that are believed to underlie learning and memory (Santoro et al., 2012)

Page 19: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

8

Activation of mGluRs triggers long term depression in the CA1 region of hippocampus

(Huber et al., 2002), which is dependent on the rapid de novo synthesis of new proteins in

synapses. FMRP serves as a brake on the synthesis of synaptic mRNAs that encode long-term

depression (LTD) proteins, including proteins that mediate α-amino-3-hydroxyl-4-isoxazole

propionic acid receptor (AMPA-R) internalisation and stabilise LTD. In Fmr1 mice, mGluR-

LTD is exaggerated and no longer dependent on new protein synthesis, suggesting that the

LTD-related proteins are already expressed at adequate levels (Hou et al., 2006; Nosyreva and

Huber, 2006). Interestingly, postnatal FMRP expression in Fmr1 KO mouse reduces the

mGluR-LTD magnitude and retrieves its dependence on new protein synthesis.

Numerous studies in mouse, zebrafish and drosophila models of FXS have supported the

validity of mGluR theory (for a review see Bhakar et al., 2012). Furthermore, the presumption

that reducing mGluR activity by pharmacological and genetic interventions can normalize FXS

behaviors has been validated in a wide range of animal studies (reviewed in Berry-Kravis, 2014;

Pop et al., 2014). Administration of a potent and selective mGluR5 antagonist, 2-methyl-6-

(phenylethynyl)-pyridine (MPEP) in Fmr1 mice, normalized susceptibility to audiogenic seizure

(Yan et al., 2005; Thomas et al., 2012), impaired eyelid conditioning (Koekkoek et al., 2005),

motor hyperactivity (Yan et al., 2005; Min et al., 2009), defective pre-pulse inhibition and

increased repetitive behavior (Thomas et al., 2012). Furthermore, MPEP corrected several

molecular phenotypes in Fmr1 mice including increased AMPA receptor internalization

(Nakamoto et al., 2007), increased protein synthesis (Osterweil et al., 2010), and increased

dendritic spine density (Su et al., 2011).

The promising results from the use of mGluR antagonists in animal models of FXS strengthened

the rationale and paved the way for the use of these agents in clinical trials. However, the

compelling pre-clinical evidence for the therapeutic benefits of mGluR antagonists in rodent

Page 20: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

9

models of the disease did not translate in human patients in clinical trials. A number of phase 2

clinical trials of mGluR5 antagonists such as fenobam demonstrated some phenotypic

improvement including reduced anxiety and hyper-arousal, improved pre-pulse inhibition, and

higher accuracy in a performance task (Berry-Kravis et al., 2009) and improved adaptive skills

(Berry-Kravis et al., 2008). From these trials, only a few advanced to phase 3, including the

studies investigating the efficacy of selective mGluR5 antagonists such as mavoglurant

(AFQ056), RG7090, and STX107. However, these trials were all terminated in 2014, mostly

due to lack of efficacy. There are a number of reasons why the clinical trials may have failed,

including inappropriate inclusion criteria, inadequate outcome measures, inadequate drug dosing

or development of drug tolerance (Mullard, 2015). The failure of these clinical trials of

mGluR5 antagonists, together with the failure of the GABAB agonist R-baclofen, has prompted

consideration of other therapeutic approaches.

1.5. Prospects of gene therapy application for FXS

Considering the extensive expression of FMRP in the developing brain, it is obvious that

FXS is not merely a bi-product of aberrant mGluR signalling. Accordingly it can be presumed

that certain defects in FXS pathophysiology might be less reversible in response to mGluR

antagonists (Thomas et al., 2011; Thomas et al., 2012). Given the plethora of genes whose

expression is regulated by FMRP, a priori, it may be expected that restoring FMRP expression

in the CNS could provide a more comprehensive reversal of the disorder compared to targeting

single molecules (e.g. mGluR5). The possibility of restoring normal brain function after

introduction of exogenous FMRP into the brain has previously been attempted. Using an AAV5

vector coding for FMRP, Zeier et al, (2009) reported that injections directly into the

hippocampus of five week old Fmr1 mice resulted in correction of the abnormally enhanced

hippocampal long-term synaptic depression (Zeier et al., 2009); however, no other analyses of

Page 21: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

10

phenotypic rescue were carried out and immunocytochemical analysis showed localized

transgene expression only in the hippocampus.

When weighing the possibility of developing a gene therapy approach for FXS, significant

barriers need to be overcome before achieving sufficient therapeutic benefits. In FXS there is

no recognizable, circumscribed locus of pathology, necessitating global delivery of the

transgene in the brain. Furthermore, it is reasonable to presume that cell-specific transgene

expression in KO mice must mimic the WT FMRP expression pattern to provide the maximum

therapeutic benefit. Although FMRP is primarily expressed in neurons of adult WT mice, it is

also present in astrocytes, oligodendrocyte precursor cells, and microglia during the early and

mid-postnatal developmental stages of brain maturation (Gholizadeh et al., 2015). Furthermore,

identifying the therapeutic window for essential levels of FMRP protein in the brain which

translates to rescue of behavioral abnormalities without causing deleterious effects is essential.

1.6. Viral vectors for gene delivery into the CNS

Viruses are among the most efficient biological agents naturally capable of entering cells

through transfection and delivering genetic cargo into the nucleus and possess properties that are

crucial for gene delivery. Recent advancements in viral vector technology have been

encouraging for the development of treatments for disorders of the central nervous system

(CNS). Gene therapy offers promise for treating monogenic neurological disorders, as it can

target the fundamental cause of the loss of functionality in affected cell types within the CNS

through gene replacement. Several viral vectors have been utilized for CNS gene delivery,

including vectors derived from adeno-associated virus (AAV), retrovirus, adeno-

virus and herpes simplex virus. Each of these vectors offers unique solutions to the challenges

of CNS gene delivery. The properties that an ideal vector should possess for gene delivery into

Page 22: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

11

the CNS include high transduction efficiency in the target tissue together with cell type specific

tropism and minimal transduction of “off-target” cells, tissues or organs (Table 3).

Table 2. Clinical trials for gene therapy using AAV vectors in neurological

disorders.

Disease Transgene Trial

Number Phase Status

Published

Results

Parkinson’s

disease

GDNF NCT01621581 1 Recruiting N/A

Parkinson’s

disease

hAADC NCT02418598 2 Recruiting N/A

Parkinson’s

disease

GAD NCT00643890 2 Terminated (LeWitt et al.,

2011)

Parkinson’s

disease

Neurturin NCT00985517 2 Ongoing (Bartus et al.,

2013)

Alzheimer’s

disease

NGF NCT00087789 1 Terminated N/A

Spinal Muscular

Atrophy Type 1

SMN NCT02122952 1 Recruiting N/A

Batten disease CLN2 NCT01414985 2 Recruiting N/A

Data obtained from http://clinicaltrials.gov/. GDNF, Glial cell-derived neurotrophic factor;

hAADC, human aromatic l-amino acid decarboxylase; GAD, Glutamic acid decarboxylase;

NGF, nerve growth factor; SMN, survival motor neuron.

Table 3. Comparison of AAV viral vectors against the ideal vector for gene

delivery to the CNS. Properties of an ideal vector for CNS gene delivery AAV vectors

Ability to transduce dividing and non-dividing cells neurons, astrocytes, glial and

ependymal cells

Minimal pathogenesis no associated pathologies

Page 23: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

12

Long-lasting expression after a single injection 6 months in brain, 6 years in

other tissues of primates

Limited host immunological response Very low or no

immunological response

Ability to pass through the blood brain barrier AAV8, AAV9, AAVrh8,

AAVrh10

Scalable production large scale production of

highly pure vector

Wide distribution in the CNS AAV9, AAVrh10

Main features of AAV vectors in comparison to the ideal vector for gene delivery to the CNS

. (Table adapted from Lentz et al., 2012)

Furthermore, depending on the particular goals of the gene delivery required, the transgene

expression needs to be maintained at a specific level, usually for a protracted period of time to

provide maximal therapeutic impact. Transgene over-expression of specific genes in the CNS

can have cytotoxic effects on target cells and consequently detrimental ramifications on

behavior. High expression levels may also cause a stronger immune response against the

therapeutic transgene in the host body.

Another challenge of targeting the brain for gene delivery, and perhaps the most important is

identifying vectors that are able to cross the blood brain barrier (BBB) which prevents global

transduction of the CNS following non-invasive peripheral delivery of the vectors (Fig. 4).

Furthermore, different viral vectors have various abilities to spread within the brain parenchyma

which may affect their application in brain disorders where a widespread global transgene

delivery is required. .

Page 24: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

13

The efficiency of a virus for gene delivery into the CNS is mainly determined by the natural

properties of the virus. These properties include the packaging capacity, the tissue tropism and

immunogenicity of the virus. Furthermore, the transgene expression level and duration may rely

on the type of the viral genome and its cellular stability. Alterations in the natural biology of

vectors, such as trans-encapsidation or pseudotyping can result in changes in the tropism of viral

vectors, and consequently improvements in the efficiency of transgene delivery in the CNS

(Ojala et al., 2015).

1.7. Adeno-associated viral vectors

Adeno-associated viruses (AAV) are the most common viral vectors now used for CNS gene

delivery, because they possess many properties of an ideal vector described in the previous

section (Table 3). AAVs are non-enveloped, helper-virus dependent parvoviruses with an

icosahedral capsid architecture of approximately 25 nm in diameter. AAVs package ~4.7 kb

genome flanked by ~145 bp inverted terminal repeats (ITRs) on the 5′ and 3′ ends. The WT

AAV genome is a linear single-stranded DNA with two open reading frames. These open

reading frames encode four replication proteins (78, 68, 52 and 40, named by their molecular

weight in kilodaltons), three capsid proteins (VP1, VP2 and VP3) and an assembly activating

protein (Agbandje-McKenna and Kleinschmidt, 2011). The capsid proteins are vital in binding

of the virus to the cell surface receptor which is different for various serotypes, whereas

replication proteins are crucial for the replication and packaging of the viral genome (Ojala et

al., 2015).

Integration of AAV vectors into host chromosome occurs infrequently, at a specific site on

chromosome 19 in most cases; however, this integration is highly dependent on the presence of

Rep 78/68 (McCarty et al., 2004). Recombinant AAV vectors appear to integrate randomly at

an infrequent rate while most genomes are maintained as episomes. It is critical to prevent the

Page 25: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

14

formation of WT AAV vectors with chromosome 19 integration, when making recombinant

AAV vectors. This is done by separating the rep and cap coding sequences from the vector

plasmid DNA.

Importantly, the duration of AAV-delivered transgene expression is essentially permanent in

non-dividing cells following a single administration. AAV vectors can also result in long-term,

stable transgene expression after a single administration in rodents and non-human primates.

AAV-delivered transgenes express for more than 18 months in the mouse brain (Miyake et al.,

2011) and can persist in the serum of rhesus monkey for at least 6 years (Rivera et al., 2005) and

in dogs for over 8 years (Niemeyer et al., 2009; Stieger et al., 2009). Interestingly, a recent

long-term follow-up report from a gene therapy trial of Canvan disease has shown that the

therapeutic effects of AAV-delivered transgenes can persist in the human brain for more than

10 years after intra-parenchymal administration (Leone et al., 2012).

Despite these advantages, the use of AAV vectors for CNS gene delivery is hindered by

several limitations. One of the challenges for the use of AAV vectors in clinical trials is the

presence of humoral responses to the wild-type (WT) AAV, which is common among humans

and the possibility of immune clearance of the vector and vector-infected cells (Boutin et al.,

2010). Approximately 70% of the human population carries neutralizing antibodies against

AAV2, the most studied serotype (Boutin et al., 2010). However, the prevalence of neutralizing

factors against other AAV serotypes such as AAV8 and AAV9 is lower (38% and 47%

respectively) indicating a higher therapeutic potential for human gene therapy trials using these

serotypes (Boutin et al., 2010).

Another constraining factor for single stranded AAV vectors is dependence of transgene

expression on second-strand synthesis (Ferrari et al., 1996). This rate-limiting step in transgene

expression has been circumvented after the development of self-complementary viral vectors

Page 26: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

15

(McCarty et al., 2001). With these vectors earlier and higher expression can be achieved

compared to single-stranded AAV vectors. However, limited coding capacity has restricted the

use of self-complementary AAV for the delivery of transgenes larger than 5 kb. To circumvent

this, dual hybrid trans-splicing vectors have been developed with transgene capacities up

to 8 kb, in which the transgene cassette is divided between two vectors and assembled into the

full-length transgene only when the two vectors infect a single cell together (Duan et al., 2001).

1.8. Subsequent steps in AAV cell entry and implications for gene

delivery into the CNS

Efficient transduction by AAV vectors relies on several essential steps including cell surface

binding, endocytosis, trafficking to the nucleus, nuclear entry, capsid uncoating, genome

release, second strand synthesis, and finally transcription. The capsid proteins (VP1, VP2, VP3)

determine the interactions with the host cell surface receptors (Huang et al., 2014). Cell surface

glycans serve as the most common receptors for the majority of natural AAVs (Asokan et al.,

2012) (Table 4). Among different AAV serotypes, AAV9 is one of the most effective vectors

for gene delivery into the CNS (Weinberg et al., 2013). AAV9 is able to induce extensive

neuronal and glial transduction through different routes of administration in a variety of animal

models (for a review see Murlidharan et al., 2014). Apart from the important features of the

capsid proteins, AAV vector mediated gene delivery can also be affected by multiple trafficking

factors within the CNS, which will be discussed further in sections 1.9 to 1.12.

1.9. Routes of AAV administration into the CNS

One of the main factors affecting the efficiency and cellular tropism of AAV is the route of

administration. Four of the most commonly used routes for CNS gene delivery are discussed

below. Different routes of administration can affect interactions of AAV vectors with different

Page 27: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

16

receptors, vector dissemination from the site of injection, directional transport of the vector and

eventually efficiency and cellular tropism of transfection (Fig. 3).

Table 4. Primary receptors and gene delivery tropism of different AAV

serotypes in the mammalian CNS Serotype Primary

receptor

Intra-CSF or intra-

parenchymal administration

Intra-vascular

administration

Axonal

transport

Neuronal

transduction

Glial

transduction

Neuronal

transduction

Glial

transduction

AAV1 α2,3/α2,6 N-

linked Sialic

acid

++ + + +

A–,R+

AAV2 Heparan

sulfate + – – –

A+,R–

AAV4 α2,3 O-linked

Sialic acid – + – –

?

AAV5 α2,3 N-linked

Sialic acid ++ + – –

?

AAV6 α2,3/α2,6 N-

linked Sialic

acid/heparan

sulfate

++ – + +

A–,R+

AAV8 ? ++ ++ ++ ++ A+, R+

AAV9 N-linked β1,4-

Galactose +++ ++ +++ +++

A+,R+

AAVRh.8 ? ++ ++ +++ +++ ?

AAVRh.10 ? +++ + +++ +++ ?

? Receptor usage/axonal transport has not been characterized; + low levels of transduction;

++ moderate levels of transduction; +++ high levels of transduction; – no transduction; ? A+ or

R+ (AAV vector undergoes axonal transport in the anterograde (A) or retrograde (R) direction

during in vivo characterization). Table adopted from (Murlidharan et al., 2014).

Page 28: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

17

Fig. 3. Major factors affecting tropism, transduction and transgene

expression properties of AAV

A. Anterograde gene transport can result in transgene expression distal to the AAV injection

site. This occurs through axonal transport of the protein product. Retrograde viral transport

(e.g. from muscle tissue to motor neuron cell bodies) requires viral transport from the delivery

site to the neuronal nucleus. B. Three major CNS delivery routes including intra-cranial, intra-

cerebroventricular, and intra-venous delivery. C. Encapsidation of different AAV serotypes can

dramatically alter both overall tissue transduction volume and specific cellular expression levels.

(Fig. adapted from Weinberg et al., 2013).

1.9.1. Intra-cranial Administration

Intra-cranial administration of AAV has been the most common route for vector

administration to the brain parenchyma (Gelfand and Kaplitt, 2013; Kantor et al., 2014;

Murlidharan et al., 2014; Ojala et al., 2015). Compared to intravenous systemic delivery, intra-

cranial administration circumvents the challenging requirement of passing through the BBB

Page 29: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

18

(Fig. 4), and minimizes the risk of vector neutralization by anti-AAV antibodies, since the levels

of neutralizing antibodies in brain parenchyma are less than 1% of their levels in the blood

(Treleaven et al., 2012).

However, intra-cranial vector delivery has significant down sides. It is perhaps the most

invasive route of vector administration to the CNS, with a risk of hemorrhage, edema and

bacterial infection. Limited vector dissemination from the injection site is another major

drawback in brain disorders where efficient transgene delivery is essential in large brain regions

such as Parkinson’s disease or Alzheimer’s disease, or the entire brain, such as Fragile X

syndrome, Rett syndrome or lysosomal storage disorders. Multiple intracranial injections

spaced throughout the brain have been used with limited success in clinical trials of lysosomal

storage disorders such as Canavan disease (Leone et al., 2000; McPhee et al., 2006) and late

infantile Batten disease (Crystal et al., 2004; Worgall et al., 2008). It is estimated that 50-350

intracranial injection tracts are required for sufficient gene delivery to the entire human brain in

these disorders, which is technically impossible (Cunningham et al., 2008).

AAV diffusion in brain parenchyma can be enhanced using a technique referred to as

convection-enhanced delivery (Hadaczek et al., 2006; Carty et al., 2010; Barua et al., 2013),

which takes advantage of the convective flow in the interstitial fluid, in addition to simple

diffusion, to accelerate the spread of viral vectors away from cannula tip at the injection site.

This technique was safely used for intracranial delivery of AAV2 in a phase 1 clinical trials of

patients with Parkinson’s disease (Mittermeyer et al., 2012), and is currently being assessed in

two ongoing large-scale double-blinded clinical trials of Parkinson’s disease (ClinicalTrials.gov

Identifiers: NCT01621581 and NCT01973543).

Page 30: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

19

1.9.2. Intra-vascular Administration

Intravascular delivery provides a non-invasive route to deliver viral vectors into the whole

brain with a single injection, by taking advantage of the dense network of capillaries in the

brain. However, the application of this approach is obviously limited by the BBB, which

consists of endothelial cells, with tight junctions that preclude the transfer of most AAV

serotypes to brain parenchyma. A few serotypes such as AAV9 and AAVrh10 have the ability

to bypass through BBB, possibly through receptor-mediated transcytosis (Di Pasquale and

Chiorini, 2006; Duque et al., 2009), but the exact para-cellular or trans-cellular mechanisms

remain to be identified (Zhang et al., 2011).

Peripheral delivery of the vector often results in off-target undesirable transduction which

might be detrimental, depending on the endogenous expression pattern of the transgene, and at

high doses can lead to liver toxicity(Chandler et al., 2015). Furthermore, as this route delivers

only a small number of virus particles to the brain and spinal cord, it is necessary to administer

higher doses of the vectors compared to direct intracranial or i.c.v. routes. This entails a higher

risk of immunological response from the body (Samaranch et al., 2014), given that almost 50%

of individuals are sero-positive for both AAV9 (Mingozzi et al., 2007) and AAVrh10 viruses

(Thwaite et al., 2015).

Page 31: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

20

Fig. 4. The brain barriers in the developing and adult brain

A. The blood-CSF barrier. A barrier between choroid plexus blood vessels and the cerebro-

spinal fluid (CSF). B. The blood-brain barrier. A barrier between the lumen of cerebral blood

vessels and the brain parenchyma. C. The inner CSF-brain barrier, present only during early

development. A barrier between the CSF and the brain parenchyma. D. The outer CSF-brain

barrier. A barrier between the CSF-filled subarachnoid space (sas) and overlying structures.

Abbreviations: arach, arachnoid membrane; cpec, choroid plexus epithelial cells; dura, dura

mater; nu., nucleus; pia, pia mater; sas, subarachnoid space (Fig. adapted from Liddelow, 2011).

Page 32: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

21

1.9.3. Intra-CSF

Direct delivery to the CSF reduces peripheral off-target transduction, and lowers vector

neutralization by circulating serum antibodies. Direct AAV administration into ventricles,

cisterna magna, or intra-vertebral lumbar puncture has resulted in extensive transduction in

multiple areas of the CNS (for a review see Murlidharan et al., 2014).

Intra-cerebroventricular (i.c.v.) administration provides widespread transduction of CNS

through transporting the viral vectors in the CSF, as well as transduction of the ependymal cell-

lining of the ventricles, from which transgene can be expressed into the CSF, and distributed

throughout the brain. Transduction of the ependymal cells, which consist of adult neural stem

cells with the ability of lifelong migration and differentiation (Alvarez-Buylla and Lim, 2004;

Lim and Alvarez-Buylla, 2014), makes i.c.v. a viable route for more widespread transduction in

the brain compared to intra-cranial injections. Among different AAV serotypes, AAV9 is one

of the most efficacious serotypes for i.c.v. delivery both in neonatal and adult mice (Haurigot et

al., 2013; Dirren et al., 2014; McLean et al., 2014; Gong et al., 2015).

Intra-thecal AAV vector delivery has also been broadly investigated, as it provides

advantages conferred by the CSF connectivity between the brain and the spinal cord. These

injections are typically performed by exposing either the sub-arachnoid space at the sub-

occipital cisterna magna region or the intra-vertebral space at lumbar region. The lumbar

puncture has been utilized in studies where efficient transduction of motor, sensory or

nociceptive neurons were required (e.g., within dorsal root ganglia). Preclinical studies utilizing

an intra-thecal route of administration for AAV vectors have been encouraging in rodents and

non-human primates (Gray et al., 2013; Hirai et al., 2014; Dirren et al., 2015; Hinderer et al.,

2015; Hordeaux et al., 2015). From different AAV serotypes, AAV6 and AAV9 yield the most

efficient transduction in the mouse brain (Snyder et al., 2011). Consistent with this, intra-thecal

AAV9 administration provided widespread transduction in the brain and spinal cord in mice

Page 33: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

22

(Hirai et al., 2014; Dirren et al., 2015), as well as dogs and rhesus monkeys (Hinderer et al.,

2015) and pigs (Federici et al., 2012). The successful scaling of the intra-CSF approach to

larger non-human primates suggests the potential for translation to human therapy.

1.9.4. Retrograde transport delivery

Retrograde axonal transport of vectors (i.e. transport of vectors from axonal terminals back to

cell bodies) can provide a non-invasive gene delivery via peripheral intra-muscular vector

administration and subsequent vector transfer to cell bodies in the spinal cord. This approach is

particularly favorable for transducing motor neurons affected in disorders such as amyotrophic

lateral sclerosis and spinal muscular atrophy. Using this approach, intra-muscular AAV9

vectors have been used to deliver the survival motor neuron gene into motor neurons in neonatal

(Foust et al., 2010) and adult (Benkhelifa-Ziyyat et al., 2013) mouse model of spinal muscular

atrophy. In both studies, more than 40% of motor neurons were transduced which resulted in

improvements in motor function, and survival (Foust et al., 2010; Benkhelifa-Ziyyat et al.,

2013).

1.10. Timing of AVV administration

In addition to capsid serotype and route of administration, other factors such as age of

animals when receiving injections also influences efficiency and CNS bio-distribution of AAV

transduction from systemic i.v. or i.c.v. injections. Injection of viral vectors in neonatal mice

(postnatal day 0 – 2) provides a wider distribution in the brain and a higher chance of passing

through the BBB (Miyake et al., 2011; Yang et al., 2014), which is more permeable due to its

immature status (Ek et al., 2012). AAV2/1 i.c.v. injections into mouse pups on their day of birth

results in widespread transduction (Passini et al., 2003; Levites et al., 2006), but vector spread

was attenuated on postnatal day 2 injections (Chakrabarty et al., 2010).

Page 34: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

23

The timing of AAV administration also governs cellular tropism of transduction after global

CNS delivery. Foust et al. (2009) demonstrated that intra-venous administration of self-

complimentary AAV9 resulted in extensive transduction of neuronal cells in neonatal mice,

while expression was predominantly restricted to astrocytes in adults (Foust et al., 2009). These

differences in transduction tropism might be explained by the ongoing development of

astrocytes in neonatal mice and the interactions between astrocytic endfeet and endothelial cells.

Considering the important contributions of astrocytic processes to the BBB biology, there are

significant alterations in the mouse BBB composition within the first postnatal week. Follow-

up studies using i.c.v. injections of AAV serotypes 1, 8, and 9 performed on postnatal day 0

mice demonstrated preferential neuronal tropism. However, from injections performed on

postnatal day 1 or later (24–84 hours postnatal), the tropism changes remarkably towards

astrocytes (Chakrabarty et al., 2013). These studies highlight how the important effects of

developmental age on AAV9 tropism and efficiency, which should be considered when

assessing the potential of these vectors for human translation.

Page 35: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

24

Fig. 5. AAV9 tropism in the CNS at different developmental stages

Transduced cells are shown in green, non-transduced cells in gray. In neonates (left), AAV9

can easily cross an immature endothelium and diffuses through a lax extracellular matrix to

transduce neurons, astrocytes and microglia. In adults (right), the brain endothelium has

become a tight barrier surrounded by a cuff of closely packed astrocyte end feet. AAV9 can still

cross the brain endothelium but does not seem able to get beyond the glial barrier, thus

transducing mainly astrocytes (presumably by infecting end feet) and endothelial cells, and

possibly pericytes and perivascular macrophages, but only very few neurons or microglia. (Fig.

taken from Lowenstein, 2009).

1.11. Promoter selection

Based on specific goals of the gene delivery, gene expression can be regulated by employing

a cell type specific promoter to restrict expression to specific transduced cell types, or a

ubiquitous promoter of the desired strength to adjust the overall level of the target transgene

expressed. However, promoter strength can also be vastly affected by the choice of 5' UTR, 3'

UTR, enhancer, and poly-adenylation signal (see Table 5 for a list of common promoters and

the elements affecting their strengths). One challenge in choosing the most appropriate

Page 36: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

25

promoter for AAV vectors is that the promoter and transgene must be small enough to fit within

the restricted packaging capacity of the AAV virus.

Two of the most commonly used ubiquitous promoters are the cytomegalovirus (CMV)

promoter and the chicken beta actin (CBA) promoter, both with an approximate size of 800 bp

(including the CMV enhancer and 5' UTR sequence) (Gray et al., 2011). The CMV promoter

is stronger than CBA, however it is more prone to silencing over time in the brain. To provide

ubiquitous long-term sustainable expression at high levels, CBh hybrid promoters have been

developed by the incorporation of MVM introns in the CBA promoter (Gray et al., 2011).

Furthermore, translation may be additionally enhanced utilizing heterologous viral mRNA

stabilization elements in AAV cassettes, such as the woodchuck hepatitis virus post-

transcriptional response element (WPRE). For neuronal transgene expression, the neuron-

specific enolase (NSE), synapsin, MeCP2 or the platelet-derived growth factor (PDGF)

promoters can be used, with sizes of 2.2 kb, 470 bp, 229 bp, and 225 bp respectively.

Astrocytic expression can be achieved using a truncated glial fibrillary acidic protein (GFAP)

promoter (Lee et al., 2008), and oligodendrocytes can be targeted using myelin basic protein

promoters.

Table 5. Promoters commonly used for gene delivery into the CNS.

Enhancer Promoter 5'UTR/intron Strength Size Specificity

CMV CMV SV40 High 800bp Ubiquitous

CMV CBA SV40 High 800bp Ubiquitous

CMV CBA CBA-MVM High 800bp Ubiquitous

None UBC None Weak 430bp Ubiquitous

None GUSB None Weak 378 bp Ubiquitous

None NSE None Strong 2.2 kb Neuron

None Synapsin None Medium 470bp Neuron

None MeCP2 None Weak 229bp Neuron

None GFAP None Medium 681 bp Astrocyte

CMV CBA CBA-MVM High 800bp Ubiquitous

CMV: cytomegalovirus; CBA: chicken Beta actin; UBC: ubiquitin C; GUSB: beta

glucuronidase; NSE: neuron-specific enolase; GFAP: glial fibrillary acidic protein; MVM:

Page 37: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

26

minute virus of mice (Table taken from Gray et al., 2011).

1.12. AAV transport within the CNS

Following vector administration and interactions with cell surface receptors, AAV vectors

undergo interstitial and intra-cellular transport throughout the CNS. The spread of AAV vectors

throughout the CNS are mainly affected by two major mechanisms, namely para-vascular CSF

transport, which is responsible for the spread of interstitial fluid within the CNS, and the axonal

transport. Recent findings by Iliff et al. (2013) showed that para-vascular movement of CSF by

the “glymphatic pathway” clears accumulations of metabolites from the brain parenchyma (Iliff

et al., 2013). These results indicate that CSF transport mechanisms can affect the extent of virus

dispersion within the CNS.

Once viruses enter the host neurons, axonal transport serves as another established pathway

affecting virus diffusion within the CNS. In case of AAV vectors, both unidirectional and

bidirectional (anterograde or retrograde transport or both) axonal transport have been

established depending on the strain of the virus (table 4). AAV9, for instance, is believed to be

able to travel in both anterograde and retrograde directions (Castle et al., 2014).

1.13. Hypotheses and objectives

There is no pharmacological cure for FXS and the currently prescribed medications only

partially alleviate selected symptoms and are associated with deleterious side effects.

Considering the plethora of genes whose expression is regulated by FMRP, a priori, it may be

expected that restoring FMRP expression in the CNS could provide a more comprehensive

reversal of the disorder compared to targeting single molecules (e.g. mGluR5). Our overarching

hypothesis was that introducing the Fmr1 gene into the brains of neonatal Fmr1 KO mice, using

i.c.v. injection of AAV vectors serotype 2/9 would provide widespread transgene dispersion

Page 38: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

27

throughout the brain, and will prevent or ameliorate the neurodevelopmental impairments in the

Fmr1 KO mice.

The specific objectives of the present study were:

1. Analysis of FMRP cell type expression: To investigate FMRP expression in neurons,

astrocytes, microglia and oligodendrocyte precursor cells in the developing and adult brain of

WT mice by immunocytochemistry (Chapter 3, Gholizadeh et al., 2015).

2. Efficiency of AAV-mediated transgene delivery: To determine the most effective route(s) of

administration, age of injection and promoter that would lead to wide-spread transduction in the

brains of neonatal mice using AAV5 and AAV9 vectors expressing enhanced green fluorescent

protein (eGFP) (Chapter 4, Gholizadeh et al., 2013).

3. FXS gene therapy-Phase 1: To assess the efficacy of administering an AAV9 vector

containing the Fmr1 gene driven by the neuron- specific human synapsin promoter directly into

the CNS of Fmr1 KO mice via i.c.v. administration to PND 5 Fmr1 KO mice (Chapter 5,

Gholizadeh et al., 2014).

4. FXS gene therapy-Phase 2: To assess level and distribution of FMRP and cell-type

specificity of transduction, after i.c.v. injection of a viral vector coding for FMRP, in PND-0

Fmr1 KO mice and also to extend the behavioral analyses to include additional tests not

employed in phase 1 (e.g. anxiety and pre-pulse inhibition) (Chapter 6).

5. To study the potential effects of FMRP over-expression on mouse behavior by i.c.v.

injections of AAV-FMRP in WT mice at PND 0-1 followed by behavioral analysis starting 60

days post-injection (Chapters 6).

Page 39: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

28

Chapter 2. Materials and Methods

2.1. Animals

All injections were carried out using C57/BL6 wild-type (WT) or Fmr1 knockout (KO) mice

on postnatal day (PND) 0-1, 5 or 21. Mice were kept in a room maintained at constant

temperature (21 ± 2 °C) and humidity (55 ± 5%) with an automatic 12 h light/dark cycle and

free access to standard laboratory diet and tap water. 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.

2.2. AAV vectors

Various single-stranded AAV vectors were used with different promoters or cDNA transgene

sequences. AAV vector containing the inverted terminal repeat DNA sequences from the

genome of serotype type 2 (AAV2), and the capsid protein genes from serotype 5 (AAV5) or

serotype 9 (AAV9) were used. The AAV9 vectors contained the cDNA for the enhanced green

fluorescent protein (eGFP), driven by either the ubiquitous cytomegalovirus (CMV) promoter,

referred to here as “AAV-CMV-eGFP”, or the human synapsin-1 (SYN) promoter, defined here

as “AAV-SYN-eGFP”. We also used AAV9 vectors containing the cDNA for the major murine

isoform of Fmr1 (isoform 1) (Brackett et al., 2013), driven by the neuron-specific human

synapsin (SYN), referred to here as “AAV-FMRP”, or empty control vectors containing no

transgene, here referred to as “AAV-null”.

Single stranded AAV5 vectors were constructed and packaged at the University of Florida

Powell Gene Therapy Center. The AAV5 vectors contained an eGFP reporter gene driven by

the chicken-β-actin (CBA) promoter (AAV5-CBA-eGFP) and the titer was 1.24 × 1013

genomes/ml. AAV9 vectors were supplied by the University of Pennsylvania Vector Core

facility (Philadelphia, PA, USA) and were titer-matched to 1 × 1013

genomes/ml. A woodchuck

Page 40: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

29

hepatitis post-transcriptional regulatory element (WPRE) sequence, was inserted in the AAV9

expression cassette downstream of the eGFP or Fmr1 gene to enhance stabilization of the

mRNAs and to eventually increase transgene expression (Loeb et al., 1999; Cederfjäll et al.,

2012). All AAV vectors were suspended in sterile phosphate-buffered saline (PBS) and stored

at -80ºC.

2.3. Vector injections

2.3.1. i.c.v. injections in neonatal mice

For injection of PND 0 or PND 5 mouse neonates, the pups were immobilized via cryo-

anesthesia for 2 minutes and the immobilized pup was grasped by the skin behind the head and

placed on a fiber-optic light to illuminate the midline and transverse sutures that were used as a

guide for injections. A 30-gauge needle attached to a 5 µl Hamilton syringe (Hamilton, Reno,

NV) through long polyethylene tubing was used. For PND 0 injections the needle was inserted

2 mm deep, perpendicular to the skull surface, at a location approximately 0.1 mm lateral to the

sagittal suture and 0.50 mm rostral to the neonatal coronary suture. PND 5 injections were

performed 2 mm deep, 0.25 mm lateral to the sagittal suture and approximately 0.75 mm rostral

to the neonatal coronary suture. 0.75 - 1 µl of the vector or vehicle (PBS or AAV-null vector)

was injected using a syringe pump (NE-1002X, New Era Pump Systems, Farmingdale, NY) at

the rate of 1 µl/min into each lateral ventricle. The needle was left in place for 1 minute after

discontinuation of plunger movement to prevent backflow. The pups were allowed to recover in

a warmed container in order to return to normal temperature and were placed back into the cage

with the dam after normal movement and general responsiveness were restored.

Page 41: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

30

2.3.2. Stereotaxic injections in juvenile mice

Juvenile PND 21 animals were anesthetized with isofluorane and secured in a stereotaxic

frame (David Kopf instruments, Tujunga, CA). The skull was exposed and holes the size of the

30 gauge injection needle were drilled into the skull. AAV5 or AAV9 vectors expressing eGFP

were injected bilaterally into the lateral ventricles with 1 μl of vector or PBS per side at the rate

of 0.5 μl/minute. Coordinates for injections were 0.22 mm caudal to bregma, ±1 mm lateral to

the midline, and 1 mm ventral to pial surface. Following infusion, the needle was left in place

for 5 minutes prior to being slowly retracted from the brain. The incision was cleaned with

sterile saline and closed with taper point non-absorbable 5-0 surgical sutures (Syneture,

Mansfield, MA). After surgery, the mice were housed individually and were monitored every

day for signs of stress or infection until euthanasia.

2.4. Behavioral analysis

All behavioral tests were conducted on aged and sex-matched WT and KO animals. All mice

were naive to the tests and tested only once in each test. The experimenters were blinded to the

treatment groups during the time of testing.

2.4.1. Locomotor activity measurements

Locomotor activity was assessed using an automated open field locomotor monitor system

(Accuscan Images, Salt Lake City, USA). Mice were acclimated to the testing room for 5

minutes, then placed in the open field and monitored for 20 minutes under low-light conditions.

The total distance covered (horizontal movement) was recorded using the Fusion software

(Fusion software, Johannesburg, South Africa). Total distance travelled was compared between

groups.

Page 42: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

31

2.4.2. Marble Burying for stereotypic behavior

Approximately 10 cm of laboratory animal bedding (Bed-o'Cobs combination bedding;

Andersons Inc, Maumee, USA) was added to empty cages and blue marbles were placed

equidistant from each other in a 4–by-5 grid covering two thirds of the surface. Mice were

acclimated to the testing room for 5 minutes, and then placed in the cage on the side devoid of

marbles. Mouse activity was left undisturbed for 30 min and then the number of buried marbles

was counted; a buried marble consisted of any marble where less than 50% of its surface was

left uncovered by bedding.

2.4.3. Ultrasonic vocalizations

Virgin adult female mice were placed in a new cage that contained only bedding for two

minutes and the cage was fitted with a customized polystyrene cover. Subsequently, a virgin

male was placed into the same cage and ultrasonic vocalizations (USV) were recorded using an

Ultrasound Detector D 1000X (Pettersson Elektronik AB, Uppsala, Sweden) for 4 minutes as

described previously (Wang et al., 2008). The distinct waveform patterns seen in the

spectrograph rendering, indicative of vocalizations, were counted. The first 5 seconds of

recording following the introduction of the male was also ignored. The total number of

vocalization contained within 4 minutes of recording was compared between animals.

2.4.4. Tube test of social dominance

The mice were tested using the tube test to measure social dominance. Each match involved

two mice of different genotype (WT or Fmr1 KO) or injection groups (PBS, AAV-null or AAV-

FMRP) that were not housed together. The experimenter was blind to the category of the

groups being tested. One mouse was placed into each end of a transparent PVC tube (2.5 cm

inner diameter, 30.5 cm length) and the mice were released simultaneously. The match ended

when one mouse placed at least two paws outside the tube with the mouse remaining inside the

Page 43: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

32

tube being deemed the “winner”. The number of wins for each group or genotype was tallied

and a chi-square analysis was used to determine whether the percent of wins in each group was

significantly different from the 50:50 win/loss outcome expected by chance. Each animal was

tested three to five times, depending on pairing availability, against animals of opposing

injection group and/or genotype.

2.4.5. Elevated plus maze test of anxiety

The elevated plus maze consisted of four arms (two open without walls and two enclosed by

15.25 cm high walls) 30 cm long and 5 cm wide. Each arm of the maze was attached to sturdy

legs such that it was elevated 40 cm off from the floor. The maze was placed in the testing

room in a way that there were similar levels of illumination on both open and closed arms. The

legs of the maze were adjusted so that the maze was perpendicular to the ground and each arm

was level. Each mouse was placed at the junction of the open and closed arms, facing the open

arm opposite to where the experimenter was. The mouse was allowed to move freely in the

maze for 5 minutes while the total number of entries in both the open and closed arms, as well

as the total time spent in each arm was measured by an observer. The observer was blind to the

experiment groups and made minimal movements and no noise during the experiment. An arm

entry was counted when all four paws of the mouse were on the arm. At the end of the 5-min

test, the mouse was removed from the plus maze and placed into its home cage. The maze was

cleaned with Virox and dried with paper towels before testing with another mouse.

2.4.6. Pre-pulse inhibition for sensorimotor gating

Sensorimotor gating was measured by pre-pulse inhibition (PPI) of the startle response, as

previously described (Thomas et al., 2011). PPI of acoustic startle responses was measured

using the SR-Lab System (San Diego Instruments, San Diego, CA). The apparatus comprised

of a sound attenuating chamber, which contained a cylindrical tube. A test session began by

Page 44: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

33

placing a mouse in the Plexiglas cylinder where it was left undisturbed for 5 min. Each test

session consisted of 48 trials comprising of 6 blocks of eight trial types each presented in a

pseudo random order such that each trial type was presented once within a block of seven trials.

One trial type was a 40 millisecond, 120 dB sound burst used as the startle stimulus. There were

five different acoustic pre-pulse plus acoustic startle stimulus trials. The pre-pulse sound was

presented 100 milliseconds before the startle stimulus. The 20 milliseconds pre-pulse sounds

were 74, 78, or 82 dB. Finally, each block had a trial where no stimulus was used to measure

baseline movement in the cylinders where no sound was presented. The inter-trial interval

ranged from 10 s to 20 s, and the startle response was recorded every 1 millisecond for

65 millisecond following the onset of the startle stimulus. The startle response was recorded for

65 milliseconds (measuring the response every 1 millisecond) starting at the onset of the startle

stimulus. The background noise was 70 dB in each chamber. The maximum startle amplitude

during the 65 milliseconds was utilized as the dependent variable. The following formula was

used to calculate percent PPI of a startle response: 100 – [(startle response on acoustic pre-pulse

+ startle stimulus trials / startle response alone trials) × 100]. Thus, a high percent PPI value

indicates that the subject showed a reduced startle response when a pre-pulse stimulus was

presented compared to when the startle stimulus was presented alone. Conversely, a low

percent PPI value indicates that the startle response was similar with and without the pre-pulse.

2.4.7. Audiogenic Seizures

For audiogenic seizure testing, the apparatus consisted of a Plexiglas mouse cage (28 × 17 ×

14 cm) with a 135-dB sound source (Piezo siren, Electrosonic; Piezo Technologies,

Indianapolis, IN) attached to the lid and extending 5 cm down into the cage. Mice (30-31 days

old) were placed individually into the testing apparatus and were allowed to explore for 2 min,

after which the bell was rung for 2 min. Seizure activity was observed and scored using a

Page 45: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

34

seizure severity score as follows: wild running = 1; clonic seizure = 2; tonic seizure = 3; status

epilepticus/respiratory arrest/death = 4 (Musumeci et al., 2000). Animals were considered to

have had a seizure if the seizure severity score was greater than 1. Animals were tested only

once. Seizure testing was carried out between 1:00 PM and 5:00 PM. All mice were

immediately euthanized at the end of the test.

2.5. Tissue preparation, immunocytochemistry and confocal microscopy

PND 10, PND 20, PND 30 and adult (PND 55-65) WT or KO mice were anesthetized with a

mixture of ketamine and xylazine (intra-peritoneal, 80 mg/kg and 8 mg/kg respectively) and

perfused transcardially with a solution of PBS (pH = 7.4) followed by 4% paraformaldehyde

(pH = 7.4). For PND 0, pups were decapitated, rinsed with PBS and the skull was fixed in 4%

PFA at room temperature for 2-4 hours. The brain was then removed from the skull and further

incubated in 4% PFA at 4 °C for 20-24 hours. Perfused brains were then removed and placed in

4% paraformaldehyde overnight at 4 °C, then transferred to 30% sucrose for cryo-protection.

Once the brains sank in the sucrose, they were mounted in optimum cutting temperature solution

(Sakura, Torrance, CA) and frozen at −20 °C until sectioning. Serial coronal or sagittal

sectioning sectioning of the entire brain was performed at a thickness of 25 μm using a cryostat

(Leica Microsystems, Wetzlar, Germany). Free-floating sections were rinsed with Tris buffered

saline and antigen retrieval was performed as described previously (Gabel et al., 2004).

Monoclonal mouse anti-FMRP (2F5; 1:1000; gift from Dr. Jennifer Darnell, or 5c2, 1:1000; gift

from Richard Kascsak) was used along with one of the following monoclonal rabbit primary

antibodies: NeuN (1:1000; Abcam) to immunolabel neurons and anti-S100β (1:1000; Abcam) to

mark astrocytes. For analyzing the cell-type specific expression of FMRP in the WT mouse

brain, sections were also stained with rabbit polyclonal anti-Iba-1 (1:2000; WAKO, Richmond,

VA, USA) to immunolabel microglia, and rabbit polyclonal anti-NG2 (1:500; Millipore,

Page 46: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

35

Temecula, CA, USA) to stain oligodendrocyte precursor cells. To assess the transduction

efficiency of AAV-eGFP vectors, polyclonal rabbit anti-GFP (1:2000; Abcam, Cambridge,

USA) was used along with each of the following monoclonal mouse primary antibodies: NeuN

(1:200; Millipore, Temecula, CA, USA) to mark neurons and anti-S100β to mark astrocytes.

Cerebellar sections were also co-incubated with a polyclonal rabbit anti-calbindin antibody

(diluted 1:10,000; Swant, Marly, Switzerland) to label Purkinje neurons. After overnight

incubation, 5 washes with Tris buffered saline for 10 minutes each were carried out and

secondary antibodies diluted in Tris buffered saline containing 5% goat serum were applied.

The sections incubated with anti-FMRP were labeled with goat anti-mouse Alexa Fluor 488 and

goat anti-rabbit Alexa Fluor 594 (1:1000; Jackson ImmunoResearch Laboratories, Inc. West

Grove, PA). The images were captured using a laser-scanning confocal microscope (Nikon A1,

Tokyo, Japan) at 10, 40, 60 or 100X magnifications and analyzed using the NIS-Elements

software (Nikon Instruments, Tokyo, Japan). The images for sagittal sections were captured

using a Zeiss Mirax slide scanner at 20X magnification.

2.5.1. Semi-quantitative and quantitative analysis of transgene expression in the

brain

For a comparative analysis of the transduction pattern in the brain, two scoring methods

were used: a semi-quantitative scoring system to analyze transduction in different brain regions

and a quantitative cell-count of transduced cells in the retrosplenial cortex. We first used a

semi-quantitative scoring system to estimate transduction efficiency of different AAV vectors in

discrete regions of the mouse brain including the prefrontal cortex, striatum, cingulate cortex,

hippocampus, retrosplenial cortex, thalamus, piriform cortex, inferior colliculus, superior

colliculus, brainstem and cerebellum. The semi-quantitative scoring system was used to analyze

the cellular tropism of the AAV-eGFP or AAV-FMRP in different brain regions. Scoring was

done by classifying the number of transduced cells in four categories: regions with no detectable

Page 47: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

36

transgene expression, regions with 0 - 100 transgene expressing cells/mm2, regions with 100 -

200 transgene expressing cells, and regions with more than 200 transgene expressing cells.

Because individual cells were most easily resolved and distinguished in the retrosplenial

cortex and cingulate cortex, these brain regions were targeted for quantitative analysis of

neuronal vs. astroglial virus transduction in photomicrographs captured at 10X or 40X

magnifications. The boundaries of the retrosplenial cortex and cingulate cortex were delineated

according to the Paxinos atlas of the mouse brain (Paxinos and Franklin, 2013) in 3 consecutive

25 µm sections stained for transduced cells via GFP or FMRP immunoreactivity. Quantification

was carried out by an individual masked to the time and type of AAV vector administration.

The total number of transgene-positive cells in each group as well as the percentage of

transgene-positive cells that co-localized with the neuronal marker NeuN or the astrocytic

marker S100β was recorded.

2.6. Quantitative western blotting

Samples of the inferior colliculus, cerebellum, cortex, frontal cortex, hippocampus and

striatum were collected and stored at – 80 oC. After electrophoresis the proteins were

transferred onto nitrocellulose membranes and then incubated overnight with primary antibodies

including mouse anti-FMRP 5c2 (1:250), (LaFauci et al., 2013) and mouse anti-GAPDH

(1:40,000; Clone GAPDH-71.1; Sigma-Aldrich, Saint Louis, Missouri, USA). The sections

were incubated with horseradish peroxidase-conjugated goat anti-mouse (Jackson

ImmunoResearch Laboratories, Inc.; West Grove, PA, USA); after washing, SuperSignal West

Pico Chemiluminescent Substrate (Thermo Scientific; Rockford, IL, USA) was added to the

membranes and the bands were revealed by exposure under Alpha Innotec Fluorochem gel

imager (Protein Simple; Toronto, ON, Canada). Images were analyzed using AlphaEase SA.

Quantification of protein expression was normalized to GAPDH expression. Results are

Page 48: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

37

presented as averages ± S.E.M. Statistical significance was determined by using one-way

ANOVA and Tukey's multiple comparison tests.

2.7. Statistical analyses

Fisher's exact test was used for statistical analysis of audiogenic seizure incidence between

the groups. In the tube test, a chi-square analysis was used to determine whether the percent of

wins were significantly different from the 50:50 win/loss outcome expected by chance. For

motor activity, marble burying, ultrasonic vocalization, elevated plus maze and pre-pulse

inhibition tests, one-way ANOVA test was performed followed by Bonferroni post-hoc test.

The GraphPad prism software (version 6) was used to perform the statistical analyses.

Page 49: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

38

Chapter 3. Expression of FMRP protein in neurons and glia of the developing and adult mouse brain

Acknowledgements: Immunocytotochemical analyses and imaging were performed with

assistance from Dr. Sebok Kumar (Post-doctoral fellow).

3.1. Specific hypotheses, objectives and rationale

The purpose of this phase of the study was to fully document the cell type expression of

FMRP in neurons and glia in the developing and mature brain of WT C57/BL6 mice. In

contrast to the expression of FMRP in neurons of the adult brain, relatively little is known about

the types of glial cells that express FMRP during CNS development. Because fragile X

syndrome is a neurodevelopmental disorder, a comprehensive analysis of its cell type

expression in the developing brain can provide a better understanding of the molecular functions

of FMRP, and the pathogenesis of the syndrome. Moreover, this information was used in the

generation of our AAV vectors. Our analysis encompassed the cerebral cortex, striatum,

hippocampus, cerebellum, and the corpus callosum, and extended from postnatal days (PNDs) 0

(day of birth), to PNDs10, 20, and adult (8-12 weeks old) WT mice. We investigated FMRP

expression in the brain by double-labeling immunocytochemistry using an anti-FMRP antibody

and cell type markers for neurons (NeuN), astrocytes (S100β), microglia (Iba-1) or

oligodendrocyte precursor cells (NG2).

3.2. Gradual reduction in the number of FMRP-positive cells from the

early postnatal period to adulthood

FMRP+ cell counts in the cingulate cortex and corpus callosum revealed that the number of

FMRP+ cells in both brain regions was highest at PND 0 (see “Total FMRP+” column in Table

6). In the cingulate cortex there was a gradual decline over time such that the adult level (8-12

weeks old) was approximately 57% of that at birth (P0). In the corpus callosum there was a

relatively minor decline whereby adult mice showed about 85% of the level at P0. Overall,

Page 50: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

39

these observations are consistent with previous reports indicating peak FMRP expression over

the first 1-2 postnatal weeks and declining thereafter (Lu et al., 2004).

Table 6. Quantitative analysis of cell type-specific expression of FMRP in the

cingulate cortex and corpus callosum. Brain region Age Total FMRP+ % NeuN+ % S100β % Iba-1 % NG2

Cingulate

cortex

PND 0 154 ± 5 85 ± 2 5 4 6

PND 10 123 ± 5 89 ± 1 2 5 3

PND 20 86 ± 5 94 ± 1 2 2 4

Adult 88 ± 7 94 ± 1 3 2 5 ± 1

Corpus

callosum

PND 0 82 ± 3 2 27 ± 2 20 ± 2 18 ± 2

PND 10 75 ± 3 2 33 ± 3 19 ± 3 14 ± 2

PND 20 76 ± 3 2 33 ± 22 10 ± 1 11 ± 2

Adult 70 ± 3 2 22 ± 3 3 7

The analysis was performed by counting the FMRP-positive neurons (NeuN), astrocytes

(S100β), microglia (Iba-1) and oligodendrocyte precursor cells (NG2) in the cingulate cortex

and corpus callosum. The results are reported as the average number of total FMRP-positive

cells per visual field, and the percentage of FMRP-positive cells that co-localized with NeuN,

S100β, Iba-1, and NG2. Data are presented as mean ± S.E.M. Standard error values smaller

than 0.5 are not reported.

3.3. Predominant neuronal expression of FMRP throughout

development

At all postnatal times examined, FMRP expression was predominantly neuronal in all of the

brain regions analyzed, except for corpus callosum (Fig. 6, Tables 6 and 7) where only a few

NeuN+ cells were present and most FMRP expression was observed in S100β+ cells (Fig. 7,

Table 6). Quantitative analysis of cell type-specific expression of FMRP revealed 85% - 94%

co-expression with NeuN in FMRP+ cells in the cingulate cortex. In stark contrast only 2%

NeuN/FMRP co-expression was detected in the corpus callosum at all of the time points studied.

Page 51: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

40

Table 7. Semi-quantitative analysis of cell type-specific FMRP expression in

the developing and adult mouse brain. Brain

region

Age of

mice

FMRP+/NeuN

+ FMRP+/S100β+

FMRP+

/Iba-1+ FMRP+/NG2+

Cingulate

cortex

PND 0 +++ + + +

PND 10 +++ + + +

PND 20 +++ + + +

Adult +++ + + +

Corpus

callosum

PND 0 + +++ +++ +++

PND 10 + +++ +++ ++

PND 20 + +++ ++ ++

Adult + +++ + +

Striatum

PND 0 +++ ++ ++ ++

PND 10 +++ + + +

PND 20 +++ + + +

Adult +++ + - +

Hippocamp

us

PND 0 +++ ++ ++ +

PND 10 +++ + + +

PND 20 +++ - - +

Adult +++ - - -

Cerebellum

PND 0 +++ + + +

PND 10 +++ + + ++

PND 20 +++ + - +

Adult +++ + - +

The level of co-localization of FMRP with NeuN-positive neurons, S100β-positive

astrocytes, Iba-1-positive microglia, and NG2-positive oligodendrocyte precursor cells in mouse

brain was assessed in the brain regions indicated at PND 0, PND 10, PND 20, and adults. The

extent of co-localization is reported as: - , none; + low; ++ moderate; and +++ high.

Page 52: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

41

Fig. 6. Expression of FMRP in neurons of the WT mouse brain.

Coronal sections from postnatal days 0, 10, 20, and adult mice were double immunolabeled

using anti-FMRP and anti-NeuN, and counterstained with DAPI (A–T). Co-localization of

FMRP in NeuN-positive neurons (FMRP, green; NeuN, red; DAPI, blue) is shown in the

cingulate cortex, hippocampus, striatum, corpus callosum, and cerebellum. Arrows indicate

examples of co-localization. The heavy dashed lines delineate the boundaries of the corpus

callosum. Scale bar = 20 μm.

3.4. Developmental expression of FMRP in astrocytes, microglia and

oligodendrocyte precursor cells

We examined FMRP expression in astrocytes by double labeling with anti-FMRP and anti-

S100β antibodies. FMRP and S100β were co-localized at PND 0 in cells in the cingulate cortex,

striatum, hippocampus, corpus callosum and cerebellum (Fig. 7, Tables 6 and 7). By PND 10,

PN

D 1

0Cingulate cortex Hippocampus Striatum Corpus callosum Cerebellum

NeuN FMRP DAPIP

ND

20

PN

D 0

Ad

ult

DA CB

N

HG

ML

SP RQ

O

J

E

K

IF

T20 μm

Page 53: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

42

PND 20 and adult mouse brain, the population of FMRP/S100β -positive cells had declined

drastically in cingulate cortex and hippocampus, (less than 3%). Compared to the cingulate

cortex and hippocampus, the decline in FMRP/S100 β co-expression occurred more slowly in

the striatum where substantial co-expression was seen at PND 10, PND 20, but not in adult

mice. Persistent co-expression of FMRP/S100β was also observed in the cerebellum at PND0,

10, 20 and adult mice, although the number of FMRP/S100β cell gradually decreased in adult

mouse brain.

Interestingly, FMRP was co-expressed in many S100β+ cells in the corpus callosum. Of the

brain regions analyzed, the corpus callosum showed by far the largest percentage of cells co-

expressing of FMRP and S100β (Fig. 7, Table 7); more than 75% of S100β positive cells in the

corpus callosum co-expressed FMRP at all time points. This is an intriguing finding as this

brain region is mainly composed of white matter. Approximately 30% of FMRP-expressing

cells in the corpus callosum also expressed S100β at all developmental stages analyzed (Tables

6 and 7). The percent of S100β/FMRP-positive cells was maintained at a moderate level from

PND 0 to adult mice, indicating a continual expression of FMRP in astrocytes from early

postnatal stages through adulthood in this brain region.

In addition to the expression of S100β in astrocytes, S100β is also expressed by a population

of oligodendrocyte precursor cells and oligodendrocytes (Hachem et al., 2005; Steiner et al.,

2008). Therefore, two population of S100β immune-positive cells may exist, one representing

astrocytes and one dedicated to oligodendrocyte precursor cells. To better characterize these

two populations of cells, we also used NG2 staining as a specific marker for oligodendrocyte

precursor cells. The NG2 proteoglycan is expressed in oligodendrocyte precursor cells, but not

in mature oligodendrocytes, both in vitro and in developing rodent brain (Polito and Reynolds,

2005). FMRP was expressed in many NG2+ oligodendrocyte precursor cells in the cingulate

Page 54: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

43

cortex, striatum, hippocampus, corpus callosum and cerebellum at PND 0 (Fig. 8, Tables 6 and

7). Thereafter, from PND 10 to adult mouse brain, FMRP expression in NG2-positive cells

progressively declined in all of the brain regions examined, reaching low co-localization in adult

mice in the cingulate cortex, corpus callosum, striatum and cerebellum and undetectable co-

localization in the hippocampus (Table 7).

FMRP expression in microglia was examined by assessing co-expression with Iba-1. FMRP/

Iba-1 double labeled cells were present in all brain regions analyzed at PND 0 (Fig. 9). At PND

0, more than 75% of the total NG2+ cells and Iba-1+ cells co-expressed FMRP in the all the

brain regions analyzed. At PND 10 and PND 20, the number of FMRP/Iba-1 co-localized cells

remained relatively constant in the cingulate cortex, corpus callosum, and striatum, but

decreased in the hippocampus and cerebellum by PND 20. In the brain regions studied here,

there was a gradual decrease in FMRP/Iba-1 co-expression after PND 20 with no detectable co-

localization in adult mice.

Page 55: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

44

Fig. 7. Expression of FMRP in the astrocytes of mouse brain.

Coronal sections from postnatal days 0, 10, 20, and adult mice were labeled using anti-

FMRP, anti-S100β and DAPI. FMRP in S100β -positive astrocytes (FMRP, green; S100β, red;

DAPI, blue) are shown in the cingulate cortex, hippocampus, striatum, corpus callosum, and

cerebellum. Arrows indicate examples of co-localization of FMRP in S100β-positive

astrocytes, whereas dashed-squares indicate the absence of FMRP in astrocytes. The heavy

dashed lines delineate the boundaries of the corpus callosum. Scale bar = 20 μm.

Cingulate cortex Hippocampus Striatum Corpus callosum Cerebellum

S100B FMRP DAPI

PN

D 1

0P

ND

20

PN

D 0

Ad

ult

IF HG

K ML

P RQ

A B C

O

T

J

N

S

D E

20 μm

Page 56: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

45

Fig. 8. Expression of FMRP in oligodendrocyte precursor cells in the mouse

brain.

Coronal sections were labeled using anti-FMRP, anti-NG2 and DAPI. Co-localization of

endogenous FMRP in NG2-positive oligodendrocyte precursor cells (FMRP, green; NG2, red;

DAPI, blue) is shown in the cingulate cortex, hippocampus, striatum, corpus callosum, and

cerebellum. Arrows indicate the co-localization of FMRP in NG2-positive oligodendrocyte

precursor cells, whereas dashed-squares indicate the absence of FMRP in oligodendrocyte

precursor cells. The heavy dashed lines delineate the boundaries of the corpus callosum. Scale

bar = 20 μm.

Cingulate cortex Hippocampus Striatum Corpus callosum Cerebellum

NG2 FMRP DAPIP

ND

10

PN

D 2

0P

ND

0A

du

lt

IF HG

NK OM

SP TR

DBA C

J

E

L

Q20 μm

Page 57: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

46

Fig. 9. Expression of FMRP in microglia.

Coronal sections were labeled with anti-FMRP and anti-Iba-1. Co-localization of FMRP in

Iba-1-positive microglia (FMRP, green; Iba1, red; DAPI, blue) is shown. Arrows indicate

examples of co-localization of FMRP in Iba-1-positive microglia, whereas dashed-squares

indicate the absence of FMRP in microglia. Scale bar = 20 μm.

Cingulate cortex Hippocampus Striatum Corpus callosum Cerebellum

Iba1 FMRP DAPIP

ND

10

PN

D 2

0P

ND

0A

du

lt

A CB

IF HG

NK ML

SP RQ T

O

J

ED

20 μm

Page 58: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

47

Chapter 4. Transduction efficiency of adeno-associated viral vectors expressing eGFP after intra-cerebroventricular

administration in neonatal and juvenile mice

Acknowledgements: Immunocytotochemical analyses and imaging were performed with some

assistance from Dr. Sujeenthar Tharmalingam (Ph.D. student) and Margarita Macaldaz

(summer student).

4.1. Specific hypotheses, objectives and rationale

We hypothesized that intra-cerebroventricular (i.c.v.) administration of single stranded AAV

vectors leads to robust widespread gene delivery to multiple regions of the brain and the

transduction efficiency and cellular tropism is affected by the AAV serotype used, time of

administration and the promoter selected in the AAV construct. The transport of viral vectors

directly in the CSF as well as transducing the ependymal cells lining the ventricles has made

i.c.v. delivery an established route of AAV administration, particularly where a global transgene

delivery in the brain is required. The main objective of this study was to evaluate the ability of

single-stranded AAV 2/9 (AAV9) and 2/5 (AAV5) encoding the enhanced green fluorescent

protein (eGFP) reporter to transduce the brain and target gene expression to specific cell types

following i.c.v. injection in mice. Major factors affecting tropism, expression level, and cell-

type specificity of AAV-mediated transgenes include encapsidation of different AAV serotypes,

promoter selection, and the timing of vector administration. Titer-matched AAV9 vectors

encoding eGFP, driven by the cytomegalovirus (CMV) promoter, or the neuron-specific

synapsin-1 promoter, were injected bilaterally into the lateral ventricles of C57/BL6 mice at

postnatal day 5 (neonatal) or 21 (juvenile). Brain sections were analyzed 25 days after injection

using immunocytochemistry and confocal microscopy.

Page 59: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

48

4.2. AAV9-CMV-eGFP targets astrocytes in neonatal mice but neurons

in juvenile mice

To evaluate virus transduction of the brain at early stages prior to CNS maturation, we

investigated transgene expression after i.c.v. injections in neonatal mice on PND 5, or juvenile

mice on PND 21. One group of mice received i.c.v. injections of 2 × 1010

particles of a single

stranded AAV9-CMV-eGFP (Fig. 10A). The animals were euthanized 25 days post-injection,

and brains were evaluated for transgene expression (Fig. 10B).

Fig. 10. Schematic diagrams depicting (A) the AAV-eGFP vector constructs

used and (B) bilateral intra-cerebroventricular injections on postnatal days 5,

and 21.

The vectors were single stranded, contained ITR elements from AAV serotype 2, and were

packaged in either serotype 5 or serotype 9 capsids. A WPRE element was inserted into the

AAV9 vectors downstream of the eGFP cDNA to enhance transcription. Twenty-five days after

AAV administrations (i.e., on postnatal day 30 or 46), animals were sacrificed and brains were

Page 60: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

49

analyzed for GFP immunoreactivity. CBA, chicken β-actin; CMV, cytomegalovirus; ITR,

inverted terminal repeat; WPRE, woodchuck hepatitis post-transcriptional regulatory element;

eGFP, enhanced green fluorescent protein; hSynapsin-1, human synapsin-1.

We observed remarkable differences between PND 5 and PND 21 injections both in the

distribution and cell type specificity of transduction. Injection of AAV-CMV-eGFP at PND 5

resulted in an extensive distribution in all brain regions examined: prefrontal cortex, striatum,

hippocampus, piriform cortex, cingulate cortex, retrosplenial cortex and cerebellum. The

transduction was primarily observed in astrocytes in several of the forebrain regions examined

including the striatum (Fig. 11), retrosplenial cortex (Fig. 12), and prefrontal, piriform, and

cingulate cortices (results not shown), as assessed by double labeling with anti-GFP and anti-

S100β antibodies. In contrast, in the hippocampus transduction was mainly neuronal, as

assessed by double labeling with anti-GFP and anti-NeuN antibodies (Fig. 13, A-F). In the

cerebellum, transgene expression was predominantly present in the dendrites and cell bodies of

Purkinje neurons as determined by double labeling of the sections with anti-GFP and anti-

calbindin antibodies (Fig. 14, A-F).

In contrast, i.c.v. injections of titer-matched AAV9-CMV-eGFP vector on PND 21 resulted

in almost exclusively neuronal transduction but with a more restricted distribution compared to

neonatal injections on PND 5. Robust eGFP expression was found in many regions in the

forebrain including the prefrontal cortex, striatum, and hippocampus (Table 8). Co-labeling for

NeuN and GFP expression in the striatum (Fig. 11, D-F), retrosplenial cortex (Fig. 12, D-F),

hippocampus (Fig. 13, D-F) and cingulate cortex (not shown) revealed many NeuN-positive

cells expressing GFP throughout all examined sections, indicating widespread neuronal

transduction. Unlike PND 5 injections, which resulted in transduction of the cerebellum, PND

21 administration of AAV9-CMV-eGFP produced virtually no GFP-positive cells in the

Page 61: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

50

cerebellum and showed very limited transduction in the piriform cortex (Table 8), suggesting a

lower vector diffusion after injection at PND 21 compared to PND 5.

In summary, the pattern of eGFP expression observed after i.c.v. administration of AAV9-

CMV-eGFP on PND 5 showed a preferential astrocytic transduction with an extensive

distribution in forebrain regions and the cerebellum, while injections on PND 21 gave a nearly

exclusive neuronal transgene expression and a more restricted distribution in forebrain regions

and no expression in the cerebellum.

4.3. Use of the synapsin-1 promoter to achieve neuron-specific

transduction

We next assessed CNS expression after i.c.v. injection of single stranded AAV9-SYN-eGFP

on PND 5 and PND 21 to compare the cell type specificity and level of transgene expression

using this vector with the same construct driven by the CMV promoter. The use of the

synapsin-1 promoter resulted in extensive neuron-specific transduction regardless of the age of

injection. Intra-cerebroventricular injections of AAV9-SYN-eGFP on PND 5 or PND 21 both

resulted in robust neuron-specific eGFP expression in the prefrontal and cingulate cortex,

striatum (Fig. 11, G-L), retrosplenial cortex (Fig. 12, G-L), hippocampus (Fig. 13, G-L).

However, transgene expression in the piriform cortex and cerebellum (Fig. 14, G-L) was only

observed in mice injected on PND 5. The fact that there was no transgene expression in the

cerebellum after i.c.v. delivery of vectors on PND 21, suggests a wider dispersion of AAV9

after i.c.v. delivery at PND 5. In the cerebellum, unlike the AAV9-CMV-eGFP which resulted

in transgene expression only in the Purkinje cells (Fig. 14, A-F), transduction of AAV9-SYN-

eGFP extended to neurons in the granule cell layer as well (Fig. 14, G-L).

Page 62: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

51

Fig. 11. AAV-eGFP transgene expression in the mouse striatum.

eGFP transgene expression in the mouse striatum. AAV9 vectors were injected on PND 5

(A-C, G-I) or on PND 21 (D-F, J-L). Different patterns of cell-specific gene expression were

observed; AAV9-CMV-eGFP predominantly transduced protoplasmic astrocytes in PND 5

injections, as assessed by double labeling with anti-GFP with the protoplasmic astrocyte-

specific anti-S100β antibody (A-C). In contrast, AAV9-CMV-eGFP vectors injected on PND

21 and AAV9-SYN-eGFP injected at either PND 5 or PND 21 resulted in primarily neuronal

transduction (D-L), as confirmed by co-staining with NeuN. Insets: cells of interest at higher

magnification. Scale bars = 50 µm.

Page 63: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

52

Fig. 12. AAV-eGFP transgene expression in the retrosplenial cortex.

In mice injected with AAV9-CMV-eGFP on PND 5, the majority of eGFP positive cells in

the retrosplenial cortex were confirmed to be protoplasmic astrocytes by double labeling with

anti-S100β (A-C). In contrast, AAV9-CMV-eGFP injected on PND 21 and AAV9-SYN-eGFP

injected at either PND 5 or PND 21 resulted in transduction of almost exclusively neurons (D-

L), as confirmed by co-staining with anti-NeuN. Insets: cells of interest at higher magnification.

Scale bars = 50 µm.

Page 64: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

53

Fig. 13. AAV-eGFP transgene expression in hippocampus.

Transgene expression in the CA1 region of the hippocampus after administration of AAV9-

CMV-eGFP (A-F) or AAV9-SYN-eGFP (G-L) vectors on PND 5 or PND 21. Brain sections

from the hippocampus of AAV-injected mice were double labeled using anti-GFP and anti-

NeuN. Insets: cells of interest shown at higher magnification. Scale bars = 50 µm.

Page 65: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

54

4.4. Low transduction efficiency of AAV5-GFP vectors

We also sought to compare AAV9 vectors with an AAV5 vector. The AAV5-eGFP vector

contained the chicken beta actin (CBA) promoter. The CBA promoter has previously been used

in AAV vectors to transduce neurons (Gray et al., 2010; Gray et al., 2011). We observed

however that unlike the AAV9 vectors, i.c.v. injections of titer-matched AAV5-CBA-eGFP

vector on PND 5 resulted in transduction of very few cells in the striatum, most of which were

located close to the lateral ventricles (data not shown). No GFP immunoreactivity was detected

in other brain regions in mice injected at PND 5 with AAV5-CBA-eGFP. Administration of

AAV5-GFP at PND 21 resulted in transduction of a few cells in the hippocampus, striatum,

prefrontal cortex, cingulate cortex and retrosplenial cortex, but no transduction in the piriform

cortex and cerebellum (Table 8). Taken together, these results indicate (a) higher transduction

efficiency was obtained with AAV5-CBA-eGFP injected on PND 21 compared to PND 5, and

(b) at both PND 5 and 21, the two AAV9 vectors showed higher diffusion in the CNS compared

to the AAV5-based vector.

Page 66: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

55

Fig. 14. AAV-eGFP transgene expression in the cerebellum.

eGFP transgene expression in the cerebellum after injection of mice with AAV9-CMV-eGFP

or AAV9-SYN-eGFP on PND 5 (A-L). Co-staining with ant-calbindin was used to confirm the

presence of GFP positive cells in Purkinje neurons. M: molecular layer; G: granular layer; P:

Purkinje cell layer. Insets: selected Purkinje neurons shown at higher magnification. Scale bars

= 200 µm (A-C, G-I), 50 µm (D-F, J-L).

Page 67: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

56

Table 8. Analysis of AAV-eGFP vector transduction patterns in discrete

areas of the brain after i.c.v. delivery in PND 5 or PND 21 mice. PND 5 PND 21

AAV5-

CBA-eGFP

AAV9-

CMV-

eGFP

AAV9-

SYN-

eGFP

AAV5-

CBA-

eGFP

AAV9-

CMV-

eGFP

AAV9-

SYN-

eGFP

N 3 5 3 2 3 3

Prefrontal cortex _ +++ +++ + ++ ++

Striatum + ++++ ++++ ++ ++++ ++++

Cingulate cortex + ++ ++++ + ++ ++++

Hippocampus _ ++++ ++++ + ++++ ++++

Retrosplenial

cortex

_ ++ ++++ + ++ ++++

Piriform cortex _ +++ +++ _ + +

Cerebellum _ ++ +++ _ _ _

AAV vector Age of

administration

GFP+ / mm2

(mean ± s.e.m.)

%

GFP+/NeuN+

(mean± s.e.m.)

%GFP+/S100β

+ (mean±

s.e.m.)

AAV9-CMV-eGFP PND 5 261 ± 91 ND 89 ± 1

AAV9-SYN-eGFP PND 5 1027 ± 91 95 ± 1 ND

AAV9-CMV-eGFP PND 21 195 ± 23 81 ± 6 ND

AAV9-SYN-eGFP PND 21 856 ± 85 92 ± 2 ND

A. Semi-quantitative analysis of vector transduction patterns in discrete areas of the brain.

The levels of transduction were graded as: (-) no transduction, (+) a few transduced cells, (++) a

moderate number of transduced cells, (+++) many transduced cells and (++++) a region that was

saturated with transduced cells. B. Quantitative analysis of eGFP transduction in the

retrosplenial cortex. The total number of eGFP-positive cells in both hemispheres was counted

in three sections from each brain and reported as average number of cells per square millimetre.

Percentages of neuronal and astrocytic transduction were calculated based on the number of

eGFP-positive cells co-localized with antibodies to NeuN and S100β.

Predominant neuronal transduction Predominant astrocytic transduction

B.

A.

Page 68: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

57

Chapter 5. FXS gene therapy-Phase 1: Reduced phenotypic severity following adeno-associated virus mediated Fmr1 gene

delivery in postnatal day 5 Fmr1 mice

Acknowledgements: Vector injections, Western blotting and behavioral experiments were

performed with assistance from Dr. Jason Arsenault (Post-doctoral fellow), Ingrid Xuan

(Graduate student) and Laura Pacey (Research Associate).

5.1. Specific hypotheses, objectives and rationale

There is no pharmacological cure for FXS and the currently prescribed medications, such as

anti-psychotics, anti-depressants, and stimulants, only partially alleviate selected symptoms and

are associated with deleterious side effects. Newer second generation drugs to treat FXS,

including metabotropic glutamate receptor 5 (mGluR5) antagonists, are being investigated in

clinical trials (Castren et al., 2012; Hampson et al., 2012; Pop et al., 2014). However,

considering the plethora of genes whose expression is regulated by FMRP, a priori, it may be

expected that restoring FMRP expression in the CNS could provide a more comprehensive

reversal of the disorder compared to targeting single molecules (e.g. mGluR5).

FMRP is widely distributed throughout most regions of the CNS; therefore a major goal of

this study was to identify conditions for AAV-FMRP administration that would provide

widespread transgene dispersion in the brain. We also sought to attain a level of expression that

was as close to WT levels as possible to achieve the most comprehensive behavioral rescue after

AAV delivery. All analytical assessments were carried out during two periods post-injection:

the first began 22 days post-injection (on PND 27) and is hereafter referred to as the “short

arm”, and the second began 50 days post-injection (on PND 55) and is referred to as the “long

arm” (see Fig. 15b).

Page 69: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

58

Fig. 15. Fmr1 gene delivery (Phase 1): Overview of viral vector construction,

experimental plan for injections, and behavioral analyses.

(a) Schematic depiction of the AAV-FMRP vector construct. The single-stranded AAV

viral vector contained the human synapsin-1 promoter and inverted terminal repeat (ITR)

elements from AAV serotype 2 and packaged in serotype 9 capsids. A woodchuck hepatitis

post-transcriptional regulatory element (WPRE) was inserted downstream of the mouse Fmr1

cDNA to induce elevation of transcripts. (b) Timelines for the short and long arms of the study

for mice injected with PBS or viral vector on postnatal day 5. The total number of mice (N)

injected for each treatment group is indicated.

Page 70: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

59

5.2. Quantification of FMRP expression levels following neonatal i.c.v.

injection of AAV-FMRP in postnatal day 5 mice

To quantify total transgene expression, samples of the cerebellum, inferior colliculus,

cerebral cortex, striatum and hippocampus were subjected to quantitative western blotting. In

samples from WT mice, three bands corresponding to FMRP isoforms were detected (Fig. 16).

In Fmr1 KO mice injected with AAV-FMRP, as expected, only isoform 1 was observed (Fig.

16). Fmr1 KO mice injected with AAV-FMRP and sacrificed at PND 31 (Fig. 16a and c),

revealed 52 ± 9% of WT FMRP expression in the hippocampus, 41±13% in the striatum, and 71

± 20% in the cerebral cortex (Fig. 16c). At PND 60 (Fig. 16b and c), the AAV-FMRP-injected

Fmr1 mice displayed a mean 47 ±15% of WT expression in the cerebral cortex, 48 ± 20% of

WT expression in the hippocampus and 18 ±5% of WT expression in the striatum (Fig. 16c).

The results shown in the western blots in Fig. 16a, b, and d, and in the error bars shown in the

summary graph in Fig. 16c clearly reveal variable expression of the transgene from mouse-to-

mouse in the brain regions examined. The variable range of FMRP transgene expression levels

in forebrain regions from mouse-to-mouse was likely the result of the ability of the vector to

diffuse from the site of the injection in the lateral ventricles. FMRP was not detected in brain

regions more distal from the lateral ventricles such as the inferior colliculus and the cerebellum;

these brain regions are likely located too far from the ventricles to acquire sufficient vector

uptake for FMRP transgene expression and detection. Importantly, transgene expression levels

in the brain regions where it was detected remained relatively constant for at least 7 months

post-injection, showing 77 ± 28% of WT expression in the cortex, 40 ±14% in the hippocampus

and 14 ± 3% in the striatum (Fig. 16c and d).

Page 71: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

60

Fig. 16. Western blots of FMRP expression in control and AAV-FMRP-

treated mice.

(a) Representative western blots of the cerebral cortex, hippocampus, and striatum from

individual mice injected with AAV-FMRP in the short arm where brain samples were collected

at PND 31. (b) FMRP transgene expression in the long arms of the study at PND 60. (c)

Quantification of samples from the short and long arms of the study as well as long term (7

months) expression. Expression in injected Fmr1 KO mice was normalized to the GAPDH

signal intensity and compared with age and sex-matched PBS-injected WT brain regions (100%

FMRP expression). The results are presented as the mean ± S.E.M. (d) Representative western

blots of Fmr1 KO mice 7 months after i.c.v. injection with AAV-FMRP.

5.3. Distribution and cellular selectivity of transgene expression

Immunocytochemical analysis revealed expression of the FMRP transgene in the striatum,

hippocampus, retrosplenial cortex and cingulate cortex at both the end of the short arm (26 days

post-injection) and the long arm (56-62 days post-injection) of the study (Figs. 17a and 18).

Page 72: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

61

Representative sagittal images of FMRP expression from PBS-injected WT and KO mice and

AAV-FMRP-injected KO mice are shown in Fig. 17b. No FMRP expression was detected in

regions more distant and caudal to the site of injection in the lateral ventricles including the

piriform cortex, cerebellum, inferior colliculus, and brainstem. The highest transduction

efficiencies were observed in the retrosplenial cortex and the cingulate cortex, while lower

FMRP expression was observed in the striatum (Figs. 17a, b and 18).

Double-labeling experiments showed that FMRP was predominantly observed in NeuN-

positive cells within the striatum, hippocampus, retrosplenial cortex and cingulate cortex,

suggesting strong preferential expression in neuronal populations (Figs. 17c, 18, and 19).

Quantitative analysis of FMRP positive cells co-expressing NeuN in the cingulate cortex

revealed over 90% neuronal transduction (Fig. 19). In contrast, we did not detect transduced

cells that were immune-positive for the glial selective marker S100β in any of the brain regions

examined (Fig. 17c), indicating no or undetectable levels of FMRP transduction in astrocytes.

The latter observation is consistent with the use of the neuron-selective synapsin promoter used

to drive transgene expression. Analysis of the cellular localization of FMRP in AAV-FMRP

transduced cells revealed predominant expression in the cytoplasm of neurons (Figs. 17c, d and

18). This cytoplasmic localization of the FMRP transgene was observed in all brain regions

where transgene expression was detected and was similar to that observed previously in neurons

of WT mice (Pacey et al., 2013; Sidorov et al., 2013).

Page 73: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

62

Fig. 17. Immunocytochemical analysis of AAV-FMRP transduction in

discrete brain regions

(a) Diagrammatic representation of FMRP transgene expression in Fmr1 KO mice treated

with AAV-FMRP. The levels of transduction were graded as indicated. (b) Representative low

magnification (top row) and higher magnification (bottom row) sagittal images showing FMRP

expression in PBS-injected WT, AAV-FMRP-injected Fmr1 KO mouse, and PBS-injected Fmr1

KO mouse brain. (c) Photomicrographs depicting neuronal specificity of the AAV-FMRP

Page 74: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

63

transduction in the cingulate cortex of mice collected at PND 61. Brain sections were double

labeled using anti-FMRP antibody and either the neuronal marker NeuN or the glial specific

marker S100β. Scale bars = 20 µm (top row), 50 µm (bottom row). (d) Higher magnification

photomicrographs illustrating similar cytosolic distributions of FMRP in the cortex of WT and

AAV-FMRP-injected KO mice. Scale bars = 20 µm in all panels.

5.4. Phase 1 - Behavioral analyses

Behavioral experiments were carried out to determine if AAV-induced restoration of FMRP

in the brains of Fmr1 mice could rescue or ameliorate pathological behaviors typically seen in

Fmr1 KO mice (and observed here in PBS-injected Fmr1 KO mice). In the behavioral tests, if a

mouse displayed an obvious abnormality that could influence the test, such as weight loss

following malocclusion, it was excluded from the analysis. Overall, this happened infrequently

and did not appear to be restricted to any particular treatment group. Additional contributions to

the variable number of mice analyzed in each test include the assessment of only male mice in

the ultrasonic vocalization analysis, and in tube test, the N values represent the total number of

pairings; this depended on the availability of age-matched groups at the time of testing.

In the short arm of the study, the tests included measurement of locomotor activity, marble

burying (as a measure of repetitive behavior), and audiogenic seizure susceptibility. Separate

groups of mice were examined in the long arm of the study (i.e. no mice studied in the short arm

were again studied in the long arm). In the long arm, motor activity and marble burying were

measured along with two additional tests: ultrasonic vocalizations to examine mouse-to-mouse

communication during courtship behavior, and the tube test which measures social dominance.

In both the short and long arms, preliminary statistical analyses were conducted using two-way

ANOVA on males and females (i.e. gender and treatment as the main effects). These results

indicated no significant interaction effect between males vs. females; therefore further statistical

analyses were only carried out on pooled data from males and females combined.

Page 75: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

64

In the motor activity test, a significant increase in the total distance travelled was seen in both

PBS-injected and AAV-FMRP injected Fmr1 KO mice compared with age and gender-matched

PBS-injected WT mice in the shorts and long arms of the study (Figs. 21a and 20). Thus, AAV-

FMRP vector administration did not rescue the motor hyperactivity in Fmr1 KO mice.

In the marble burying test, used as a measure of repetitive behavior, a significant increase

was observed in the number of marbles buried by PBS-injected Fmr1 KO mice compared to

PBS-injected WT mice (Fig. 21b; F = 29.79, p < 0.001), similar to previous observations made

in some strains of the Fmr1 KO mouse (Spencer et al., 2011). There was a significant decrease

in the number of buried marbles in Fmr1 mice injected with AAV-FMRP compared to PBS-

injected Fmr1 KO mice (Fig. 21b), indicating that the introduction of FMRP rescued

stereotypical behavior. In the short arm of the study this trend was also observed but did not

reach statistical significance due to the lower number of marbles buried by mice in all groups at

PND 32 (Fig. 20; F = 4.01, p > 0.05). The variable level of FMRP transgene expression in

AAV–FMRP-injected animals provided us an opportunity to examine potential correlations

between FMRP expression and behavioral rescue. A preliminary analysis based on a small set

of data points (n = 10), suggested a possible inverse correlation between the number of marbles

buried and cortical levels of FMRP protein expression (Fig. 22; r2

= 0.37, p = 0.06). To

summarize, the trend towards reversal of elevated repetitive behavior seen in the short arm was

more robust and statistically significant in the long arm of the study.

In summary, FMRP transgene expression in the phase 1 study resulted in full reversal of the

elevated repetitive behavior and the deficit in social dominance behavior seen in

PBS-injected Fmr1 KO mice. In other behavioral tests such as audiogenic seizure, motor

activity and ultrasonic vocalizations, there was no statistically significant difference between the

Fmr1 KO mice treated with PBS or AAV-FMRP.

Page 76: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

65

Fig. 18. FMRP transgene expression in the striatum, hippocampus, cingulate

cortex, and retrosplenial cortex 61 days after i.c.v. administration of AAV-

FMRP on PND 5.

FMRP transgene expression in the striatum, hippocampus, cingulate cortex, and retrosplenial

cortex 61 days after i.c.v. administration of AAV-FMRP on PND 5. Brain sections were double

labeled using anti-FMRP (green) and anti-NeuN (red). Scale bars = 50 µm.

Page 77: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

66

Fig. 19. Quantitative analysis of AAV-FMRP transduction in cingulate

cortex.

The cells were counted in 3 consecutive coronal 25 μm thick sections (100 μm apart) of the

cingulate cortex from each brain (total of 9 sections from 3 mice) and reported as the average

number of cells per square millimeter ± S.E.M.

For the analysis of audiogenic seizure susceptibility, as expected from previous observations

in our laboratory WT C57/BL6J mice were not susceptible to audiogenic seizures (Pacey et al.,

2009). In PBS-injected Fmr1 KO mice, 36% of the females and 31% of the males had seizures.

In the combined male plus female data, the incidence of audiogenic seizures in both PBS-

injected Fmr1 KO mice and AAV-FMRP-injected mice was significantly elevated compared to

PBS-injected WT mice (Fig. 20c; p < 0.05). However, the AAV-FMRP group was not different

than the PBS-injected Fmr1 group.

Animal # FMRP+ cells/mm2

NeuN+/FMRP+

cells/mm2

% of neuronal

transduction

1 1714 ± 470 1588 ± 403 94 ± 2

2 2154 ± 734 2097 ± 731 96 ± 2

3 2042 ± 666 2003 ± 656 98 ± 1

Page 78: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

67

Fig. 20. Summary of behavioral analyses of the short arm of the study.

Summary of the behavioral analyses in the short arm of the phase 1 of the study. PBS-

injected Fmr1 KO mice exhibited (a) Elevated motor activity (b) Increased repetitive behavior

and (c) Higher susceptibility to audiogenic seizures compared to PBS-injected WT mice. There

was a trend towards rescue in the marble burying test, but not in motor activity or seizure

susceptibility. Each bar represents the average ± SEM. For panel c, Fisher’s exact test was

performed; each bar represents the percentage of mice that had seizures (* p < 0.05; *** p <

0.001).

Page 79: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

68

Fig. 21. Summary of the behavioral results from the long arm of the study.

Summary of the behavioral results from the long arm of the study at postnatal days 55 to 61.

PBS-injected Fmr1 KO mice exhibited hyperactivity, increased repetitive behavior, decreased

ultrasonic vocalizations, and a reduction in social dominant behavior compared to PBS-injected

WT mice. (a) Total horizontal activity over 20 min. (b) Rescue of stereotypical behavior as

seen by total number of marbles buried during 30 min. (c) Total number of ultrasonic

vocalization during a 4 min encounter in naive (left) and treated mice (right). (a-c) Each bar

represents the average ± SEM (*** p < 0.001). (d) Rescue of impaired social dominance in the

tube test in AAV-FMRP Fmr1 KO mice compared to PBS-injected Fmr1 KO mice. Columns

indicate total % of wins for each group. The tube test was administered to PBS-treated WT

paired with PBS-Fmr1 KO mice (top), PBS-treated Fmr1 KO mice paired with AAV-FMRP-

treated Fmr1 KO mice (middle), and WT PBS paired with AAV-FMRP mice (bottom). The

number of wins for each group was tallied and a chi-square analysis was used to determine

whether the scores were significantly different from the 50:50 win/loss outcome expected by

chance. n = the number of pairings. ** p < 0.01, *** p < 0.001.

Ultrasonic vocalizations were recorded from PND 59 male mice. In a preliminary

experiment, we tested adult naïve uninjected WT and Fmr1 KO and observed a significant

Page 80: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

69

decrease in the number of ultrasonic vocalized calls in Fmr1 KO mice compared to WT mice

(Fig. 21c left panel; F = 2.53, p < 0.01). This result confirmed a previous finding showing

reduced vocalizations in Fmr1 mice (Rotschafer et al., 2012). We then conducted a second

study comparing male PBS-injected WT mice with male PBS-injected and AAV-FMRP-

injected Fmr1 mice. Although the number of ultrasonic vocalizations was lower in the PBS-

Fmr1 KO group compared to AAV-FMRP-Fmr1 KOs and PBS-treated WT mice, there was no

statistically significant difference among the 3 groups (Fig. 21c, right panel; F = 0.32, p > 0.05).

Impaired social dominance is an established characteristic of Fmr1 KO mice (Spencer et al.,

2005; Pacey et al., 2011). The tube test measures social dominance and aggressive tendencies

without allowing mice to injure one another. Dominant versus submissive behaviors were

scored for two age and sex-matched mice during a brief pairing. For each trial the more

dominant mouse pushes the other out of the tube and is deemed the winner while the second

(subordinate) mouse is the loser. All combinations of WT-PBS, Fmr1-PBS and Fmr1-AAV-

FMRP were evaluated and tallied as the percent of wins by each group against their opponents.

As expected based on previous studies (Spencer et al., 2005; Pacey et al., 2011), Fmr1 KO mice

injected with PBS won significantly fewer matches than expected by chance against WT mice

injected with PBS (Fig. 21d; p < 0.001). Male and female Fmr1 animals injected with AAV-

FMRP won significantly more matches against gender-matched PBS-injected Fmr1 KO mice

(Fig. 21d, p < 0.01). Thus, the deficit in social dominance in Fmr1 KO mice was rescued in the

AAV-FMRP- treated mice.

Page 81: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

70

Fig. 22. Correlation between the number of marbles buried and FMRP

transgene expression levels in the cerebral cortex.

The location of each point on the graph depicts the level of FMRP transgene expression as

determined by western blotting from an individual mouse mapped to the marble burying

behavior of that mouse. Linear regression of the data points showed a noticeable, albeit non-

significant, correlation of r2

= 0.37, p = 0.06, F = 4.61, n = 10.

Page 82: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

71

Chapter 6. FXS gene therapy-Phase 2:

Fmr1 transgene delivery in postnatal day 0 Fmr1 mice

Acknowledgements: Vector injections, Western blotting and behavioral experiments were

performed with assistance from Dr. Jason Arsenault (Post-doctoral fellow).

6.1. Specific hypotheses, objectives and rationale

We hypothesized that i.c.v. delivery of viral vectors at PND 0-1 may facilitate AAV

distribution in the brain, and therefore provide a more comprehensive behavioral rescue,

compared to PND 5 injections, as conducted in phase 1 of this work. Our rationale for

delivering the AAV viral vectors at PND 0-1 is based on (a) our own observations from pilot

experiments (see below), and (b) a previous study by Kim, et al (2013) both indicating the most

widespread diffusion of AAV vectors after i.c.v. delivery at PND 0-1 and diminished spread of

the virus with age (PND 2-5), particularly in distant structures from the site of injection,

including the brainstem and cerebellum. These authors also demonstrated that the widespread

expression of virally-encoded transgene is apparent within one day post-injection, during early,

critical stages of neuronal development, and it is also stable for up to 12 months post-injection

(Kim et al., 2013b). Here, we re-introduced the FMRP protein at an earlier time point in order

to elevate the transduction levels as well as heighten the overall brain distribution of the

transgene, while avoiding potentially pathological over-expression. Since the ependymal cell

lining surrounding the ventricles of the brain is still immature during the first 24 hours after

birth, the AAV vectors can more easily transfer from the CSF to the brain parenchyma (Passini

and Wolfe, 2001; Kim et al., 2014; Yamazaki et al., 2014). Moreover, while taking advantage

of the innate mouse-to-mouse variability of transduction levels seen in previous studies

(Gholizadeh et al., 2014), we sought to investigate what range of FMRP transduction and

expression levels would be most adequate for long-term behavioral correction in Fmr1 KO

mice.

Page 83: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

72

Another objective in this project was to evaluate the potential effects of FMRP over-

expression in the brain on mouse behavior. Therefore, WT C57/BL6 mice were also injected

with a range of AAV-FMRP vector particles (0.75 to 1 µl of 1010

genome/µl) to investigate the

effects of over-expression. These variable levels of transgene expression could give us better

insight into the proper range of FMRP needed in different regions of the brain to achieve a

proper rescue. The effects of FMRP over-expression in humans and mice are presently unclear.

To date, few patients with duplication of the entire FMR1 gene have been reported (Rio et al.,

2010; Nagamani et al., 2012; Vengoechea et al., 2012; Hickey et al., 2013) . FMRP duplication

in these case reports has been associated with severe developmental delay, fine motor and

speech delay, and progressive seizures, which support the notion that regulated Fmr1 gene

dosage is critical for normal neurocognitive function. To our knowledge, only one study

investigated the effects of FMRP over-expression on mouse behavior, suggesting that over-

expression may be pathological in some but not all of behavioral phenotypes associated with

Fmr1 mice (Peier et al., 2000). These conclusions were based on the finding that while KO

mice demonstrate hyperactivity and reduced anxiety, transgenic mice that over-express Fmr1,

demonstrate hypo-activity and elevated anxiety. However, these conclusions might have been

premature since the transgenic mouse used in these experiments expressed the human FMRP

protein at levels 10 times greater than average FMRP levels in healthy individuals. Although

the mouse and human FMRP share 97% homology, there are important differences in their

mRNA binding properties (Denman and Sung, 2002), and over-expression of the human FMRP

isoform at immensely high levels in the brains of transgenic mice, might have obvious

detrimental effects on their behavior.

Page 84: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

73

Table 9. Summary of the main differences in the experimental design between

Phase 1 and Phase 2 Main differences Phase 1 Phase 2

Age of mice receiving

viral injections

PND 5 PND 0-1

Control injections PBS AAV-null

Injection groups 1. Fmr1 KO : PBS

2. Fmr1 KO: AAV-FMRP

3. WT : PBS

1. Fmr1 KO: AAV-null

2. Fmr1 KO: AAV-FMRP

3. WT: AAV-null

4. WT: AAV-FMRP

Behavioral analysis Short arm (starting on PND 27) and

Long arm (starting on PND 55)

Only long arm (starting on PND 55)

The two phases of the gene therapy study differed in the age of injection, control injections,

and behavioral analysis.

Fig. 23. Fmr1 gene delivery (Phase 2): Overview of experimental plan for

injections, and behavioral analyses

Overview of tests following AAV injections; The behavioral tests started on PND 55, which

is 54-55 days after bilateral i.c.v. injections of viral vectors on PND 0-1. The behavioral tests

were performed at least 2 days apart and all animals were tested only once in each behavioral

paradigm.

6.2. Quantification of FMRP expression levels following neonatal i.c.v.

injection of AAV-FMRP in postnatal day 0 mice

A total of 206 neonatal WT and Fmr1 mice were included in the phase 2 study. All mice

were injected under blinded condition with either AAV-null or AAV-FMRP. Among the 206

injected mice, 27 displayed hydrocephalus and were immediately euthanized or, following post

mortem analysis, were seen to have had brain damage from the procedure and were excluded

from the study, on the assumption that their behavior would be adversely affected. Brain

damage was not restricted to any one treatment group or genotype, but was evident across all

Page 85: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

74

groups and both genotypes. The absence in elevated glial fibrillary acidic protein (GFAP)

staining following AAV administrations into the CNS suggested a lack of astrocyte activation

and neuroinflammation in response to AAV injections (Fig. 24).

Fig. 24. Analysis of GFAP following AAV-FMRP injections in WT animals.

GFAP was probed to investigate astrocyte activation. A-B) Western immunoblotting and

protein quantification of GFAP in 2-month-old naive WT (n = 4) or WT-AAV-FMRP (n = 5)

mice, shows no elevation in GFAP levels in the striatum (A) and hippocampus (B).

Western immunoblotting was used to determine the relative expression rates (compared to

WT-null) found in the mouse brain. Five brain regions were assessed for FMRP transgene

quantification. Fig. 25A shows a topographical view of the brain regions examined. The cortex

(C) comprises the retrosplenial area, the anterior cingulate area, the visual area, and the posterior

somatomotor area. The frontal cortex (FC) comprises the anterior somatomotor area, the pre-

limbic area, the orbital area and the inferior limbic area. The cerebellums, hippocampus and

striatum were also collected separately for analysis. Fig. 25B shows these same brain regions as

seen by a sagittal view. Fig. 25C shows exemplified western blotting of the cortex (top left),

Page 86: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

75

frontal cortex (top right) and hippocampus (bottom left) of Fmr1 KO mice injected with AAV-

FMRP (KO + FMRP). AAV-null injected WT (WT-null) was used to normalize protein

expression while AAV-null injected Fmr1 KO was used as a negative control to confirm

antibody specificity. The cortex of WT mice injected with AAV-FMRP (WT+FMRP) can also

be seen (bottom right). Fig. 25D shows the bar charts representing the average values of each of

these brain regions. We observed a 54 ± 7% expression rate in the cortex of Fmr1 KO mice

injected with AAV-FMRP, 115 ± 18% in the frontal cortex, 80 ± 15% in the hippocampus,

34±8% in the striatum and 2±1% in the cerebellum. The variable range of FMRP transgene

expression levels in forebrain regions was likely the result of mouse-to-mouse variation in the

ability of the vector to diffuse from the site of the injection in the lateral ventricles. FMRP was

detected at very low levels in the cerebellum, which likely reflects the longer distance from the

site of injection in the lateral ventricles compared to forebrain regions.

In the WT mice injected with AAV-FMRP, we observed an average expression level of 235

± 47% in the cortex, 182 ± 30% in the frontal cortex, 209 ± 45% in the hippocampus, 113 ±

24% in the striatum and 91 ± 4% in the cerebellum, compared to WT-null-injected mice. The

quantitative western immunoblotting results showed that in addition to achieving a wider

distribution of the FMRP transgene compared to that seen in phase 1, the overall levels of CNS

expression were higher in the mice analyzed in phase 2.

Page 87: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

76

Fig. 25. Western blots of FMRP expression in AAV-FMRP-treated KO and

WT mice.

AAV-FMRP transgene, and PSD-95 protein expression levels. A. Schematic representation

of the brain regions investigated (top view). The regions comprise the cortex (C; see text for

anatomical description), the frontal cortex (FC), the cerebellum (CB), the striatum (S) and the

hippocampus (H). B. Sagittal representation of the brain regions investigated. C.

Representative western blots of FMRP expression in the mouse brain. The FMRP transgene

expression level in Fmr1 KO mice injected with AAV-FMRP shown in the cortex (top left), the

frontal cortex (top right) and the hippocampus (lower left). The lower right panel shows the

FMRP expression levels in the cortex of WT mice injected with AAV-FMRP. D. Summary of

quantification of FMRP levels in different brain regions of Fmr1 KO mice (left) and WT

(middle) injected with AAV-FMRP. E. Quantification of PSD-95 in the cortex. All protein

levels were normalized to WT-null expression rates and corrected with Gapdh protein levels.

Results are shown as the average ± SEM. *p<0.05.

Page 88: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

77

6.2.1. Levels of FMRP substrates in AAV-FMRP-treated mice

Expression of the FMRP transgene, at or near WT levels in the KO mouse brain, is expected

to restore the natural functions of the WT protein, and normalize the translation of its substrate

mRNAs. To investigate this, we analyzed the protein expression levels of two mRNA substrates

of FMRP, namely PSD-95 and MeCP2 in the brains of AAV-FMRP-injected KO mice.

PSD-95 (post-synaptic density-95) is a synaptic adapter and transducer protein that is highly

expressed throughout the CNS, and also functions as a scaffold protein to link downstream

signaling components to mGluRs. PSD-95 mRNA is a well-established target for FMRP, and

previous studies have reported reduced PSD-95 protein levels in the Fmr1 KO mouse brain

(Zalfa et al., 2007). This down-regulation is at least partly explained by the well-established

role of FMRP in maintaining PSD-95 mRNA stability (Zalfa et al., 2007). PSD-95 protein in

the cerebral cortex of 2-month-old KO-null mice was reduced by 28±7% compared to that

observed in the WT-null group (Fig. 25E). In the KO+FMRP-treated group, PSD-95 levels

were restored to the level seen in the WT-null mice. A small elevation of PSD-95 was observed

in the WT+FMRP group compared to the WT-null group, indicating that FMRP over-expression

over native WT levels had relatively little impact on PSD-95.

MeCP2 is a transcription factor that is severely down-regulated due to gene mutation in Rett

syndrome, another monogenic autism spectrum disorder. MeCP2 was listed as a substrate for

FMRP in a previous study (Ascano et al., 2012). To confirm MeCP2 as an mRNA substrate for

FMRP, we initially examined cellular co-localization of FMRP and MeCP2. FMRP, which is

principally localized in the cytosol, and MeCP2, a nuclear protein, are co-expressed within the

same cells in the cortex (Fig. 26A), the striatum (Fig. 26B) and the hippocampus (Fig. 26C).

Despite the different compartmentalization of the two proteins, almost all MeCP2 positive cells

were also positive for FMRP. Subsequently, an mRNA pull down followed by quantitative RT-

Page 89: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

78

PCR was performed to determine if MeCP2 mRNA would be enriched in the anti-FMRP

immunoprecipitate. There was a modest enrichment of MeCP2 mRNA in whole forebrain

isolates from WT C57/BL6 mice, where PSD-95 mRNA served as a positive control (Fig. 26D).

A significant MeCP2 enrichment was also seen in FMRP immunoprecipitate in the human

embryonic kidney 293 cell line (Fig. 26E). These results indicate that both homo sapiens and

mus musculus MeCP2 mRNA interacts with FMRP.

To ascertain the functional relevance of this finding, we quantified MeCP2 protein

expression levels in the cortex (Fig. 26F). In contrast to reduced PSD-95 protein levels in Fmr1

KO mice, MeCP2 expression levels were elevated by 32 ± 8% in the cortex compared to WT-

null levels (*p<0.05). The introduction of FMRP significantly reduced the MeCP2 protein

levels compared to KO-null (Fig. 26F, **p<0.01). WT+FMRP group also had a significant

reduction in MeCP2 protein levels compared to KO-null (Fig. 26F, ***p<0.001). Since micro

gene duplications of MeCP2 was previously reported to cause severe intellectual disabilities in

humans (Fieremans et al., 2014), it is plausible to assume that high levels of MeCP2 in Fmr1

KO mice might be a contributing factor to at least some of the pathological mechanisms of this

disorder. Overall, these results showed that the AAV-FMRP was able to correct the up-

regulation in MeCP2 protein levels and partially ameliorate the down-regulation of PSD-95

levels seen in Fmr1 KO animals. The FMRP transgene thus recapitulated the native

translational modulation of endogenous FMRP.

Page 90: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

79

Fig. 26. Characterization of MeCP2 as an mRNA substrate for FMRP and

quantification of MeCP2 levels in AAV-treated WT and KO mice.

A-C: High magnification of WT coronal sections labeled with anti-FMRP 5c2 (green) and

anti-MeCP2 antibodies (red) of the A. cortex, B. Striatum and C. Hippocampus. D-E: FMRP

immunoprecipitation and mRNA quantification. D. Total mRNA enrichment of Gapdh, PSD-

95, and MeCP2 from whole mouse forebrain. E. Total mRNA enrichment of Gapdh and

MeCP2 from Hek 293 cell lysates. Results were normalized to Gapdh mRNA levels. Results

are shown ± S.E.M. *p<0.05. F. Quantification of MeCP2 levels in the cortex of WT-null,

WT+FMRP, KO-null, and KO+FMRP. MeCP2 protein levels were normalized to WT-null

expression rates and corrected for Gapdh protein levels. Results are shown as the average ±

SEM. *p<0.05, **p<0.01, *** p<0.001.

6.3. Distribution and cellular selectivity of transgene expression

Immunocytochemical analysis of AAV-injected KO mice sacrificed on PND 65 revealed

expression of the FMRP transgene in the striatum, hippocampus, retrosplenial cortex, cingulate

cortex, piriform cortex, prefrontal cortex, amygdala, thalamus, and at lower levels in dispersed

cells in superior colliculus, inferior colliculus and cerebellum (Fig. 27). A representative

sagittal image of FMRP expression from AAV-FMRP-injected KO mice is shown in Fig. 27A

emphasizing the brain regions with higher levels of FMRP transgene expression. The highest

Page 91: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

80

transduction efficiencies were observed in the hippocampus and most cortical regions. Unlike

phase 1, FMRP transgene expression was detected in the anterior forebrain such as the

orbitofrontal cortex and the anterior olfactory nucleus, which were not transduced in PND 5-

treated animals (Gholizadeh et al., 2014). Fig. 27B shows higher magnification coronal images

at different brain regions of 2-month-old Fmr1 KO animals injected with AAV-FMRP.

Compared to the vector tropism in phase 1, here FMRP transgene is expressed more extensively

in regions more distal from the site of i.c.v. injection including the amygdala, inferior colliculus,

thalamus and some cells in the cerebellum.

Fig. 27. Immunocytochemical analysis of AAV-FMRP transduction in

discrete brain regions of AAV-treated KO mice.

A

Page 92: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

81

Immunocytochemical analysis of AAV-FMRP transduction in discrete brain regions. (A)

Representative sagittal image showing FMRP expression in AAV-FMRP-injected Fmr1 KO

mouse brain on PND 65. (B) FMRP transgene expression in different brain regions 65 days

after i.c.v. administration of AAV-FMRP on PND 0-1. Brain sections were double labeled

using anti-FMRP (green) and anti-NeuN (red).

As we previously reported, the synapsin promoter endows a neuronal specific tropism of the

FMRP protein in the CNS (Gholizadeh et al., 2013; Gholizadeh et al., 2014). This was again

confirmed in phase 2 study where double-labeling with the neuron-specific marker NeuN

showed that FMRP was predominantly observed in NeuN-positive cells, in all of the brain

regions analyzed, indicating strong preferential expression in neuronal populations (Fig. 27B).

Quantitative analysis of FMRP positive cells co-expressing NeuN in the cingulate cortex

revealed over 90% neuronal transduction (Table 10).

B

Page 93: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

82

Table 10. Quantitative analysis of AAV-FMRP transduction in cingulate

cortex.

FMRP+ cells were counted in 3 consecutive coronal 25 μm thick sections (100 μm apart) of

the cingulate cortex from each brain (total of 9 sections from 3 mice) and reported as the

average number of cells per visual field. Percent of FMRP+ cells that expressed co-expressed

NeuN, total DAPI+ cells and NeuN+ neurons that expressed FMRP are calculated separately.

Analysis of FMRP over-expression in brains of WT mice injected with AAV-FMRP and

sacrificed on PND 65 revealed obvious FMRP over-expression in hippocampus, striatum,

retrosplenial cortex, cingulate cortex and motor cortex but no obvious over-expression in

cerebellum, compared to matched brain sections from AAV-null injected WT mice (Fig. 28).

These results suggest similar transduction efficiencies of AAV-FMRP in WT and KO neonatal

brain. We also quantified FMRP expression in the cingulate cortex and striatum of AAV-null or

AAV-FMRP-treated WT mice by measuring the total number of FMRP+ cells, average cell size,

and the mean intensity of the FMRP signal in each cell. As expected from the high abundance

and ubiquitous expression of FMRP protein in the brain, there was no difference in the number

of FMRP+ cells and size of the cells in AAV-null compared to AAV-FMRP-treated brains in

cingulate cortex or striatum. However, AAV-FMRP injections resulted in a 67% increase in

mean FMRP expression level in each cell (FMRP signal intensity/cell) in cingulate cortex and

Animal #

FMRP+ cells/mm2

% FMRP+/NeuN+

% DAPI+/FMRP+

% NeuN+/FMRP+

1 173 96 87 98

2 184 99 92 97

3 171 92 92 98

4 168 99 89 97

5 172 92 91 96

Average 173.6 95.6 90.2 97.2

Page 94: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

83

52% increase in striatum, compared to AAV-null injected mice (Fig. 29). There was a

remarkable variability in FMRP levels in different neurons both in the striatum and cingulate

cortex of AAV-treated WT brain, which possibly stems from a. the natural variability of FMRP

expression levels in different neurons in the WT brain, and b. the fact that more than one virion

can transduce a given neuron resulting in various FMRP levels in different cells.

Page 95: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

84

Fig. 28. Immunocytochemical analysis of FMRP over-expression in different

brain regions of AAV-SYN-Fmr1-treated WT mice.

NeuN NeuN

FMRP FMRP

DAPI DAPI

Merge Merge

NeuN NeuN

FMRP FMRP

DAPI DAPI

Merge Merge

NeuN NeuN

FMRP FMRP

DAPI DAPI

Merge Merge

WT AAV-null WT AAV-syn

Cingulate cortex Retrosplenial cortex Motor cortex

WT AAV-null WT AAV-syn WT AAV-null WT AAV-syn

Matched brain sections of AAV-null or AAV-SYN-Fmr1 treated WT mice were

double labeled using anti-FMRP (green) and anti-NeuN (red).

Page 96: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

85

Fig. 29. Quantitative analysis of FMRP over-expression in AAV-SYN- Fmr1-

treated WT mice.

Total number of FMRP+ cells per visual field, the area encompassing each cell (µm2) and

the mean intensity of FMRP+ signal per cell per µm2 is shown in the cingulate cortex or

striatum of AAV-FMRP or AAV-null-treated WT mice.

Page 97: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

86

6.4. Phase 2 - Behavioral Analyses

The first behavioral analysis was assessment of the reversibility of the well-established

locomotor hyperactivity in Fmr1 KO mice (Kazdoba et al., 2014; Wrenn et al., 2015) after

AAV-FMRP administration. As expected, KO-null compared to WT-null mice displayed a

significant elevation in the horizontal activity (Fig. 30A) and ambulatory activity (Fig. 30B),

while showing a reduction in the number of rest episodes (Fig. 30C). There was also a moderate

but not significant decrease in the total rest time (Fig. 30D). The total distance travelled

between groups was also not significantly different (data not shown). The KO+FMRP group

showed reduced horizontal activity, ambulatory activity and an elevation of the number of rest

episodes, all characteristic of a reduction in hyperactive behavior. Thus, the locomotor

hyperactivity was partially rescued in KO+FMRP mice, with no statistically significant

difference compared to the WT-null group. FMRP over-expression (WT+FMRP) did not

induce significant changes in the motor activity parameters.

Based on the high mouse-to-mouse variability of FMRP transduction rates seen in AAV-

FMRP injected mice, a potential correlation may exist between the total distance travelled and

FMRP or MeCP2 protein levels. There was an inverse trend observed between motor activity

and FMRP levels (Fig. 30E), indicating that FMRP levels might be inversely correlated with the

severity of the hyperactive phenotype. Considering the high number of data points, the innate

variability of the mice, and the high variability of FMRP transduction levels from the vector

delivery, this trend did not reach statistical significance (n = 56, r2= 0.055; p<0.0806).

However, a significant positive correlation was found between MeCP2 levels, and total distance

travelled. (n = 57, r2=0.1231, p<0.0074, Fig. 30F).

In summary, we conclude that (a) FMRP and MeCP2 are co-expressed in many if not most

neurons in the CNS, (b) MeCP2 mRNA is a substrate for FMRP, (c) MeCP2 is intrinsically

Page 98: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

87

over-expressed in Fmr1 KO mice (d) the combined analysis of all four treatments groups

demonstrated that cortical MeCP2 expression correlated with the severity of the hyperactive

phenotype, and (e) that FMRP transgene expression eliminated the pathological over-expression

of brain MeCP2 levels observed in Fmr1 KO-null-treated mice and concomitantly rectified

motor hyperactivity.

Page 99: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

88

Fig. 30. Analysis of locomotor activity.

A. Horizontal activity (beam breaks) for the WT-null, WT+FMRP, KO-null, and KO+FMRP

groups during locomotor testing. B. Ambulatory activity shown as total number of events; C.

Rest episodes as measured by the number of pauses (absence of beam breaks for ≥1s); and D.

Total rest time. Results are presented as an average ± SEM. *p<0.05. E. Linear correlation

between the FMRP protein levels as a function of the total distance travelled for the WT-null,

WT+FMRP and KO+FMRP groups combined. F. Correlation between the MeCP2 protein

levels and total distance travelled for WT-null, WT+FMRP, KO-null, and KO+FMRP groups.

Page 100: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

89

Impaired sensorimotor gating and auditory hyper-excitability are also among the robust,

well-established behavioral impairments in Fmr1 KO mice (Paylor et al., 2008; Rotschafer et

al., 2012), therefore pre-pulse inhibition and the base startle response were investigated. The

KO-null group had a significantly higher base startle response than the WT-null group (Fig.

31A). Furthermore, the KO+FMRP group showed a full rescue with a significant reduction in

this parameter compared KO-null and rendered no different than the WT-null group. The

WT+FMRP group, having the same trend as the KO+FMRP, showed a slight but not significant

reduction in the startle response compared to the WT-null group. The pre-pulse inhibition at 72

dB was not significantly different between null groups (Fig. 31B). Pre-pulse inhibition at 78 dB

(Fig. 31C) and 82 dB (Fig. 31D) also did not show any difference between AAV-null injected

groups, but interestingly, this difference was accentuated between AAV-FMRP injected groups

showing a statistical elevation of pre-pulse inhibition in the KO+FMRP group compared to the

reduced inhibition seen in WT+FMRP. Linear regression did not show any statistical

significance when compared to FMRP expression levels in the quantified mice (data not shown).

Page 101: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

90

Fig. 31. Analysis of sensorimotor gating.

A. Basal startle reaction corresponding to the pulse alone for the WT-null, WT+FMRP, KO -

null, and KO+FMRP groups. B. Percentage of inhibition to the startle response by submitting

the mice to a 74 dB pre-pulse; C. Percentage of inhibition to a 78 dB pre-pulse, and D.

Percentage of inhibition to a 82 dB pre-pulse. Results are presented as an average ± SEM.

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

In the elevated plus maze, which measures anxiety, the Fmr1 KO mice spent significantly

more time in the open arms (Fig. 32A), which is consistent with a previous report (Qin et al.,

2011). Intriguingly, this behavioral abnormality was fully reversed in the KO+FMRP treatment

group (Fig. 32A). Furthermore, the KO-null mice showed a significantly higher number of open

Page 102: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

91

arm entries compared to WT-null, which was also reversed in KO+FMRP group (Fig. 32C).

FMRP over-expression in the WT+FMRP group showed a significant increase in the number of

open arm entries, compared to WT-null (Fig. 32C), but no difference in the time spent in open

arms (Fig. 32A). The total number of entries was not different among the four groups (Fig.

32D). No significant correlation was detected in the elevated plus maze parameters when

compared to FMRP transgene levels (data not shown).

In the marble burying test, which measures stereotypic behavior in mice, our phase 1 results

(Gholizadeh et al., 2014), together with a previous report (Spencer et al., 2011), showed a

significant elevation in the number of marbles buried in Fmr1 KO mice. In the second phase of

this study, the KO-null and KO+FMRP groups displayed a modest but non-significant increase

in marbles buried compared to the WT-null group (Fig. 32E). The WT+FMRP over-expression

group, however, buried significantly less marbles compared to both KO groups, indicating that

higher levels of FMRP contribute to lowering the total number of marbles buried, as was

previously observed (Gholizadeh et al., 2014).

The number of ultrasonic vocalizations during courtship behavior was found to be reduced in

Fmr1 KO mice in phase 1 (Gholizadeh et al., 2014), as well as a previous study (Rotschafer et

al., 2012). Here, the KO-null mice and the WT+FMRP showed non-significant reductions in

the number of vocalizations (Fig. 32F). The number of vocalizations in the KO+FMRP group

was significantly lower than the number in the WT-null group, indicating no reversal of the

trend towards reduced vocalization in the KO-null mice.

Page 103: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

92

Fig. 32. Analysis of anxiety, stereotypical behavior, and courtship behavior.

.

A. Elevated plus maze trial showing the total time spent in the open arm for the WT-null,

WT+FMRP, KO-null, and KO+FMRP groups. B. Elevated plus maze trial showing the number

of closed arm entries, C. the number of open arm entries, and D. the total number of entries. E.

Marble burying assay for repetitive, stereotyped behavior showing the total number of marbles

buried during a 30 min. interval. F. Measurement of the total number of ultrasonic vocalizations

of male mice during courtship behavior. Results are presented as an average ± SEM. *p<0.05,

**p<0.01, *** p<0.001.

Page 104: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

93

Chapter 7. Discussion

7.1. FMRP expression in the developing and adult brain

The purpose of this part of my research was to fully document the cell type expression of

FMRP in neurons and glia in selected brain regions of the developing and mature WT mouse

CNS. Our results demonstrated a predominantly neuronal expression of FMRP in all of the

brain regions analyzed, except the corpus callosum, where only a small number of neurons were

present and FMRP expression was mainly observed in astrocytes. Our data also revealed the

presence of FMRP in cells expressing markers for astrocytes (S100β), microglia (Iba-1) and

oligodendrocyte precursor cells (NG2) in the developing brain. In general, glial expression of

FMRP diminished to low or undetectable levels in the adult brain following different trajectories

in different brain regions.

FMRP expression was abundant in astrocytes at PND 0 in all of the brain regions analyzed;

thereafter, expression in astrocytes showed a gradual decline in striatum and hippocampus,

reaching negligible numbers of FMRP+/S100β+ cells in the adult mouse brain. Pacey and

Doering (2007) reported FMRP expression in cells expressing the astrocytic marker GFAP in

the hippocampus and the ependymal cells surrounding the third ventricle at PND 1 and PND 7,

but no FMRP/GFAP co-localization in the brains of adult (2-month-old) mice. Our results

confirmed the findings of Pacey and Doering, (2007) in the hippocampus using a different

astrocytic marker (S100β), and extended these findings to other brain regions including the

cingulate cortex, corpus callosum, striatum, and cerebellum. Quantitative analysis of cell-type

expression of FMRP indicated that more than 20% of FMRP+ cells in the corpus callosum co-

expressed S100β and this co-localization level remained relatively constant from PND 0 to adult

mice. In contrast, there were few NeuN+ cells detected in the corpus callosum.

Page 105: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

94

Expression of FMRP in astrocytes could be of crucial importance in regulating the

established functions of astrocytes in synapse formation and neural plasticity. More

specifically, during the early postnatal weeks in rodents when astrocytes play a critical role in

regulation of neuronal growth and synapse formation, the lack of FMRP could contribute to the

abnormal dendritic morphology and synapse development seen in FXS (Jacobs and Doering,

2010). Astrocytes dynamically regulate synaptic transmission, partly by releasing soluble

synaptogenic molecules such as the astrocyte-secreted matricellular proteins, including Hevin,

and thrombospondin-1 (Jones and Bouvier, 2014), or neurotrophic factors such as Neurotrophin-

3 (Yang et al., 2012), which are highly expressed within first few weeks of postnatal

development and decrease as the brain matures. Interestingly, Hevin, also known as SPARC-

like 1, and Neurotrophin-3 have been identified as mRNA substrates for FMRP (Darnell et al.,

2011) and excessive neurotrophin-3, secreted from Fmr1 KO astrocytes, has been suggested to

contribute to abnormal neuronal dendritic development in Fmr1 KO mouse model (Yang et al.,

2012). Furthermore, thrombospondin-1 protein is reduced in astrocytes of Fmr1 KO mice and

synaptic deficits are corrected by its protein replacement in Fmr1 KO mouse hippocampal

neurons (Cheng et al., 2014). These findings highlight the important role for FMRP expression

in astrocytes during early postnatal weeks, which coincides with the peak of synaptogenesis.

FMRP expression has been reported in oligodendrocyte precursors in vitro (Wang et al.,

2004) and in the early postnatal cerebellum (Pacey et al., 2013). Our data extend these findings

by reporting a more comprehensive analysis of FMRP/NG2 co-localization in different regions

of the mouse brain at multiple time points during postnatal CNS development. FMRP

expression is also present in mature MBP+ oligodendrocytes of rodents as well as human

oligodendrocytes (Giampetruzzi et al., 2013). FMRP was previously shown to bind to MBP

mRNA and down-regulate its translation in vitro (Wang et al., 2004). Moreover, the lack of

Page 106: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

95

FMRP is associated with delayed myelination in the Fmr1 KO mouse cerebellum (Pacey et al.,

2013). In the present study, we observed a gradual decline in the number of FMRP+ cells co-

expressing NG2 from PND 0 to adult mice; this pattern was observed in all of the brain regions

examined, but was most prominent in the corpus callosum and striatum. A decline in FMRP

levels during oligodendrocyte differentiation was previously reported in primary cultures of

oligodendrocytes derived from neonatal rat brain (Wang et al., 2004). This gradual decline

indicates that FMRP likely plays a more important role in myelin formation during CNS

maturation, rather than in mature CNS.

FMRP has been previously reported in cultures of microglial cells (Yuskaitis and Jope, 2009;

Yuskaitis et al., 2010a). However, the findings reported here provide the first direct evidence

for FMRP expression in microglia in brain tissue. Post mortem studies have identified

neuroinflammation and glial activation in the brains of individuals with idiopathic autism

(Tetreault et al., 2012), and other studies suggest a similar pathology may be present in FXS

(Jacobs et al., 2012). FMRP expression was reported in the BV-2 microglial cell line (Yuskaitis

and Jope, 2009). However, despite microglial expression of FMRP, there was no difference in

the production of pro-inflammatory cytokines IL-6 and TNFα after acute lipopolysaccharide-

induced activation of microglia in WT or Fmr1 KO mice (Yuskaitis and Jope, 2009). Recent

results from our laboratory demonstrated that a persistent activation of astrocytes exists in the

adult Fmr1 KO mouse brain; however, no evidence was observed for activated microglia in

Fmr1 mice (Pacey et al., 2015).

In the current study, FMRP expression in microglia in WT mice was detected most

prominently during the first 10 postnatal days and declined thereafter. Interestingly, the number

of microglial cells in mice is immensely increased during the first two postnatal weeks when the

vast majority (>95%) of microglia are born (Alliot et al., 1999). These findings suggest a

Page 107: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

96

potential regulatory role for FMRP in the cell lineage and maturation of microglia during early

development. However, the exact molecular function of FMRP in these cells during brain

development and its contribution to behavioral abnormalities in Fmr1 KO mice remains to be

elucidated.

Quantitative analysis of FMRP+ cells in the cingulate cortex and corpus callosum of the WT

mouse revealed the highest number of FMRP+ cells at PND 0 which diminished steadily

throughout brain development. The abundant FMRP expression at birth and during the first 1-2

postnatal weeks indicates that the functional requirement of FMRP in glia is highest during this

critical period of early brain development. In the drosophila nervous system, FMRP appears to

be present sequentially, first in neuroblasts and then in glia, and glial expression was required

for neuroblast reactivation (Callan et al., 2012). Similar to the mouse model, the expression of

the drosophila FMRP homolog is developmentally regulated (Tessier and Broadie, 2008) and its

re-introduction during brain development rescues the dendritic impairment in the drosophila

model (Gatto and Broadie, 2009), indicating a causal relationship between FMRP expression

during a distinct developmental time-window and normal synapse formation. In the mouse

CNS, the most intense period of synaptogenesis (Zito and Svoboda, 2002) and the most severe

abnormalities in dendritic spine morphology converge at the end of the first postnatal week,

during which FMRP is abundantly expressed (Nimchinsky et al., 2001).

7.2. Gene therapy as a potential therapeutic avenue for the treatment of

Fragile X syndrome

There is no pharmacological cure for FXS and the currently prescribed medications, such

as anti-psychotics, anti-depressants, and stimulants, only partially alleviate selected symptoms

and are associated with deleterious side effects. Recently failed clinical trials of metabotropic

glutamate receptor antagonists and a GABAB receptor agonist to treat FXS relied on single

Page 108: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

97

molecular targets that were expected to alleviate certain symptoms, but did not address the core

underlying issue—the pathological absence or reduction of FMRP in the brain (Hampson et al.,

2012). Given that FMRP regulates the translation and stability of hundreds of mRNAs in the

CNS (Darnell et al., 2011; Ascano et al., 2012), it is not surprising that manipulation of single

molecular targets, such as metabotropic glutamate receptors, p21-activated kinase, and

membrane metalloproteinase-9, failed to produce long-term benefits (Choi et al., 2011; Dolan et

al., 2013; Gkogkas et al., 2014). In fact, to date none of the phase 3 clinical trials of small

molecules against single molecular targets have been successful, in spite of promising results in

pre-clinical or early clinical stage testing (Paribello et al., 2010; Scharf et al., 2014). However,

considering the plethora of genes whose expression is regulated by FMRP, a priori, it may be

expected that restoring FMRP expression in the CNS could provide a more comprehensive

reversal of the disorder compared to targeting single molecules.

7.3. Major factors affecting cellular tropism, efficiency and distribution

of AAV-induced transduction

Our initial results demonstrated that i.c.v. administration of AAV9- eGFP on PND 5,

compared to PND 21, resulted in broader distribution of the transgene throughout the brain.

Transgene delivery at early postnatal days is also favourable for the development of gene

therapy strategies for neurodevelopmental disorders, such as FXS, as earlier re-introduction of

the missing protein very early in postnatal period at the peak of neurogenesis is expected to

translate into a higher level of phenotypic rescue.

A goal of this study was to compare cell type specificity of virus transduction after treatment

of AAV9-eGFP as directed by the CMV vs. the synapsin-1 promoter (Gholizadeh et al., 2013).

AAV9-CMV-eGFP vector preferentially targeted astrocytes in neonatal mice and neurons in

juvenile mice, whereas treatment with the same AAV9 vector construct, but swapping the CMV

Page 109: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

98

promoter for the synapsin 1 promoter (AAV9-SYN-eGFP), resulted in extensive neuron-specific

transduction, regardless of the time of administration. Differences in cell-type-specific

transduction profiles at different stages in development are important considerations when

planning gene therapy studies for monogenic neurological disorders, where the aim of the study

is to introduce the missing protein specifically to either neurons or glia.

Our results demonstrated that i.c.v. treatment with AAV9 vectors displayed very good

diffusion properties in the mouse brain, especially when administered during the early postnatal

period. Another advantage of delivering AAV vectors into the CSF is the possibility of

targeting ependymal cells that line the inside perimeter of the ventricles and form the interface

between the ventricles and brain parenchyma. For transgenes that produce soluble proteins,

uptake into ependymal cells, which are susceptible to a wide range of viral vectors, provides the

potential to secrete the transgene into the cerebrospinal fluid, where it can diffuse continuously

into the brain; this strategy has been successfully employed to treat mouse models of lysosomal

storage disorders (Ghodsi et al., 1999; Liu et al., 2005; Fu et al., 2007; Haurigot et al., 2013;

Yamazaki et al., 2014).

Based on our initial results, we expected that administration of the AAV9 vector driven by

the SYN promoter at early postnatal days would result in wide diffusion of the vectors, and

consequently predominant expression of the therapeutic protein in neurons, where it is most

critically required in many neurological disorders. Early postnatal administration of AAV-based

therapeutics would be applicable not only to FXS but may also be a useful strategy in other

neurodevelopmental disorders such as Angelman Syndrome and Rett Syndrome where the

disease pathology is caused primarily by the absence or near absence of a missing protein in

neurons (Hampson et al., 2012). Together, these findings highlight the important effects of

Page 110: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

99

AAV serotype, the promoter, and the age of administration on the intensity, distribution and cell

type specificity of AAV transduction.

7.4. FMRP transgene expression in the brains of Fmr1 KO mice

As outlined in Chapter 3, FMRP is widely distributed in most neurons throughout the adult

mammalian CNS. This wide distribution throughout the CNS presents a challenge from the

perspective of virally mediated gene therapy in terms of achieving sufficiently wide distribution

of the viral vector. Other groups investigating approaches for treating CNS disorders in animal

models had previously reported success after direct CNS injections (Daily et al., 2011; Gadalla

et al., 2012), and after intra-venous injection of AAV vectors in the mouse (Garg et al., 2013)

and monkey (Samaranch et al., 2012). Of the various AAV vectors currently available, AAV9

was used in the current study because of its documented propensity to disseminate within the

brain parenchyma and its neuronal tropism (Nonnenmacher and Weber, 2012; Rothermel et al.,

2013).

We chose i.c.v. administration into the lateral ventricles for several reasons including (a) the

desire to avoid possible off-target tissue effects such as the liver, a known AAV-targeted organ,

(b) to minimize the amount of vector given to the animal, and (c) to specifically scrutinize the

effects of FMRP restoration in the brain. The results of our early work indicated substantial

diffusion of a vector coding for eGFP from the ventricles and transgene expression in multiple

forebrain regions of the mouse CNS (Gholizadeh et al., 2013). Relevant conclusions gleaned

from that AAV9-eGFP study were that the age of injection and the promoter employed were

critical factors in determining the distribution and cell type transduction specificity of the

transgene. We speculate that one parameter that may promote the spread of the viral vector in

the brain after i.c.v. administration is the recently characterized “glymphatic system”. Similar in

function to the peripheral lymphatic system, in brain parenchyma, para-vascular exchange

Page 111: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

100

between cerebrospinal fluid and interstitial fluid has been shown to be facilitated by astroglial

water transport mediated in part by the aquaporin-4 water channel (Iliff et al., 2013).

In the adult mammalian CNS, FMRP is highly expressed in neurons (Cruz-Martín et al.,

2010; Harlow et al., 2010). In the present study, double labeling immunocytochemical analysis

demonstrated that AAV-FMRP transduction occurred primarily in neurons rather than glia, as

expected from the incorporation of the neuron-selective synapsin promoter in the AAV-FMRP

vector construct. In the first phase of the study where mice were treated at PND5, analysis of

the anatomical distribution of the recombinant FMRP transgene in AAV-FMRP-treated KO

mice indicated that it was present in several forebrain regions including the retrosplenial and

cingulate cortices, the hippocampus, and the striatum. FMRP was not detected in brain regions

located more distal from the lateral ventricles such as the piriform cortex, inferior colliculus,

cerebellum, and brainstem. However, in the second phase of the study where mice received

AAV-FMRP on PND 0 or 1, immunocytochemical analyses revealed increased distribution of

the vectors in brain regions devoid of FMRP expression in phase 1, such as amygdala, thalamus,

inferior colliculus, superior colliculus, and the cerebellum. Quantitative WB analysis further

confirmed more efficient FMRP transgene expression after PND 0-1 vector injections, with

higher FMRP levels in the cerebral cortex, frontal cortex and hippocampus, detected

approximately two months after PND 0-1 injections compared to PND 5 AAV delivery at phase

1.

Because severely low or elevated levels of FMRP expression have both been associated with

pathological outcomes, the correct level of expression of FMRP was previously suggested to be

of critical concern (Nagamani et al., 2012). In phase 2 we examined four different treatment

groups with disparate levels of FMRP in the forebrain. The KO-null group with none, the

KO+FMRP group had a midpoint average expression level compared to WT levels, the WT-null

Page 112: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

101

had normal expression levels, while the WT+FMRP showed an almost two fold increase in the

expression of largest isoform of FMRP. In addition, the innate mouse-to-mouse variability in

the degree of vector diffusion and cellular transduction also facilitated the delineation of the

effects of a wide spectrum of FMRP expression levels on behavior.

7.5. Behavioral effects of FMRP restoration in the brains of Fmr1 KO

mice

In the first phase of the research, we assumed one potential reason that might explain the

absence of reversal of abnormal behavior on some tests might be the limited FMRP expression

in brain regions located distally from the injection site in the lateral ventricles. This may be

particularly pertinent to more caudal regions such as the cerebellum and brainstem, where very

limited FMRP transgene expression was detected. The presence of the FMRP transgene in the

observed brain regions (cerebral cortex, hippocampus, and striatum), and its absence or very low

expression in other regions provided an opportunity to link the restoration of FMRP expression

in specific brain regions to corrected pathological behaviors observed in Fmr1 KO mice. Our

data from phase 1 suggested that the presence of FMRP in the cortex and/or the striatum of the

AAV-FMRP-treated mice could have been a contributing factor for the reduction in repetitive

behavior, as assessed by the marble burying test. Several lines of evidence have shown that

lesions of the dorsal or ventral striatum in rodents (Aliane et al., 2011) and non-human primates

(Saka et al., 2004) block stereotyped behavior. Moreover, in a longitudinal study of brain

development in children, it was shown that an abnormally increased growth rate of the striatum

was correlated with more severe repetitive behavior in autistic children compared to control

subjects (Langen et al., 2013).

It could also be speculated that FMRP transgene expression in neurons of the cingulate

cortex might have contributed to the rescue of impaired social dominance (as assessed using the

Page 113: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

102

“tube test”). The medial prefrontal cortex, which includes the cingulate cortex, sub-callosal

cortex, and the medial frontal gyrus, has been shown to be linked with social dominance in

mice. Based on recordings from pyramidal neurons of the medial prefrontal cortex, Wang et al,

2011 reported that social rank in mice directly correlates with the synaptic strength of pyramidal

neurons in the medial prefrontal cortex (Wang et al., 2011). It has also been reported that levels

of key proteins involved in regulating synaptic function, including the NMDA receptor subunits

NR1, NR2A, and NR2B, are decreased in the medial prefrontal cortex of Fmr1 KO mice

(Krueger et al., 2011).

Our findings in the elevated plus maze test (phase 2) demonstrated that AAV-null injected

Fmr1 KO mice exhibited lower non-social anxiety (more time in the open arms). Abnormally

low anxiety has been reported in previous studies of FXS KO mice (Liu et al., 2011; Heulens et

al., 2012), as well as another mouse model of autism lacking the mu-opioid receptor (Ide et al.,

2010; Becker et al., 2014). In the present work, we speculate that FMRP transgene expression

in the amygdala of the AAV-FMRP-treated mice could have been a contributing factor for

normalizing the reduction in anxiety levels as assessed by the elevated plus maze test. Several

lines of evidence have shown the pivotal role of amygdala in regulating the emotional symptoms

associated with FXS such as anxiety and unstable mood (for a review see Suvrathan and

Chattarji, 2011). Whole-cell recordings in brain slices from adult Fmr1 KO mice revealed a

decrease in mGluR-dependent long-term potentiation at thalamic inputs to principal neurons in

the lateral amygdala (Suvrathan et al., 2010).

Analysis of pre-pulse inhibition test in phase 2 showed a higher basal startle response in

Fmr1 KO mice compared to WT mice which was rescued in the AAV-FMRP-treated mice.

Fmr1 KO AAV-null-treated mice displayed abnormal responses to auditory stimuli, as seen in

increased incidence of audiogenic seizure, and auditory startle response. Abnormal pre-pulse

Page 114: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

103

inhibition responses to auditory stimuli has also been reported in FXS patients (Frankland et al.,

2004; Hessl et al., 2009; Yuhas et al., 2011; Kohl et al., 2013). In our study, FMRP transgene

expression in medial prefrontal cortex of AAV-FMRP-treated KO mice might have contributed

to the rescue of the elevated startle response. Lesions in the medial prefrontal cortex of rodents

result in reduced pre-pulse inhibition responses (Bubser and Koch, 1994) and pharmacological

blockade of dopaminergic D1 receptors in the medial prefrontal cortex reduces pre-pulse

inhibition in a rat model of schizophrenia (Swerdlow et al., 2006).

In contrast to the reversal of behavioral abnormalities described above, there was no rescue

of other pathological behaviors including motor hyperactivity, reduced ultrasonic vocalizations

and susceptibility to audiogenic seizures. There are several possible explanations for this

outcome. One possibility is that undetectable or very low levels of FMRP transgene expression

in the cerebellum of AAV-treated KO mice, may be a factor contributing to the inability to

achieve full behavioral recovery of motor hyperactivity. FMRP is expressed extensively in all

neuronal populations of the cerebellum (Pacey et al., 2013), and the cerebellum has been

consistently associated with neuropathology in FXS (Koekkoek et al., 2005; Ellegood et al.,

2010; Fatemi et al., 2012), and autism (Whitney et al., 2008). The cerebellum has also been

linked with motor hyperactivity in Fmr1 KO mice (Rogers et al., 2013). Similarly, the absence

of FMRP in the inferior colliculus and the brainstem of the AAV-FMRP-treated Fmr1 KO mice

might explain the lack of reversal of audiogenic seizures in phase 1. These brain regions have

been linked to auditory processing and are thought to be important for propagation of auditory

seizures (Garcia-Cairasco, 2002; Byrns et al., 2014).

Perplexingly, increased repetitive behavior was rescued in phase 1 but not in phase 2 of the

gene therapy study. There are several potential explanations for the lack of statistically

significant reversal of this phenotype in phase 2. The number of animals tested in each group in

Page 115: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

104

phase 2 and the use of AAV-null injections instead of PBS as control groups can both

potentially contribute to differences in statistical analyses of behavioral results. Also,

differences in FMRP transgene expression levels, delivered at PND 0-1 instead of PND 5 in the

striatum, a brain region closely associated to modulating repetitive behavior in mice, might

attribute to differences in number of marbles buried in the two phases of the study. For

instance, in the long arm of phase 1 project, FMRP was detected at18 ±5% of WT expression in

the striatum; however in phase 2, FMRP transgene expression levels reached 33% of WT

expression, which is almost two times higher than FMRP levels in the AAV-FMRP group in

phase 1. Based on a preliminary analysis from a small set of data points in the phase 1 (n = 10),

a possible inverse correlation exists between the number of marbles buried and FMRP

expression level (r2 = 0.37 p = 0.06), which may explain differences in repetitive behavior in

AAV-FMRP-treated KO mice in the different phases of the study.

7.6. Possible contributions of aberrant MeCP2 expression to

pathological abnormalities in FXS

We established that MeCP2 mRNA is a substrate for FMRP and that MeCP2 protein levels

are elevated (about 1.5 to 2-fold over-expression) in the brains of adult naive Fmr1 KO mice

compared to WT mice. Moreover, in the WT mouse brain, most neurons co-expressed MeCP2

and FMRP in the brain regions examined. Finally, elevated MeCP2 levels in the cortex of Fmr1

KO mice were normalized after FMRP transgene expression. Intriguingly, we detected a

correlation between elevated MeCP2 expression in the forebrain and increased motor activity.

Evidence supporting a relationship between MeCP2 and motor activity comes from transgenic

mouse lines over-expressing MeCP2 at levels comparable to what we observed in Fmr1 mice

(about 1.5 to 2-fold over-expression), where increased vertical activity (rearing) and increased

aggressive behavior have been reported (Collins et al., 2004; Bodda et al., 2013). Moreover,

Page 116: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

105

similar to our results with male Fmr1 mice, brain MeCP2 expression in female MeCP2

heterozygous mice was also reported to be correlated with motor activity (Wither et al., 2013).

Further ancillary evidence of a link between MeCP2 and FMRP derives from the demonstration

that MeCP2-repressed genes overlap extensively with genes that are FMRP mRNA substrates

(Gabel et al., 2015). The relationship observed between increased MeCP2 levels and motor

hyperactivity in the Fmr1 mouse, together with the phenotypic overlap between Fmr1 KO mice

and mouse models over-expressing MeCP2, raises the question as to whether elevated brain

MeCP2 might also contribute to other features of the fragile X phenotype. Mice over-

expressing MeCP2 also display impairments in anxiety, hippocampal long term potentiation,

deficits in learning and memory (Shang et al., 2009; Na et al., 2012; Neuhofer et al., 2015), and

increased seizures and stereotypic behavior (Collins et al., 2004). Moreover, patients with

fragile X syndrome, duplication of the FMR1 gene, or MeCP2 gene duplication syndrome all

display several over-lapping phenotypes (Fig. 33). Gene duplication in either FMR1 or MeCP2

is associated with developmental delay, mental retardation, and seizures (Smyk et al., 2008).

Furthermore, cerebellar abnormalities such as Purkinje cell loss has been reported in MeCP2

duplication patients (Reardon et al., 2010), as well as elderly males with FXS (Greco et al.,

2011). The relationship observed between increased MeCP2 levels and motor hyperactivity in

the Fmr1 mouse, together with the phenotypic overlap between Fmr1 KO mice and mouse

models over-expressing MeCP2, raises the intriguing possibility that elevated brain MeCP2

might also contribute to other features of the fragile X phenotype and may provide a common

mechanistic link across some populations of autism spectrum disorders.

Page 117: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

106

Fig. 33. Overlapping characteristics of FXS, Rett syndrome, FMR1

duplication syndrome, and MeCP2 duplication syndrome.

Page 118: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

107

7.7. Concluding remarks and future directions

Our results demonstrated a predominantly neuronal expression profile of native FMRP in all

of the brain regions analyzed of the adult WT mouse, except in the corpus callosum, where only

a small number of neurons were present and FMRP expression was mainly observed in

astrocytes. Our data also revealed the presence of FMRP in cells expressing markers for

astrocytes, microglia and oligodendrocyte precursor cells in the developing brain. In general,

glial expression of FMRP diminished to low or undetectable levels in the adult brain following

different trajectories in different brain regions, with the exception of the corpus callosum in

which predominant FMRP expression in astrocytes remained almost unaffected from birth to

adulthood.

The findings gleaned from our viral vector gene therapy work provide a proof-of-principle

demonstrating that expression of recombinant FMRP in specific regions of the CNS can reverse

or alleviate selected neurological abnormalities in the Fmr1 KO mouse. The exceptionally

protracted expression of AAV-mediated transgenes as shown here (at least 7 months post-

transfection), and in other studies using AAV transduction in the brain (up to 1 year post-

injection), makes this approach especially attractive for treating neurodevelopmental disorders

where the presence of the missing protein is likely required over an extended time frame.

Conceivably, the introduction of FMRP into Fmr1 KO mouse neurons could indirectly result

in correction of disrupted neuron-to-astrocyte signaling in FXS at a critical time during early

developmental stages during which astrocyte support of neuronal growth and synaptic formation

is critical (Cheng et al., 2012). Since glia play indispensable roles in synaptic development,

developmentally regulated FMRP expression in glia might regulate synaptic development of

contiguous neurons. Future efforts could be directed towards determining whether FMRP re-

introduction in astrocytes alone, or concomitantly in both astrocytes and neurons, may provide

Page 119: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

108

enhanced benefit (correction of abnormal behaviors) over that seen when expressed only in

neurons, as done in the current research.

One potential reason for limited transgene expression in cerebellum might be low expression

levels of the synapsin gene within the first two postnatal weeks in the mouse cerebellum

(Melloni et al., 2004), which could have contributed to low levels of promoter-driven FMRP

transgene expression in our study. This issue might be resolved by using other promoters in the

AAV construct such as the Fmr1 promoter (which has not yet been fully mapped out), or by

using one of the more established ubiquitous promoters such as cytomegalovirus, or the chicken

beta-acting promoter in our future attempts. A potential problem from the use of ubiquitous

promoters could be ectopic expression in non-neuronal cells such as glia in adults, where FMRP

is almost exclusively expressed in neurons. Therefore another alternative is the use of combined

cytomegalovirus mini-promoter and synapsin, as used in a recent study to provide widespread

neuronal transduction (Huda et al., 2014). A better understanding of the brain regions FMRP

needs to be delivered to and levels of FMRP needed to cause significant changes on

pathological behavior will facilitate development of more efficacious gene therapies for FXS.

Another potential parameter to consider is delivery of AAV vectors containing other Fmr1

isoforms. First, it must be established whether expression of isoform 1, which encodes for the

longest isoform of FMRP in the CNS, is sufficient to restore normal function. Once the most

functionally essential FMRP isoforms are fully characterized, co-administration of multiple

AAV vectors, each containing a different isoform, might potentially lead to more favorable

outcomes compared to using vectors expressing isoform 1 alone. It has been shown that only

two FMRP isoforms (isoforms 1 and 7) have the major phosphorylation site at Ser-499, which is

believed to affect the ability of the protein to bind to stalled ribosomes and inhibit translation

(Brackett et al., 2013). Therefore, in the next phase of the study, we might consider delivering

Page 120: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

109

isoform 7 using viral vectors on its own and in combination with isoform 1. Expression of each

isoform individually would enable the characterization of the functional impact of each isoform

in modulating behavior, independent from the contribution of other isoforms.

Based on our AAV-FMRP experiments in the WT mouse, forebrain over-expression was

associated with a reduction in ultrasonic vocalizations, stereotyped behavior, and pre-pulse

inhibition. Of course, how these abnormalities in mice might relate to human FMR1 gene

duplication syndrome remains unknown. FMR1 gene duplication in humans is characterized

with symptoms including mental retardation, developmental delay and seizures (Rio et al., 2010;

Nagamani et al., 2012; Vengoechea et al., 2012; Hickey et al., 2013). The present work

provides the first evidence supporting the notion that modest (1.5- 2-fold) postnatal over-

expression of the dominant mouse isoform of FMRP in the brain results in behavioral

abnormalities. Overall our findings indicate that establishing the therapeutic window for Fmr1

gene delivery and careful regulation of the level of transgene expression, are critical for

achieving successful rescue of this disorder.

In conclusion, this work has established a proof-of-principle by demonstrating that abnormal

behaviors in Fmr1 KO mice can be reversed after treatment with AAV-FMRP and transduction

of forebrain neurons. PND 0-1 bilateral i.c.v. injection of AAV-FMRP resulted in wider

distribution compared to AAV treatment at PND 5. We believe that the neurodevelopmental

effects of early FMRP introduction may also promote normal neurogenesis and neuronal

maturation in the Fmr1 brain and that this might be a contributing factor in the behavioral rescue

seen in the adult mice. In addition, brain levels of PSD-95 and MeCP2 proteins (two key FMRP

substrates) were normalized in the cortex and likely contributed to establishing proper synaptic

integrity. It remains to be seen whether FMRP expression in more distal brain regions, such as

the cerebellum, would further correct additional pathological and behavioral abnormalities in

Page 121: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

110

Fmr1 KO mice. The findings of this study contribute useful new information in the quest for

eventually translating our work here in the mouse model of FXS to human gene therapy.

Page 122: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

111

References

Akins MR, Leblanc HF, Stackpole EE, Chyung E, Fallon JR (2012) Systematic mapping of

fragile X granules in the mouse brain reveals a potential role for presynaptic FMRP in

sensorimotor functions. The Journal of comparative neurology 520:3687-3706.

Aliane V, Perez S, Bohren Y, Deniau JM, Kemel ML (2011) Key role of striatal cholinergic

interneurons in processes leading to arrest of motor stereotypies. Brain 134:110-118.

Alliot F, Godin I, Pessac B (1999) Microglia derive from progenitors, originating from the yolk

sac, and which proliferate in the brain. Brain research Developmental brain research

117:145-152.

Alvarez-Buylla A, Lim DA (2004) For the long run: maintaining germinal niches in the adult

brain. Neuron 41:683-686.

Ascano M, Jr., Mukherjee N, Bandaru P, Miller JB, Nusbaum JD, Corcoran DL, Langlois C,

Munschauer M, Dewell S, Hafner M, Williams Z, Ohler U, Tuschl T (2012) FMRP

targets distinct mRNA sequence elements to regulate protein expression. Nature

492:382-386.

Asokan A, Schaffer DV, Samulski RJ (2012) The AAV vector toolkit: poised at the clinical

crossroads. Mol Ther 20:699-708.

Bagni C, Greenough WT (2005) From mRNP trafficking to spine dysmorphogenesis: the roots

of fragile X syndrome. Nature reviews Neuroscience 6:376-387.

Bagni C, Tassone F, Neri G, Hagerman R (2012) Fragile X syndrome: causes, diagnosis,

mechanisms, and therapeutics. The Journal of clinical investigation 122:4314-4322.

Bartus RT, Baumann TL, Siffert J, Herzog CD, Alterman R, Boulis N, Turner DA, Stacy M,

Lang AE, Lozano AM, Olanow CW (2013) Safety/feasibility of targeting the substantia

nigra with AAV2-neurturin in Parkinson patients. Neurology 80:1698-1701.

Barua NU, Woolley M, Bienemann AS, Johnson D, Wyatt MJ, Irving C, Lewis O, Castrique E,

Gill SS (2013) Convection-enhanced delivery of AAV2 in white matter--a novel method

for gene delivery to cerebral cortex. Journal of neuroscience methods 220:1-8.

Bechara E, Didiot M, Melko M, Davidovic L, Bensaid M, Martin P, Castets M, Pognonec P,

Khandjian E, Moine H, Bardoni B (2009) A novel function for fragile X mental

retardation protein in translational activation. PLoS biology 7.

Page 123: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

112

Becker JA, Clesse D, Spiegelhalter C, Schwab Y, Le Merrer J, Kieffer BL (2014) Autistic-like

syndrome in mu opioid receptor null mice is relieved by facilitated mGluR4 activity.

Neuropsychopharmacology 39:2049-2060.

Benkhelifa-Ziyyat S, Besse A, Roda M, Duque S, Astord S, Carcenac R, Marais T, Barkats M

(2013) Intramuscular scAAV9-SMN injection mediates widespread gene delivery to the

spinal cord and decreases disease severity in SMA mice. Mol Ther 21:282-290.

Berry-Kravis E (2014) Mechanism-based treatments in neurodevelopmental disorders: fragile X

syndrome. Pediatr Neurol 50:297-302.

Berry-Kravis E, Sumis A, Hervey C, Nelson M, Porges SW, Weng N, Weiler IJ, Greenough WT

(2008) Open-label treatment trial of lithium to target the underlying defect in fragile X

syndrome. J Dev Behav Pediatr 29:293-302.

Berry-Kravis E, Hessl D, Coffey S, Hervey C, Schneider A, Yuhas J, Hutchison J, Snape M,

Tranfaglia M, Nguyen DV, Hagerman R (2009) A pilot open label, single dose trial of

fenobam in adults with fragile X syndrome. J Med Genet 46:266-271.

Bhakar AL, Dolen G, Bear MF (2012) The pathophysiology of fragile X (and what it teaches us

about synapses). Annu Rev Neurosci 35:417-443.

Blackwell E, Zhang X, Ceman S (2010) Arginines of the RGG box regulate FMRP association

with polyribosomes and mRNA. Human molecular genetics 19:1314-1323.

Bodda C, Tantra M, Mollajew R, Arunachalam JP, Laccone FA, Can K, Rosenberger A,

Mironov SL, Ehrenreich H, Mannan AU (2013) Mild overexpression of Mecp2 in mice

causes a higher susceptibility toward seizures. The American journal of pathology

183:195-210.

Bolduc FV, Bell K, Cox H, Broadie KS, Tully T (2008) Excess protein synthesis in Drosophila

fragile X mutants impairs long-term memory. Nature neuroscience 11:1143-1145.

Bolduc FV, Bell K, Rosenfelt C, Cox H, Tully T (2010) Fragile x mental retardation 1 and

filamin a interact genetically in Drosophila long-term memory. Front Neural Circuits

3:22.

Boutin S, Monteilhet V, Veron P, Leborgne C, Benveniste O, Montus MF, Masurier C (2010)

Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV)

types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using

AAV vectors. Hum Gene Ther 21:704-712.

Page 124: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

113

Brackett DM, Qing F, Amieux PS, Sellers DL, Horner PJ, Morris DR (2013) FMR1 transcript

isoforms: association with polyribosomes; regional and developmental expression in

mouse brain. PloS one 8:e58296.

Bubser M, Koch M (1994) Prepulse inhibition of the acoustic startle response of rats is reduced

by 6-hydroxydopamine lesions of the medial prefrontal cortex. Psychopharmacology

(Berl) 113:487-492.

Byrns CN, Pitts MW, Gilman CA, Hashimoto AC, Berry MJ (2014) Mice lacking selenoprotein

P and selenocysteine lyase exhibit severe neurological dysfunction, neurodegeneration,

and audiogenic seizures. The Journal of biological chemistry 289:9662-9674.

Callan MA, Clements N, Ahrendt N, Zarnescu DC (2012) Fragile X Protein is required for

inhibition of insulin signaling and regulates glial-dependent neuroblast reactivation in

the developing brain. Brain research 1462:151-161.

Carty N, Lee D, Dickey C, Ceballos-Diaz C, Jansen-West K, Golde TE, Gordon MN, Morgan

D, Nash K (2010) Convection-enhanced delivery and systemic mannitol increase gene

product distribution of AAV vectors 5, 8, and 9 and increase gene product in the adult

mouse brain. Journal of neuroscience methods 194:144-153.

Castle MJ, Perlson E, Holzbaur EL, Wolfe JH (2014) Long-distance axonal transport of AAV9

is driven by dynein and kinesin-2 and is trafficked in a highly motile Rab7-positive

compartment. Mol Ther 22:554-566.

Castren E, Elgersma Y, Maffei L, Hagerman R (2012) Treatment of neurodevelopmental

disorders in adulthood. The Journal of neuroscience : the official journal of the Society

for Neuroscience 32:14074-14079.

Cederfjäll E, Sahin G, Kirik D, Björklund T (2012) Design of a Single AAV Vector for

Coexpression of TH and GCH1 to Establish Continuous DOPA Synthesis in a Rat

Model of Parkinson's Disease. Molecular therapy : the journal of the American Society

of Gene Therapy 20:1315-1326.

Ceman S, O'Donnell WT, Reed M, Patton S, Pohl J, Warren ST (2003) Phosphorylation

influences the translation state of FMRP-associated polyribosomes. Human molecular

genetics 12:3295-3305.

Chakrabarty P, Ceballos-Diaz C, Beccard A, Janus C, Dickson D, Golde TE, Das P (2010) IFN-

gamma promotes complement expression and attenuates amyloid plaque deposition in

amyloid beta precursor protein transgenic mice. Journal of immunology 184:5333-5343.

Page 125: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

114

Chakrabarty P, Rosario A, Cruz P, Siemienski Z, Ceballos-Diaz C, Crosby K, Jansen K,

Borchelt DR, Kim JY, Jankowsky JL, Golde TE, Levites Y (2013) Capsid serotype and

timing of injection determines AAV transduction in the neonatal mice brain. PloS one

8:e67680.

Chandler RJ, LaFave MC, Varshney GK, Trivedi NS, Carrillo-Carrasco N, Senac JS, Wu W,

Hoffmann V, Elkahloun AG, Burgess SM, Venditti CP (2015) Vector design influences

hepatic genotoxicity after adeno-associated virus gene therapy. J Clin Invest 125:870-

880.

Chen L, Toth M (2001) Fragile X mice develop sensory hyperreactivity to auditory stimuli.

Neuroscience 103:1043-1050.

Cheng C, Sourial M, Doering L (2012) Astrocytes and developmental plasticity in fragile X.

Neural plasticity 2012:197491.

Cheng C, Zhu L, Doering LC (2014) Astrocyte-secreted TSP-1 corrects synaptic and spine

phenotypes in the fragile X mouse model. Poster presented at 14th International Fragile

X Conference, Orange County, CA.

Choi CH, Schoenfeld BP, Bell AJ, Hinchey P, Kollaros M, Gertner MJ, Woo NH, Tranfaglia

MR, Bear MF, Zukin RS, McDonald TV, Jongens TA, McBride SM (2011)

Pharmacological reversal of synaptic plasticity deficits in the mouse model of fragile X

syndrome by group II mGluR antagonist or lithium treatment. Brain research 1380:106-

119.

Colak D, Zaninovic N, Cohen MS, Rosenwaks Z, Yang WY, Gerhardt J, Disney MD, Jaffrey

SR (2014) Promoter-bound trinucleotide repeat mRNA drives epigenetic silencing in

fragile X syndrome. Science 343:1002-1005.

Collins A, Levenson J, Vilaythong A, Richman R, Armstrong D, Noebels J, David Sweatt J,

Zoghbi H (2004) Mild overexpression of MeCP2 causes a progressive neurological

disorder in mice. Human molecular genetics 13:2679-2689.

Cruz-Martín A, Crespo M, Portera-Cailliau C (2010) Delayed stabilization of dendritic spines in

fragile X mice. The Journal of neuroscience : the official journal of the Society for

Neuroscience 30:7793-7803.

Crystal RG, Sondhi D, Hackett NR, Kaminsky SM, Worgall S, Stieg P, Souweidane M, Hosain

S, Heier L, Ballon D, Dinner M, Wisniewski K, Kaplitt M, Greenwald BM, Howell JD,

Strybing K, Dyke J, Voss H (2004) Clinical protocol. Administration of a replication-

Page 126: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

115

deficient adeno-associated virus gene transfer vector expressing the human CLN2 cDNA

to the brain of children with late infantile neuronal ceroid lipofuscinosis. Hum Gene

Ther 15:1131-1154.

Cunningham J, Pivirotto P, Bringas J, Suzuki B, Vijay S, Sanftner L, Kitamura M, Chan C,

Bankiewicz KS (2008) Biodistribution of adeno-associated virus type-2 in nonhuman

primates after convection-enhanced delivery to brain. Mol Ther 16:1267-1275.

Daily J, Nash K, Jinwal U, Golde T, Rogers J, Peters M, Burdine R, Dickey C, Banko J, Weeber

E (2011) Adeno-associated virus-mediated rescue of the cognitive defects in a mouse

model for Angelman syndrome. PloS one 6.

Darnell J, Van Driesche S, Zhang C, Hung K, Mele A, Fraser C, Stone E, Chen C, Fak J, Chi S,

Licatalosi D, Richter J, Darnell R (2011) FMRP stalls ribosomal translocation on

mRNAs linked to synaptic function and autism. Cell 146:247-261.

Darnell JC, Fraser CE, Mostovetsky O, Stefani G, Jones TA, Eddy SR, Darnell RB (2005)

Kissing complex RNAs mediate interaction between the Fragile-X mental retardation

protein KH2 domain and brain polyribosomes. Genes & development 19:903-918.

Denman RB, Sung YJ (2002) Species-specific and isoform-specific RNA binding of human and

mouse fragile X mental retardation proteins. Biochemical and biophysical research

communications 292:1063-1069.

Devys D, Lutz Y, Rouyer N, Bellocq JP, Mandel JL (1993) The FMR-1 protein is cytoplasmic,

most abundant in neurons and appears normal in carriers of a fragile X premutation.

Nature genetics 4:335-340.

Di Pasquale G, Chiorini JA (2006) AAV transcytosis through barrier epithelia and endothelium.

Mol Ther 13:506-516.

Dirren E, Towne CL, Setola V, Redmond DE, Jr., Schneider BL, Aebischer P (2014)

Intracerebroventricular injection of adeno-associated virus 6 and 9 vectors for cell type-

specific transgene expression in the spinal cord. Hum Gene Ther 25:109-120.

Dirren E, Aebischer J, Rochat C, Towne C, Schneider BL, Aebischer P (2015) SOD1 silencing

in motoneurons or glia rescues neuromuscular function in ALS mice. Ann Clin Transl

Neurol 2:167-184.

Dolan BM, Duron SG, Campbell DA, Vollrath B, Shankaranarayana Rao BS, Ko HY, Lin GG,

Govindarajan A, Choi SY, Tonegawa S (2013) Rescue of fragile X syndrome

phenotypes in Fmr1 KO mice by the small-molecule PAK inhibitor FRAX486.

Page 127: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

116

Proceedings of the National Academy of Sciences of the United States of America

110:5671-5676.

Dolen G, Osterweil E, Rao BS, Smith GB, Auerbach BD, Chattarji S, Bear MF (2007)

Correction of fragile X syndrome in mice. Neuron 56:955-962.

Duan D, Yue Y, Engelhardt JF (2001) Expanding AAV packaging capacity with trans-splicing

or overlapping vectors: a quantitative comparison. Mol Ther 4:383-391.

Duque S, Joussemet B, Riviere C, Marais T, Dubreil L, Douar AM, Fyfe J, Moullier P, Colle

MA, Barkats M (2009) Intravenous administration of self-complementary AAV9 enables

transgene delivery to adult motor neurons. Mol Ther 17:1187-1196.

Dury AY, El Fatimy R, Tremblay S, Rose TM, Cote J, De Koninck P, Khandjian EW (2013)

Nuclear Fragile X Mental Retardation Protein is localized to Cajal bodies. PLoS Genet

9:e1003890.

Eberhart DE, Malter HE, Feng Y, Warren ST (1996) The fragile X mental retardation protein is

a ribonucleoprotein containing both nuclear localization and nuclear export signals.

Human molecular genetics 5:1083-1091.

Eichler EE, Richards S, Gibbs RA, Nelson DL (1993) Fine structure of the human FMR1 gene.

Human molecular genetics 2:1147-1153.

Ek CJ, Dziegielewska KM, Habgood MD, Saunders NR (2012) Barriers in the developing brain

and Neurotoxicology. Neurotoxicology 33:586-604.

Ellegood J, Pacey L, Hampson D, Lerch J, Henkelman R (2010) Anatomical phenotyping in a

mouse model of fragile X syndrome with magnetic resonance imaging. NeuroImage

53:1023-1029.

Fatemi SH, Folsom TD (2011) The role of fragile X mental retardation protein in major mental

disorders. Neuropharmacology 60:1221-1226.

Fatemi SH et al. (2012) Consensus paper: pathological role of the cerebellum in autism.

Cerebellum 11:777-807.

Federici T, Taub JS, Baum GR, Gray SJ, Grieger JC, Matthews KA, Handy CR, Passini MA,

Samulski RJ, Boulis NM (2012) Robust spinal motor neuron transduction following

intrathecal delivery of AAV9 in pigs. Gene therapy 19:852-859.

Feng Y, Gutekunst CA, Eberhart DE, Yi H, Warren ST, Hersch SM (1997) Fragile X mental

retardation protein: nucleocytoplasmic shuttling and association with somatodendritic

Page 128: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

117

ribosomes. The Journal of neuroscience : the official journal of the Society for

Neuroscience 17:1539-1547.

Ferrari FK, Samulski T, Shenk T, Samulski RJ (1996) Second-strand synthesis is a rate-limiting

step for efficient transduction by recombinant adeno-associated virus vectors. J Virol

70:3227-3234.

Fieremans N, Bauters M, Belet S, Verbeeck J, Jansen AC, Seneca S, Roelens F, De Baere E,

Marynen P, Froyen G (2014) De novo MECP2 duplications in two females with

intellectual disability and unfavorable complete skewed X-inactivation. Human genetics

133:1359-1367.

Foust K, Nurre E, Montgomery C, Hernandez A, Chan C, Kaspar B (2009) Intravascular AAV9

preferentially targets neonatal neurons and adult astrocytes. Nature biotechnology 27:59-

65.

Foust KD, Wang X, McGovern VL, Braun L, Bevan AK, Haidet AM, Le TT, Morales PR, Rich

MM, Burghes AH, Kaspar BK (2010) Rescue of the spinal muscular atrophy phenotype

in a mouse model by early postnatal delivery of SMN. Nature biotechnology 28:271-

274.

Frankland PW, Wang Y, Rosner B, Shimizu T, Balleine BW, Dykens EM, Ornitz EM, Silva AJ

(2004) Sensorimotor gating abnormalities in young males with fragile X syndrome and

Fmr1-knockout mice. Mol Psychiatry 9:417-425.

Fridell RA, Benson RE, Hua J, Bogerd HP, Cullen BR (1996) A nuclear role for the Fragile X

mental retardation protein. EMBO J 15:5408-5414.

Fu H, Kang L, Jennings JS, Moy SS, Perez A, Dirosario J, McCarty DM, Muenzer J (2007)

Significantly increased lifespan and improved behavioral performances by rAAV gene

delivery in adult mucopolysaccharidosis IIIB mice. Gene therapy 14:1065-1077.

Gabel LA, Won S, Kawai H, McKinney M, Tartakoff AM, Fallon JR (2004) Visual experience

regulates transient expression and dendritic localization of fragile X mental retardation

protein. The Journal of neuroscience : the official journal of the Society for

Neuroscience 24:10579-10583.

Gadalla K, Bailey M, Spike R, Ross P, Woodard K, Kalburgi S, Bachaboina L, Deng J, West A,

Samulski R, Gray S, Cobb S (2012) Improved Survival and Reduced Phenotypic

Severity Following AAV9/MECP2 Gene Transfer to Neonatal and Juvenile Male Mecp2

Page 129: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

118

Knockout Mice. Molecular therapy : the journal of the American Society of Gene

Therapy.

Garcia-Cairasco N (2002) A critical review on the participation of inferior colliculus in

acoustic-motor and acoustic-limbic networks involved in the expression of acute and

kindled audiogenic seizures. Hear Res 168:208-222.

Garg S, Lioy D, Cheval H, McGann J, Bissonnette J, Murtha M, Foust K, Kaspar B, Bird A,

Mandel G (2013) Systemic Delivery of MeCP2 Rescues Behavioral and Cellular

Deficits in Female Mouse Models of Rett Syndrome. The Journal of neuroscience : the

official journal of the Society for Neuroscience 33:13612-13620.

Gatto CL, Broadie K (2009) Temporal requirements of the fragile x mental retardation protein

in modulating circadian clock circuit synaptic architecture. Front Neural Circuits 3:8.

Gelfand Y, Kaplitt MG (2013) Gene therapy for psychiatric disorders. World neurosurgery

80:S32 e11-38.

Ghodsi A, Stein C, Derksen T, Martins I, Anderson RD, Davidson BL (1999) Systemic

hyperosmolality improves beta-glucuronidase distribution and pathology in murine MPS

VII brain following intraventricular gene transfer. Exp Neurol 160:109-116.

Gholizadeh S, Halder SK, Hampson DR (2015) Expression of fragile X mental retardation

protein in neurons and glia of the developing and adult mouse brain. Brain research

1596:22-30.

Gholizadeh S, Tharmalingam S, Macaldaz ME, Hampson DR (2013) Transduction of the central

nervous system after intracerebroventricular injection of adeno-associated viral vectors

in neonatal and juvenile mice. Human gene therapy methods 24:205-213.

Gholizadeh S, Arsenault J, Xuan IC, Pacey LK, Hampson DR (2014) Reduced phenotypic

severity following adeno-associated virus-mediated FMR1 gene delivery in fragile x

mice. Neuropsychopharmacology : official publication of the American College of

Neuropsychopharmacology 39:3100-3111.

Giampetruzzi A, Carson JH, Barbarese E (2013) FMRP and myelin protein expression in

oligodendrocytes. Molecular and cellular neurosciences 56:333-341.

Gkogkas CG, Khoutorsky A, Cao R, Jafarnejad SM, Prager-Khoutorsky M, Giannakas N,

Kaminari A, Fragkouli A, Nader K, Price TJ, Konicek BW, Graff JR, Tzinia AK,

Lacaille JC, Sonenberg N (2014) Pharmacogenetic inhibition of eIF4E-dependent Mmp9

Page 130: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

119

mRNA translation reverses fragile X syndrome-like phenotypes. Cell reports 9:1742-

1755.

Gong Y, Mu D, Prabhakar S, Moser A, Musolino P, Ren J, Breakefield XO, Maguire CA,

Eichler FS (2015) Adenoassociated virus serotype 9-mediated gene therapy for x-linked

adrenoleukodystrophy. Mol Ther 23:824-834.

Gray S, Foti S, Schwartz J, Bachaboina L, Taylor-Blake B, Coleman J, Ehlers M, Zylka M,

McCown T, Samulski R (2011) Optimizing promoters for recombinant adeno-associated

virus-mediated gene expression in the peripheral and central nervous system using self-

complementary vectors. Human gene therapy 22:1143-1153.

Gray SJ, Nagabhushan Kalburgi S, McCown TJ, Jude Samulski R (2013) Global CNS gene

delivery and evasion of anti-AAV-neutralizing antibodies by intrathecal AAV

administration in non-human primates. Gene therapy 20:450-459.

Gray SJ, Blake BL, Criswell HE, Nicolson SC, Samulski RJ, McCown TJ, Li W (2010)

Directed evolution of a novel adeno-associated virus (AAV) vector that crosses the

seizure-compromised blood-brain barrier (BBB). Mol Ther 18:570-578.

Greco CM, Navarro CS, Hunsaker MR, Maezawa I, Shuler JF, Tassone F, Delany M, Au JW,

Berman RF, Jin LW, Schumann C, Hagerman PJ, Hagerman RJ (2011) Neuropathologic

features in the hippocampus and cerebellum of three older men with fragile X syndrome.

Mol Autism 2:2.

Guo W, Allan A, Zong R, Zhang L, Johnson E, Schaller E, Murthy A, Goggin S, Eisch A,

Oostra B, Nelson D, Jin P, Zhao X (2011) Ablation of Fmrp in adult neural stem cells

disrupts hippocampus-dependent learning. Nature medicine 17:559-565.

Hachem S, Aguirre A, Vives V, Marks A, Gallo V, Legraverend C (2005) Spatial and temporal

expression of S100B in cells of oligodendrocyte lineage. Glia 51:81-97.

Hadaczek P, Kohutnicka M, Krauze MT, Bringas J, Pivirotto P, Cunningham J, Bankiewicz K

(2006) Convection-enhanced delivery of adeno-associated virus type 2 (AAV2) into the

striatum and transport of AAV2 within monkey brain. Hum Gene Ther 17:291-302.

Hampson D, Gholizadeh S, Pacey LK (2012) Pathways to drug development for autism

spectrum disorders. Clinical pharmacology and therapeutics 91:189-200.

Harlow EG, Till SM, Russell TA, Wijetunge LS, Kind P, Contractor A (2010) Critical period

plasticity is disrupted in the barrel cortex of FMR1 knockout mice. Neuron 65:385-398.

Page 131: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

120

Haurigot V, Marco S, Ribera A, Garcia M, Ruzo A, Villacampa P, Ayuso E, Anor S, Andaluz

A, Pineda M, Garcia-Fructuoso G, Molas M, Maggioni L, Munoz S, Motas S, Ruberte J,

Mingozzi F, Pumarola M, Bosch F (2013) Whole body correction of

mucopolysaccharidosis IIIA by intracerebrospinal fluid gene therapy. J Clin Invest.

Hessl D, Berry-Kravis E, Cordeiro L, Yuhas J, Ornitz EM, Campbell A, Chruscinski E, Hervey

C, Long JM, Hagerman RJ (2009) Prepulse inhibition in fragile X syndrome: feasibility,

reliability, and implications for treatment. American journal of medical genetics Part B,

Neuropsychiatric genetics : the official publication of the International Society of

Psychiatric Genetics 150B:545-553.

Heulens I, D'Hulst C, Van Dam D, De Deyn PP, Kooy RF (2012) Pharmacological treatment of

fragile X syndrome with GABAergic drugs in a knockout mouse model. Behav Brain

Res 229:244-249.

Heulens I, Suttie M, Postnov A, De Clerck N, Perrotta CS, Mattina T, Faravelli F, Forzano F,

Kooy RF, Hammond P (2013) Craniofacial characteristics of fragile X syndrome in

mouse and man. Eur J Hum Genet 21:816-823.

Hickey SE, Walters-Sen L, Mosher TM, Pfau RB, Pyatt R, Snyder PJ, Sotos JF, Prior TW

(2013) Duplication of the Xq27.3-q28 region, including the FMR1 gene, in an X-linked

hypogonadism, gynecomastia, intellectual disability, short stature, and obesity

syndrome. Am J Med Genet A 161A:2294-2299.

Hinderer C, Bell P, Louboutin JP, Zhu Y, Yu H, Lin G, Choa R, Gurda BL, Bagel J, O'Donnell

P, Sikora T, Ruane T, Wang P, Tarantal AF, Casal ML, Haskins ME, Wilson JM (2015)

Neonatal Systemic AAV Induces Tolerance to CNS Gene Therapy in MPS I Dogs and

Nonhuman Primates. Mol Ther 23:1298-1307.

Hinds HL, Ashley CT, Sutcliffe JS, Nelson DL, Warren ST, Housman DE, Schalling M (1993)

Tissue specific expression of FMR-1 provides evidence for a functional role in fragile X

syndrome. Nature genetics 3:36-43.

Hirai T, Enomoto M, Kaburagi H, Sotome S, Yoshida-Tanaka K, Ukegawa M, Kuwahara H,

Yamamoto M, Tajiri M, Miyata H, Hirai Y, Tominaga M, Shinomiya K, Mizusawa H,

Okawa A, Yokota T (2014) Intrathecal AAV serotype 9-mediated delivery of shRNA

against TRPV1 attenuates thermal hyperalgesia in a mouse model of peripheral nerve

injury. Mol Ther 22:409-419.

Page 132: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

121

Hordeaux J, Dubreil L, Deniaud J, Iacobelli F, Moreau S, Ledevin M, Le Guiner C, Blouin V,

Le Duff J, Mendes-Madeira A, Rolling F, Cherel Y, Moullier P, Colle MA (2015)

Efficient central nervous system AAVrh10-mediated intrathecal gene transfer in adult

and neonate rats. Gene therapy 22:316-324.

Hou L, Antion MD, Hu D, Spencer CM, Paylor R, Klann E (2006) Dynamic translational and

proteasomal regulation of fragile X mental retardation protein controls mGluR-

dependent long-term depression. Neuron 51:441-454.

Huang LY, Halder S, Agbandje-McKenna M (2014) Parvovirus glycan interactions. Current

opinion in virology 7:108-118.

Huang T, Li L, Shen Y, Qin X, Pang Z, Wu G (1996) Alternative splicing of the FMR1 gene in

human fetal brain neurons. American journal of medical genetics 64:252-255.

Huber K, Gallagher S, Warren S, Bear M (2002) Altered synaptic plasticity in a mouse model of

fragile X mental retardation. Proceedings of the National Academy of Sciences of the

United States of America 99:7746-7750.

Huda F, Konno A, Matsuzaki Y, Goenawan H, Miyake K, Shimada T, Hirai H (2014) Distinct

transduction profiles in the CNS via three injection routes of AAV9 and the application

to generation of a neurodegenerative mouse model. Mol Ther Methods Clin Dev

1:14032.

Ide S, Sora I, Ikeda K, Minami M, Uhl GR, Ishihara K (2010) Reduced emotional and

corticosterone responses to stress in mu-opioid receptor knockout mice.

Neuropharmacology 58:241-247.

Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, Deane R, Nedergaard M

(2013) Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in

the murine brain. The Journal of neuroscience : the official journal of the Society for

Neuroscience 33:18190-18199.

Jacobs S, Doering LC (2010) Astrocytes prevent abnormal neuronal development in the fragile

x mouse. The Journal of neuroscience : the official journal of the Society for

Neuroscience 30:4508-4514.

Jacobs S, Cheng C, Doering LC (2012) Probing astrocyte function in fragile X syndrome.

Results Probl Cell Differ 54:15-31.

Jones EV, Bouvier DS (2014) Astrocyte-secreted matricellular proteins in CNS remodelling

during development and disease. Neural Plast 2014:321209.

Page 133: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

122

Kanellopoulos AK, Semelidou O, Kotini AG, Anezaki M, Skoulakis EM (2012) Learning and

memory deficits consequent to reduction of the fragile X mental retardation protein

result from metabotropic glutamate receptor-mediated inhibition of cAMP signaling in

Drosophila. The Journal of neuroscience : the official journal of the Society for

Neuroscience 32:13111-13124.

Kantor B, McCown T, Leone P, Gray SJ (2014) Clinical applications involving CNS gene

transfer. Advances in genetics 87:71-124.

Kazdoba TM, Leach PT, Silverman JL, Crawley JN (2014) Modeling fragile X syndrome in the

Fmr1 knockout mouse. Intractable & rare diseases research 3:118-133.

Kelley K, Chang SJ, Lin SL (2012) Mechanism of repeat-associated microRNAs in fragile X

syndrome. Neural Plast 2012:104796.

Kim H, Gibboni R, Kirkhart C, Bao S (2013a) Impaired critical period plasticity in primary

auditory cortex of fragile X model mice. The Journal of neuroscience : the official

journal of the Society for Neuroscience 33:15686-15692.

Kim J-Y, Ash R, Ceballos-Diaz C, Levites Y, Golde T, Smirnakis S, Jankowsky J (2013b) Viral

transduction of the neonatal brain delivers controllable genetic mosaicism for visualising

and manipulating neuronal circuits in vivo. The European journal of neuroscience

37:1203-1220.

Kim JY, Grunke SD, Levites Y, Golde TE, Jankowsky JL (2014) Intracerebroventricular viral

injection of the neonatal mouse brain for persistent and widespread neuronal

transduction. Journal of visualized experiments : JoVE:51863.

Kim M, Bellini M, Ceman S (2009) Fragile X mental retardation protein FMRP binds mRNAs

in the nucleus. Molecular and cellular biology 29:214-228.

Koekkoek S et al. (2005) Deletion of FMR1 in Purkinje cells enhances parallel fiber LTD,

enlarges spines, and attenuates cerebellar eyelid conditioning in Fragile X syndrome.

Neuron 47:339-352.

Kohl S, Heekeren K, Klosterkotter J, Kuhn J (2013) Prepulse inhibition in psychiatric disorders-

-apart from schizophrenia. J Psychiatr Res 47:445-452.

Krueger DD, Osterweil EK, Chen SP, Tye LD, Bear MF (2011) Cognitive dysfunction and

prefrontal synaptic abnormalities in a mouse model of fragile X syndrome. Proceedings

of the National Academy of Sciences of the United States of America 108:2587-2592.

Page 134: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

123

LaFauci G, Adayev T, Kascsak R, Nolin S, Mehta P, Brown WT, Dobkin C (2013) Fragile X

screening by quantification of FMRP in dried blood spots by a Luminex immunoassay.

The Journal of molecular diagnostics : JMD 15:508-517.

Langen M, Bos D, Noordermeer SD, Nederveen H, van Engeland H, Durston S (2013) Changes

in the Development of Striatum Are Involved in Repetitive Behavior in Autism.

Biological psychiatry.

Lee Y, Messing A, Su M, Brenner M (2008) GFAP promoter elements required for region-

specific and astrocyte-specific expression. Glia 56:481-493.

Lentz TB, Gray SJ, Samulski RJ (2012) Viral vectors for gene delivery to the central nervous

system. Neurobiology of disease 48:179-188.

Leone P, Janson CG, Bilaniuk L, Wang Z, Sorgi F, Huang L, Matalon R, Kaul R, Zeng Z,

Freese A, McPhee SW, Mee E, During MJ (2000) Aspartoacylase gene transfer to the

mammalian central nervous system with therapeutic implications for Canavan disease.

Ann Neurol 48:27-38.

Leone P, Shera D, McPhee SW, Francis JS, Kolodny EH, Bilaniuk LT, Wang DJ, Assadi M,

Goldfarb O, Goldman HW, Freese A, Young D, During MJ, Samulski RJ, Janson CG

(2012) Long-term follow-up after gene therapy for canavan disease. Sci Transl Med

4:165ra163.

Levites Y, Jansen K, Smithson LA, Dakin R, Holloway VM, Das P, Golde TE (2006)

Intracranial adeno-associated virus-mediated delivery of anti-pan amyloid beta, amyloid

beta40, and amyloid beta42 single-chain variable fragments attenuates plaque pathology

in amyloid precursor protein mice. The Journal of neuroscience : the official journal of

the Society for Neuroscience 26:11923-11928.

LeWitt PA et al. (2011) AAV2-GAD gene therapy for advanced Parkinson's disease: a double-

blind, sham-surgery controlled, randomised trial. Lancet Neurol 10:309-319.

Liddelow SA (2011) Fluids and barriers of the CNS: a historical viewpoint. Fluids Barriers CNS

8:2.

Lim DA, Alvarez-Buylla A (2014) Adult neural stem cells stake their ground. Trends Neurosci

37:563-571.

Liu G, Martins I, Wemmie JA, Chiorini JA, Davidson BL (2005) Functional correction of CNS

phenotypes in a lysosomal storage disease model using adeno-associated virus type 4

Page 135: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

124

vectors. The Journal of neuroscience : the official journal of the Society for

Neuroscience 25:9321-9327.

Liu ZH, Chuang DM, Smith CB (2011) Lithium ameliorates phenotypic deficits in a mouse

model of fragile X syndrome. Int J Neuropsychopharmacol 14:618-630.

Loeb JE, Cordier WS, Harris ME, Weitzman MD, Hope TJ (1999) Enhanced expression of

transgenes from adeno-associated virus vectors with the woodchuck hepatitis virus

posttranscriptional regulatory element: implications for gene therapy. Hum Gene Ther

10:2295-2305.

Lowenstein PR (2009) Crossing the rubicon. Nature biotechnology 27:42-44.

Lu R, Wang H, Liang Z, Ku L, O'Donnell W T, Li W, Warren ST, Feng Y (2004) The fragile X

protein controls microtubule-associated protein 1B translation and microtubule stability

in brain neuron development. Proceedings of the National Academy of Sciences of the

United States of America 101:15201-15206.

Ludwig AL, Espinal GM, Pretto DI, Jamal AL, Arque G, Tassone F, Berman RF, Hagerman PJ

(2014) CNS expression of murine fragile X protein (FMRP) as a function of CGG-repeat

size. Human molecular genetics.

Luo Y, Shan G, Guo W, Smrt R, Johnson E, Li X, Pfeiffer R, Szulwach K, Duan R, Barkho B,

Li W, Liu C, Jin P, Zhao X (2010) Fragile x mental retardation protein regulates

proliferation and differentiation of adult neural stem/progenitor cells. PLoS genetics 6.

McCarty DM, Monahan PE, Samulski RJ (2001) Self-complementary recombinant adeno-

associated virus (scAAV) vectors promote efficient transduction independently of DNA

synthesis. Gene therapy 8:1248-1254.

McCarty DM, Young SM, Jr., Samulski RJ (2004) Integration of adeno-associated virus (AAV)

and recombinant AAV vectors. Annual review of genetics 38:819-845.

McLean JR, Smith GA, Rocha EM, Hayes MA, Beagan JA, Hallett PJ, Isacson O (2014)

Widespread neuron-specific transgene expression in brain and spinal cord following

synapsin promoter-driven AAV9 neonatal intracerebroventricular injection.

Neuroscience letters 576:73-78.

McPhee SW, Janson CG, Li C, Samulski RJ, Camp AS, Francis J, Shera D, Lioutermann L,

Feely M, Freese A, Leone P (2006) Immune responses to AAV in a phase I study for

Canavan disease. J Gene Med 8:577-588.

Page 136: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

125

Michalon A, Bruns A, Risterucci C, Honer M, Ballard TM, Ozmen L, Jaeschke G, Wettstein JG,

von Kienlin M, Kunnecke B, Lindemann L (2014) Chronic metabotropic glutamate

receptor 5 inhibition corrects local alterations of brain activity and improves cognitive

performance in fragile x mice. Biological psychiatry 75:189-197.

Min WW, Yuskaitis CJ, Yan Q, Sikorski C, Chen S, Jope RS, Bauchwitz RP (2009) Elevated

glycogen synthase kinase-3 activity in Fragile X mice: key metabolic regulator with

evidence for treatment potential. Neuropharmacology 56:463-472.

Mingozzi F, Maus MV, Hui DJ, Sabatino DE, Murphy SL, Rasko JE, Ragni MV, Manno CS,

Sommer J, Jiang H, Pierce GF, Ertl HC, High KA (2007) CD8(+) T-cell responses to

adeno-associated virus capsid in humans. Nature medicine 13:419-422.

Mittermeyer G, Christine CW, Rosenbluth KH, Baker SL, Starr P, Larson P, Kaplan PL,

Forsayeth J, Aminoff MJ, Bankiewicz KS (2012) Long-term evaluation of a phase 1

study of AADC gene therapy for Parkinson's disease. Hum Gene Ther 23:377-381.

Miyake N, Miyake K, Yamamoto M, Hirai Y, Shimada T (2011) Global gene transfer into the

CNS across the BBB after neonatal systemic delivery of single-stranded AAV vectors.

Brain research 1389:19-26.

Mullard A (2015) Fragile X disappointments upset autism ambitions. Nat Rev Drug Discov

14:151-153.

Murlidharan G, Samulski RJ, Asokan A (2014) Biology of adeno-associated viral vectors in the

central nervous system. Frontiers in molecular neuroscience 7:76.

Musumeci S, Bosco P, Calabrese G, Bakker… C (2000) Audiogenic seizures susceptibility in

transgenic mice with fragile X syndrome. Epilepsia.

Na ES, Nelson ED, Adachi M, Autry AE, Mahgoub MA, Kavalali ET, Monteggia LM (2012) A

mouse model for MeCP2 duplication syndrome: MeCP2 overexpression impairs learning

and memory and synaptic transmission. The Journal of neuroscience : the official journal

of the Society for Neuroscience 32:3109-3117.

Nagamani SC, Erez A, Probst FJ, Bader P, Evans P, Baker LA, Fang P, Bertin T, Hixson P,

Stankiewicz P, Nelson D, Patel A, Cheung SW (2012) Small genomic rearrangements

involving FMR1 support the importance of its gene dosage for normal neurocognitive

function. Neurogenetics 13:333-339.

Nakamoto M, Nalavadi V, Epstein MP, Narayanan U, Bassell GJ, Warren ST (2007) Fragile X

mental retardation protein deficiency leads to excessive mGluR5-dependent

Page 137: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

126

internalization of AMPA receptors. Proceedings of the National Academy of Sciences of

the United States of America 104:15537-15542.

Neuhofer D, Henstridge CM, Dudok B, Sepers M, Lassalle O, Katona I, Manzoni OJ (2015)

Functional and structural deficits at accumbens synapses in a mouse model of Fragile X.

Frontiers in cellular neuroscience 9:100.

Neuwirth LS, Volpe NP, Ng S, Marsillo A, Corwin C, Madan N, Ferraro AM, El Idrissi A

(2015) Taurine recovers mice emotional learning and memory disruptions associated

with fragile x syndrome in context fear and auditory cued-conditioning. Adv Exp Med

Biol 803:425-438.

Ng MC, Yang YL, Lu KT (2013) Behavioral and synaptic circuit features in a zebrafish model

of fragile X syndrome. PloS one 8:e51456.

Niemeyer GP, Herzog RW, Mount J, Arruda VR, Tillson DM, Hathcock J, van Ginkel FW,

High KA, Lothrop CD, Jr. (2009) Long-term correction of inhibitor-prone hemophilia B

dogs treated with liver-directed AAV2-mediated factor IX gene therapy. Blood 113:797-

806.

Nimchinsky E, Oberlander A, Svoboda K (2001) Abnormal development of dendritic spines in

FMR1 knock-out mice. The Journal of neuroscience : the official journal of the Society

for Neuroscience 21:5139-5146.

Nonnenmacher M, Weber T (2012) Intracellular transport of recombinant adeno-associated

virus vectors. Gene therapy 19:649-658.

Nosyreva ED, Huber KM (2006) Metabotropic receptor-dependent long-term depression

persists in the absence of protein synthesis in the mouse model of fragile X syndrome.

Journal of neurophysiology 95:3291-3295.

Ojala DS, Amara DP, Schaffer DV (2015) Adeno-associated virus vectors and neurological

gene therapy. Neuroscientist 21:84-98.

Osterweil EK, Krueger DD, Reinhold K, Bear MF (2010) Hypersensitivity to mGluR5 and

ERK1/2 leads to excessive protein synthesis in the hippocampus of a mouse model of

fragile X syndrome. The Journal of neuroscience : the official journal of the Society for

Neuroscience 30:15616-15627.

Ouwenga RL, Dougherty J (2015) Fmrp targets or not: long, highly brain-expressed genes tend

to be implicated in autism and brain disorders. Mol Autism 6:16.

Page 138: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

127

Pacey LK, Heximer S, Hampson D (2009) Increased GABA(B) receptor-mediated signaling

reduces the susceptibility of fragile X knockout mice to audiogenic seizures. Molecular

pharmacology 76:18-24.

Pacey LK, Doss L, Cifelli C, van der Kooy D, Heximer SP, Hampson DR (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 neurosciences

46:563-572.

Pacey LKK, Doering LC (2007) Developmental expression of FMRP in the astrocyte lineage:

implications for fragile X syndrome. Glia 55:1601-1609.

Pacey LKK, Xuan IC, Guan S, Sussman D, Henkelman RM, Chen Y, Thomsen C, Hampson

DR (2013) Delayed myelination in a mouse model of fragile X syndrome. Human

molecular genetics 22:3920-3930.

Pacey LKK, Guan A, Tharmalingham S, Thomsen C, Hampson DR (2015) Persistent Astrocyte

Activation in the Fragile X Mouse Cerebellum. Brain and Behavior (in press).

Paribello C, Tao L, Folino A, Berry-Kravis E, Tranfaglia M, Ethell IM, Ethell DW (2010)

Open-label add-on treatment trial of minocycline in fragile X syndrome. BMC neurology

10:91.

Passini M, Watson D, Vite C, Landsburg D, Feigenbaum A, Wolfe J (2003) Intraventricular

brain injection of adeno-associated virus type 1 (AAV1) in neonatal mice results in

complementary patterns of neuronal transduction to AAV2 and total long-term

correction of storage lesions in the brains of beta-glucuronidase-deficient mice. Journal

of virology 77:7034-7040.

Passini MA, Wolfe JH (2001) Widespread gene delivery and structure-specific patterns of

expression in the brain after intraventricular injections of neonatal mice with an adeno-

associated virus vector. Journal of virology 75:12382-12392.

Paxinos G, Franklin K (2013) The Mouse Brain in Stereotaxic Coordinates, 4th Edition. San

Diego.

Paylor R, Yuva-Paylor LA, Nelson DL, Spencer CM (2008) Reversal of sensorimotor gating

abnormalities in Fmr1 knockout mice carrying a human Fmr1 transgene. Behavioral

neuroscience 122:1371-1377.

Page 139: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

128

Peier AM, McIlwain KL, Kenneson A, Warren ST, Paylor R, Nelson DL (2000)

(Over)correction of FMR1 deficiency with YAC transgenics: behavioral and physical

features. Human molecular genetics 9:1145-1159.

Polito A, Reynolds R (2005) NG2-expressing cells as oligodendrocyte progenitors in the normal

and demyelinated adult central nervous system. Journal of anatomy 207:707-716.

Pop AS, Gomez-Mancilla B, Neri G, Willemsen R, Gasparini F (2014) Fragile X syndrome: a

preclinical review on metabotropic glutamate receptor 5 (mGluR5) antagonists and drug

development. Psychopharmacology (Berl) 231:1217-1226.

Qin M, Xia Z, Huang T, Smith CB (2011) Effects of chronic immobilization stress on anxiety-

like behavior and basolateral amygdala morphology in Fmr1 knockout mice.

Neuroscience 194:282-290.

Reardon W, Donoghue V, Murphy AM, King MD, Mayne PD, Horn N, Birk Moller L (2010)

Progressive cerebellar degenerative changes in the severe mental retardation syndrome

caused by duplication of MECP2 and adjacent loci on Xq28. European journal of

pediatrics 169:941-949.

Reeb-Sutherland BC, Fox NA (2015) Eyeblink conditioning: a non-invasive biomarker for

neurodevelopmental disorders. Journal of autism and developmental disorders 45:376-

394.

Rio M, Malan V, Boissel S, Toutain A, Royer G, Gobin S, Morichon-Delvallez N, Turleau C,

Bonnefont JP, Munnich A, Vekemans M, Colleaux L (2010) Familial interstitial

Xq27.3q28 duplication encompassing the FMR1 gene but not the MECP2 gene causes a

new syndromic mental retardation condition. Eur J Hum Genet 18:285-290.

Rivera VM, Gao GP, Grant RL, Schnell MA, Zoltick PW, Rozamus LW, Clackson T, Wilson

JM (2005) Long-term pharmacologically regulated expression of erythropoietin in

primates following AAV-mediated gene transfer. Blood 105:1424-1430.

Rogers TD, Dickson PE, McKimm E, Heck DH, Goldowitz D, Blaha CD, Mittleman G (2013)

Reorganization of circuits underlying cerebellar modulation of prefrontal cortical

dopamine in mouse models of autism spectrum disorder. Cerebellum 12:547-556.

Rothermel M, Brunert D, Zabawa C, Diaz-Quesada M, Wachowiak M (2013) Transgene

expression in target-defined neuron populations mediated by retrograde infection with

adeno-associated viral vectors. The Journal of neuroscience : the official journal of the

Society for Neuroscience 33:15195-15206.

Page 140: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

129

Rotschafer S, Trujillo M, Dansie L, Ethell I, Razak K (2012) Minocycline treatment reverses

ultrasonic vocalization production deficit in a mouse model of Fragile X Syndrome.

Brain research 1439:7-14.

Rotschafer SE, Razak KA (2014) Auditory processing in fragile x syndrome. Frontiers in

cellular neuroscience 8:19.

Saka E, Goodrich C, Harlan P, Madras BK, Graybiel AM (2004) Repetitive behaviors in

monkeys are linked to specific striatal activation patterns. The Journal of neuroscience :

the official journal of the Society for Neuroscience 24:7557-7565.

Samaranch L, Salegio EA, San Sebastian W, Kells AP, Foust KD, Bringas JR, Lamarre C,

Forsayeth J, Kaspar BK, Bankiewicz KS (2012) Adeno-associated virus serotype 9

transduction in the central nervous system of nonhuman primates. Hum Gene Ther

23:382-389.

Samaranch L, San Sebastian W, Kells AP, Salegio EA, Heller G, Bringas JR, Pivirotto P,

DeArmond S, Forsayeth J, Bankiewicz KS (2014) AAV9-mediated expression of a non-

self protein in nonhuman primate central nervous system triggers widespread

neuroinflammation driven by antigen-presenting cell transduction. Mol Ther 22:329-337.

Santoro MR, Bray SM, Warren ST (2012) Molecular mechanisms of fragile X syndrome: a

twenty-year perspective. Annual review of pathology 7:219-245.

Scharf SH, Jaeschke G, Wettstein JG, Lindemann L (2014) Metabotropic glutamate receptor 5

as drug target for Fragile X syndrome. Current opinion in pharmacology.

Shang Y, Wang H, Mercaldo V, Li X, Chen T, Zhuo M (2009) Fragile X mental retardation

protein is required for chemically-induced long-term potentiation of the hippocampus in

adult mice. J Neurochem 111:635-646.

Sidorov MS, Auerbach BD, Bear MF (2013) Fragile X mental retardation protein and synaptic

plasticity. Molecular brain 6:15.

Sittler A, Devys D, Weber C, Mandel JL (1996) Alternative splicing of exon 14 determines

nuclear or cytoplasmic localisation of fmr1 protein isoforms. Human molecular genetics

5:95-102.

Smyk M, Obersztyn E, Nowakowska B, Nawara M, Cheung SW, Mazurczak T, Stankiewicz P,

Bocian E (2008) Different-sized duplications of Xq28, including MECP2, in three males

with mental retardation, absent or delayed speech, and recurrent infections. American

Page 141: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

130

journal of medical genetics Part B, Neuropsychiatric genetics : the official publication of

the International Society of Psychiatric Genetics 147B:799-806.

Snyder BR, Gray SJ, Quach ET, Huang JW, Leung CH, Samulski RJ, Boulis NM, Federici T

(2011) Comparison of adeno-associated viral vector serotypes for spinal cord and motor

neuron gene delivery. Hum Gene Ther 22:1129-1135.

Spencer C, Alekseyenko O, Serysheva E, Yuva-Paylor L, Paylor R (2005) Altered anxiety-

related and social behaviors in the Fmr1 knockout mouse model of fragile X syndrome.

Genes, brain, and behavior 4:420-430.

Spencer C, Alekseyenko O, Hamilton S, Thomas A, Serysheva E, Yuva-Paylor L, Paylor R

(2011) Modifying behavioral phenotypes in Fmr1KO mice: genetic background

differences reveal autistic-like responses. Autism research : official journal of the

International Society for Autism Research 4:40-56.

Steiner J, Bernstein HG, Bogerts B, Gos T, Richter-Landsberg C, Wunderlich MT, Keilhoff G

(2008) S100B is expressed in, and released from, OLN-93 oligodendrocytes: Influence

of serum and glucose deprivation. Neuroscience 154:496-503.

Stieger K, Lheriteau E, Moullier P, Rolling F (2009) AAV-mediated gene therapy for retinal

disorders in large animal models. ILAR journal / National Research Council, Institute of

Laboratory Animal Resources 50:206-224.

Su T, Fan HX, Jiang T, Sun WW, Den WY, Gao MM, Chen SQ, Zhao QH, Yi YH (2011) Early

continuous inhibition of group 1 mGlu signaling partially rescues dendritic spine

abnormalities in the Fmr1 knockout mouse model for fragile X syndrome.

Psychopharmacology (Berl) 215:291-300.

Suvrathan A, Chattarji S (2011) Fragile X syndrome and the amygdala. Curr Opin Neurobiol

21:509-515.

Suvrathan A, Hoeffer CA, Wong H, Klann E, Chattarji S (2010) Characterization and reversal

of synaptic defects in the amygdala in a mouse model of fragile X syndrome.

Proceedings of the National Academy of Sciences of the United States of America

107:11591-11596.

Swerdlow NR, Shoemaker JM, Kuczenski R, Bongiovanni MJ, Neary AC, Tochen LS, Saint

Marie RL (2006) Forebrain D1 function and sensorimotor gating in rats: effects of D1

blockade, frontal lesions and dopamine denervation. Neuroscience letters 402:40-45.

Page 142: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

131

Tamanini F, Meijer N, Verheij C, Willems PJ, Galjaard H, Oostra BA, Hoogeveen AT (1996)

FMRP is associated to the ribosomes via RNA. Human molecular genetics 5:809-813.

Tassone F, De Rubeis S, Carosi C, La Fata G, Serpa G, Raske C, Willemsen R, Hagerman PJ,

Bagni C (2011) Differential usage of transcriptional start sites and polyadenylation sites

in FMR1 premutation alleles. Nucleic acids research 39:6172-6185.

Tessier CR, Broadie K (2008) Drosophila fragile X mental retardation protein developmentally

regulates activity-dependent axon pruning. Development 135:1547-1557.

Tetreault NA, Hakeem AY, Jiang S, Williams BA, Allman E, Wold BJ, Allman JM (2012)

Microglia in the cerebral cortex in autism. Journal of autism and developmental

disorders 42:2569-2584.

Thomas A, Bui N, Graham D, Perkins J, Yuva-Paylor L, Paylor R (2011) Genetic reduction of

group 1 metabotropic glutamate receptors alters select behaviors in a mouse model for

fragile X syndrome. Behavioural brain research 223:310-321.

Thomas AM, Bui N, Perkins JR, Yuva-Paylor LA, Paylor R (2012) Group I metabotropic

glutamate receptor antagonists alter select behaviors in a mouse model for fragile X

syndrome. Psychopharmacology (Berl) 219:47-58.

Thwaite R, Pages G, Chillon M, Bosch A (2015) AAVrh.10 immunogenicity in mice and

humans. Relevance of antibody cross-reactivity in human gene therapy. Gene therapy

22:196-201.

Tobia MJ, Woodruff-Pak DS (2009) Delay eyeblink classical conditioning is impaired in Fragile

X syndrome. Behavioral neuroscience 123:665-676.

Treleaven CM, Tamsett TJ, Bu J, Fidler JA, Sardi SP, Hurlbut GD, Woodworth LA, Cheng SH,

Passini MA, Shihabuddin LS, Dodge JC (2012) Gene transfer to the CNS is efficacious

in immune-primed mice harboring physiologically relevant titers of anti-AAV

antibodies. Mol Ther 20:1713-1723.

Vengoechea J, Parikh AS, Zhang S, Tassone F (2012) De novo microduplication of the FMR1

gene in a patient with developmental delay, epilepsy and hyperactivity. Eur J Hum Genet

20:1197-1200.

Wang F, Zhu J, Zhu H, Zhang Q, Lin Z, Hu H (2011) Bidirectional control of social hierarchy

by synaptic efficacy in medial prefrontal cortex. Science 334:693-697.

Page 143: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

132

Wang H, Liang S, Burgdorf J, Wess J, Yeomans J (2008) Ultrasonic vocalizations induced by

sex and amphetamine in M2, M4, M5 muscarinic and D2 dopamine receptor knockout

mice. PloS one 3:e1893.

Wang H, Ku L, Osterhout DJ, Li W, Ahmadian A, Liang Z, Feng Y (2004) Developmentally-

programmed FMRP expression in oligodendrocytes: a potential role of FMRP in

regulating translation in oligodendroglia progenitors. Human molecular genetics 13:79-

89.

Wang T, Bray S, Warren S (2012) New perspectives on the biology of fragile X syndrome.

Current opinion in genetics & development 22:256-263.

Weinberg MS, Samulski RJ, McCown TJ (2013) Adeno-associated virus (AAV) gene therapy

for neurological disease. Neuropharmacology 69:82-88.

Whitney ER, Kemper TL, Bauman ML, Rosene DL, Blatt GJ (2008) Cerebellar Purkinje cells

are reduced in a subpopulation of autistic brains: a stereological experiment using

calbindin-D28k. Cerebellum 7:406-416.

Wijetunge LS, Chattarji S, Wyllie DJ, Kind PC (2013) Fragile X syndrome: from targets to

treatments. Neuropharmacology 68:83-96.

Wither RG, Lang M, Zhang L, Eubanks JH (2013) Regional MeCP2 expression levels in the

female MeCP2-deficient mouse brain correlate with specific behavioral impairments.

Exp Neurol 239:49-59.

Worgall S, Sondhi D, Hackett NR, Kosofsky B, Kekatpure MV, Neyzi N, Dyke JP, Ballon D,

Heier L, Greenwald BM, Christos P, Mazumdar M, Souweidane MM, Kaplitt MG,

Crystal RG (2008) Treatment of late infantile neuronal ceroid lipofuscinosis by CNS

administration of a serotype 2 adeno-associated virus expressing CLN2 cDNA. Hum

Gene Ther 19:463-474.

Wrenn CC, Heitzer AM, Roth AK, Nawrocki L, Valdovinos MG (2015) Effects of clonidine

and methylphenidate on motor activity in Fmr1 knockout mice. Neuroscience letters

585:109-113.

Yamazaki Y, Hirai Y, Miyake K, Shimada T (2014) Targeted gene transfer into ependymal cells

through intraventricular injection of AAV1 vector and long-term enzyme replacement

via the CSF. Sci Rep 4:5506.

Page 144: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

133

Yan QJ, Rammal M, Tranfaglia M, Bauchwitz RP (2005) Suppression of two major Fragile X

Syndrome mouse model phenotypes by the mGluR5 antagonist MPEP.

Neuropharmacology 49:1053-1066.

Yang B, Li S, Wang H, Guo Y, Gessler DJ, Cao C, Su Q, Kramer J, Zhong L, Ahmed SS,

Zhang H, He R, Desrosiers RC, Brown R, Xu Z, Gao G (2014) Global CNS transduction

of adult mice by intravenously delivered rAAVrh.8 and rAAVrh.10 and nonhuman

primates by rAAVrh.10. Mol Ther 22:1299-1309.

Yang Q, Feng B, Zhang K, Guo YY, Liu SB, Wu YM, Li XQ, Zhao MG (2012) Excessive

astrocyte-derived neurotrophin-3 contributes to the abnormal neuronal dendritic

development in a mouse model of fragile X syndrome. PLoS Genet 8:e1003172.

Yuhas J, Cordeiro L, Tassone F, Ballinger E, Schneider A, Long JM, Ornitz EM, Hessl D

(2011) Brief report: Sensorimotor gating in idiopathic autism and autism associated with

fragile X syndrome. Journal of autism and developmental disorders 41:248-253.

Yuskaitis CJ, Jope RS (2009) Glycogen synthase kinase-3 regulates microglial migration,

inflammation, and inflammation-induced neurotoxicity. Cellular signalling 21:264-273.

Yuskaitis CJ, Beurel E, Jope RS (2010a) Evidence of reactive astrocytes but not peripheral

immune system activation in a mouse model of Fragile X syndrome. Biochim Biophys

Acta 1802:1006-1012.

Yuskaitis CJ, Mines MA, King MK, Sweatt JD, Miller CA, Jope RS (2010b) Lithium

ameliorates altered glycogen synthase kinase-3 and behavior in a mouse model of fragile

X syndrome. Biochem Pharmacol 79:632-646.

Zalfa F, Eleuteri B, Dickson KS, Mercaldo V, De Rubeis S, di Penta A, Tabolacci E, Chiurazzi

P, Neri G, Grant SG, Bagni C (2007) A new function for the fragile X mental retardation

protein in regulation of PSD-95 mRNA stability. Nature neuroscience 10:578-587.

Zeier Z, Kumar A, Bodhinathan K, Feller J, Foster T, Bloom D (2009) Fragile X mental

retardation protein replacement restores hippocampal synaptic function in a mouse

model of fragile X syndrome. Gene therapy 16:1122-1129.

Zhang H, Yang B, Mu X, Ahmed SS, Su Q, He R, Wang H, Mueller C, Sena-Esteves M, Brown

R, Xu Z, Gao G (2011) Several rAAV vectors efficiently cross the blood-brain barrier

and transduce neurons and astrocytes in the neonatal mouse central nervous system. Mol

Ther 19:1440-1448.

Page 145: Virus-Mediated Delivery of the Fmr1 Gene as a Tool for the Treatment of Fragile X Syndrome · 2016-07-12 · Treatment of Fragile X Syndrome Shervin Gholizadeh Moghaddam ... 2.4.5

134

Zhao MG, Toyoda H, Ko SW, Ding HK, Wu LJ, Zhuo M (2005) Deficits in trace fear memory

and long-term potentiation in a mouse model for fragile X syndrome. The Journal of

neuroscience : the official journal of the Society for Neuroscience 25:7385-7392.

Zito K, Svoboda K (2002) Activity-dependent synaptogenesis in the adult Mammalian cortex.

Neuron 35:1015-1017.