95
MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons by Wesley Lai A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Molecular Genetics University of Toronto © Copyright by Wesley Lai 2014

MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons

by

Wesley Lai

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

University of Toronto

© Copyright by Wesley Lai 2014

Page 2: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

ii

MECP2e1 Mutation Reduces the Dendritic Complexity of Rett

Syndrome Patient iPS Cell-Derived Neurons

Wesley Lai

Master of Science

Department of Molecular Genetics University of Toronto

2014

Abstract

The gene MECP2 that is commonly mutated in Rett Syndrome (RTT) can be alternatively

spliced in neurons into MeCP2e1 and MECP2e2 isoforms. Fibroblasts from a patient with a

MECP2e1 mutation were reprogrammed into induced pluripotent stem cells (iPS) and

differentiated into neurons with an EB-based directed differentiation protocol. I employed

methods to analyze neuronal complexity with respect to numbers of terminal dendrites, higher

order dendrites, intersections by Sholl analysis, and total dendrite length in RTTe1 compared to

wildtype neurons. My results show that neuronal complexity is reduced in the RTTe1 neurons,

indicating that the MECP2e1 isoform is essential for healthy neuronal morphology. Wildtype and

RTT iPS cells from a MECP2 null and R306C patient were also differentiated into neurons with

a one-step protocol by exogenous expression of Ngn2 in iPS cells. RTT neurons did not

recapitulate dendritic defects, suggesting that these phenotypes may be masked by Ngn2

overexpression.

Page 3: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

iii

Acknowledgments

Thank you, Dr. James Ellis, for being a superb supervisor who provided me with the resources

and platform for success. You assembled a phenomenal team of scientists who have taught me

much over the two years. I have come to appreciate the art of precise and clear writing, which is

something you continually emphasized, and I will not cease to hone this skill. Your assistance in

the completion of this thesis has not gone unnoticed and I am very grateful that you readily

provided honest thoughts, insights, and encouragements. Your honesty is a valuable trait and I

hope you will continue to exercise it with much wisdom as you supervise current and future

trainees. Peter Pasceri, your pleasant demeanor really helped me to integrate into the lab. Thank

you for your willingness to teach me new things from the realms of molecular biology and

biochemistry to that of engineering. Those mind-boggling life-hacks will not be forgotten. My

mentor and friend, Dr. Deivid Rodrigues, thank you for being a rock that grounded me

throughout my graduate studies. Your ability to provide uplifting encouragements, kind

hospitality, and truthful yet gentle critiques on my hypotheses, your willingness to teach me the

same principles repeatedly though with great patience, and your drive for excellence is a standard

I hope to embody. You have been a phenomenal role model and I hope other students will

benefit from you as much as I have. Dr. Dae-sung Kim, you too have been a great friend and

mentor over these years and I will not forget the numerous scientific discussions that we had on

our commutes after work. Alina Piekna, Wei Wei, and Zhanna Konovalova, the three technicians

in our lab, thank you for your hard work and patience. Each one of you should be recognized for

working tirelessly, being self-motivated, and dedicated to being on the front lines of scientific

inquiry alongside the post-doctoral fellows and graduate students in our lab. Dr. Joel Ross, the

sparse labeling technique was an essential technique used in my experiments and I am grateful

for your willingness to share it. Thank you also for facilitating some meaningful discussions

regarding my experiments at lab meeting. My committee members, Drs. Freda Miller, Sabine

Cordes, and Jim Eubanks, thank you for taking the time to participate in my supervisory

committee and to oversee my project. I was very fortunate to learn from the three of you. To the

Mau family, thank you for your kind hospitality. Thank you Emily Mau, for your support

through life. To my family, words cannot express the infinite support and love I have received

from you. I am fortunate to have nurturing parents, and caring brothers, all of whom have been

integral in the successful completion of this thesis.

Page 4: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

iv

Table of Contents

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

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

List  of  Tables  ........................................................................................................................................  vii  

List  of  Figures  .....................................................................................................................................  viii  

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

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

1.1   Rett  Syndrome  diagnosis  and  current  treatments  ....................................................................  1  1.2   The  genetic  etiology  of  RTT:  MECP2  mutations  .........................................................................  2  1.3   General  properties  of  MeCP2  and  its  functions  on  methylated  DNA  ..................................  3  1.4   MeCP2  isoforms,  properties,  and  domains  ..................................................................................  4  1.5   Cellular  phenotypes  in  RTT  post-­‐mortem  tissue  .......................................................................  6  1.6   The  most  common  RTT  mouse  models:  behavioural  deficits  and  cellular  phenotypes  

of  neurons  and  glia  .........................................................................................................................................  7  1.7   Rescue  of  disease  models  and  limitations  of  the  model  .......................................................  11  1.8   iPS  cells  in  studying  disease  ..........................................................................................................  12  1.9   Pertinent  neuronal  differentiation  techniques  and  subtypes  made  ...............................  13  1.10   Existing  RTT  iPS  and  ES  cell  models  and  their  limitations  ...............................................  23  1.11   Establishment  and  characterization  of  the  RTTe1  cell  lines  ...........................................  25  1.12   Thesis  Rationale  and  Hypothesis  ..............................................................................................  27  

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

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

2.1   Cell  Culture  ..........................................................................................................................................  28  2.2   Neuronal  Differentiation  ................................................................................................................  28  2.3   Immunocytochemical  (ICC)  Staining  ..........................................................................................  30  2.4   Neuronal  Morphometric  Analyses  ...............................................................................................  31  2.5   Western  Blot  .......................................................................................................................................  32  2.6   Statistics  ...............................................................................................................................................  33  

Chapter  3  Results  ...............................................................................................................................  34  

Page 5: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

v

3   Results  ............................................................................................................................................  34  

3.1   RTT  neurons  were  obtained  by  directing  the  differentiation  of  iPS  cells  into  neurons

  34  3.2   Establishing  neuronal  morphometric  analyses  to  quantify  neurite  complexity  .........  36  3.3   RTT  neurons  differentiated  for  two  weeks  in  vitro  have  reduced  neurite  complexity

  39  3.4   Experimental  set  up  for  neuronal  morphometric  assays  performed  on  neurons  

differentiated  for  6  weeks  in  vitro  ..........................................................................................................  42  3.5   RTTΔ3-­‐4#37  neurons  and  NPCs  obtained  from  my  differentiation  should  be  excluded

  46  3.6   RTTe1  neurons  differentiated  for  6  weeks  in  vitro  have  reduced  dendritic  complexity

  53  3.7   The  overexpression  of  Ngn2  for  induction  of  iPS  cells  into  neurons  may  mask  RTT  

phenotypes  .....................................................................................................................................................  57  

Chapter  4  Discussion  ........................................................................................................................  62  

4   Discussion  .....................................................................................................................................  62  

4.1   Potential  consequences  of  reduced  neuronal  dendritic  complexity  on  behaviour  and  

neuronal  function  .........................................................................................................................................  62  4.2   Targeting  Ras-­‐PI3K-­‐AKT,  Ras-­‐MAPK,  and  Rho-­‐family  GTPases  signaling  pathways  

may  reverse  defects  of  dendritic  complexity  ......................................................................................  63  4.3   Experimental  Limitations  ...............................................................................................................  64  4.3.1   A  method  to  equalize  cell  density  needs  to  be  established  to  account  for  potential  

density-­‐dependent  effects  on  dendritic  arborization  ...................................................................................  64  4.3.2   No  RTTe1  isogenic  wildtype  controls  were  isolated  from  reprogramming  .......................  65  4.3.3   RTTΔ3-­‐4#37  healthy  control  neurons  consistently  underperformed  ..................................  66  4.3.4   Loss  of  MeCP2e1  and  continued  expression  of  MeCP2e2  in  RTTe1  neurons  need  to  be  

confirmed  by  isoform  specific  antibodies  .........................................................................................................  66  4.3.5   EB-­‐based  neuronal  differentiation  protocol  lacks  heterogeneity  that  is  often  found  in  

the  brain  ...........................................................................................................................................................................  67  4.3.6   One-­‐step  induction  neurons  should  be  cocultured  with  both  wildtype  and  Mecp2-­‐

deficient  astrocytes  to  faithfully  model  disease  ..............................................................................................  68  4.3.7   Lack  of  experimental  replicates  for  morphology  assays  performed  on  neurons  obtained  

from  the  one-­‐step  induction  protocol  preclude  biological  conclusions  ...............................................  68  

Page 6: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

vi

Chapter  5  Future  Experiments  ......................................................................................................  70  

5   Future  Experiments  ...................................................................................................................  70  

5.1   Exploring  genetic  and  pharmacological  approaches  to  rescue  RTT  dendritic  

complexity  defects  .......................................................................................................................................  70  5.1.1   Background  and  Rationale  ........................................................................................................................  70  5.1.2   Rescuing  RTTe1  dendritic  defects  with  MeP-­‐MECP2e1  and  IGF-­‐1  .........................................  71  

5.2   Exploring  RTT  phenotypes  in  RTT  neurons  derived  by  the  overexpression  of  Ngn2  73  5.3   Conclusions  ..........................................................................................................................................  75  

References  ............................................................................................................................................  76  

Page 7: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

vii

List of Tables

Table 1. Summarized characterizations of RTT and healthy control cell lines.

Table 2. Summary of antibodies used in various experiments.

Table 3. Summary of cell lines included in experiment to determine if RTT neurons I derived

recapitulate the disease phenotype of reduced neurite complexity.

Table 4. Summary of cell lines used and assays performed on neurons differentiated for 6 weeks.

Table 5. Summary of assays conducted and their results suggesting that RTTΔ3-4#37 and its

isogenic RTTΔ3-4#20 null cell line should be excluded from dendritic complexity assays

performed on neurons differentiated for 6 weeks.

Table 6. Neurons obtained from the one-step induction protocol may be more complex than

neurons differentiated for 6 weeks with the directed differentiation protocol.

Table 7. Summary of proposed experimental conditions for rescue experiments.

Table 8. Summary of cell lines included in study to determine whether Ngn2 overexpression

masks RTT phenotypes.

Page 8: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

viii

List of Figures

Figure 1. Schematic of MECP2 gene with mutational hot spots indicated.

Figure 2. Splice map of MECP2e1 and MECP2e2.

Figure 3. MeCP2e1 and MeCP2e2 have different N-termini but share the same remaining

domains.

Figure 4. Studying neurological diseases with iPS cell technology.

Figure 5. Schematic overview of the neuronal differentiation protocol described by Zhang et al.,

2001.

Figure 6. Schematic overview of the neuronal differentiation protocol described by Li et al.,

2009.

Figure 7. Schematic overview of the neuronal differentiation protocol described by Chambers et

al., 2009.

Figure 8. Schematic overview of the neuronal differentiation protocol described by Brennand et

al., 2011.

Figure 9. Directed differentiation protocol of iPS cells into neurons used in my thesis.

Figure 10. One-step induction protocol can generate functional neurons after 2-3weeks of

differentiation.

Figure 11. Test RTT neurons were differentiated into MAP2 positive neurons.

Figure 12. Schematic represents sparse labeling technique for the visualization of single neurons.

Figure 13. GFP positive cells counterstained with MAP2 verified the identity of sparse labeled

neurons.

Figure 14. GFP positive neurons can be reconstructed to assay neurite complexity.

Page 9: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

ix

Figure 15. Two-week-old RTT neurons have reduced neurite tip number compared to healthy

control neurons.

Figure 16. The axon of a neuron could be easily identified by its long morphology.

Figure 17. Overview of experimental design performed on neurons differentiated for 6 weeks.

Figure 18. Nuclear MeCP2 was detected in healthy control neurons but not in RTT neurons.

Figure 19. RTTΔ3-4#37 neurons had less MeCP2 than the 3 other healthy controls.

Figure 20. Graphical representation of MeCP2 protein levels normalized to β-actin loading

control.

Figure 21. Graphical representation of MeCP2 protein levels normalized to βIII-tubulin loading

control.

Figure 22. RTTΔ3-4#37 isogenic healthy control neurons to RTTΔ3-4#20 null neurons have

smaller soma sizes.

Figure 23. DAPT treatment was ineffective at equalizing cellular density after 6 weeks of

differentiation.

Figure 24. RTTe1 neurons differentiated for 6 weeks in vitro have reduced dendritic complexity.

Figure 25. RTTe1 neurons differentiated for 6 weeks in vitro have reduced number of higher

order dendrites.

Figure 26. RTT neurons derived from the one-step induction protocol for 2 weeks in vitro do not

recapitulate disease phenotypes.

Figure 27. RTT neurons derived from the one-step induction protocol for 3 weeks in vitro do not

recapitulate disease phenotypes.

Page 10: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

1

Chapter 1 Introduction

1 Introduction

1.1 Rett Syndrome diagnosis and current treatments

Rett Syndrome (RTT) is a neurodevelopmental disorder that affects mostly girls at a prevalence

of 1 in 10,000 (1). The core features of RTT are normal development until 6-18 months of age,

after which the child loses acquired abilities of speech, purposeful hand movements, motor

abilities, and develops stereotypical hand movements. There are additional supportive diagnostic

features that are commonly present in RTT children. Some examples include: microcephaly,

seizures, breathing abnormalities, autonomic dysfunction, and reduced cognitive ability (2). The

presence of the core symptoms alone in the absence of exclusion factors, which are disease

caused by secondary brain injury or clinical manifestations before 6 months of age, is sufficient

for the diagnosis of RTT. After a period of developmental regression, the condition of a child

usually stabilizes and can improve in some cases (3).

There is currently no cure for RTT. The majority of treatments aim to manage symptoms such as

anxiety and seizures but have limited success. An ongoing Phase 2 clinical trial is testing the

efficacy of mecasermin (recombinant human IGF-1) in ameliorating RTT phenotypes. In the

Phase 1 clinical trial, mecasermin was well tolerated by treated participants. 9 out of 12 patients

had RTT causing MECP2 mutations. Treatment with mecasermin appeared to moderately reduce

breathing abnormalities and partially ameliorate behavioural symptoms associated with RTT

such as anxiety (4).

A randomized, double-blinded, placebo-controlled trial of L-Carnitine was conducted and though

it did not show significant advantages to improving hand apraxia, L-Carnitine did improve some

aspects of motor function and behavior as well as overall patient wellbeing. The trial reported

reduced constipation in treatment groups and increased energy, awareness, and physical activity

demonstrating its potential utility in improving the lifestyles of both patients and caregivers. The

authors noted that the long-term effects of L-Carnitine were not studied and that it may be

beneficial for only a small cohort of patients who have low plasma L-Carnitine levels (5).

Page 11: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

2

The use of bromocriptine, a dopamine agonist, in the clinic had been explored in a double-blind

trial. Ten RTT girls with a mean age of 8 years and 1 month were partitioned into two groups,

one receiving bromocriptine and the other placebo. Treatments lasted for 8 weeks and the

developmental progression of children was assessed before treatment, 4 weeks into treatment,

and 8 weeks after treatment. Two of the 5 girls receiving the drug benefited significantly from

bromocriptine, and 4 of 5 girls receiving placebo did not. This study demonstrated that a

subgroup of RTT children might benefit from dopamine agonist treatment though its mechanism

remains unknown (6).

Naltrexone, an opiate antagonist, has also been explored in clinical trials but only improved

breathing abnormalities found in RTT patients. 6 out of 10 girls treated with naltrexone

worsened after 4 months of treatment, advancing to higher stages of disease and showing poorer

motor function and no changes in the frequency of characteristic hand movements associated

with RTT (7).

Few therapies have been explored in clinical trials. Identifying novel therapeutics that may

improve patient symptoms or overall condition will greatly help them to achieve greater

autonomy while improving not only the patients’ lifestyle, but also the lifestyle of their

caregivers. It is evident that some drugs are effective in improving a patient’s condition.

Therefore, efforts to identify better compounds, small molecules, or drugs that can maintain,

improve, or restore the patients’ health should not cease.

1.2 The genetic etiology of RTT: MECP2 mutations

Genetically, the most common causes of RTT are mutations in the gene methyl-CpG-binding

protein 2 (MECP2) (8). MECP2 encodes the ubiquitously expressed protein MeCP2, which was

first shown to be a repressor of transcription (9,10). Over 200 RTT causing MECP2 mutations

have been described including: whole deletions, missense, nonsense, and synonymous mutations

(11-14). The majority of MECP2 mutations are single C>T transitions that localize to mutational

hotspots in functional protein domains. The methyl-CpG-binding domain (MBD), inter-domain

region (ID), transcriptional repression domain (TRD), nuclear localization signal (NLS), and C-

terminal domain of MECP2 are particularly affected. Genetic mutations occurring in the 5’UTR,

3’UTR, N-terminus, and intronic regions of MECP2 are rare (Figure 1) (15,16).

Page 12: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

3

Figure 1. Schematic of MECP2 gene with mutational hot spots indicated. MECP2 contains

four exons. Exons 3 and 4 encode functional domains including the methyl-CpG-binding domain

(MBD), inter-domain (ID), transcription repression domain (TRD), and C-terminal domain.

Mutations commonly occur in domains indicated with asterisks. Examples of the most common

mutations and their localization to domains are indicated (mutations with ≥ 1% prevalence were

included based on RettBASE).

1.3 General properties of MeCP2 and its functions on methylated DNA

MeCP2 is a ubiquitously expressed protein but is most abundant in the brain, where expression is

highest in neurons and increases throughout neurogenesis (9,17). Quantification of MeCP2 levels

in neuronal nuclei estimates the abundance of MeCP2 to be near histone octamer levels (18). In

this same study, it was demonstrated that MeCP2 tracks methylated DNA across the genome and

might modulate chromatin structure to regulate transcription.

Earlier studies aiming to elucidate the function of MeCP2 suggested that it was as a

transcriptional repressor that binds methylated-CpG-dinucleotides in vitro (19). A follow up

report showed that the Sin3A-HDAC corepressor complex interacts with a region of MeCP2 now

called the transcriptional repression domain (TRD), when bound to methylated DNA, to facilitate

the repression of transcription (20). Its mechanism of repression was further delineated by a

study showing that MeCP2 interacts with the NCoR/SMRT co-repressor complex, which

interacts with HDAC3 for transcriptional repression (21). Evidence for the repressive function of

MeCP2 was demonstrated by microarray experiments that showed that MeCP2 subtly repressed

gene expression (22-25,27).

Page 13: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

4

An important function of MeCP2 in post-mitotic neurons was defined when it was shown that

MeCP2 could play a role in gene regulation in response to neuronal activity. Activity dependent

transcription is important for converting short-term stimuli into long-term changes in brain

function. Using a candidate gene approach, it was discovered that MeCP2 participates in activity

dependent transcription of brain-derived neurotrophic factor (BDNF) (28). Membrane

depolarization of neurons increased calcium influx, which resulted in the phosphorylation of

MeCP2 and subsequently reduced its affinity for methylated-CpG-dinucleotides. This correlated

with Bdnf promoter activation (28). The resultant induction of Bdnf would facilitate neuronal

remodeling in response to activity that could promote cell survival and brain plasticity. This is an

example of the functional utility of MeCP2 in the brain.

However, the notion that MeCP2 functions as a transcriptional repressor only was questioned

when discordant data from microarray experiments were published suggesting that MeCP2 might

act as a transcriptional activator (22-27,29). It was later shown in neurons that MeCP2 binds to

5-hydroxymethycytosine (5hmC) and strongly correlated with active transcription (30). Further

evidence supporting the role of MeCP2 in activating transcription was reported when a global

down-regulation of gene expression was observed in a study conducted on MECP2-deficient

human embryonic stem (ES) cell-derived neurons (31). It is unclear how MeCP2 functions as

both a repressor and activator of transcription. Further research needs to be conducted to

understand the multi-faceted role of MeCP2.

1.4 MeCP2 isoforms, properties, and domains

An additional layer of complexity to MeCP2 function is added when the different isoforms are

considered. MECP2 can be alternatively spliced into two isoforms: MECP2e1 and MECP2e2,

(Figure 2) (32,33).

MeCP2e1 is expressed at much higher levels than MeCP2e2 in neurons (33). Two reasons

account for this difference in protein isoform levels. The first reason is that there are higher

levels of MECP2e1 transcripts than MECP2e2. The second reason is because MECP2e2 contains

an upstream open reading frame (upORF) in exon 1 that interferes with the translation of the

down stream ORF beginning at exon 2. Mutating the ATG of the upORF increases MeCP2e2

levels (Figure 2) (33).

Page 14: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

5

Figure 2. Splice map of MECP2e1 and MECP2e2. MECP2e1 and MECP2e2 have distinct N-

terminal domains because they are spliced differently. MECP2e1 excludes exon 2 while

MECP2e2 includes exon 2. MECP2e1 translates from the ATG codon in exon 1. MECP2e2 is

translated by initiating translation at the second ATG site. It is hypothesized that the upstream

open reading frame (upORF) present in MECP2e2 interferes with translation at the second ATG

start site in exon 2. Both isoforms of MeCP2 include exon 3 and 4.

MeCP2e1 and MeCP2e2 have a different N-terminal domain, but share the same MBD, ID,

TRD, and C-terminal sequences (Figure 3). It is uncertain whether the two isoforms share

similar or distinct functions. It has been reported that MeCP2e2 but not MeCP2e1 is neurotoxic

to cells when overexpressed in cerebellar granule cells (34). However, it has also been reported

that the reintroduction of MeCP2e2 in Mecp2-deficient mice can fully reverse reduced brain

weight, and nuclear size phenotypes, suggesting that there may be some overlapping function

between the two MeCP2 isoforms (35). More experiments need to be conducted in order to

uncover the unique or common functions of both protein isoforms.

Page 15: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

6

Figure 3. MeCP2e1 and MeCP2e2 have different N-termini but share the same remaining

domains. Both MeCP2e1 and MeCP2e2 have an N-terminus (NTD), methyl-CpG binding

domain (MBD), inter-domain (ID), transcriptional repression domain (TRD), nuclear localization

signal (NLS), and C-terminal domains. The two protein isoforms have distinct N-terminal

domains. Numerical values represent amino acid number. MeCP2e1 is larger in size than

MeCP2e2.

1.5 Cellular phenotypes in RTT post-mortem tissue

Before the development of rodent models to study RTT, clinicians and researchers relied on the

study of post-mortem tissue to gain insight into the neurobiology of RTT. Studying RTT with

post-mortem tissue was a useful tool to gain insight into the cellular phenotypes present during

disease stabilization. By measuring the weight of brains from RTT patients and comparing them

to age-matched controls, researchers discovered that RTT brains were lighter (36-38). When

trying to understand the underlying mechanism for smaller and lighter brains, Golgi staining of

layer III and layer V neurons in the frontal and motor cortex showed that arborization of both

apical and basal dendrites was reduced (36,37). Furthermore, increased cellular density was

reported in various regions of the brain such as the cerebral cortex and hippocampus (38). In this

same study, the authors showed that neuronal soma size in the hippocampus was also reduced.

By creating 3-dimensional reconstructions of frontal cortical neurons labeled with Lucifer yellow

and imaged by laser scanning confocal microscopy, Belichenko et al., provided evidence for

reduced number of dendritic spines in RTT brains (39). This study suggested that patients had

Page 16: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

7

reduced number of synapses, which was later supported when a reduced number of

synaptophysin puncta was observed after immunocytochemical staining of neurons from layer II,

III, and V of the frontal, temporal, and motor cortices of RTT patients (40). In summary, these

studies seemed to provide preliminary evidence to support the hypothesis that RTT was a

neurodevelopmental disorder with defects in neural circuitry and function.

There were a few limitations to using post-mortem tissue to study disease. One limitation that

was acknowledged by researchers was that post-mortem tissue reflected more closely the stage

of RTT where disease had stabilized and did not offer a representation of disease progression

(38,40). Other limitations included the inability to closely match age and gender, and acquire the

appropriate controls. Some studies used brains from patients with existing medical conditions

such as Trisomy 21 or lupus erythematosus as healthy controls (36-38,40). The issue of tissue

accessibility prompted the need for an alternative method to study RTT.

1.6 The most common RTT mouse models: behavioural deficits and cellular phenotypes of neurons and glia

The earliest attempts to generate Mecp2-deficient mice were unsuccessful when researchers

engineered the loss of Mecp2 in mouse embryonic stem cells and injected them into blastocyst

stage embryos for in utero development. Mecp2-/y chimeric mice were highly abnormal and the

majority of chimeras did not survive beyond weaning age. In contrast to Mecp2-/y chimeric mice,

control mice developed normally (41).

To circumvent this obstacle, the same research group created a conditional knockout mouse

model of RTT. To do this, mice with exon 3 and 4 of Mecp2 flanked by loxP sites were crossed

with mice, containing ubiquitous Cre or nestin-Cre. This created Mecp2-deficient mice with a

ubiquitous deletion of Mecp2 or targeted deletion of Mecp2 in neurons and glia, respectively.

For simplicity of nomenclature in my thesis, I called these mice Mecp2B-/y for male mice and

Mecp2B-/B- or Mecp2B-/+ mice for homozygous and heterozygous female mice, respectively (42).

When these Mecp2-null mice were studied for aberrant phenotypes, abnormal gait, reduced

spontaneous movement, hind limb clasping, and irregular breathing was observed after a period

of normalcy. The aberrant phenotypes were also observed in Mecp2-null mice with targeted

deletions in neurons and glia. Since RTT is a disorder predominantly affecting girls with a

Page 17: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

8

heterozygous MECP2 mutation, the authors rationalized that studying heterozygous female mice

would be the ideal genotype (Mecp2B-/+). Interestingly, it was reported that heterozygous female

mice showed symptoms much later (9 months) than viable male mice (3 months). This

developmental time line of RTT did not correspond with what has been observed in humans and

suggested that mouse models may be an underrepresentation of disease phenotypes (42).

Another research group conducted a similar study using the Cre-loxP system to generate Mecp2-

deficient mice harboring a deletion in exon 3 (43). For simplicity of nomenclature in my thesis,

these mice are called Mecp2J-/y male, Mecp2J-/J- female homozygous, and Mecp2J-/+ female

heterozygous deficient mice. Mecp2-deficient mice had behavioural defects: increased anxiety,

body tremors, pila erection, and respiratory irregularities. These deficits were accompanied by

neuroanatomical defects such as lighter brains, increased cellular densities in different regions of

the brain that were accompanied by reduced cellular size and nuclear size. These observations

have been previously described in studies conducted on human RTT post-mortem brain samples

(36,38). To determine if these phenotypes were caused by Mecp2 deficiencies in the brain, Chen

et al. targeted the loss of Mecp2 in neurons and glia, and neurons only and found that their

observations from null-mice were recapitulated in mice with targeted deletions of Mecp2. This

suggested that RTT was a neurological disorder and that the loss of Mecp2 in post-mitotic

neurons was sufficient to cause disease (43).

The behavioural phenotypes of RTT in mice were more carefully studied when a different

research group generated another mouse model of RTT that maintained partial function of

MeCP2 (44). Mice were engineered to translate truncated MeCP2 that contained its functional

MBD and TRD domains but lost a significant portion of its C-terminal tail. The authors reasoned

that in order to observe behavioural phenotypes in RTT-male mice, a less severe mutation should

be introduced to ameliorate the premature death phenotype that was observed in Mecp2-null

male mice (42,43). Male mice expressing truncated MeCP2 was termed Mecp2308/y mice and

demonstrated general phenotypes such as tremors that were readily visible at 4 months of age

and seizures by 8 months of age, and paw wringing when suspended by their tail. The motor

ability of Mecp2308/y mice was tested through a number of assays. Mecp308/y performed poorly on

the rotarod experiment, where mice are challenged to maintain their position on an elevated

rotating rod. Mecp308/y mice also fell down more during the vertical pole test, where mice are

positioned at the top of a vertical pole and need to climb down on their own. They also

Page 18: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

9

performed poorly on the wire suspension test, falling down more frequently in comparison to

control mice (44).

In the three aforementioned mouse models of RTT, all authors reported delayed onset of disease

progression that did not correspond to the RTT developmental time line in humans. Specifically,

female Mecp2-deficient mice had a tendency to develop disease symptoms in adulthood while

male Mecp2-deficient mice manifested symptoms early in development that corresponded with

the human condition (42-44). Additionally, Mecp2-null mice had increased incidences of obesity

depending on the selected background (42,43). These observations were unexpected since male

RTT patients do not usually survive into adulthood, female RTT patients develop symptoms

early in their childhood, and RTT children are usually underweight. Despite these limitations, it

was evident that Mecp2-deficient mice could recapitulate certain disease phenotypes and were a

valid model for RTT. This created an opportunity to study the cellular phenotypes associated

with Mecp2-deficiency in mice as well understand the biological mechanisms underlying RTT.

Chen et al., was the first research group to show that Mecp2-deficient mice had the neuronal

phenotype of reduced soma size and nuclear size (43). Since then, other research groups have

used Mecp2-deficient mice to determine if other neuronal morphological phenotypes are present.

Using postnatal 3 week old Mecp2-deficient male mice harbouring a deletion of exon 3 and 4

(Mecp2B-/y mice) (42) and an exon 3 deletion (Mecp2J-/y mice) (43), Belichenko et al.

demonstrated that variability in the distribution of dendritic spines on dendrites was present in

both mouse models. The observed variability in the distribution of dendritic spines along

dendrites was quantified as a greater number of spineless dendrites in both Mecp2B-/y and

Mecp2J-/y mice in all compartments of the brain examined (45). Mecp2B-/y and Mecp2J-/y mice

both had a greater number of dendrites with swollen segments in comparison to control mice.

Interestingly, overall dendrite width was measured to be thinner than controls. Axonal

organization was also examined in Mecp2B-/y mice and was visibly disorganized when compared

to wildtype mice (45).

Dendrite and dendritic spine morphology was also examined in Mecp2B-/+ female mice at

postnatal 6-7 months of age and male mice at postnatal 3 weeks of age (46). Reduced dendrite

width and dendritic spine number, increased frequency of spineless dendrites and dendrites with

long spines in cortical neurons was reported. Interestingly, this report provided preliminary

Page 19: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

10

evidence to suggest that neurons expressing MeCP2 within the cortex could exert a positive non-

cell autonomous effect on MeCP2 deficient neuronal growth (46).

Previous studies on post-mortem brain tissue had provided evidence for reduced dendrite

branching and complexity (36,37). Dendritic phenotypes in layer V pyramidal neurons from the

motor cortex of Mecp2J-/y mice were identified to have reduced total dendrite length and

branching (47). This finding was congruent with an earlier report demonstrating reduced dendrite

branching and soma size in layer II/III pyramidal neurons in Mecp2B-/y and Mecp2J-/y mice (17).

This defect was influenced predominately by cell-autonomous means but weakly influenced by a

non-cell-autonomous mechanism. This was observed when the distal complexity of dendrites

was reduced in wildtype neurons injected into Mecp2-deficient brains but not when wildtype

neurons were injected into the brains of wildtype mice (48).

It was commonly held that MeCP2 was absent in astrocytes and that their contributions to RTT

were unknown (17,49). Greater clarity into the contributions of non-cell-autonomous effects on

disease was gained when various research groups independently showed that MeCP2 is

endogenously expressed in astrocytes and that mutant astrocytes can exert a negative influence

on wildtype neurons through the reduced secretion of soluble factors such as BDNF (50-52).

The targeted deletion of Mecp2 from oligodendrocytes has some consequences as well.

Oligodendrocytes are generally known as the myelinating cells in the brain. This function is

critical for proper neuronal signaling and function. Mice with a targeted deletion of Mecp2 in

oligodendrocytes and oligodendrocyte precursors cells have more severe hind limb clasping

compared to control mice. It was noted that the Mecp2-deficient mice in this study had dull fur

and that some exhibited piloerection. These results demonstrated that the non-cell autonomous

effects of other cells might partially influence disease phenotypes (53).

The reported changes in dendritic spine number and dendritic morphology and cellular soma size

support the hypothesis that MeCP2 regulates neuronal circuitry and function since these

structures process electrical information for communication between neurons (54). Supporting

this hypothesis is evidence of electrophysiological measurements conducted on layer V

pyramidal cortical neurons from the somatosensory cortex of Mecp2J-/y mice, which showed

reduced spontaneous action potential firing in brain slices from symptomatic Mecp2-deficient

mice (postnatal 3-4 weeks of age). This reduction in spontaneous action potential firing was

Page 20: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

11

shown to be a result of shifted synaptic balance towards greater inhibition and reduced

excitation. The overall net effect was reduced excitatory synaptic activity, which in part accounts

for the observed altered electrophysiological properties in Mecp2-null neurons (55).

1.7 Rescue of disease models and limitations of the model

The first rescue experiments were performed by Luikenhuis et al. where the reintroduction of

Mecp2 in post-mitotic neurons of developing mice increased longevity, brain weight, and

improved behavioural phenotypes observed in Mecp2-deficient mice (56). Early administration

of BDNF could partially abolish RTT phenotypes (57). In this study, Mecp2-deficient mice

overexpressing BDNF had delayed onset of disease, improved locomotor ability and partially

restored brain weight, which is usually reduced. Of particular interest was the functional

restoration of spontaneous action potential firing deficits observed in mutants. These two studies

demonstrated that RTT was reversible so long as Mecp2 was reintroduced early in the

developing embryo. What remained unknown at the time was the reversibility of RTT after

disease onset and progression.

The principle of RTT reversibility was demonstrated in an elegant study where the reexpression

of MeCP2 in symptomatic Mecp2-deficient mice resulted in the amelioration of RTT

phenotypes. Male and Female symptomatic Mecp2-deficient mice (containing a floxed STOP-

Neo cassette in intron 2 of Mecp2 and ubiquitously expressing Cre-ER transgene had irregular

breathing, hind limb clasping, abnormal gait, and electrophysiological changes in long term

potentiation. Tamoxifen treatment resulted in the excision of the floxed STOP-Neo cassette,

which reactivated Mecp2 postnatally and could rescue changes in electrophysiological properties

(58). This study was reproduced soon after when cellular reexpression of Mecp2e2 isoform

postnatally partially reversed the RTT mouse body weight phenotypes, reduced brain weight

phenotype, and reduced nuclear size defect (35). These genetic rescue experiments demonstrated

the revocable nature of RTT by reintroducing wildtype Mecp2 in Mecp2-deficient mice after

disease onset and progression. These studies led to future attempts at aiming to reverse RTT

phenotypes in symptomatic mice by using small molecules, drugs, and compounds such as: TrkB

agonists, Insulin growth factor 1 (IGF-1), and Fingolimod (59-61). Interestingly, the compounds

listed above targeted and activated the PI3K/AKT and MAPK pathways, which are critical for

Page 21: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

12

neuronal development, synaptic maturation, and plasticity. Evidence for reduced PI3K/AKT

signaling in Mecp2-deficient mice had been described previously (59,62).

Mecp2-deficient mice have demonstrated their utility for studying RTT. Since RTT is a complex

disease that alters behaviour and motor ability, a clear read-out can be obtained from mice that is

not be possible from post-mortem studies. However, it was clear from the initial use of Mecp2-

deficient mice that they may be an underrepresentation of disease phenotypes since male mice

reflect the human condition of girls more accurately than female mice. In addition to this caveat,

another limitation is that mouse models may display supplemental phenotypes that are not

commonly present in patients (42,43). One example of this scenario is that Mecp2-deficient mice

are sometimes overweight, which is not a condition seen in patients who are usually underweight

(43). Therefore, to overcome these obstacles, some researchers have turned to the application of

induced pluripotent stem (iPS) cell and embryonic stem (ES) cell technology to study RTT

disease phenotypes (63-67).

1.8 iPS cells in studying disease

iPS cells were discovered by Shinya Yamanaka’s research group (68). His research group

demonstrated that embryonic-like cells could be generated from murine and human somatic cells

such as fibroblasts by the exogenous expression of four transcription factors OCT4, SOX2, KLF-

4, and c-MYC (68,69). One advantage of this technology is its ability to generate any cell type in

the body, which may be useful for studying disease mechanisms, identifying disease phenotypes,

drug discovery, and cell transplantation (70).

Soon after the discovery of iPS cells, several research groups were able to generate disease

specific iPS cells but were unable to identify disease phenotypes (71-74). A group studying

familial dysautonomia (FD) obtained FD-iPS cells and was the first group to successfully show

that disease phenotypes can be elucidated from iPS derived cells and that they are useful for drug

discovery (75). FD is a hereditary disease characterized by the degeneration of sensory and

autonomic neurons. FD iPS cell-derived neural crest precursors had distinct gene expression

profiles in comparison to control neural crest precursor cells. By comparing multiple cell lines

from different FD patients, they showed that these changes in gene expression were correlated

with both a differentiation defect and a migration defect. They also demonstrated that this

technology could be used for drug discovery and reported a candidate compound that was able to

Page 22: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

13

partially rescue some disease phenotypes. This study identified novel disease phenotypes and a

potentially beneficial therapeutic compound that had not been previously identified in FD mouse

models. This validated the use of iPS cells for studying early onset disease and drug discovery

(Figure 4) (75).

Figure 4. Studying neurological diseases with iPS cell technology. Patient fibroblasts can be

reprogrammed into iPS cells by the viral expression of four transcription factors: OCT4 (O),

SOX2 (S), KLF-4 (K), and c-MYC (M). iPS cells can be differentiated in vitro to generate an

affected cell type to study disease. The efficacy of drugs, small molecules, or compounds can be

tested for their ability reverse disease phenotypes.

1.9 Pertinent neuronal differentiation techniques and subtypes made

For the effective identification of disease phenotypes with iPS cell derived neurons, in vitro

differentiation protocols must be carefully crafted so that the specific affected cell type of

interest is obtained (76). Zhang et al., was one of the earlier groups to successfully differentiate

human ES cells into neurons (77). ES cells were detached from their adherent culture conditions

and cultured in suspension for four days. This gave rise to embryoid bodies (EBs), which were

then cultured in adherent conditions for 7 days in the presence of FGF-2. This yielded neural

rosettes with neural progenitor cells (NPCs) localized at the core of rosettes and contaminating

cells at the periphery. Zhang et al., isolated neural precursors by differential enzymatic treatment

and adhesion and expanded neural precursors in suspension cultures with the continued presence

of FGF-2. To induce the differentiation of NPCs into neurons, NPCs were dissociated with

trypsin and seeded onto ornithine/laminin coated plates and cultured in the absence of FGF-2 but

in the presence of N2 supplement, BDNF, and cAMP (Figure 5).

Page 23: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

14

Figure 5. Schematic overview of the neuronal differentiation protocol described by Zhang

et al., 2001. hES cells were differentiated into EBs by withdrawing FGF-2 from ES cell media

and detaching them from adherent plates. EBs were transferred to ornithine/laminin coated plates

and cultured in suspension with media containing insulin, transferrin, putrescine, sodium

selenite, heparin, and FGF-2 for 10 days to generate neural rosettes. Neural progenitor cells were

enzymatically isolated and cultured as neurospheres with neural progenitor media containing

FGF-2. NPCs were differentiated into neurons by culturing cells with media containing N2

supplement, cAMP, and BDNF. Neurons were studied at approximately 1-2 weeks after

differentiation from the NPC stage.

This research group conducted immunocytochemistry experiments and determined that the

majority of cell types generated were positive for the neuronal marker MAP2 or βIII-tubulin.

They also determined that the majority of cells were glutamatergic neurons with fewer

GABAergic neurons present. Astrocytes could also be made with this differentiation protocol.

Most importantly, the authors demonstrated that NPCs could incorporate into various regions of

neonatal mice brains and differentiate into both neurons and astrocytes (77).

Li et al., also used an EB-based directed differentiation protocol that was based on the protocol

developed by Zhang et al., but with a few modifications (77,78). hES cells were differentiated

into EBs by detaching ES cells and culturing them in suspension with ES cell media without

Page 24: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

15

FGF-2 for 4 days. EBs were transferred to plastic surface plates and cultured in neural media

without FGF-2 for 10-12 days to obtain neural rosettes. NPCs were isolated from neural rosettes

and expanded in suspension cultures as neurospheres with neural media lacking FGF-2. To

induce the differentiation of NPCs into neurons, NPCs were dissociated into single cells with

Accutase and transferred to ornithine/laminin coated plates so that NPCs could grow in adherent

conditions with nutrient rich neuronal media containing BDNF, GDNF, IGF-1, N2 supplement,

and B27 supplement without FGF-2 (Figure 6) (78).

Figure 6. Schematic overview of the neuronal differentiation protocol described by Li et al.,

2009. The protocol described by Li et al., was based on the protocol reported by Zhang et al.,

2001 with a few modifications. ES cells were induced to form EBs by withdrawing FGF-2 from

ES cell media and detaching ES cells from adherent plates. EBs were plated after 4 days of

suspension culture to plastic surface plates and cultured for 10 more days under adherent

conditions in neural media lacking FGF-2. NPCs were obtained from neural rosettes and

maintained in suspension cultures. To obtain neurons, NPCs were seeded onto ornithine/laminin

coated plates and cultured in neuronal differentiation media consisting of neurobasal media

supplemented with N2, B27, BDNF, GDNF, and IGF1 without FGF-2.

Page 25: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

16

EB-based methods can be protracted, inefficient, and generate heterogeneous population of cells

(79). To overcome these limitations, Chambers et al., provided evidence that the use of dual

SMAD inhibitors, Noggin and SB431542, can increase the efficiency of neural rosette formation,

reduce the time needed to obtain neurons, and remove the need for transient formation of EBs

(79). Noggin is a bone morphogenic protein (BMP) signaling inhibitor and SB431542 is a

Lefty/Activin/TGF-β signaling inhibitor. For the induction of hES or iPS cells into neural

rosettes, ES cells were plated at high density and cultured in knockout serum replacement (KSR)

media supplemented with Noggin for 11 days. Cells were cultured with SB431542 for the first 5

days of neural induction and withdrawn for the remaining days. Starting on day 5, KSR media

was supplemented with sequential increases of N2 (25, 50, 75, 100%) with feedings and

increasing dosages of N2 occurring every other day. By day 11, robust neural rosette formation

was observed (Figure 7A). It was reported that neural rosettes were positive for PAX6, OTX2,

and FOXG1B, which are markers for anterior fate neural precursors. This research group was

interested in generating dopaminergic neurons and motor neurons. To generate dopaminergic

neurons, ES cells were cultured in KSR media with Noggin and SB431542 for the first 5 days,

then switched to N2 media containing BDNF, AA, sonic hedgehog (SHH). On day 9, FGF8 was

added to culture media for an additional 3 days, after which neurons were cultured in N2 media

supplemented with BDNF, GDNF, cAMP, ascorbic acid, TGF-β1 without SHH and FGF8. To

generate motor neurons, identical culture conditions to generating dopaminergic neurons were

used, but after day 9, neurons were cultured in N2 media containing BDNF, ascorbic acid, SHH,

and retinoic acid (RA) for two weeks (79) (Figure 7B).

Page 26: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

17

A

B

Figure 7. Schematic overview of the neuronal differentiation protocol described by

Chambers et al., 2009. (A) The differentiation of ES cells into neural rosettes. The

differentiation of ES cells was induced by dual SMAD inhibition in KSR media. SB431542

Lefty/Activin/ TGF-β inhibitor was withdrawn after 5 days of treatment and levels of N2

supplement was sequentially increased in culture media. (B) The culture conditions for obtaining

dopaminergic neurons and motor neurons were identical until day 12 of differentiation. ES cells

were induced by dual SMAD inhibition and then cultured in N2 media supplemented with BDNF

and ascorbic acid (AA) after 5 days of culture. SHH is added to media on day 5 and FGF8 is

introduced to media on day 9. Both SHH and FGF8 were maintained in culture until day 12. To

obtain dopaminergic neurons, neural rosettes were switched to neuronal differentiation media

containing BDNF, GDNF, AA, cAMP, and TGF-β1. To obtain motor neurons, neural rosettes

were switched to BDNF, AA, SH, and retinoic acid (RA) rich media.

Page 27: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

18

One of the challenges to the aforementioned neuronal differentiation protocols is synchronizing

the time course of differentiation for multiple cell lines for disease modeling. This is because the

induction of iPS cells into neurons may often fail and result in incomplete experiments that lack

important controls. The expansion and passaging of NPCs as neurospheres is also cumbersome

and technically demanding. Brennand et al., created a neuronal differentiation protocol that

addressed some of the limitations to the aforementioned protocols (77-80). iPS cells were

detached with collagenase and cultured as EBs for 7 days in N2 media then transferred to

polyornithine (PORN)/Laminin coated plates and cultured in N2 media with laminin for another

7 days. Neural rosettes were manually isolated and then cultured in NPC media with N2, B27-

RA supplement, laminin, and FGF2. Neural rosettes were dissociated into single cells when

cultures became confluent and grown on PORN/Laminin coated plates in NPC media. At this

point of differentiation, these cells could be frozen so that the neuronal differentiation of multiple

cell lines could be synchronized or seeded immediately for neuronal induction of NPCs into

neurons. For the differentiation of NPCs into neurons, NPCs were seeded onto PORN/Laminin

coated plates and cultured in neural differentiation medium containing N2, B27-RA, BDNF,

GDNF, cAMP, and ascorbic acid. For their study, Brennand et al. differentiated neurons for 1-3

months before performing their experiments (Figure 8) (80).

The advantages of this protocol are that NPCs can be expanded in adherent monolayer culture

and frozen for future use (80). Freezing NPCs for future use is advantageous especially if

multiple cell lines need to be obtained for one experiment. After NPCs for all cell lines have

been acquired, NPCs can be seeded simultaneously and differentiated into neurons. Generating

neurons from the iPS cell stage is very difficult and inefficient, but generating neurons from the

NPC stage is much easier, more efficient, and more reliable.

Page 28: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

19

Figure 8. Schematic overview of the neuronal differentiation protocol described by

Brennand et al., 2011. iPS cells were detached from plates and induced to form EBs by

culturing iPS cells with media supplemented with N2. EBs were cultured for 7 days then

transferred to PORN/laminin coated plates and cultured with N2 media supplemented with

laminin for an additional 7 days. Neural rosettes were manually isolated and dissociated into

single cells and maintained as monolayer cultures with N2 media supplemented with B27

without vitamin A (B27-RA), laminin, and FGF2. To induce NPC differentiation into neurons,

NPCs were cultured with N2/B27 media with BDNF, GDNF, cAMP, and ascorbic acid (AA).

To improve the protocol described by Brennand et al., so that neural rosettes could be obtained

more efficiently, our lab modified the Brennand protocol to include dual SMAD inhibitors that

were similarly used in the protocol described by Chambers et al. (79,80). EBs were cultured in

suspension for 7 days in vitro with dual SMAD inhibitors then transferred to PORN/laminin

coated plates and cultured in adherent conditions for an additional 7 days to generate neural

epithelial cells that cluster together to form neural rosettes. NPCs were manually isolated from

neural rosettes and propagated as monolayer cultures on PORN/laminin coated plates or

cryopreserved until needed. To obtain neurons, NPCs were seeded onto PORN/laminin coated

plates and cultured in neuronal differentiation media rich with BDNF, IGF-1, GDNF, cAMP, and

Page 29: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

20

ascorbic acid. In our hands, this protocol required approximately 8-9 weeks of culture to obtain

electrophysiologically active neurons for experiments (Figure 9).

Figure 9. Directed differentiation protocol of iPS cells into neurons used in my thesis. iPS

cells were induced to form embryoid bodies (EBs) that were cultured in suspension conditions

with media containing BMP and Lefty/Activin/TGF-β signaling inhibitors (Dorsomorphin

(DSM) and SB431542, respectively). After 1 week of culture, EBs were plated onto

PORN/laminin coated plates and neural rosettes were cultured in adherent conditions for 1 to 2

weeks before NPCS were manually isolated and cultured in the presence of FGF2. NPCs at this

stage can be frozen for later use, expanded as monolayer NPCs, or seeded for neuronal

differentiation. Supplementing media with BDNF, GDNF, IGF-1, cAMP, and ascorbic acid

facilitates neuronal differentiation. Neurons can be identified as early as two weeks after

differentiation but do not have robust electrophysiological read outs until 6 weeks of

differentiation.

These protocols are technically challenging and require a lot of time before neurons are ready for

experimentation. Another limitation to these protocols is the heterogeneity in neuronal cultures.

Experiments conducted by another member of our lab observed through single cell expression

analyses that our lab’s modified Brennand protocol (described below) generates glutamatergic

Page 30: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

21

and GABAergic neurons, and upper layer and lower layer cortical neurons (81). Alternative

methods that could obtain functionally active neurons with greater efficiency and higher purity

would be favourable.

The one-step induction protocol can yield a homogenous population of excitatory,

electrophysiologically active neurons within 3 weeks of induction from the iPS cell stage (82).

Neurons are generated directly from iPS cells to make iNeurons through the overexpression of

transcription factor neurogenin-2 (Ngn2). To obtain iNeurons, iPS cells or ES cells are infected

with lentiviruses containing Ngn2 gene, a GFP reporter, and puromycin selection cassette

(Figure 10A). A tetracycline inducible promoter controls transgene expression. Treating iPS

cells with doxycycline induces gene expression and only cells expressing the lentiviruses are

selected for by puromycin selection. Resistant cells are cultured with glial cells to facilitate

neuronal differentiation and synapse formation. This protocol can yield electrophysiologically

active neurons within 2-3 weeks of differentiation (Figure 10B). This method yields neurons

much faster and is less technically demanding than EB-based methods. Unlike EB-based

methods, this protocol has not been used to study RTT phenotypes. It is unclear whether neurons

derived from this protocol can recapitulate disease phenotypes.

Page 31: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

22

A

B

Figure 10. One-step induction protocol can generate functional neurons after 2-3 weeks of

differentiation. (A) iPS cells were infected with viruses expressing rtTA and viruses expressing

Ngn2/EGFP/puromycin fusion protein linked by P2A and T2A sequences. A TetO inducible

promoter that is sensitive to doxycycline treatment controls the expression of Ngn2. (B)

Lentiviruses were infected into iPS cells. Cells were treated with doxycycline to induce

transgene expression. Doxycycline treatment remained for the duration of the experiment. Cells

were treated with puromycin for 1 day only to select for resistant cells. Mouse astrocytes were

added to neuronal cultures after puromycin selection to facilitate synapse formation and neuronal

maturation. Neurons were cultured for 2-3 weeks in vitro.

Page 32: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

23

1.10 Existing RTT iPS and ES cell models and their limitations

The first RTT iPS cells were described by Hotta et al., who obtained iPS cells from Mecp2308

mice and an 8-year-old RTT girl harboring a MECP2R306C mutation (74). No disease phenotypes

were reported, but it demonstrated that iPS cells could be isolated from both murine and human

patients. Follow up studies conducted on Mecp2308/y iPS cells demonstrated the utility of mouse

iPS cell technology for recapitulating disease phenotypes that have been previously described

from post-mortem tissue and Mecp2-deficient mouse models (65). miPS cells were differentiated

using the retinoic acid-mediated differentiation protocol that generates glutamatergic neurons. In

this study, our lab demonstrated that iPS-cell derived neurons from mice with disease relevant

Mecp2 mutations had reduced soma size and changes in electrophysiological properties (65).

Another research group used mouse ES-cell derived neurons with an engineered MECP2 exon 3-

4 mutation to study disease phenotypes. This research group identified reduced nuclear size in

MECP2-deficient ES cell-derived neurons (67). These studies validated the use of neuronally

differentiated iPS and ES cells from Mecp2-deficient mice for studying RTT.

Marchetto et al., was the first research group to show that human RTT iPS cell derived neurons

have disease phenotypes and can be used for studying the efficacy of drugs at reversing disease

related phenotypes (63). Fibroblasts from four female patients with distinct mutations were

reprogrammed into iPS cells and differentiated into neurons for morphological analysis.

Marchetto et al., first determined whether X chromosome-inactivation was erased after

reprogramming patient fibroblasts and found that their iPS cell lines either had two active X

chromosomes or retained an inactive X chromosome. They chose to model the disease using

only the reactivated iPS cell lines because they were randomly inactivated upon differentiation to

produce a mixture of wildtype and mutant neurons for phenotyping. An EB-based method of

neuronal differentiation was used to obtain neurons from iPS cells. Treatment with retinoic acid

induced neuronal differentiation. The neuronal subtypes made were not studied extensively but

glutamatergic neurons were identified by immunocytochemistry. RTT neurons had reduced soma

size and fewer synapse numbers, which was quantified by the number of VGLUT1 puncta on

MAP2 positive dendrites. The reduced number of synapses was correlated with altered activity-

dependent calcium transients that reflected functional deficits present in RTT neurons.

Remarkably, the synapse deficit phenotype was reversed by IGF-1 treatment. This seminal

Page 33: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

24

research paper demonstrated that iPS cell derived neurons from RTT patients can effectively

reveal RTT phenotypes and be applied to drug discovery (63).

Our lab obtained human fibroblasts from a female 6-year-old RTT patient with a deletion in exon

3 and 4 of MECP2 (RTTΔ3-4). The fibroblasts were reprogrammed into iPS cells for neuronal

induction and morphological phenotyping (64). When fibroblasts were reprogrammed into iPS

cells, the inactive X-chromosome was retained in a non-random pattern that was also maintained

after neuronal differentiation. This allowed for the isolation of an isogenic wildtype control

(RTTΔ3-4#37) that shared an identical genetic background to a RTT cell line (RTTΔ3-4#20), but

differed by the deletion of exon 3 and 4 of MECP2. The isolation of isogenic cell lines is

advantageous since variability between cell lines due to genetic background is lost. iPS cells

were differentiated into neurons using the protocol described by Li et al., and the soma sizes of

iPS cell derived neurons from both cell lines were measured (78). Reduced soma size was

reported for RTT neurons. Our study signified the potential to generate isogenic pairs of cell

lines from RTT patients, which can be utilized to study disease phenotypes (64).

To study the impact of MECP2 mutations in its various domains, Kim et al., obtained fibroblasts

from 5 different patients with point mutations affecting the three main domains of MECP2:

MBD, TRD, and CTD. RTT patient fibroblasts were obtained and reprogrammed into iPS cells.

The X-inactivation status of iPS cells was also studied and this research group isolated some iPS

cell lines with two active X-chromosomes and some with only one active X-chromosome. They

differentiated iPS cells into neurons using an EB-based differentiation protocol and studied the

expression level of candidate genes and found that the expression level of both TuJ and markers

for sodium channels were reduced. The authors concluded that RTT neurons had a neuronal

maturation defect, which they alluded to as in agreement with the study conducted by Marchetto

et al. (63,66).

Ananiev et al., obtained fibroblasts from a RTT patient with a MECP2 R294X mutation and

reprogrammed them into iPS cells (83). This research group also studied the X-inactivation

status of iPS cells and neuronal derivatives and showed that the X-inactivation mark was not

erased upon reprogramming of patient fibroblasts into iPS cells. When these cells were

differentiated into neurons they retained the inactive X chromosome status in a nonrandom

manner. This explains why isogenic cell lines could be isolated from this one patient. The iPS

Page 34: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

25

cell lines were differentiated into neurons using a slightly modified version of the protocol

described by Zhang et al., for morphological analyses (77). It was reported that R294X RTT

neurons had reduced nuclear size compared to isogenic wildtype controls. This study supported

previous findings that Mecp2-deficient mice and neurons from post-mortem RTT patients had

reduced nuclear size (83). This reaffirmed the notion that RTT iPS cell derived neurons can

exhibit clinical features observed in post-mortem tissue and Mecp2-deficient mice.

Pluripotent stem cell derived systems have great utility for studying the cellular morphology of

specific cells that may be otherwise difficult to obtain. In the context of RTT, further

optimizations of differentiation protocols to yield highly homogenous cultures of neurons will

facilitate more accurate characterizations of affected cells. However, this same advantage may be

a disadvantage since the brain itself is a highly heterogeneous organ. Glial cells are required for

proper neuronal development in living organisms, and the in vivo environmental niche of the

brain may not be easily recreated in vitro. Neurons can be cocultured with glia, but it is still

challenging to recreate an in vivo context that can faithfully reconstruct the disease

environmental niche. Cell line variability is also a concern for iPS cell-based studies, and can be

countered by evaluating multiple cell lines obtained from the same patient (84). The genetic

background of cell lines can also affect the phenotypic read out of interest. Therefore, if

comparisons are made between disease cells and healthy cells, where possible, the employment

of isogenic controls will reduce artifacts of genetic background (84).

1.11 Establishment and characterization of the RTTe1 cell lines

Since the discovery of MECP2 isoforms e1 and e2, there has been greater interest in identifying

their functions and relevance to RTT. At the beginning of my graduate studies in 2012, it was

unclear whether the two isoforms had unique or similar function. Work from Rudolph Jaenisch’s

research group suggested that they had overlapping function and that RTT phenotypes could be

partially rescued by the delayed overexpression of MeCP2e2 in symptomatic Mecp2-deficient

mice, and fully rescued if reintroduced early in the neural development of Mecp2-deficient mice

(35,56). Patients with MECP2e1 mutations have been previously described, whereas patients

with MECP2e2 mutations have not (32,85-88). This suggests that MECP2e1 is the disease

relevant isoform of RTT.

Page 35: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

26

No one has characterized the functional role of MeCP2e1 in humans. To study the function of

this isoform, our lab acquired and reprogrammed fibroblasts into iPS cells from a RTT patient

with an 11 base pair (bp) deletion in exon 1 of MECP2 (RTTe1) (32). This deletion causes a

premature stop codon resulting in truncated MeCP2e1 while leaving MeCP2e2 intact.

From the reprogramming event, a total of 4 RTTe1 iPS cell lines were extensively characterized.

Three of those cell lines were included in my study. The characterized RTTe1 cell lines were

karyotypically normal, and demonstrated pluripotent properties in vitro and in vivo via teratoma

assay formation (Table 1). cDNA from RTTe1 iPS cells and neurons were sequenced to verify

the presence of their MECP2 deletion. Androgen receptor assays were also performed to

demonstrate that RTTe1 iPS cell lines and neurons were skewed towards the mutant allele.

Neurons were obtained by the modified Brennand protocol described in Figure 9. Based on

single cell gene expression profiling comparing control neurons to RTTe1 neurons, RTTe1

neurons generated similar numbers of upper layer and lower layer neurons, and similar numbers

of excitatory and inhibitory neurons, but fewer cells expressing neuron markers: DCX, NCAM,

and MAP2 compared to controls. The soma sizes of RTTe1 neurons were also measured and

found to be smaller. Electrophysiological recordings of RTTe1 neurons showed that they had

reduced capacitance, and reduced frequency and amplitude of both evoked action potentials and

spontaneous action potentials. These assays were performed by other members of our lab and

demonstrated that the RTTe1 cell lines were pluripotent, carried MECP2 mutations, and can

display RTT phenotypes. No isogenic controls were isolated from the reprogramming of

fibroblasts into iPS cells (81).

In my thesis, additional wildtype and RTT iPS cell lines were employed as controls for assaying

dendritic complexity. The characterizations of iPS cell lines were completed by other members

of our lab and are summarized in Table 1. The additional cell lines expressed pluripotency genes

and made cells from the three germ layers in vitro and in vivo. All iPS cell lines were

karyotypically normal. All iPS cells were differentiated into neurons using the protocol described

in Figure 9.

Page 36: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

27

Healthy Female Control RTT

Cell line H9 SK0186_ 001#2.14

SK441_ 001#39

RTTΔ3-4 #37

RTTΔ3-4 #20

RTTe1 #27

RTTe1 #39

RTTe1 #96

RTT R306C

ES or iPS ES iPS iPS iPS iPS iPS iPS iPS iPS

Pluripotency Gene Expression C C C C C C C C C

Functional Pluripotency

In vitro (Embryoid Body) N/A C C C C C C C C

In Vivo (Teratoma) N/A C C C C N/A C C C

Karyotype N/A Normal Normal Normal Normal Normal Normal Normal Normal

C – Completed ES – Embryonic Stem Cell iPS – Induced Pluripotent Stem Cell N/A – Not available

Table 1. Summarized characterizations of RTT and healthy control cell lines. Nine cell lines

were included in my thesis. iPS and ES cell lines were characterized for pluripotency by lab

members. All iPS cell lines were karyotypically normal.

1.12 Thesis Rationale and Hypothesis

MECP2 mutations affecting both isoforms have been described in human patients and results in

defects of neural circuitry and function (63). These findings were also reported in Mecp2-

deficient mice containing a mutation affecting both isoforms of MeCP2 (55). Both lines of

evidence strengthen the current perspective that communication between neurons in RTT is

defective. MeCP2e1 is approximately 10-fold more abundant than MeCP2e2 in the brain (33).

No research group has studied the effects of losing only the predominant isoform of MeCP2,

MeCP2e1, on neural circuitry and function in humans. It is unclear whether a disease causing

mutation affecting MeCP2e1 and leaving MeCP2e2 intact is sufficient to cause perturbed

dendritic complexity, which would partially affect intercellular communication. I hypothesize

that a MECP2e1 mutation in RTT iPS cell derived neurons is sufficient to cause defects in

dendritic complexity.

Page 37: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

28

Chapter 2 Materials and Methods

2 Materials and Methods

2.1 Cell Culture

Human patient derived iPS cells and hES cells were cultured under the approval of the SickKids

Research Ethics Board and Canadian Institute of Health Research Stem Cell Oversight

Committee. Pluripotent cells (iPS and ES cells) were cultured on matrigel-coated plates

(STEMCELL Technologies) and maintained in iPS media [mTESRTM1 media containing 5X

supplement (STEMCELL Technologies) and 100X penicillin and streptomycin (Gibco)].

Spontaneously differentiating cell types surrounding pluripotent colonies were manually

removed with a plastic micropipette tip under a dissection microscope. Pluripotent cells were

enzymatically detached with Dispase (STEMCELL Technologies) and passaged every 7 days.

2.2 Neuronal Differentiation

Pluripotent stem cells were differentiated into neurons using the directed differentiation protocol

described by Brennand et al. with a few modifications or recently acquired one-step induction

protocol (80,82).

To obtain neurons by the directed differentiation protocol, pluripotent cell colonies were

enzymatically detached from matrigel-coated plates using Collagenase IV (Invitrogen),

transferred to 6 well low cluster plates for embryoid body formation (Corning). Embryoid bodies

(EBs) were cultured in EB media [Dulbecco Modified Eagle Medium Nutrient Mixture F-12

(DMEM/F12; Gibco) supplemented with N-2 (100X; Gibco), MEM Non-essential Amino Acids

(NEAA; 100X; Gibco), Penicillin and Streptomycin (100X; Gibco), Heparin (2 µg/ml; Sigma),

Fibroblast Growth Factor 2 (FGF2; 10 ng/ml; R&D), Dorsomorphin (2 µM; Sigma), and

SB431542 (10 µM; Stemgent)] for 7 days in vitro then transferred to Poly-L-Ornithine (0.1

mg/ml; Sigma) and Laminin (20 µg/ml; Roche) (PORN/Laminin) coated plates for the formation

of primary neural rosettes. Primary neural rosettes were cultured in adherent conditions and

maintained in Neural Rosette (NR) media [Dulbecco Modified Eagle Medium Nutrient Mixture

F-12 (DMEM/F12; Gibco) supplemented with N-2 (100X; Gibco), MEM Non-essential Amino

Page 38: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

29

Acids (NEAA; 100X; Gibco), Penicillin and Streptomycin (100X; Gibco), Heparin (2 µg/ml;

Sigma), Fibroblast Growth Factor 2 (FGF2; 10 ng/ml; R&D), and Laminin (1 µg/ml; Roche)] for

7 days in vitro. Primary Neural Rosettes were manually resected using a sterile needle and P20

micropipette tip, and transferred to PORN/Laminin-coated plates for an additional 7 days of in

vitro culture in NR media. Secondary NRs were manually resected and enzymatically dissociated

to single cells with Accutase (Innovative Cell Technologies). Dissociated Neural Progenitor

Cells (NPCs) were transferred to PORN/Laminin-coated plates and were cultured in NPC media

[Dulbecco Modified Eagle Medium Nutrient Mixture F-12 (DMEM/F12; Gibco) supplemented

with N-2 (100X; Gibco), MEM Non-essential Amino Acids (NEAA; 100X; Gibco), Penicillin

and Streptomycin (100X; Gibco), Heparin (2 µg/ml; Sigma), Fibroblast Growth Factor 2 (FGF2;

10 ng/ml; R&D), Laminin (1 µg/ml; Roche), and B27 without Vitamin A (50X; Gibco)]. NPCs

were enzymatically passaged with Accutase every 7 days and were discarded after 10 passages.

For assays performed on 2-week-old neurons, NPCs were seeded together at a density of 5.0x104

cells/well of 24-well plates with PORN/Laminin-coated glass cover slips. NPCs were cultured in

complete Neuronal Differentiation (cND) media [Neurobasal (Gibco) supplemented with N-2

(100X; Gibco), MEM Non-essential Amino Acids (NEAA; 100X; Gibco), Penicillin and

Streptomycin (100X; Gibco), Laminin (1 µg/ml; Roche), B27 without Vitamin A (50X; Gibco),

BDNF, GDNF, IGF-1 (all 10 ng/ml; Peprotech), ascorbic Acid (200ng/ml; Sigma) and cAMP (1

µM; Sigma)] for two weeks in vitro.

For 6-week-old neuronal morphology assays, RTT neurons were seeded at equivalent densities

(10x104 cells/well of 24-well plate) and maintained in cND media with the addition of N-[N-(3,5-

Difluorophenacetyl-L-alanyl)]-(S)-phenylglycine t-butyl ester (DAPT) to promote neuronal

induction and manage cell density. Cells were treated with DAPT for the first 10 days of NPC

induction into neurons and then maintained in cND without DAPT for the remaining 32 days.

For western blot experiments performed on neurons differentiated for 6 weeks, NPCs were

seeded at a density of 6.0x105 cells/well of 6-well PORN/Laminin-coated plates and maintained

as described above.

Neurons derived from the one-step induction protocol were obtained from Dae-Sung Kim, a

post-doctoral fellow in our lab (82). Briefly, pluripotent cells were infected with Ngn-2

lentiviruses while being maintained in iPS media. One day after infection, cells were treated with

Page 39: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

30

doxycycline to induce transgene expression. Ngn2 expressing cells were isolated by puromycin

selection then cocultured with mouse astrocytes on the following day. Neurons were

differentiated for two or three weeks in vitro before fixation, immunocytochemical staining, and

morphological analyses.

2.3 Immunocytochemical (ICC) Staining

Neuronal cultures were fixed with 4% formaldehyde (EMD Biosciences) diluted in phosphate

buffer saline (PBS) for 10 min at room temperature and permeabilized with 0.1% Triton X-100

diluted in PBS for 10 min at room temperature. Neuronal cultures were incubated in 5% normal

goat serum diluted in PBS (blocking solution) for 1 hour at room temperature before incubating

neuronal cultures in primary antibodies diluted in blocking solution overnight at 40C. Excess

primary antibody was removed by washing with PBS and neuronal cultures were incubated with

the appropriate fluorescence conjugated secondary antibody for detection. After staining nuclei

with DAPI (2 µg/ml), coverslips were mounted onto glass slides and imaged with a Leica DMI

4000B microscope equipped with a Leica DFC340 Fx camera. Images were acquired using Leica

Application Suite V3.6 software. Information regarding primary antibodies and their pairings

with secondary antibodies are described in Table 2.

Page 40: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

31

Experiment 1o Ab/DAPI 1o Ab/DAPI Dilution Catalog # 2o Ab 2o Ab

Dilution Catalog #

Detecting MeCP2

Levels by ICC

DAPI 2 µg/ml N/A ✕ ✕ ✕

Ms Anti- MeCP2 1:1000 Sigma #M6818 Anti-Ms-488 1:500 Molecular

Probes #A11001

Rb Anti-MAP2 1:500 Millipore #AB5622 Anti-Rb-555 1:500 Molecular

Probes #A21429

Dendrite Morphology (Directed)

Rb Anti-GFP 1:1000 Invitrogen #A11122 Anti-Rb-488 1:500 Invitrogen #A11008

Ms Anti-MAP2 1:3000 Sigma #M1406 Anti-Ms-555 1:500 Molecular

Probes #A21422

Dendrite Morphology (One-step)

DAPI 2 µg/ml N/A ✕ ✕ ✕

Ms Anti-MAP2 1:3000 Sigma #M1406 Anti-Ms-555 1:500 Molecular

Probes #A21422

Table 2. Summary of antibodies used in various experiments. All antibodies were diluted in

blocking solution (5% normal goat serum diluted in PBS).

2.4 Neuronal Morphometric Analyses

Neurite morphology assays were performed on neurons differentiated for 2 weeks and 6 weeks in

vitro. One day before fixation, EF1α-EGFP plasmids were transfected into neurons with

Lipofectamine 2000 (Invitrogen). To obtain low efficiency transfections, cultures were exposed

to the transfection mixture for 2.5 – 4 hours only and then transfection mixture was replaced with

fresh cND media. Neurons were given 24 ± 3 hours for transgene expression. After

immunocytochemical staining, GFP positive cells that costained with MAP2 were imaged. Grey-

scaled, colour-inverted GFP images were manually traced using NeuronJ software. Since axons

were not easily distinguished from dendrites at 2 weeks of differentiation, neurites were

measured. Terminal neurites less than 10µm in length were excluded from tracings. In contrast

to morphology assays performed on 2-week-old neurons, axons were easily distinguished from

dendrites at 6 weeks of differentiation. Therefore axons were excluded from manual tracings and

only dendrites were traced. Terminal dendrites less than 10 µm in length were excluded from

final reconstructions.

Page 41: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

32

Dendrite order was labeled in NeuronJ software. The first projection from the soma was labeled

order 1 (ie. primary dendrite) and dendrites branching from primary dendrites were labeled order

2. This method of dendrite labeling continued until all dendrites were labeled. Sholl analysis

was performed to measure neurite complexity through ImageJ software. Conceptually, a series of

concentric circles are drawn irradiating outward in defined increments from the soma and the

number of intersections between neurite and concentric circle at each increment is quantified.

The Sholl radius was set to 10 µm increments and the number of intersections between

reconstructed neurites and concentric circles was computed in ImageJ software.

Dendrite morphology assays were also performed on neurons differentiated by the one-step

induction protocol after 2 and 3 weeks of differentiation with one modification; neurons were

reconstructed based on grey-scaled, colour-inverted MAP2 images instead of GFP images since

no transfections were necessary.

Soma size based on MAP2 stains of neurons differentiated for 6 weeks in vitro by the directed

differentiation protocol was measured using ImageJ software. The perimeters of neuronal somas

were traced and the area was determined by ImageJ.

2.5 Western Blot

Neurons differentiated for 6 weeks in vitro were lysed with Radio Immuno Precipitation Assay

(RIPA) buffer [150 mM NaCl, 1% Triton X-100, 0.5% Sodium Deoxycholate, 0.1% SDS, 50

mM Tris (pH 8.0)] containing protease and phosphatase inhibitors. Lysates were incubated on

ice for 30 min with periodic vortexing before sonication. Protein concentration was quantified

and 80 µg of whole cell extract was mixed with 4X Laemmli buffer [20% glycerol, 0.125 M

Tris-HCl pH 6.8, 8% SDS, 0.02% Bromophenol blue, 20% β-mercapto ethanol] before resolving

by 8% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). Resolved

proteins were transferred to Hybond membranes (GE Healthcare Life Sciences). Membranes

were blocked with 5% Milk in 0.05% Tween-Phosphate Buffer Saline (T-PBS) for 1 hour at

room temperature and probed with rabbit anti-MeCP2 (1:1000; Millipore #07-013) primary

antibody overnight at 40C. Blots were washed several times with T-PBS and incubated with anti-

rabbit-HRP conjugated (1:5000) secondary antibody for 1 hour at room temperature. Blots were

washed several times with T-PBS and imaged with Chemidoc MP Imaging System and Image

Lab 4.1 software. Blots were stripped twice and reprobed overnight at 40C with anti-β-actin

Page 42: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

33

(1:5000; Sigma #A5441) or anti-βIII-tubulin (1:5000; Millipore #MAB1637) loading controls.

HRP-conjugated anti-mouse (1:5000) secondary antibodies were used to detect both loading

controls. Densitometry was performed using ImageJ software.

2.6 Statistics

All datasets were managed in Microsoft Excel 2011 and imported into GraphPad Prism 6 for

statistical analyses. For experiments where only two groups were compared, Student’s t-test was

used with the significance p-value set to 0.05. When multiple comparisons had to be made

between 2 groups (ie. Sholl analysis and comparing the number of dendrites per order in 6-week-

old dendrite complexity assays), multiple t-tests were conducted with a false discovery rate set to

5%. When 3 or more groups were compared, Analysis of Variance (ANOVA) statistics were

used with post-hoc correction.

Page 43: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

34

Chapter 3 Results

3 Results

3.1 RTT neurons were obtained by directing the differentiation of iPS cells into neurons

As a first step towards phenotyping the morphology of RTT neurons, I performed test

differentiations on 2 well-characterized human RTT cell lines (RTTΔ3-4#37 wildtype and

RTTΔ3-4#20 isogenic null mutant). I chose to employ the well established directed

differentiation technique described by Brennand et al., with a few modifications, under the

guidance of Alina Piekna, a technician in our lab, because NPCs could be frozen and stored until

all cell lines were acquired for experiments, NPCs could be seeded as single cells making it

easier to analyze neuronal morphology after differentiating NPCs into neurons, and this protocol

has been successfully used for studying disease phenotypes (Figure 10) (80). All NPCs used in

my thesis were obtained from this neuronal differentiation protocol and were performed by more

experienced lab members or me. I performed the induction of all NPCs into neurons for my

experiments.

I induced iPS colonies to form embryoid bodies (EBs) and cultured EBs for 7 days in

suspension. After 7 days of suspension culture, EBs were transferred to PORN/Laminin coated

plates and cultured in adherent conditions for an additional 7 days. During this period, neural

rosettes formed and comprised the multipotent neural progenitor cell (NPC) population, which

was manually isolated and expanded as monolayers to obtain a pure population of NPCs. I

stained NPCs for the NPC marker, Nestin, and the neuronal marker, Microtubule associated

protein 2 (MAP2), and observed an abundance of Nestin positive cells and few MAP2 positive

cells. I seeded RTTΔ3-4#37 healthy and RTTΔ3-4#20 null NPCs into separate vessels with

media rich in BDNF, IGF-1, GDNF, cAMP, and ascorbic acid for NPC differentiation into

neurons. Neuronal cultures were stained for MAP2 and DAPI after two weeks of culture. MAP2

positive neurons were visible by this time (Figure 11).

Page 44: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

35

Figure 11. Test RTT neurons were differentiated into MAP2 positive neurons. RTTΔ3-4#37

healthy isogenic control and RTTΔ3-4#20 null iPS cells were induced to form embryoid bodies

(EBs) and then cultured in adherent conditions. Neural progenitor cells (NPCs) within neural

rosettes were manually isolated and stained for Nestin and MAP2. NPCs were differentiated into

neurons for two weeks in vitro and stained for MAP2. All images were obtained from my own

differentiation. Scale bar = 100 µm.

Page 45: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

36

3.2 Establishing neuronal morphometric analyses to quantify neurite complexity

To visualize an individual neuron and its neurites from adjacent neurons for morphological

analyses, I tested a sparse labeling technique developed in the lab by Dr. Joel Ross, a post-

doctoral fellow in our lab. In brief, 50,000 NPCs were seeded in one well of a 4-well or 24-well

plate and cultured in neuron differentiation media for 2 weeks in vitro. One day before fixing

neurons, neuronal cultures were lipofected with 0.5 µg of EF1α-EGFP plasmid for four hours to

obtain low efficiency transfection (ie. sparse labeling). The transfection mix was replaced with

regular neuron differentiation media and transfected neurons were given 24 hours for transgene

expression before being fixed. I optimized this transfection protocol so that the neurites from an

individual neuron can be distinguished from neurites of an adjacent neuron (Figure 12).

Figure 12. Schematic represents sparse labeling technique for the visualization of single

neurons. NPCs were seeded at a defined density and cultured in neuronal differentiation media

for 2 weeks in vitro. One day before fixing neuronal cultures, cells were lipofected with EGFP

plasmids controlled by an EF1α promoter. Neurons were transfected for 4 hours to obtain low

efficiency transfection so that after 24 hours for transgene expression, single neurons and its

neurites can be easily distinguished from adjacent neurons.

After twenty-four hours post-transfection, neuronal cultures were fixed and stained for the

neuronal marker microtubule-associated protein 2 (MAP2) and GFP. Staining for GFP was used

to amplify its signal. Staining for MAP2 was employed to determine if GFP positive cells had

neuronal identity (Figure 13-arrow) or did not (Figure 13-arrowhead), since GFP was

expressed in neuronal and non-neuronal cells as expected for an EF1α promoter reporter.

Page 46: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

37

Figure 13. GFP positive cells counterstained with MAP2 verified the identity of sparse

labeled neurons. Twenty-four hours after transgene expression, neuronal cultures were fixed,

permeabilized, and stained for anti-GFP (green) and anti-MAP2 (red). GFP staining was

essential for signal amplification. The purpose of MAP2 staining was to verify that GFP positive

cells were neurons (ie. MAP2+; Arrow) and to distinguish them from non-neuronal cells

(MAP2-; arrowhead). Nuclei were stained with DAPI. Scale bar = 50 µm.

To examine neuronal complexity, I manually reconstructed MAP2 positive and GFP positive

cells with NeuronJ software (Figure 14A). Grey-scaled, color-inverted GFP images were

imported into NeuronJ software and traced using the tracing tool. After tracings were completed,

the “measure tracings” option was selected to calculate total neurite length. Tracings were

exported into ImageJ software for Sholl analysis, which measures neurite complexity as distance

increased from the soma. In principle, Sholl analysis is conducted by drawing a series of

concentric circles irradiating outwards at defined increments from the soma, and the number of

intersections between concentric circles and traced neurites are quantified (Figure 14B). The

Sholl radius was set to 10 µm for my experiments. From the exported tracings into ImageJ

software, the number of terminal neurites was manually counted. In total, 3 measures of neurite

complexity were employed: Total neurite length, number of terminal neurites, and number of

intersections per 10 µm away from the soma (ie. Sholl analysis).

Page 47: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

38

A

B

Figure 14. GFP positive neurons can be reconstructed to assay neurite complexity. (A) A

GFP positive cell that was positive for the neuronal marker MAP2 was reconstructed with

NeuronJ software. The neurites from a grey-scaled, colour-inverted GFP image was manually

traced. (B) Schematic illustrates the principle of Sholl analysis. To perform Sholl analysis, a

series of concentric circles were drawn irradiating outward from the soma at defined increments

of 10 µm. To measure neurite complexity, the number of intersections between concentric circle

at each increment and neurites was quantified.

Page 48: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

39

3.3 RTT neurons differentiated for two weeks in vitro have reduced neurite complexity

To test whether the RTT neurons I derived recapitulate RTT phenotypes, I analyzed the

morphology of 2-week-old neurons from two RTT patients (RTTΔ3-4#20 null and RTTe1#27)

and two healthy controls (SK0186_001#2.14 and RTTΔ3-4#37). RTTΔ3-4#20 null and its

RTTΔ3-4#37 isogenic control NPCs were obtained from my differentiation while RTTe1#27

affected patient and SK0186_001#2.14 unrelated female healthy control NPCs were obtained

from experienced technicians in our lab (Table 3).

Genotype Cell Line iPS ! NPC

differentiation by:

NPC ! Neuron

differentiation by:

Wildtype RTTΔ3-4#37 WL WL

SK0186_001#2.14 Technician WL

RTT RTTΔ3-4#20 WL WL

RTTe1#27 Technician WL

Table 3. Summary of cell lines included in experiment to determine if RTT neurons I

derived recapitulate the disease phenotype of reduced neurite complexity. Two wildtype and

two RTT cell lines were selected for this experiment. The RTTΔ3-4#37 and RTTΔ3-4#20 NPCs

were obtained from my own differentiation (highlighted in orange). SK0186_001#2.14 and

RTTe1#27 NPCs were acquired from a differentiation performed by different technicians. I

differentiated all NPCs into neurons (highlighted in orange). WL are initials for Wesley Lai.

Healthy control and RTT NPCs were differentiated for two weeks in vitro for morphological

analysis. I manually counted the number of terminal neurites and observed a reduced number of

terminal neurites in RTT null neurons and RTTe1 neurons compared to healthy control neurons

(Figure 15A). Differences in total neurite length were quantified by NeuronJ software from

morphological reconstructions and a shorter overall neurite length was observed from both

patients and one healthy control cell line (SK0186_001#2.14). Conclusions could not be made

because of inconsistencies between the two healthy control cell lines (Figure 15B). I observed a

Page 49: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

40

similar inconsistency by Sholl analysis. Neuronal reconstructions were exported into ImageJ

software and Sholl analysis was performed. The Sholl radius was set to 10 µm, so that a series of

concentric circles irradiating outward at defined increments from the soma were drawn and the

number of intersections between neurite and circle at each level were quantified. RTT neurons

had fewer intersections compared to one healthy control (SK0186_001#2.14) but not the other

(RTTΔ3-4#37) healthy control, thus making my results inconclusive (Figure 15C). These results

suggest that the WT neurons that I derived from my NPCs (RTTΔ3-4#37) behave differently

than the WT neurons that I derived from NPCs given to me by a technician (SK0186_001#2.14).

Page 50: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

41

A B

SK01

86_0

01#2

.14

RTTΔ

3-4#

37

RTTΔ

3-4#

20

RTT

e1#2

70

5

10

15

Neu

rite

Tip

Num

ber

n=97n=101 n=94 n=89

WT MUT

**

**

SK

0186

_001

#2.1

4

RTTΔ

3-4#

37

RTTΔ

3-4#

20

RTT

e1#2

70

200

400

600

Tota

l Neu

rite

Len

gth

(µm

)

n=101 n=98 n=93 n=89

*

WT MUT

*

*

C

Figure 15. Two-week-old RTT neurons have reduced neurite tip number compared to

healthy control neurons. (A) Two-week-old RTT neurons have reduced number of neurite tips.

(B) Results for differences in total neurite length were inconclusive because of inconsistencies

between control cell lines. (C) Results for number of intersections per 10 µm away from the

soma were inconclusive due to inconsistencies between healthy control cell lines. All values are

mean ± SEM, *p<0.05. This experiment was performed in triplicate.

Page 51: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

42

3.4 Experimental set up for neuronal morphometric assays performed on neurons differentiated for 6 weeks in vitro

I was interested in testing whether RTTe1 neurons had defects in dendritic complexity and

morphological assays conducted on neurons differentiated for 2 weeks in vitro did not provide

clear results that addressed my hypothesis. This was because dendrites and axons were not easily

distinguished at the 2 week time point. I rationalized that because other members of our lab have

electrophysiology data suggesting that functional neurons can be obtained after 6 weeks of

differentiation, the axons and dendrites of neurons should be easily identified. Indeed, axons

were easily identified from dendrites by their long morphology (Figure 16). Therefore, I chose

to repeat my neuronal morphometric analyses on neurons differentiated for 6 weeks.

Figure 16. The axon of a neuron could be easily identified by its long morphology. Neurons

were differentiated for 6 weeks, sparse labeled with GFP, and imaged. The axon of a neuron

(arrows) was much longer than the dendrites of a neuron. Scale bar = 50 µm.

Page 52: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

43

Inconsistencies between the two healthy controls were present in neuronal morphology assays

conducted on neurons differentiated for 2 weeks. Two possible reasons for the difference

between the WT neurons is that the RTTΔ3-4#37 cells expressed less MeCP2 than the other or

were mislabeled null cells. To test these hypotheses, I performed immunocytochemistry on 6-

week-old neurons and stained them for MAP2, MeCP2, and DAPI to determine if MeCP2 was

present in wildtype and mutant cell lines. This experiment was performed three times and I was

blinded to the identity of the cell line each time.

In parallel to the immunocytochemistry experiments performed for the detection of MeCP2, I

prepared 6-week-old neurons for western blot experiments to quantify MeCP2 levels. One 3 cm

dish per cell line was prepared. Whole cell lysates were obtained and western blotted. Blots were

probed for MeCP2, and two loading controls: β-actin and βIII-tubulin. The β-actin loading

control was used to control for total protein. The βIII-tubulin loading control was used to

determine if there were large differences in the differentiation efficiency that may affect my

results.

Neurons for dendritic morphology assays were simultaneously prepared with neurons purposed

for MeCP2 detection by immunocytochemistry and western blots. After 6 weeks of

differentiation, neurons were sparse labeled with GFP and stained for MAP2 and GFP. GFP+

and MAP2+ neurons were imaged. The dendrites of GFP+ neurons from grey-scaled, colour-

inverted GFP images were traced and quantified for defects in dendritic complexity using

NeuronJ software.

Since the soma sizes of the RTTΔ3-4#37 wildtype and RTTΔ3-4#20 null neurons have been

characterized previously, I also performed soma size measurements on RTTΔ3-4#37 wildtype

and RTTΔ3-4#20 null neurons from my experiments to determine if they were behaving as

described previously (64). Soma size from grey-scaled MAP2 images obtained from sparse

labeled neurons was measured by tracing the perimeter around the soma using ImageJ software.

The area was automatically calculated by ImageJ software after the tracing was performed. I was

blinded to the identity of the cell line.

Page 53: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

44

To ensure that any differences I observed were not due to cell line differences, I obtained NPCs

of two additional female WT control cell lines and two additional RTTe1 cell lines and included

them in the immunocytochemistry, western blot, and dendritc complexity assays described

above.

In summary, 8 cell lines were included in the assays performed after 6 weeks of differentiation

(Table 4, Figure 17). I performed all differentiations of NPCs into neurons. NPCs from 8 cell

lines were seeded simultaneously. Three experimental replicates were prepared one week apart.

Healthy Female Control RTT

Cell line H9 SK0186_ 001#2.14

SK441_ 001#39

RTTΔ3-4 #37

RTTΔ3-4 #20

RTTe1 #27

RTTe1 #39

RTTe1 #96

NPC generated by: other other other WL WL other other other

Immunocytochemistry ✓ ✓ ✓ ✓ ✓ ✓ ✓ ✓

Western Blot ✓ ✓ ✓ ✓ ✓ ✓ ✓ ✓

Soma size Directed (6 weeks) ✕ ✕ ✕ ✓ ✓ ✕ ✕ ✕

Dendrite morphology

Directed (6 weeks) ✓ ✓ ✓ ✓ ✓ ✓ ✓ ✓

✓ - Completed ✕ - Not completed WL - Wesley Lai

Table 4. Summary of cell lines used and assays performed on neurons differentiated for 6

weeks. Four WT female cell lines and four RTT cell lines (1 null and 3 RTTe1) were

differentiated into neurons for 6 weeks. Neurons were prepared for immunocytochemistry to

determine if MeCP2 was present, western blot to quantify MeCP2 levels, and dendritic

morphology assays to test if defects were present. The soma sizes of RTTΔ3-4#37 and RTTΔ3-

4#20 null neurons obtained from my differentiation were also measured to determine if they

reproduced previously published results. NPCs obtained from my differentiation are highlighted

in orange.

Page 54: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

45

Figure 17. Overview of experimental design performed on neurons differentiated for 6

weeks. NPCs from 8 different cell lines were differentiated into neurons. Two coverslips per

cell line was stained for MAP2, MeCP2, and DAPI, and then imaged. One 3 cm plate per cell

line was prepared for western blot experiments. For neuronal morphology assays, 5-6 coverslips

per cell line was sparse labeled with GFP and stained for GFP and MAP2. GFP+ and MAP2+

neurons were imaged. Dendrites were traced based on grey-scaled, colour inverted GFP images

and analyzed for morphological defects. Soma sizes of the RTTΔ3-4#37 and RTTΔ3-4#20

neurons was measured from grey-scaled MAP2 images. Neurons within the field of view were

randomly selected for soma size measurements. All experiments involving

immunocytochemistry were performed in a blinded manner. Three experimental replicates were

performed for each assay described.

Page 55: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

46

3.5 RTTΔ3-4#37 neurons and NPCs obtained from my differentiation should be excluded

One possible explanation for the morphological difference between the two wildtype cell lines in

neurite morphology assays performed on neurons differentiated for 2 weeks is that RTTΔ3-4#37

neurons were mislabeled RTTΔ3-4#20 null cells. To determine if this was true, I performed

immunocytochemical staining of 6-week-old neurons for MeCP2, MAP2, and DAPI. The

presence of MeCP2 in neurons was assessed in 8 cell lines (4 healthy controls, 1 RTT null, 3

RTTe1). This experiment was performed three times from separate differentiations. Nuclear

MeCP2 was detected in 6-week-old neurons derived from all four healthy control cell lines and

not detected in RTT null (RTTΔ3-4#20) and RTTe1 cell lines (Figure 18). Based on

immunocytochemistry experiments, MeCP2 was clearly present in the RTTΔ3-4#37 neurons and

absent in the RTTΔ3-4#20 null neurons. This indicates that RTTΔ3-4#37 cells were not

mislabeled RTTΔ3-4#20 null cells.

Page 56: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

47

Figure 18. Nuclear MeCP2 was detected in healthy control neurons but not in RTT

neurons. Neurons differentiated for 6 weeks in vitro from the NPC stage were fixed and probed

for anti-MeCP2 (green) and anti-MAP2 (red). DAPI stained nuclei (blue). MeCP2 was detected

in healthy control neurons but not detected in RTT neurons. This experiment was performed in

triplicate. Scale bars = 10 µm.

A second possible explanation for why differences were present between the two wildtype cell

lines is because RTTΔ3-4#37 neurons produced lower levels of MeCP2 than the other wildtype

neurons. To quantify differences in MeCP2 protein levels, whole cell lysates from RTT and

healthy control neurons differentiated for 6 weeks were collected for western blot experiments. I

established a reproducible western blot protocol for detecting MeCP2 protein, which was not

previously reliable to our lab. Whole cell lysates were collected and the total concentration of

protein was quantified. 80 µg of lysate were subject to 8% SDS-PAGE and transferred to

Hybond membranes. Blots were probed for MeCP2, and then stripped twice to probe blots for β-

actin and βIII-tubulin loading controls.

Page 57: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

48

Using an antibody that recognizes the C-terminus of MeCP2, protein was detected by western

blot in all four healthy control neurons. MeCP2 was not detected in RTTΔ3-4#20 null neurons,

which was expected since these neurons harbor a MECP2 deletion spanning exon 3 and 4.

Interestingly, MeCP2 was detected by western blot in neurons from all RTTe1 cell lines. This

finding is consistent with our hypothesis that the MECP2e1 mutation affects MeCP2e1 only and

does not alter the integrity of MeCP2e2. The presence of MeCP2 in RTTe1 neurons is likely

from the continued expression of MeCP2e2. For quantification purposes, two loading controls

were used: β-actin and βIII-tubulin. The βIII-tubulin loading control was also used to account for

potential differences in differentiation efficiency, which may alter the levels of MeCP2.

Quantification of MeCP2 protein levels normalized to loading controls were indicated by

numerical values. MeCP2 levels appeared to be lower in RTTΔ3-4#37 neurons compared to the

3 other healthy control neurons regardless of loading control selected for normalization. This

also indicated that the differentiation efficiencies were similar between cell lines (Figure 19).

This experiment was performed in triplicate with lysates from three separate differentiations.

Page 58: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

49

Figure 19. RTTΔ3-4#37 neurons had less MeCP2 than the 3 other healthy controls. Western

blots were probed with an antibody that recognizes the C-terminus of MeCP2. MeCP2 was

detected in healthy control neurons. MeCP2 was not detected in RTTΔ3-4#20 null neurons.

MeCP2 was detected in RTTe1 neurons, which is consistent with the continued expression of

MeCP2e2 though MeCP2e1 is lost. Blots were stripped twice and probed for β-actin and βIII-

tubulin loading controls, respectively. MeCP2 levels in RTTΔ3-4#37 and RTTe1 neurons were

visibly reduced when compared to the other unrelated healthy controls. Quantifications above

loading controls reflected this observation. This experiment was performed in triplicate with

lysates obtained from different differentiations.

Page 59: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

50

I quantified MeCP2 levels by normalizing to β-actin loading control. This analysis showed that

RTTΔ3-4#37 neurons consistently yielded approximately 3-fold less MeCP2 compared to other

healthy control neurons, and that RTTe1 neurons consistently yielded approximately 8-fold less

MeCP2 compared to healthy controls (Figure 20). This observation is consistent with our

hypothesis that the RTTe1 mutation affects MeCP2e1 but leaves MeCP2e2 intact. This

experiment was performed in triplicate.

Figure 20. Graphical representation of MeCP2 protein levels normalized to β-actin loading

control. Normalized MeCP2 protein levels were compared to RTTe1 neurons. RTTe1 neurons

consistently had a 8-fold decrease in MeCP2 protein levels compared to unrelated healthy

controls. RTTΔ3-4#37 neurons were excluded from the other healthy control group because it

consistently yielded 4-fold lower levels of MeCP2. Values are mean ± SEM from three

experimental replicates. * p < 0.05.

Page 60: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

51

MeCP2 protein levels were also normalized to βIII-tubulin loading control and quantified.

Similar to the quantification obtained from measuring MeCP2 levels normalized to β-actin,

RTTΔ3-4#37 neurons consistently yielded 3-fold lower levels of MeCP2 compared to other

unrelated healthy controls. RTTe1 neurons yielded 9-fold lower levels of MeCP2 compared to

healthy control neurons. This result is consistent with the continued expression of MeCP2e2 in

RTTe1 neurons (Figure 21). This experiment was performed in triplicate.

Figure 21. Graphical representation of MeCP2 protein levels normalized to βIII-tubulin

loading control. A 3-fold lower level of MeCP2 was detected in RTTΔ3-4#37 healthy control

neurons compared to other unrelated healthy controls. Quantification of MeCP2 levels in RTTe1

neurons showed a 9-fold reduction compared to unrelated healthy controls. Values are mean ±

SEM from three experimental replicates. * p < 0.05.

It was evident that RTTΔ3-4#37 neurons were underperforming in western blot experiments in

addition to morphology assays performed on neurons differentiated for 2 weeks. Our lab

previously published results demonstrating that isogenic RTTΔ3-4#37 wildtype neurons had

larger soma sizes than RTTΔ3-4#20 null neurons. To determine whether RTTΔ3-4#37 and

RTTΔ3-4#20 neurons were misbehaving, I performed soma size measurements with ImageJ

software. The soma was identified by MAP2 staining and the perimeter of the soma was traced

Page 61: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

52

(Figure 22A). The area of soma was calculated automatically by ImageJ software after the

tracings were performed. RTTΔ3-4#37 isogenic healthy control neurons had smaller soma sizes

than RTT null neurons, demonstrating that RTTΔ3-4#37 was not performing as expected

(Figure 22B). Over a hundred neurons from each cell line was measured between three

independent experiments.

A B

Figure 22. RTTΔ3-4#37 isogenic healthy control neurons to RTTΔ3-4#20 null neurons have

smaller soma sizes. (A) The perimeters of neuronal somas were traced as demonstrated based on

MAP2 staining obtained from neurons differentiated for 6 weeks in vitro. Scale bar = 10 µm. (B)

Bar graph shows that RTTΔ3-4#37 isogenic control to RTTΔ3-4#20 null neurons had reduced

soma size. The total number of neurons analyzed were 130 for RTTΔ3-4#37 isogenic control and

142 for RTTΔ3-4#20 null neurons between three experimental replicates. Values are mean ±

SEM. * p < 0.05.

From the accumulation of evidence suggesting that RTTΔ3-4#37 healthy control neurons are not

behaving as expected, I concluded that it would be appropriate to exclude this and its isogenic

null cell line from my morphometric assays conducted on neurons differentiated for 6 weeks in

vitro (Table 5). The reason for their underperformance is unknown but appears to be an isolated

event of faulty NPCs from my differentiation since other members of our lab were able to

reproduce the published soma size defect using their own NPCs differentiated from the same iPS

cells I used.

Page 62: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

53

Healthy Female Control RTT

Cell line H9 SK0186_ 001#2.14

SK441_ 001#39

RTTΔ3-4 #37

RTTΔ3-4 #20

RTTe1 #27

RTTe1 #39

RTTe1 #96

NPC generated by: other other other WL WL other other other

Immunocytochemistry (MeCP2 detected?) ✓ ✓ ✓ ✓ ✕ ✕ ✕ ✕

Western Blot (MeCP2 detected?) ✓ ✓ ✓ ✓ ✕ ✓ ✓ ✓

Soma Size N/A N/A N/A S B N/A N/A N/A

✓ - Yes ✕ - No N/A - Not available S - Smaller B - Bigger

Table 5. Summary of assays conducted and their results suggesting that RTTΔ3-4#37 and

its isogenic RTTΔ3-4#20 null cell line should be excluded from dendritic complexity assays

performed on neurons differentiated for 6 weeks. NPCs obtained from my differentiation are

indicated in orange. MeCP2 was present in wildtype but undetected in RTT neurons by ICC.

MeCP2 was present in wildtype neurons and RTTe1 neurons but undetected in RTT null

neurons. Lower levels of MeCP2 were detected in the RTTΔ3-4#37 and RTTe1 neurons

compared to the 3 other healthy control neurons. RTTΔ3-4#37 neurons had smaller soma size

than RTTΔ3-4#20 null neurons.

3.6 RTTe1 neurons differentiated for 6 weeks in vitro have reduced dendritic complexity

Morphology assays performed on neurons differentiated for two weeks showed that cellular

density was unequal amongst cell lines and that seeding the same number of NPCs at the

beginning of neuronal differentiation was ineffective at equalizing culture density. In attempts to

overcome this limitation in assays performed on neurons differentiated for 6 weeks, NPCs were

treated with DAPT for the first 10 days of differentiation into neurons. This approach has been

routinely used in our lab. DAPT promotes cell cycle exit and neuronal differentiation. It

appeared to be ineffective at equalizing cellular density in my hands (Figure 23).

Page 63: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

54

Figure 23. DAPT treatment was ineffective at equalizing cellular density after 6 weeks of

differentiation. Control and RTTe1 NPCs were seeded simultaneously and differentiated for 6

weeks in vitro before being transfected with GFP one day before fixation. Neurons were stained

for MAP2 (red) and GFP (green). NPCs were treated with DAPT for 10 days at the beginning of

neuronal induction in attempts to equalize cellular density. DAPT treatment appeared to be

ineffective. Only GFP positive and MAP2 positive cells were manually reconstructed. Grey-

scaled, colour-inverted GFP images were used for neuronal reconstructions. This experiment was

performed in triplicate. Scale bars = 50 µm.

I measured the dendritic complexity of neurons from 3 RTTe1 lines and 3 healthy control lines

after 6 weeks of in vitro differentiation. Total dendrite length, dendrite tip number, and the

number of intersections by Sholl analysis were quantified from reconstructed neurons as

described previously. RTTe1 neurons had significantly shorter total dendrite length than WT

neurons (Figure 24A). The average total dendrite length of wildtype neurons and RTTe1

neurons was 359.7 ± 10.41 µm and 266.9 ± 13.13 µm, respectively. RTTe1 neurons had reduced

number of terminal dendrites that was significantly different (Figure 24B). The mean number of

dendrite tips for wildtype neurons and RTTe1 neurons was 6.26 ± 0.35 and 5.09 ± 0.16,

respectively. RTTe1 neurons also had reduced number of intersections by Sholl analysis that

reached significance as distance increased from the soma (Figure 24C). These observations

support my hypothesis that RTTe1 neurons have reduced dendritic complexity.

Page 64: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

55

A B

C

Figure 24. RTTe1 neurons differentiated for 6 weeks in vitro have reduced dendritic

complexity. (A) RTTe1 neurons have reduced total dendrite length. Values are mean ± SEM. *

p <0.05. (B) RTTe1 neurons have reduced number terminal dendrites. Values are mean ± SEM.

* p < 0.05. (C) RTTe1 neurons have reduced number of dendrites per Sholl radii irradiating

outward from the soma at 10 µm increments. Values are mean ± SEM from three experimental

replicates. * FDR = 5%.

Page 65: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

56

To obtain an additional measure of dendritic complexity, I quantified the number of primary

dendrites and higher order dendrites from neuronal reconstructions. Primary, secondary, and

tertiary dendrites were labeled following the principle of centrifugal order of branch labeling

until all dendrites were labeled. The centrifugal order of branch labeling states that primary

dendrites projecting immediately from the soma are labeled “order 1”, secondary dendrites

branching from “order 1” dendrites are labeled “order 2”, and tertiary dendrites branching from

“order 2” dendrites are labeled “order 3”. This system applies to all dendrites until all of them

have been assigned an order. Dendrite order was manually labeled using the “label tracing”

option in NeuronJ software (Figure 25A). No difference in the number of primary dendrites was

observed, but a significant reduction in the number of higher order dendrites in RTTe1 neurons

compared to healthy control neurons was observed (Figure 25B).

A B

Figure 25. RTTe1 neurons differentiated for 6 weeks in vitro have reduced number of

higher order dendrites. (A) Dendrites were labeled by following the principle of centrifugal

order of branch labeling. Primary dendrites projecting immediately from the soma were labeled

“order 1”. Dendrite branches from “order 1” dendrites were labeled “order 2” and dendrites

branching from “order 2” dendrites were labeled “order 3” dendrites. This method of dendrite

labeling was applied until all branches were labeled. (B) No differences in the number of primary

dendrites were observed. RTTe1 neurons had reduced numbers of higher order dendrites. Values

are mean ± SEM from three experimental replicates. * FDR = 5%.

Page 66: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

57

The findings from neuronal morphology experiments performed on neurons differentiated for 6

weeks in vitro support my hypothesis that RTT neurons with a MECP2e1 mutation have reduced

dendritic complexity.

3.7 The overexpression of Ngn2 for induction of iPS cells into neurons may mask RTT phenotypes

Dae-Sung Kim, a post-doctoral fellow in our lab, differentiated two patient derived RTT iPS cell

lines and two healthy control cell lines into neurons using the one-step neuronal induction

protocol developed in the lab of Thomas Sudhof (82). Dae-Sung stained neuronal cultures for

MAP2 and acquired fluorescent images. I obtained the images from him for experiments

performed after 2 and 3 weeks of neuronal induction and conducted dendrite morphology assays

to determine whether RTT neurons derived by the one-step induction protocol can recapitulate

disease phenotypes. Preliminary experiments performed on RTT null neurons and RTT R306C

patient derived neurons from the one step-induction protocol after two weeks of in vitro

differentiation did not have shorter total dendrite length or reduced number of dendrite tips

(Figure 26A and B). The average total dendrite length for wildtype neurons and RTT neurons

was 395.35 ± 30.19 µm and 394.05 ± 25.93 µm, respectively. The average number of dendrite

tips for wildtype neurons and RTT neurons was 10.91 ± 0.86 and 10.36 ± 0.80, respectively. I

observed a significant reduction in the number of second order dendrites between RTT neurons

and one healthy control, which was not reproduced in a second healthy control (Figure 26C). No

differences in dendritic complexity between RTT neurons and healthy control neurons were

observed by Sholl analysis (Figure 26D). These data may suggest that the overexpression of

Ngn2 for neuronal differentiation might mask RTT phenotypes that were previously described.

This conclusion cannot be made however, since this experiment was performed once with a

small sample size.

Page 67: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

58

A B

C D

Figure 26. RTT neurons derived from the one-step induction protocol for 2 weeks in vitro

do not recapitulate disease phenotypes. (A) RTT neurons do not have reduced total dendrite

length. (B) RTT neurons do not have reduced number of dendrite tips. (C) RTT neurons do not

have differences in the number of primary dendrites. Differences in the number of secondary

dendrites were observed between RTT neurons and one healthy control but not the other.

Inconsistencies preclude any conclusions. (D) No differences in dendrite complexity were

observed by Sholl analysis. All values are represented as mean ± SEM from one experimental

replicate.

Page 68: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

59

Similar results were obtained when I analyzed 3-week-old neurons derived from the one-step

induction protocol. No significant difference in total dendrite length was observed between

wildtype neurons and RTT neurons (Figure 27A). The average total dendrite length of wildtype

neurons and RTT neurons was 365.3 ± 37.61 µm and 405.7 ± 44.53, respectively. No significant

difference in the number of dendrite tips was observed between RTT neurons and WT neurons

(Figure 27B). The mean number of dendrite tips for wildtype neurons and RTT neurons was

9.75 ± 0.75 and 10.22 ± 0.86, respectively. Differences in the number of primary and higher

order dendrites were present when I quantified the number of dendrites per order, however, none

of the differences were consistent between cell lines. Therefore, the results from this assay were

inconclusive (Figure 27C). I assayed dendrite complexity by Sholl analysis, but no differences

were observed between healthy control neurons and RTT neurons (Figure 27D).

These results demonstrate that the reduced dendritic complexity defect previously observed in

neurons obtained from the EB-based directed differentiation protocol was not present in neurons

obtained from the one-step induction protocol. I hypothesize that this is because the continued

overexpression of Ngn2 in neurons obtained from the one-step induction protocol masks RTT

phenotypes. More experiments needed to be conducted to determine whether my observations

from this first replicate can be reproduced.

Page 69: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

60

A B

C D

Figure 27. RTT neurons derived from the one-step induction protocol for 3 weeks in vitro

do not recapitulate disease phenotypes. (A) RTT neurons do not have differences total

dendrite length. (B) RTT neurons do not have differences in dendrite tip number. (C) Differences

in the number of dendrites per order were observed but lacked consistency. (D) No differences in

dendritic complexity determined by Sholl analysis were observed. Values are mean ± SEM.

Page 70: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

61

When the number of terminal dendrites was compared between neurons obtained from the EB-

based directed differentiation protocol after 6 weeks of differentiation and neurons obtained from

the one-step induction protocol after 2 or 3 weeks of differentiation, it was evident that neurons

obtained from the one-step protocol had a greater number of tips. The mean dendrite tip number

for wildtype neurons obtained from the directed differentiation protocol was 6.26 ± 0.35

compared to 10.91 ± 0.86 (2 week) and 9.75 ± 0.75 (3 week), from the one-step induction

dendrite length in comparison to neurons obtained from the EB-based directed differentiation

protocol. The increased number of dendrite tips might reflect a potential increase in complexity,

which is likely due to the non-cell autonomous effects of astrocyte culture, which facilitate

neuronal maturation. It is also possible that the overexpression of Ngn2 may increase neuronal

complexity as well, but this hypothesis requires further testing.

Assay Directed (6 weeks) One-step (2 weeks) One-step (3 weeks)

Dendrite tip number 6.26 ± 0.35 10.91 ± 0.86 9.75 ± 0.75

Total dendrite length (µm) 359.1 ± 10.41 395.35 ± 30.19 365.3 ± 37.61

Table 6. Neurons obtained from the one-step induction protocol may be more complex than

neurons differentiated for 6 weeks with the directed differentiation protocol. The mean

value of total dendrite length between wildtype cell lines was compared between neurons

obtained from the one-step induction protocol and directed differentiation protocol. No

difference in total dendrite length was observed. The mean number of dendrite tips from

wildtype cell lines was also compared between neurons obtained from the one-step protocol and

directed differentiation protocol. Neurons obtained from the one step induction protocol had

more dendrite tips than neurons obtained from the directed differentiation protocol. All values

are displayed as mean ± SEM.

Page 71: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

62

Chapter 4 Discussion

4 Discussion

4.1 Potential consequences of reduced neuronal dendritic complexity on behaviour and neuronal function

In this thesis, I provide evidence that RTTe1 neurons have reduced dendritic complexity. I

employed methods to reconstruct individual neurons for morphological analysis and used four

assays that demonstrate reduced dendritic complexity in RTTe1 patient neurons. RTTe1 iPS cell

derived neurons have reduced total dendrite length, number of terminal dendrites, higher order

dendrites, and number of intersections per 10 µm increment away from the soma by Sholl

analysis. The observed reduction in dendritic complexity supports the current notion that neurons

with mutant MeCP2 are less mature and form disrupted neural networks in RTT brains. What is

unclear is whether the reduced dendritic complexity is a result of delayed growth where neurons

continue to grow but at a slower rate or are stunted in growth. Reduced dendritic complexity has

been described in previous studies conducted on post-mortem brain tissue and Mecp2-deficient

mouse models (17,36,37,47,48). A recent report using a human RTT ES cell derived neuronal

system tested whether or not RTT ES cell derived neurons had reduced neurite complexity by

Sholl analysis (31). In this study ES cells containing an exon 3 deletion of MECP2 were

differentiated into neurons and shown to have reduced number of neurite intersections compared

to isogenic controls. My results are the first to document a reduced dendritic complexity

phenotype in RTTe1 patient derived neurons. This complements the RTTe1 small neuron

phenotype and changes in electrophysiological properties reported by Aaron Cheung in our lab

and Wenbo Zhang from Mike Salter lab, respectively.

The changes in electrophysiological properties include reduced amplitude, increased time course,

and reduced frequency of evoked action potentials. RTTe1 neurons also displayed lower

frequency and amplitude of spontaneous action potential (miniature excitatory postsynaptic

currents), and reduced ability to store up electrical charges in their cell bodies (ie. reduced

capacitance) (81). These alterations in electrophysiological properties provide direct evidence

that there is reduced excitability in RTT neurons as was previously described (55,65). The

Page 72: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

63

electrophysiology data and my dendritic complexity assays support the notion that MECP2e1

mutations can alter brain function. Evidence for this is supported by mice lacking Mecp2e1,

which show behavioural symptoms similar to Mecp2 null mice (89). Since dendrites and

synapses are the functional units of information exchange between neurons, changes in dendritic

arborization of cortical neurons can in part account for the behavioural phenotypes observed in

RTT patients (54,90). The small neuron phenotype, the changes in electrophysiological

properties, and reduced dendritic complexity demonstrate that a MECP2e1 mutation is sufficient

to cause RTT.

4.2 Targeting Ras-PI3K-AKT, Ras-MAPK, and Rho-family GTPases signaling pathways may reverse defects of dendritic complexity

Mechanistically, the reason for reduced dendritic complexity is likely due to aberrant AKT

signaling. Mecp2-deficient mice have defects in the AKT/mTOR signaling (62). The effects of

aberrant AKT/mTOR signaling were demonstrated in human ES cell derived neurons harbouring

a deletion of exon 3 of MECP2 (31). RTT ES cell derived neurons had reduced levels of

phosphorylated AKT (p-AKT) and phosphorylated S6 (p-S6; a downstream effector of mTOR),

which could be elevated by BDNF and IGF-1 treatment. Both BDNF and IGF-1 can rescue RTT

phenotypes (57,60). In the study conducted by Li et al., the administration of BDNF and IGF-1

both elevated p-AKT and p-S6 levels to near wildtype levels and was sufficient to rescue both

the soma size and neurite complexity defect that was identified in RTT ES cell derived neurons

(31). Knockdown of PTEN, an upstream negative regulator of AKT signaling, effectively

increased neuronal soma size and neurite complexity by Sholl analysis. Based on these

experiments however, it was unclear whether PTEN knockdown completely or only partially

restored neuronal morphology to wildtype standards (31). Blocking the phosphorylation of AKT

using p-AKT inhibitors (LY294002) can significantly reduce total dendrite length, number of

terminal tips, number of intersections by Sholl analysis, and soma size, the same phenotypes

identified in RTTe1 neurons (90,91). Interestingly, in the study conducted by Kumar et al., they

demonstrated that the selective activation of Ras-AKT signaling only and Ras-MAPK signaling

only could modulate total dendrite length with similar efficacy (90). In this study, selective

activation of MAPK signaling only increased the number of terminal dendrite tips more than the

selective activation of PI3K/AKT signaling only. Soma size however, was altered only by

Page 73: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

64

selective activation of PI3K/AKT signaling. These data suggest that in addition AKT signaling,

the MAPK signaling pathway may be an effective target for rescuing certain RTT phenotypes

like reduced number of terminal tips even though the MAPK pathway may not be affected in

RTT (59). This unaffected pathway might be one reason for the lack of observed difference in

the number primary dendrites in RTTe1 neurons. An upstream affector of MAPK is GSK-3β and

its phosphorylation by integrin-linked-kinase (ILK) has been shown to result in the inactivation

of GSK-3β and an increase in dendrite initiation (92).

The Rho-family of GTPases: RhoA, Rac1, and Cdc42 are also regulators of dendritic complexity

(93). Specifically, RhoA is a negative regulator of dendritc complexity and when mutated to be

constitutively active, reduces dendrite length (94-96). RhoA activation has also been shown to

reduce the number of primary and secondary dendrites by reducing the levels of its downstream

effector cypin, which increases dendritic number when active (97). Rac1 and Cdc42 are positive

regulators of dendrite initiation. When active, they increase dendrite outgrowth from the soma

(98). The activation of both Rac1 and Cdc42 also promote dendrite branching (95,96,99)

The reason for the observed lack of difference in the number of primary dendrites in RTTe1

neuron may be due to the effects of Rac1 and/or Cdc42 signaling. It is possible that the ILK-

GSK-3β-MAPK pathway may also be responsible for the lack of observed difference in the

number of primary dendrites. I hypothesize that one or more of these signaling pathways is

responsible for maintaining the number of primary dendrites in RTTe1 neurons and that the

reason for the observed differences in the number of higher order dendrites is due aberrant AKT

signaling, which has been studied before (31).

4.3 Experimental Limitations

4.3.1 A method to equalize cell density needs to be established to account for potential density-dependent effects on dendritic arborization

Cellular density is not easily maintained in neuronal cultures and can have major effects on

dendritic arborization. It is known that neurite arborization can be modulated by contact-

dependent activation of Notch signaling (100). In this study, the authors cultured neurons at low

and high density. They showed that neurons grown at low density had reduced activation and

translocation of Notch to the nucleus, whereas neurons grown at high density had greater

Page 74: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

65

activation and translocation of Notch to the nucleus. They drew the correlation that interneuronal

contact in high-density cultures facilitated the rapid growth of neurites through Notch activation,

which stopped after 9 days in vitro in contrast to low-density cultures with lower Notch

activation, which extended neurites much slower but continued to grow in total length. An

additional observation made was that individual neurites of neurons grown in low-density

cultures were longer and smoother in comparison to high-density cultures, where individual

neurites were shorter and bushier. Another study reported that Notch1 activation inhibited

average dendrite length but not the number of dendrite branches (101). Based on the raw values

within individual replicates from the 6-week-old neuronal morphology assays, I examined

whether there were consistent correlations between cellular density and total dendrite length but

did not find a consistent cell density dependent effect. However, these are crude observations and

I acknowledge that the effects of cellular density cannot be underestimated and future

experiments should aim to manage this better.

Using DAPT early in the neuronal differentiation process (ie. first ten days), which has been

shown to promote cell cycle exit and neuronal differentiation, was ineffective at equalizing

cellular density between cell lines (102). Others members in our lab are currently exploring the

use of Ara C, also known as cytarabine, which is a potent chemotherapeutic agent that kills

proliferating cells. This compound was used to equalize cell density between cell lines in

experiments conducted by another research group who studied RTT phenotypes using ES cell

derived neurons (31). The explored approach is to differentiate neurons for a time, treat them

with Ara C overnight, pass a defined number of neurons to a new dish, and allow neurons to

recover for 1 week before proceeding to experimentation. This agent should remove proliferating

cells and leave post-mitotic cells unharmed, which would allow us to study RTT phenotypes in

culture conditions with equal densities after treatment and passaging.

4.3.2 No RTTe1 isogenic wildtype controls were isolated from reprogramming

Isogenic wildtype control cells to RTTe1 cells were not isolated when fibroblasts were

reprogrammed into iPS cells. When comparing phenotypes between iPS derived mutant cells and

unrelated wildtype cells, the potential effects associated with genetic background or modifier

genes were unaccounted for. To compensate for unavailable isogenic controls and the potential

variability due to genetic background, I included 3 unrelated wildtype controls into my

Page 75: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

66

experimental design. Recently, other members of our lab were able to improve the situation by

generating a rescue RTTe1 cell line by infecting NPCs with MECP2e1 lentiviruses controlled by

a mouse Mecp2 promoter (MeP). The expression of MeP-MECP2e1 reversed an identified

RTTe1 neuronal soma size defect, demonstrating that it would be a good control for future

studies conducted on neurons obtained from the RTTe1 patient. This does not replace the

advantages to having isogenic wildtype controls however. With strategies such as TALEN and

CRISPR/Cas-mediated genome engineering, it may be possible to create isogenic wildtype

controls from RTTe1 iPS cells.

4.3.3 RTTΔ3-4#37 healthy control neurons consistently underperformed

An observation from my experiments was that RTTΔ3-4#37 neurons did not behave as described

previously (64). My colleagues and I have high confidence in this cell line since it has been

characterized extensively and performed well in assays for other lab members. But for an

unknown reason, NPCs obtained from my differentiations only, behaved similar to the isogenic

RTTΔ3-4#20 null cell line in many assays. I provided evidence for the detection of MeCP2 in

RTTΔ3-4#37 wildtype neurons by both immunofluorescence staining and western blot

experiments. Based on western blot experiments, RTTΔ3-4#37 neurons had lower levels of

MeCP2 relative to other unrelated healthy controls. RTTΔ3-4#37 healthy control neurons had

smaller soma sizes than RTTΔ3-4#20 null neurons, which was contrary to what had been

previously published by our lab (64). Since none of my colleagues have had issues with RTTΔ3-

4#37 cell lines, I rationalized that something peculiar must have happened when I differentiated

iPS cells into NPCs, though the precise cause of its deviation from norm is unknown. It is

possible that NPCs were misidentified neural crest cells, which make peripheral neurons instead

of cortical neurons. This could be tested by immunocytochemical staining of cells with AP2,

PAX7, and p75 markers for neural crest. Additionally, I could stain cells for NPC markers: Sox2,

Sox1, and Musashi1.

4.3.4 Loss of MeCP2e1 and continued expression of MeCP2e2 in RTTe1 neurons need to be confirmed by isoform specific antibodies

Based on western blot experiments, RTTe1 neurons have detectable levels of MeCP2. I

attributed the detection of MeCP2 to the continued expression of MeCP2e2 isoform, which

Page 76: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

67

should be unaffected by the 11 bp deletion in exon 1 of MECP2. To test this hypothesis however,

I would need to employ the use of MeCP2 isoform specific antibodies, which do exist (103,104).

I obtained these antibodies and attempted to use them to demonstrate the continued expression of

MeCP2e2 but could not obtain clear results because a large number of non-specific bands were

present.

4.3.5 EB-based neuronal differentiation protocol lacks heterogeneity that is often found in the brain

The selected neuronal differentiation protocol employed for my experiments lack the degree of

heterogeneity found within the brain. Glial cells are important contributors to normal brain

function. Several lines of evidence suggest that glia contribute to RTT pathophysiology. MeCP2

is expressed in astrocytes and oligodendrocytes albeit at lower levels than neurons (50). When

hippocampal neurons from wildtype mice are cocultured in vitro with wildtype astrocytes as a

source of nutrients and factors for growth, wildtype neurons remain healthy and are able to

extend projections that appear normal. However, when hippocampal neurons from wildtype mice

are cultured in vitro with RTT mouse astrocytes that lack MeCP2, wildtype neurons appear

stunted in growth with shorter projections and compromised soma size (50). The inability of

RTT astrocytes to support normal neuronal growth likely occurs because RTT astrocytes are

unable to secrete soluble factors that are necessary for neuronal growth such as BDNF (51).

The loss of MeCP2 in oligodendrocytes also contributes to RTT pathology. Mice lacking MeCP2

in oligodendrocytes have a hindlimb clasp, which is a characteristic phenotype analogous to hand

clasping that occurs in some RTT patients (53).

The contributions of glia in RTT disease progression is fortified by a recent study demonstrating

that bone marrow transplants to RTT mice can significantly rescue RTT phenotypes (105).

Wildtype and Mecp2-deficient mice were given lethal doses of irradiation to eradicate all nascent

immunity and then a bone marrow transplant from wildtype donors. Mecp2-deficient mice that

did not receive bone marrow transplants presented with RTT phenotypes such as reduced

longevity, body weight, and brain weight, which were rescued by transplantation. The rescue

effect was attributed to the transplantation of wildtype microglia and their ability to respond to

immunologic stimuli and carry out phagocytosis, both of which are altered in mice lacking

Mecp2 (105).

Page 77: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

68

The culture system I used to demonstrate morphological deficits in neurons does not integrate

supporting cell types for neuronal growth. Therefore, future experiments should seek to apply

coculture systems that include both wildtype and RTT glial cells so that a more faithful

representation of the brain can be obtained.

4.3.6 One-step induction neurons should be cocultured with both wildtype and Mecp2-deficient astrocytes to faithfully model disease

Our lab has begun integrating coculture systems in some of our experiments. For neuronal

morphology experiments performed on neurons obtained from the one-step induction protocol,

wildtype mouse astrocytes were added to culture conditions to facilitate neuronal maturation. By

comparing just the Sholl analysis profiles of neurons differentiated by the directed differentiation

protocol and one-step induction protocol, it was evident that neurons from the one-step induction

protocol were much more complex. Astrocytes cocultured with neurons obtained from the one-

step method were the likely contributors to the observed increase in dendritic complexity. Since

RTT patients are mosaic for wildtype and mutant cells, a coculture system that includes both

wildtype and mutant glia is ideal and methods to include representative proportions of glia in

culture needs to be further developed.

It cannot be ruled out however, that the increased complexity of neurons obtained from the one-

step induction protocol was also due to the overexpression of Ngn2, which can activate the

AKT/mTOR pathway, to increase dendrite complexity, and/or reduce RhoA activity, which will

also increase dendritic complexity (94-97,99,106,107). This hypothesis needs to be tested more

thoroughly however.

4.3.7 Lack of experimental replicates for morphology assays performed on neurons obtained from the one-step induction protocol preclude biological conclusions

No conclusions could be made from experiments performed on RTT neurons derived by the one-

step induction protocol. This protocol is appealing to us since it offers the potential to shorten the

time required for obtaining functional neurons. Our current directed differentiation protocol

commonly used in our lab requires approximately 9 weeks to obtain electrophysiologically active

neurons from iPS cells. The one-step induction protocol reports the acquisition of functional

neurons by 3 weeks from the iPS cell stage (82). It would be advantageous if this protocol were

Page 78: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

69

successful at recapitulating disease phenotypes. However, preliminary experiments suggest that

it does not. I performed neuronal morphometric analyses on neurons from two RTT patients and

two healthy controls, studied at two different time points. At both time points (2 and 3 weeks),

no consistent differences in dendritic complexity were observed. Experiments were performed

once at both time points and require more replicates before making any conclusions about the

rescue effects of Ngn2. If Ngn2 does rescue RTT dendritic morphology defects, this may be a

novel pathway that might have some therapeutic value.

Page 79: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

70

Chapter 5 Future Experiments

5 Future Experiments

5.1 Exploring genetic and pharmacological approaches to rescue RTT dendritic complexity defects

5.1.1 Background and Rationale

RTT phenotypes caused by Mecp2 mutations in mice can be reversed by the reintroduction of

wildtype Mecp2 after disease onset (58). However, it is unclear whether the reintroduction of

MECP2 in human neurons can reverse morphological defects present in patients. Our lab has

evidence suggesting that reintroducing MECP2e1 in human iPS cell derived neurons obtained

from the RTTe1 patient can rescue the soma size defect (81). MECP2e1 lentiviruses controlled

under the endogenous mouse Mecp2 promoter (MeP) were infected into NPCs of one RTTe1 cell

line and differentiated into neurons (52). RTTe1 neurons expressing MeP-MECP2e1 had larger

soma sizes that resembled unrelated wildtype control neurons. This suggests that neuronal

morphological defects found in human RTT patients can be rescued by reintroducing functional

MECP2. No attempts to evaluate RTTe1 dendritic complexity defects in the rescued neurons

have been made and it would be interesting to test whether gene therapy can effectively reverse

dendritic phenotypes. If successful, this would also validate the use of rescue RTTe1 neurons as

controls for future experiments aiming to identify other RTTe1 phenotypes. Rescued RTTe1

neurons would also be a good positive control for exploring pharmacological agents and their

efficacy in reversing disease phenotypes.

For Mecp2-deficient mice, the administration of BDNF and IGF-1 can alleviate their behavioural

symptoms (57,60). BDNF and IGF-1 can also reverse both the reduced soma size defect and

reduced neurite complexity defect observed in RTT ES cell derive neurons (31). Our lab has not

been able to reproduce the published results likely because our neuronal differentiation protocol

uses high levels of neurotrophic factors, BDNF and IGF-1, for neuronal induction. In order for us

to observe an effective rescue effect, it may be necessary to remove exogenous BDNF and IGF-1

from culture conditions.

Page 80: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

71

Media lacking BDNF and IGF-1 may not be able to support neuronal induction. Therefore,

coculturing both wildtype and RTT cells with wildtype mouse astrocytes may be a viable source

of neurotrophic factors (50). These astrocytes are readily available in our lab since others have

been using them for their own experiments. By relying on astrocytes for the secretion of BDNF,

IGF-1 and other soluble factors, this will establish a baseline of neuronal complexity for

wildtype and mutant neurons. To test the rescue effects of IGF-1, I would propose to supplement

wildtype and RTT neurons with exogenous IGF-1 and compare them to untreated cells. I

hypothesize that exogenous expression of MECP2e1 and treatment with IGF-1 can rescue

RTTe1 dendritic phenotypes.

5.1.2 Rescuing RTTe1 dendritic defects with MeP-MECP2e1 and IGF-1

To test this hypothesis, I would propose to infect NPCs of the three RTTe1 cell lines used in my

experiment with MeP-MECP2e1 (RTTe1 rescue neurons), which will serve as a positive control

for pharmacological rescue experiments. NPCs of the 3 RTTe1 rescue cell lines, 2 unrelated iPS

cell lines described in 6-week-old morphometric assays, and 3 RTTe1 lines will be differentiated

with the EB-based directed differentiation protocol (80). Since DAPT was ineffective in

controlling cellular density, the more potent compound Ara C will be used to eliminate

proliferating cells after a period of neuronal differentiation. An equal number of NPCs will be

seeded across all cell lines and cocultured with wildtype astrocytes. Neurons will be

differentiated for five weeks in vitro before being treated with Ara C overnight. After overnight

treatment, neurons will be passed into a new flask, reseeded with wildtype astrocytes, and

cultured for an additional one-week recovery period. To test whether RTTe1 dendritic

phenotypes can be rescued by exogenous IGF-1 treatment, some uninfected RTTe1 and wildtype

neurons will be cultured in media supplemented with IGF-1, in addition to coculture with

astrocytes (see Table 7 for summary of experimental setup).

One day before fixation, neurons will be transfected with EF1α-EGFP and analyzed for dendritic

phenotypes as described in my thesis (see Section 2.4). Three experimental replicates should be

performed as was done previously. The novelty of this experiment is in the combination of IGF-1

treatment and coculturing neurons with astrocytes that would represent the in vivo environment

more faithfully than coculturing neurons without astrocytes. This study would also test whether

Page 81: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

72

overexpressing MECP2 under the MeP promoter in RTTe1 neurons could reverse dendritic

complexity defects, which has not been examined previously.

Genotype (WT or MUT) Cell line Astrocyte Condition

WT

Untreated

SK0186_001#2.14 WT

SK441_001#39 WT

Treated

SK0186_001#2.14 + IGF-1 (10ng/ml) WT

SK441_001#39 + IGF-1 (10ng/ml) WT

MUT

Untreated

RTTe1#27 WT

RTTe1#39 WT

RTTe1#96 WT

Virus

RTTe1#27 MeP-MECP2e1 WT

RTTe1#39 MeP-MECP2e1 WT

RTTe1#96 MeP-MECP2e1 WT

Treated

RTTe1#27 + IGF-1 (10 ng/ml) WT

RTTe1#39 + IGF-1 (10 ng/ml) WT

RTTe1#96 + IGF-1 (10 ng/ml) WT

Table 7. Summary of proposed experimental conditions for rescue experiments. All cell

lines described above are derived from iPS cell lines obtained and reprogrammed from patient

biopsies. In total there are 2 wildtype (WT) iPS cell lines, 2 WT iPS cell lines treated with IGF-1

(10 ng/ml), 3 RTTe1 cell lines, 3 RTTe1 iPS cell derived lines infected with MeP-MECP2e1,

and 3 RTTe1 cell lines treated with IGF-1 (10 ng/ml). All cells are cocultured with mouse

wildtype astrocytes.

Page 82: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

73

5.2 Exploring RTT phenotypes in RTT neurons derived by the overexpression of Ngn2

Preliminary experiments performed on neurons derived from the overexpression of Ngn2 suggest

that RTT dendritic phenotypes that have been previously identified from neurons generated from

a directed differentiation protocol may be masked by transgene expression. Ngn2 is a

transcription factor that mediates both RhoA and AKT/mTOR signaling pathways. The

advantage to treating patients with Ngn2 is that it would activate these two pathways, both of

which, have been reported to mediate dendritic complexity. However, it may not be feasible to

administer Ngn2 via gene therapy. The finding of masked phenotypes was unexpected, but

conclusions were inadmissible due to the lack of experimental replicates and small sample size.

To improve on the experiment that was conducted previously, I would propose to introduce one

more control cell line and 3 RTTe1 cell lines (See Table 8 for complete list of cell lines and

experimental conditions). All cell lines would be cocultured with mouse astrocytes so that in vivo

conditions could be replicated more faithfully. Experiment should be performed in triplicate

Genotype Cell line Astrocytes

WT

H9 (ES cell) WT

RTTΔ3-4#37 WT

SK0186_001#2.14 WT

MUT

RTTΔ3-4#20 WT

RTT R306C WT

RTTe1#27 WT

RTTe1#39 WT

RTTe1#96 WT

WT – Wildtype MUT – Mutant

Table 8. Summary of cell lines included in study to determine whether Ngn2

overexpression masks RTT phenotypes. Three wildtype cell lines (1 ES cell line, 2 iPS cell

lines) and 5 RTT cell lines from 3 different patients would be included in this study. All cell

Page 83: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

74

lines would be cocultured with mouse astrocytes. This experiment should be performed in

triplicate with iPS cells seeded and infected with lentiviruses one week apart.

Once neurons have matured for 3 weeks in vitro, neurons would be fixed and stained with

MAP2, dendritic marker. Neurons would be reconstructed based on MAP2 staining using

NeuronJ software. Total dendrite length, number of terminal dendrites, number of primary and

higher order dendrites, and number of intersections by Sholl analysis would be performed as

described previously (see Section 2.4).

I predict that there would be no differences in the level of dendritic complexity between RTT cell

lines and healthy control cell lines and that the lack of difference would be due to a masking

effect by Ngn2 overexpression. The rescue effect of Ngn2 would be tested by infecting RTTe1

NPCs with lentiviruses containing Ngn2 and comparing their neuronal morphology to RTTe1

neurons and healthy control neurons, after 6 weeks of in vitro differentiation using the EB-based

differentiation protocol.

I predict that Ngn2 overexpression would mask RTTe1 dendritic morphology defects. There is

evidence suggesting that Ngn2 transcription factor acts upstream of the AKT/mTOR signaling

pathway (106,107). Ngn2 is a transcription factors that associates with Brn3A, which regulates

the expression of TrkA, TrkB, and TrkC receptors. TrkB receptors are activated by BDNF, a

small molecule that is able to reverse RTT phenotypes by stimulating the AKT pathway.

To test this hypothesis, RTTe1 cell lines overexpressing Ngn2 should have higher levels of

phosphorylated AKT (p-AKT) than uninfected RTTe1 cell lines. This can be tested by western

blot analysis, where cultures would be treated with Ara C to eliminate astrocytes, cellular debris

washed away, and lysates obtained from remaining neurons. Western blots would be probed for

p-AKT and the relative amounts would be compared between groups. The downstream effector

phosphorylated S6 (p-S6) would also be probed to determine if elevated p-AKT correlates with

an increase of p-S6. This study would determine whether the overexpression of Ngn2 has the

potential to restore RTT phenotypes to wildtype conditions. If there is potential therapeutic value

to stimulating Ngn2 expression, it may be worthwhile to seek pharmacological inducers or

activators of Ngn2.

Page 84: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

75

5.3 Conclusions

In this thesis, I employed morphology based in vitro methods to assess the complexity of

neuronal dendrites from RTTe1 patient neurons with a MECP2e1 mutation. RTTe1 neurons from

3 iPS cell lines had significantly reduced total dendrite length, reduced number of terminal

dendrites, reduced number of higher order dendrites, and reduced complexity by Sholl analysis

compared to wildtype neurons from 3 healthy iPS cell lines. In conjunction with the small soma

size phenotype of RTTe1 neurons and the genetic rescue experiments by our lab, we provide

strong evidence that the MECP2e1 isoform is essential for healthy neuronal morphology.

Preliminary experiments suggest that RTT neurons obtained from the one-step induction

protocol do not recapitulate disease phenotypes. I hypothesize that this might be because Ngn2

overexpression masks RTT morphological defects and I propose future studies that could be

done to test this hypothesis.

Page 85: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

76

References

1. Hagberg, B. (1985). Rett's syndrome: Prevalence and impact on progressive severe mental retardation in girls. Acta Paediatrica Scandinavica, 74(3), 405-408.

2. Chahrour, M., & Zoghbi, H. Y. (2007). The story of rett syndrome: From clinic to neurobiology. Neuron, 56(3), 422-437. doi:10.1016/j.neuron.2007.10.001

3. Neul, J. L., Kaufmann, W. E., Glaze, D. G., Christodoulou, J., Clarke, A. J., Bahi-Buisson, N., . . . RettSearch Consortium. (2010). Rett syndrome: Revised diagnostic criteria and nomenclature. Annals of Neurology, 68(6), 944-950. doi:10.1002/ana.22124; 10.1002/ana.22124

4. Khwaja, O. S., Ho, E., Barnes, K. V., O'Leary, H. M., Pereira, L. M., Finkelstein, Y., . . . Kaufmann, W. E. (2014). Safety, pharmacokinetics, and preliminary assessment of efficacy of mecasermin (recombinant human IGF-1) for the treatment of rett syndrome. Proceedings of the National Academy of Sciences of the United States of America, 111(12), 4596-4601. doi:10.1073/pnas.1311141111; 10.1073/pnas.1311141111

5. Ellaway, C., Williams, K., Leonard, H., Higgins, G., Wilcken, B., & Christodoulou, J. (1999). Rett syndrome: Randomized controlled trial of L-carnitine. Journal of Child Neurology, 14(3), 162-167.

6. Zappella, M. (1990). A double blind trial of bromocriptine in the rett syndrome. Brain & Development, 12(1), 148-150.

7. Percy, A. K., Glaze, D. G., Schultz, R. J., Zoghbi, H. Y., Williamson, D., Frost, J. D.,Jr, . . . Waring, S. (1994). Rett syndrome: Controlled study of an oral opiate antagonist, naltrexone. Annals of Neurology, 35(4), 464-470. doi:10.1002/ana.410350415

8. Amir, R. E., Van den Veyver, I. B., Wan, M., Tran, C. Q., Francke, U., & Zoghbi, H. Y. (1999). Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nature Genetics, 23(2), 185-188. doi:10.1038/13810

9. Shahbazian, M. D., Antalffy, B., Armstrong, D. L., & Zoghbi, H. Y. (2002). Insight into rett syndrome: MeCP2 levels display tissue- a14nd cell-specific differences and correlate with neuronal maturation. Human Molecular Genetics, 11(2), 115-124.

10. Boyes, J., & Bird, A. (1991). DNA methylation inhibits transcription indirectly via a methyl-CpG binding protein. Cell, 64(6), 1123-1134.

11. Miltenberger-Miltenyi, G., & Laccone, F. (2003). Mutations and polymorphisms in the human methyl CpG-binding protein MECP2. Human Mutation, 22(2), 107-115. doi:10.1002/humu.10243

Page 86: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

77

12. Archer, H. L., Whatley, S. D., Evans, J. C., Ravine, D., Huppke, P., Kerr, A., . . . Clarke, A. J. (2006). Gross rearrangements of the MECP2 gene are found in both classical and atypical rett syndrome patients. Journal of Medical Genetics, 43(5), 451-456. doi:10.1136/jmg.2005.033464

13. Scala, E., Longo, I., Ottimo, F., Speciale, C., Sampieri, K., Katzaki, E., . . . Ariani, F. (2007). MECP2 deletions and genotype-phenotype correlation in rett syndrome.American Journal of Medical Genetics.Part A, 143A(23), 2775-2784. doi:10.1002/ajmg.a.32002

14. Hardwick, S. A., Reuter, K., Williamson, S. L., Vasudevan, V., Donald, J., Slater, K., . . . Christodoulou, J. (2007). Delineation of large deletions of the MECP2 gene in rett syndrome patients, including a familial case with a male proband. European Journal of Human Genetics : EJHG, 15(12), 1218-1229. doi:10.1038/sj.ejhg.5201911

15. Trappe, R., Laccone, F., Cobilanschi, J., Meins, M., Huppke, P., Hanefeld, F., & Engel, W. (2001). MECP2 mutations in sporadic cases of rett syndrome are almost exclusively of paternal origin. American Journal of Human Genetics, 68(5), 1093-1101. doi:10.1086/320109

16. Christodoulou, J., Grimm, A., Maher, T., & Bennetts, B. (2003). RettBASE: The IRSA MECP2 variation database-a new mutation database in evolution. Human Mutation,21(5), 466-472. doi:10.1002/humu.10194

17. Kishi, N., & Macklis, J. D. (2004). MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions. Molecular and Cellular Neurosciences, 27(3), 306-321. doi:10.1016/j.mcn.2004.07.006

18. Skene, P. J., Illingworth, R. S., Webb, S., Kerr, A. R., James, K. D., Turner, D. J., . . . Bird, A. P. (2010). Neuronal MeCP2 is expressed at near histone-octamer levels and globally alters the chromatin state. Molecular Cell, 37(4), 457-468. doi:10.1016/j.molcel.2010.01.030; 10.1016/j.molcel.2010.01.030

19. Nan, X., Campoy, F. J., & Bird, A. (1997). MeCP2 is a transcriptional repressor with abundant binding sites in genomic chromatin. Cell, 88(4), 471-481.

20. Nan, X., Ng, H. H., Johnson, C. A., Laherty, C. D., Turner, B. M., Eisenman, R. N., & Bird, A. (1998). Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature, 393(6683), 386-389. doi:10.1038/30764

21. Lyst, M. J., Ekiert, R., Ebert, D. H., Merusi, C., Nowak, J., Selfridge, J., . . . Bird, A. (2013). Rett syndrome mutations abolish the interaction of MeCP2 with the NCoR/SMRT co-repressor. Nature Neuroscience, 16(7), 898-902. doi:10.1038/nn.3434; 10.1038/nn.3434

22. Tudor, M., Akbarian, S., Chen, R. Z., & Jaenisch, R. (2002). Transcriptional profiling of a mouse model for rett syndrome reveals subtle transcriptional changes in the brain.Proceedings of the National Academy of Sciences of the United States of America, 99(24), 15536-15541. doi:10.1073/pnas.242566899

Page 87: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

78

23. Kriaucionis, S., Paterson, A., Curtis, J., Guy, J., Macleod, N., & Bird, A. (2006). Gene expression analysis exposes mitochondrial abnormalities in a mouse model of rett syndrome. Molecular and Cellular Biology, 26(13), 5033-5042. doi:10.1128/MCB.01665-05

24. Ben-Shachar, S., Chahrour, M., Thaller, C., Shaw, C. A., & Zoghbi, H. Y. (2009). Mouse models of MeCP2 disorders share gene expression changes in the cerebellum and hypothalamus. Human Molecular Genetics, 18(13), 2431-2442. doi:10.1093/hmg/ddp181; 10.1093/hmg/ddp181

25. Jordan, C., Li, H. H., Kwan, H. C., & Francke, U. (2007). Cerebellar gene expression profiles of mouse models for rett syndrome reveal novel MeCP2 targets. BMC Medical Genetics, 8, 36. doi:10.1186/1471-2350-8-36

26. Nuber, U. A., Kriaucionis, S., Roloff, T. C., Guy, J., Selfridge, J., Steinhoff, C., . . . Bird, A. (2005). Up-regulation of glucocorticoid-regulated genes in a mouse model of rett syndrome. Human Molecular Genetics, 14(15), 2247-2256. doi:10.1093/hmg/ddi229

27. Urdinguio, R. G., Lopez-Serra, L., Lopez-Nieva, P., Alaminos, M., Diaz-Uriarte, R., Fernandez, A. F., & Esteller, M. (2008). Mecp2-null mice provide new neuronal targets for rett syndrome. PloS One, 3(11), e3669. doi:10.1371/journal.pone.0003669; 10.1371/journal.pone.0003669

28. Chen, W. G., Chang, Q., Lin, Y., Meissner, A., West, A. E., Griffith, E. C., . . . Greenberg, M. E. (2003). Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science (New York, N.Y.), 302(5646), 885-889. doi:10.1126/science.1086446

29. Chahrour, M., Jung, S. Y., Shaw, C., Zhou, X., Wong, S. T., Qin, J., & Zoghbi, H. Y. (2008). MeCP2, a key contributor to neurological disease, activates and represses transcription. Science (New York, N.Y.), 320(5880), 1224-1229. doi:10.1126/science.1153252; 10.1126/science.1153252

30. Mellen, M., Ayata, P., Dewell, S., Kriaucionis, S., & Heintz, N. (2012). MeCP2 binds to 5hmC enriched within active genes and accessible chromatin in the nervous system.Cell, 151(7), 1417-1430. doi:10.1016/j.cell.2012.11.022; 10.1016/j.cell.2012.11.022

31. Li, Y., Wang, H., Muffat, J., Cheng, A. W., Orlando, D. A., Loven, J., . . . Jaenisch, R. (2013). Global transcriptional and translational repression in human-embryonic-stem-cell-derived rett syndrome neurons. Cell Stem Cell, 13(4), 446-458. doi:10.1016/j.stem.2013.09.001; 10.1016/j.stem.2013.09.001

32. Mnatzakanian, G. N., Lohi, H., Munteanu, I., Alfred, S. E., Yamada, T., MacLeod, P. J., . . . Minassian, B. A. (2004). A previously unidentified MECP2 open reading frame defines a new protein isoform relevant to rett syndrome. Nature Genetics, 36(4), 339-341. doi:10.1038/ng1327

Page 88: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

79

33. Kriaucionis, S., & Bird, A. (2004). The major form of MeCP2 has a novel N-terminus generated by alternative splicing. Nucleic Acids Research, 32(5), 1818-1823. doi:10.1093/nar/gkh349

34. Dastidar, S. G., Bardai, F. H., Ma, C., Price, V., Rawat, V., Verma, P., . . . D'Mello, S. R. (2012). Isoform-specific toxicity of Mecp2 in postmitotic neurons: Suppression of neurotoxicity by FoxG1. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 32(8), 2846-2855. doi:10.1523/JNEUROSCI.5841-11.2012; 10.1523/JNEUROSCI.5841-11.2012

35. Giacometti, E., Luikenhuis, S., Beard, C., & Jaenisch, R. (2007). Partial rescue of MeCP2 deficiency by postnatal activation of MeCP2. Proceedings of the National Academy of Sciences of the United States of America, 104(6), 1931-1936. doi:10.1073/pnas.0610593104

36. Armstrong, D., Dunn, J. K., Antalffy, B., & Trivedi, R. (1995). Selective dendritic alterations in the cortex of rett syndrome. Journal of Neuropathology and Experimental Neurology, 54(2), 195-201.

37. Armstrong, D. D., Dunn, K., & Antalffy, B. (1998). Decreased dendritic branching in frontal, motor and limbic cortex in rett syndrome compared with trisomy 21. Journal of Neuropathology and Experimental Neurology, 57(11), 1013-1017.

38. Bauman, M. L., Kemper, T. L., & Arin, D. M. (1995). Pervasive neuroanatomic abnormalities of the brain in three cases of rett's syndrome. Neurology, 45(8), 1581-1586.

39. Belichenko, P. V., & Dahlstrom, A. (1995). Studies on the 3-dimensional architecture of dendritic spines and varicosities in human cortex by confocal laser scanning microscopy and lucifer yellow microinjections. Journal of Neuroscience Methods, 57(1), 55-61.

40. Belichenko, P. V., Hagberg, B., & Dahlstrom, A. (1997). Morphological study of neocortical areas in rett syndrome. Acta Neuropathologica, 93(1), 50-61.

41. Tate, P., Skarnes, W., & Bird, A. (1996). The methyl-CpG binding protein MeCP2 is essential for embryonic development in the mouse. Nature Genetics, 12(2), 205-208. doi:10.1038/ng0296-205

42. Guy, J., Hendrich, B., Holmes, M., Martin, J. E., & Bird, A. (2001). A mouse Mecp2-null mutation causes neurological symptoms that mimic rett syndrome. Nature Genetics, 27(3), 322-326. doi:10.1038/85899

43. Chen, R. Z., Akbarian, S., Tudor, M., & Jaenisch, R. (2001). Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a rett-like phenotype in mice. Nature Genetics, 27(3), 327-331. doi:10.1038/85906

44. Shahbazian, M., Young, J., Yuva-Paylor, L., Spencer, C., Antalffy, B., Noebels, J., . . . Zoghbi, H. (2002). Mice with truncated MeCP2 recapitulate many rett syndrome features and display hyperacetylation of histone H3. Neuron, 35(2), 243-254.

Page 89: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

80

45. Belichenko, P. V., Wright, E. E., Belichenko, N. P., Masliah, E., Li, H. H., Mobley, W. C., & Francke, U. (2009). Widespread changes in dendritic and axonal morphology in Mecp2-mutant mouse models of rett syndrome: Evidence for disruption of neuronal networks. The Journal of Comparative Neurology, 514(3), 240-258. doi:10.1002/cne.22009; 10.1002/cne.22009

46. Belichenko, N. P., Belichenko, P. V., & Mobley, W. C. (2009). Evidence for both neuronal cell autonomous and nonautonomous effects of methyl-CpG-binding protein 2 in the cerebral cortex of female mice with Mecp2 mutation. Neurobiology of Disease, 34(1), 71-77. doi:10.1016/j.nbd.2008.12.016; 10.1016/j.nbd.2008.12.016

47. Stuss, D. P., Boyd, J. D., Levin, D. B., & Delaney, K. R. (2012). MeCP2 mutation results in compartment-specific reductions in dendritic branching and spine density in layer 5 motor cortical neurons of YFP-H mice. PloS One, 7(3), e31896. doi:10.1371/journal.pone.0031896; 10.1371/journal.pone.0031896

48. Kishi, N., & Macklis, J. D. (2010). MeCP2 functions largely cell-autonomously, but also non-cell-autonomously, in neuronal maturation and dendritic arborization of cortical pyramidal neurons. Experimental Neurology, 222(1), 51-58. doi:10.1016/j.expneurol.2009.12.007; 10.1016/j.expneurol.2009.12.007

49. Jung, B. P., Jugloff, D. G., Zhang, G., Logan, R., Brown, S., & Eubanks, J. H. (2003). The expression of methyl CpG binding factor MeCP2 correlates with cellular differentiation in the developing rat brain and in cultured cells. Journal of Neurobiology, 55(1), 86-96. doi:10.1002/neu.10201

50. Ballas, N., Lioy, D. T., Grunseich, C., & Mandel, G. (2009). Non-cell autonomous influence of MeCP2-deficient glia on neuronal dendritic morphology. Nature Neuroscience,12(3), 311-317. doi:10.1038/nn.2275; 10.1038/nn.2275

51. Maezawa, I., Swanberg, S., Harvey, D., LaSalle, J. M., & Jin, L. W. (2009). Rett syndrome astrocytes are abnormal and spread MeCP2 deficiency through gap junctions. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 29(16), 5051-5061. doi:10.1523/JNEUROSCI.0324-09.2009; 10.1523/JNEUROSCI.0324-09.2009

52. Rastegar, M., Hotta, A., Pasceri, P., Makarem, M., Cheung, A. Y., Elliott, S., . . . Ellis, J. (2009). MECP2 isoform-specific vectors with regulated expression for rett syndrome gene therapy. PloS One, 4(8), e6810. doi:10.1371/journal.pone.0006810; 10.1371/journal.pone.0006810

53. Nguyen, M. V., Felice, C. A., Du, F., Covey, M. V., Robinson, J. K., Mandel, G., & Ballas, N. (2013). Oligodendrocyte lineage cells contribute unique features to rett syndrome neuropathology. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 33(48), 18764-18774. doi:10.1523/JNEUROSCI.2657-13.2013; 10.1523/JNEUROSCI.2657-13.2013

54. Li, Z., & Sheng, M. (2003). Some assembly required: The development of neuronal synapses. Nature Reviews.Molecular Cell Biology, 4(11), 833-841. doi:10.1038/nrm1242

Page 90: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

81

55. Dani, V. S., Chang, Q., Maffei, A., Turrigiano, G. G., Jaenisch, R., & Nelson, S. B. (2005). Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of rett syndrome. Proceedings of the National Academy of Sciences of the United States of America, 102(35), 12560-12565. doi:10.1073/pnas.0506071102

56. Luikenhuis, S., Giacometti, E., Beard, C. F., & Jaenisch, R. (2004). Expression of MeCP2 in postmitotic neurons rescues rett syndrome in mice. Proceedings of the National Academy of Sciences of the United States of America, 101(16), 6033-6038. doi:10.1073/pnas.0401626101

57. Chang, Q., Khare, G., Dani, V., Nelson, S., & Jaenisch, R. (2006). The disease progression of Mecp2 mutant mice is affected by the level of BDNF expression. Neuron,49(3), 341-348. doi:10.1016/j.neuron.2005.12.027

58. Guy, J., Gan, J., Selfridge, J., Cobb, S., & Bird, A. (2007). Reversal of neurological defects in a mouse model of rett syndrome. Science (New York, N.Y.), 315(5815), 1143-1147. doi:10.1126/science.1138389

59. Schmid, D. A., Yang, T., Ogier, M., Adams, I., Mirakhur, Y., Wang, Q., . . . Katz, D. M. (2012). A TrkB small molecule partial agonist rescues TrkB phosphorylation deficits and improves respiratory function in a mouse model of rett syndrome. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 32(5), 1803-1810. doi:10.1523/JNEUROSCI.0865-11.2012; 10.1523/JNEUROSCI.0865-11.2012

60. Tropea, D., Giacometti, E., Wilson, N. R., Beard, C., McCurry, C., Fu, D. D., . . . Sur, M. (2009). Partial reversal of rett syndrome-like symptoms in MeCP2 mutant mice.Proceedings of the National Academy of Sciences of the United States of America, 106(6), 2029-2034. doi:10.1073/pnas.0812394106; 10.1073/pnas.0812394106

61. Deogracias, R., Yazdani, M., Dekkers, M. P., Guy, J., Ionescu, M. C., Vogt, K. E., & Barde, Y. A. (2012). Fingolimod, a sphingosine-1 phosphate receptor modulator, increases BDNF levels and improves symptoms of a mouse model of rett syndrome. Proceedings of the National Academy of Sciences of the United States of America, 109(35), 14230-14235. doi:10.1073/pnas.1206093109; 10.1073/pnas.1206093109

62. Ricciardi, S., Boggio, E. M., Grosso, S., Lonetti, G., Forlani, G., Stefanelli, G., . . . Broccoli, V. (2011). Reduced AKT/mTOR signaling and protein synthesis dysregulation in a rett syndrome animal model. Human Molecular Genetics, 20(6), 1182-1196. doi:10.1093/hmg/ddq563; 10.1093/hmg/ddq563

63. Marchetto, M. C., Carromeu, C., Acab, A., Yu, D., Yeo, G. W., Mu, Y., . . . Muotri, A. R. (2010). A model for neural development and treatment of rett syndrome using human induced pluripotent stem cells. Cell, 143(4), 527-539. doi:10.1016/j.cell.2010.10.016; 10.1016/j.cell.2010.10.016

64. Cheung, A. Y., Horvath, L. M., Grafodatskaya, D., Pasceri, P., Weksberg, R., Hotta, A., . . . Ellis, J. (2011). Isolation of MECP2-null rett syndrome patient hiPS cells and isogenic controls through X-chromosome inactivation. Human Molecular Genetics, 20(11), 2103-2115. doi:10.1093/hmg/ddr093; 10.1093/hmg/ddr093

Page 91: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

82

65. Farra, N., Zhang, W. B., Pasceri, P., Eubanks, J. H., Salter, M. W., & Ellis, J. (2012). Rett syndrome induced pluripotent stem cell-derived neurons reveal novel neurophysiological alterations. Molecular Psychiatry, 17(12), 1261-1271. doi:10.1038/mp.2011.180; 10.1038/mp.2011.180

66. Kim, K. Y., Hysolli, E., & Park, I. H. (2011). Neuronal maturation defect in induced pluripotent stem cells from patients with rett syndrome. Proceedings of the National Academy of Sciences of the United States of America, 108(34), 14169-14174. doi:10.1073/pnas.1018979108; 10.1073/pnas.1018979108

67. Yazdani, M., Deogracias, R., Guy, J., Poot, R. A., Bird, A., & Barde, Y. A. (2012). Disease modeling using embryonic stem cells: MeCP2 regulates nuclear size and RNA synthesis in neurons. Stem Cells (Dayton, Ohio), 30(10), 2128-2139. doi:10.1002/stem.1180; 10.1002/stem.1180

68. Takahashi, K., & Yamanaka, S. (2006). Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 126(4), 663-676. doi:10.1016/j.cell.2006.07.024

69. Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., & Yamanaka, S. (2007). Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell, 131(5), 861-872. doi:10.1016/j.cell.2007.11.019

70. Robinton, D. A., & Daley, G. Q.The promise of induced pluripotent stem cells in research and therapy. Nature, 481(7381), 295-305. doi:10.1038/nature10761

71. Dimos, J. T., Rodolfa, K. T., Niakan, K. K., Weisenthal, L. M., Mitsumoto, H., Chung, W., . . . Eggan, K. (2008). Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science (New York, N.Y.), 321(5893), 1218-1221. doi:10.1126/science.1158799; 10.1126/science.1158799

72. Park, I. H., Arora, N., Huo, H., Maherali, N., Ahfeldt, T., Shimamura, A., . . . Daley, G. Q. (2008). Disease-specific induced pluripotent stem cells. Cell, 134(5), 877-886. doi:10.1016/j.cell.2008.07.041; 10.1016/j.cell.2008.07.041

73. Ebert, A. D., Yu, J., Rose, F. F.,Jr, Mattis, V. B., Lorson, C. L., Thomson, J. A., & Svendsen, C. N. (2009). Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature, 457(7227), 277-280. doi:10.1038/nature07677; 10.1038/nature07677

74. Hotta, A., Cheung, A. Y., Farra, N., Vijayaragavan, K., Seguin, C. A., Draper, J. S., . . . Ellis, J. (2009). Isolation of human iPS cells using EOS lentiviral vectors to select for pluripotency. Nature Methods, 6(5), 370-376. doi:10.1038/nmeth.1325; 10.1038/nmeth.1325

75. Lee, G., Papapetrou, E. P., Kim, H., Chambers, S. M., Tomishima, M. J., Fasano, C. A., . . . Studer, L. (2009). Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature, 461(7262), 402-406. doi:10.1038/nature08320; 10.1038/nature08320

Page 92: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

83

76. Cherry, A. B., & Daley, G. Q. (2013). Reprogrammed cells for disease modeling and regenerative medicine. Annual Review of Medicine, 64, 277-290. doi:10.1146/annurev-med-050311-163324; 10.1146/annurev-med-050311-163324

77. Zhang, S. C., Wernig, M., Duncan, I. D., Brustle, O., & Thomson, J. A. (2001). In vitro differentiation of transplantable neural precursors from human embryonic stem cells.Nature Biotechnology, 19(12), 1129-1133. doi:10.1038/nbt1201-1129

78. Li, X. J., Zhang, X., Johnson, M. A., Wang, Z. B., Lavaute, T., & Zhang, S. C. (2009). Coordination of sonic hedgehog and wnt signaling determines ventral and dorsal telencephalic neuron types from human embryonic stem cells. Development (Cambridge, England), 136(23), 4055-4063. doi:10.1242/dev.036624

79. Chambers, S. M., Fasano, C. A., Papapetrou, E. P., Tomishima, M., Sadelain, M., & Studer, L. (2009). Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nature Biotechnology, 27(3), 275-280. doi:10.1038/nbt.1529; 10.1038/nbt.1529

80. Brennand, K. J., Simone, A., Jou, J., Gelboin-Burkhart, C., Tran, N., Sangar, S., . . . Gage, F. H. (2011). Modelling schizophrenia using human induced pluripotent stem cells. Nature, 473(7346), 221-225. doi:10.1038/nature09915; 10.1038/nature09915

81. Djuric, U., Cheung, A.Y., Zhang, W., Mok, R., Lai, W., Piekna, A., Hendry, J. A., Ross, P. J., Pasceri, P., Kim, D., Salter, M. W., Ellis, J. (TBD). The E1 isoform of MECP2 controls neuronal form and function in Rett syndrome patient neurons. In Preparation.

82. Zhang, Y., Pak, C., Han, Y., Ahlenius, H., Zhang, Z., Chanda, S., . . . Sudhof, T. C. (2013). Rapid single-step induction of functional neurons from human pluripotent stem cells.Neuron, 78(5), 785-798. doi:10.1016/j.neuron.2013.05.029; 10.1016/j.neuron.2013.05.029

83. Ananiev, G., Williams, E. C., Li, H., & Chang, Q. (2011). Isogenic pairs of wild type and mutant induced pluripotent stem cell (iPSC) lines from rett syndrome patients as in vitro disease model. PloS One, 6(9), e25255. doi:10.1371/journal.pone.0025255; 10.1371/journal.pone.0025255

84. Saha, K., & Jaenisch, R. (2009). Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell, 5(6), 584-595. doi:10.1016/j.stem.2009.11.009; 10.1016/j.stem.2009.11.009

85. Gianakopoulos, P. J., Zhang, Y., Pencea, N., Orlic-Milacic, M., Mittal, K., Windpassinger, C., . . . Vincent, J. B. (2012). Mutations in MECP2 exon 1 in classical rett patients disrupt MECP2_e1 transcription, but not transcription of MECP2_e2. American Journal of Medical Genetics.Part B, Neuropsychiatric Genetics : The Official Publication of the International Society of Psychiatric Genetics, 159B(2), 210-216. doi:10.1002/ajmg.b.32015; 10.1002/ajmg.b.32015

Page 93: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

84

86. Quenard, A., Yilmaz, S., Fontaine, H., Bienvenu, T., Moncla, A., des Portes, V., . . . Philippe, C. (2006). Deleterious mutations in exon 1 of MECP2 in rett syndrome.European Journal of Medical Genetics, 49(4), 313-322. doi:10.1016/j.ejmg.2005.11.002

87. Fichou, Y., Nectoux, J., Bahi-Buisson, N., Rosas-Vargas, H., Girard, B., Chelly, J., & Bienvenu, T. (2009). The first missense mutation causing rett syndrome specifically affecting the MeCP2_e1 isoform. Neurogenetics, 10(2), 127-133. doi:10.1007/s10048-008-0161-1; 10.1007/s10048-008-0161-1

88. Saunders, C. J., Minassian, B. E., Chow, E. W., Zhao, W., & Vincent, J. B. (2009). Novel exon 1 mutations in MECP2 implicate isoform MeCP2_e1 in classical rett syndrome. American Journal of Medical Genetics.Part A, 149A(5), 1019-1023. doi:10.1002/ajmg.a.32776; 10.1002/ajmg.a.32776

89. Yasui, D. H., Gonzales, M. L., Aflatooni, J. O., Crary, F. K., Hu, D. J., Gavino, B. J., . . . Lasalle, J. M. (2014). Mice with an isoform-ablating Mecp2 exon 1 mutation recapitulate the neurologic deficits of rett syndrome. Human Molecular Genetics, doi:10.1093/hmg/ddt640

90. Kumar, V., Zhang, M. X., Swank, M. W., Kunz, J., & Wu, G. Y. (2005). Regulation of dendritic morphogenesis by ras-PI3K-akt-mTOR and ras-MAPK signaling pathways. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 25(49), 11288-11299. doi:10.1523/JNEUROSCI.2284-05.2005

91. Jaworski, J., Spangler, S., Seeburg, D. P., Hoogenraad, C. C., & Sheng, M. (2005). Control of dendritic arborization by the phosphoinositide-3'-kinase-akt-mammalian target of rapamycin pathway. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 25(49), 11300-11312. doi:10.1523/JNEUROSCI.2270-05.2005

92. Naska, S., Park, K. J., Hannigan, G. E., Dedhar, S., Miller, F. D., & Kaplan, D. R. (2006). An essential role for the integrin-linked kinase-glycogen synthase kinase-3 beta pathway during dendrite initiation and growth. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 26(51), 13344-13356. doi:10.1523/JNEUROSCI.4462-06.2006

93. Redmond, L., & Ghosh, A. (2001). The role of notch and rho GTPase signaling in the control of dendritic development. Current Opinion in Neurobiology, 11(1), 111-117

94. Lee, T., Winter, C., Marticke, S. S., Lee, A., & Luo, L. (2000). Essential roles of drosophila RhoA in the regulation of neuroblast proliferation and dendritic but not axonal morphogenesis. Neuron, 25(2), 307-316.

95. Ruchhoeft, M. L., Ohnuma, S., McNeill, L., Holt, C. E., & Harris, W. A. (1999). The neuronal architecture of xenopus retinal ganglion cells is sculpted by rho-family GTPases in vivo. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 19(19), 8454-8463.

Page 94: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

85

96. Li, Z., Van Aelst, L., & Cline, H. T. (2000). Rho GTPases regulate distinct aspects of dendritic arbor growth in xenopus central neurons in vivo. Nature Neuroscience, 3(3), 217-225. doi:10.1038/72920

97. Chen, H., & Firestein, B. L. (2007). RhoA regulates dendrite branching in hippocampal neurons by decreasing cypin protein levels. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 27(31), 8378-8386. doi:10.1523/JNEUROSCI.0872-07.2007

98. Threadgill, R., Bobb, K., & Ghosh, A. (1997). Regulation of dendritic growth and remodeling by rho, rac, and Cdc42. Neuron, 19(3), 625-634.

99. Wong, W. T., Faulkner-Jones, B. E., Sanes, J. R., & Wong, R. O. (2000). Rapid dendritic remodeling in the developing retina: Dependence on neurotransmission and reciprocal regulation by rac and rho. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 20(13), 5024-5036.

100. Sestan, N., Artavanis-Tsakonas, S., & Rakic, P. (1999). Contact-dependent inhibition of cortical neurite growth mediated by notch signaling. Science (New York, N.Y.),286(5440), 741-746.

101. Redmond, L., Oh, S. R., Hicks, C., Weinmaster, G., & Ghosh, A. (2000). Nuclear Notch1 signaling and the regulation of dendritic development. Nature Neuroscience,3(1), 30-40. doi:10.1038/71104

102. Borghese, L., Dolezalova, D., Opitz, T., Haupt, S., Leinhaas, A., Steinfarz, B., . . . Brustle, O. (2010). Inhibition of notch signaling in human embryonic stem cell-derived neural stem cells delays G1/S phase transition and accelerates neuronal differentiation in vitro and in vivo. Stem Cells (Dayton, Ohio), 28(5), 955-964. doi:10.1002/stem.408; 10.1002/stem.408

103. Zachariah, R. M., Olson, C. O., Ezeonwuka, C., & Rastegar, M. (2012). Novel MeCP2 isoform-specific antibody reveals the endogenous MeCP2E1 expression in murine brain, primary neurons and astrocytes. PloS One, 7(11), e49763. doi:10.1371/journal.pone.0049763; 10.1371/journal.pone.0049763

104. Olson, C. O., Zachariah, R. M., Ezeonwuka, C. D., Liyanage, V. R., & Rastegar, M. (2014). Brain region-specific expression of MeCP2 isoforms correlates with DNA methylation within Mecp2 regulatory elements. PloS One, 9(3), e90645. doi:10.1371/journal.pone.0090645; 10.1371/journal.pone.0090645

105. Derecki, N. C., Cronk, J. C., Lu, Z., Xu, E., Abbott, S. B., Guyenet, P. G., & Kipnis, J. (2012). Wild-type microglia arrest pathology in a mouse model of rett syndrome. Nature,484(7392), 105-109. doi:10.1038/nature10907; 10.1038/nature10907

106. Huang, E. J., Liu, W., Fritzsch, B., Bianchi, L. M., Reichardt, L. F., & Xiang, M. (2001). Brn3a is a transcriptional regulator of soma size, target field innervation and axon pathfinding of inner ear sensory neurons. Development (Cambridge, England), 128(13), 2421-2432.

Page 95: MECP2e1 Mutation Reduces the Dendritic Complexity of Rett ... · MECP2e1 Mutation Reduces the Dendritic Complexity of Rett Syndrome Patient iPS Cell-Derived Neurons Wesley Lai Master

86

107. Mattar, P., Britz, O., Johannes, C., Nieto, M., Ma, L., Rebeyka, A., . . . Schuurmans, C. (2004). A screen for downstream effectors of Neurogenin2 in the embryonic neocortex. Developmental Biology, 273(2), 373-389. doi:10.1016/j.ydbio.2004.06.013