Top Banner
EXPERT REVIEW Modeling psychiatric disorders at the cellular and network levels KJ Brennand, A Simone, N Tran and FH Gage Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number of brain regions and produce a complex array of clinical symptoms, basic phenotypes likely exist at the level of single neurons and simple networks. Being highly heritable, it is hypothesized that these disorders are amenable to cell-based studies in vitro. Using induced pluripotent stem cell-derived neurons and/or induced neurons from fibroblasts, limitless numbers of live human neurons can now be generated from patients with a genetic background permissive to the disease state. We predict that cell-based studies will ultimately contribute to our understanding of the initiation, progression and treatment of these psychiatric disorders. Molecular Psychiatry advance online publication, 3 April 2012; doi:10.1038/mp.2012.20 Keywords: autism spectrum disorders; bipolar disorder; neurons; schizophrenia; stem cells Introduction Autism spectrum disorders (ASDs), schizophrenia (SCZD) and bipolar disorder (BD) combine to affect nearly 1 in 30 adults throughout the global popula- tion. 1 While these psychiatric disorders are character- ized by markedly different clinical phenotypes, recent genetic studies have suggested that they may share common underlying molecular causes. ASD, SCZD and BD are believed to be developmental in origin, resulting from events that occur in fetal development or early childhood. The molecular mechanism of these disorders is difficult to study in patients or animal models because of the complex genetic etiologies and varying environmental effects contri- buting to disease. Cell-based models produce live human neurons with genetic backgrounds permissive to the disease state. Temporal analysis of disease initiation and progression can be studied in the cell type relevant to disease. Human cell-based models can be ideal experimental paradigms with which to investigate disease mechanisms; for example, studies of amyo- trophic lateral sclerosis have revealed a non-cell autonomous contribution of glial cells to this neuronal disease. 2,3 In order to be studied using an in vitro model, a given disease must (1) be highly genetic, ensuring that cultured cells are afflicted by disease in the absence of any potentially unresolved environ- mental factors and (2) affect a cell type that can survive and, ideally, be robustly expanded when cultured in vitro. With respect to the first criterion, twin studies have calculated the heritability of ASD, SCZD and BD to be between 70 and 90%. 4–6 Our hypothesis is that this genetic predisposition to psychiatric illness is sufficient that cultured neurons will consistently undergo disease initiation and progression. Regarding the second criterion, while mature neurons are post- mitotic and cannot be expanded in culture, conditions for the survival of human neurons are well described, 7 and robust quantities of neurons for study can be generated through the growth and subsequent differ- entiation of proliferative neural progenitor cells. The ability to compare cellular and network proper- ties of live human neurons in vitro represents an important new approach with which to study psy- chiatric disease because live human neurons from patients or controls are exceedingly rare. Recently, three new sources of live human neurons have been reported: primary olfactory neural precursors, neurons differentiated from human-derived induced pluripo- tent stem cells (hiPSC neurons) and induced neurons (iNeurons) generated from primary patient fibroblasts. Although olfactory neural precursors are capable of self-renewal and differentiation to mature neurons, 8,9 olfactory neural precursors cannot yield cells from the neural lineages specifically implicated in psychiatric disorders, such as GABAergic or dopaminergic neurons. Because we believe it is critical that the relevant cell type affected in the disease state be studied, we will therefore focus on in vitro models of psychiatric disease utilizing hiPSC neurons and iNeurons (Figure 1). While generally considered to be whole-brain disorders, we suggest that ASD, SCZD and BD can be broken down to component aberrations at the Received 18 November 2011; revised 26 January 2012; accepted 16 February 2012 Correspondence: FH Gage, PhD, Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA. E-mail: [email protected] Molecular Psychiatry (2012), 1–15 & 2012 Macmillan Publishers Limited All rights reserved 1359-4184/12 www.nature.com/mp
15

Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

Feb 21, 2021

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

EXPERT REVIEW

Modeling psychiatric disorders at the cellular andnetwork levelsKJ Brennand, A Simone, N Tran and FH Gage

Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA

Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolardisorder affect a number of brain regions and produce a complex array of clinical symptoms,basic phenotypes likely exist at the level of single neurons and simple networks. Being highlyheritable, it is hypothesized that these disorders are amenable to cell-based studies in vitro.Using induced pluripotent stem cell-derived neurons and/or induced neurons from fibroblasts,limitless numbers of live human neurons can now be generated from patients with a geneticbackground permissive to the disease state. We predict that cell-based studies will ultimatelycontribute to our understanding of the initiation, progression and treatment of thesepsychiatric disorders.Molecular Psychiatry advance online publication, 3 April 2012; doi:10.1038/mp.2012.20

Keywords: autism spectrum disorders; bipolar disorder; neurons; schizophrenia; stem cells

Introduction

Autism spectrum disorders (ASDs), schizophrenia(SCZD) and bipolar disorder (BD) combine to affectnearly 1 in 30 adults throughout the global popula-tion.1 While these psychiatric disorders are character-ized by markedly different clinical phenotypes, recentgenetic studies have suggested that they may sharecommon underlying molecular causes. ASD, SCZDand BD are believed to be developmental in origin,resulting from events that occur in fetal developmentor early childhood. The molecular mechanism ofthese disorders is difficult to study in patients oranimal models because of the complex geneticetiologies and varying environmental effects contri-buting to disease.

Cell-based models produce live human neuronswith genetic backgrounds permissive to the diseasestate. Temporal analysis of disease initiation andprogression can be studied in the cell type relevantto disease. Human cell-based models can be idealexperimental paradigms with which to investigatedisease mechanisms; for example, studies of amyo-trophic lateral sclerosis have revealed a non-cellautonomous contribution of glial cells to this neuronaldisease.2,3 In order to be studied using an in vitromodel, a given disease must (1) be highly genetic,ensuring that cultured cells are afflicted by disease inthe absence of any potentially unresolved environ-mental factors and (2) affect a cell type that can survive

and, ideally, be robustly expanded when culturedin vitro. With respect to the first criterion, twin studieshave calculated the heritability of ASD, SCZD and BDto be between 70 and 90%.4–6 Our hypothesis is thatthis genetic predisposition to psychiatric illness issufficient that cultured neurons will consistentlyundergo disease initiation and progression. Regardingthe second criterion, while mature neurons are post-mitotic and cannot be expanded in culture, conditionsfor the survival of human neurons are well described,7

and robust quantities of neurons for study can begenerated through the growth and subsequent differ-entiation of proliferative neural progenitor cells.

The ability to compare cellular and network proper-ties of live human neurons in vitro represents animportant new approach with which to study psy-chiatric disease because live human neurons frompatients or controls are exceedingly rare. Recently,three new sources of live human neurons have beenreported: primary olfactory neural precursors, neuronsdifferentiated from human-derived induced pluripo-tent stem cells (hiPSC neurons) and induced neurons(iNeurons) generated from primary patient fibroblasts.Although olfactory neural precursors are capable ofself-renewal and differentiation to mature neurons,8,9

olfactory neural precursors cannot yield cells from theneural lineages specifically implicated in psychiatricdisorders, such as GABAergic or dopaminergicneurons. Because we believe it is critical that therelevant cell type affected in the disease state bestudied, we will therefore focus on in vitro models ofpsychiatric disease utilizing hiPSC neurons andiNeurons (Figure 1).

While generally considered to be whole-braindisorders, we suggest that ASD, SCZD and BD canbe broken down to component aberrations at the

Received 18 November 2011; revised 26 January 2012; accepted16 February 2012

Correspondence: FH Gage, PhD, Laboratory of Genetics, SalkInstitute for Biological Studies, 10010 North Torrey Pines Road,La Jolla, CA 92037, USA.E-mail: [email protected]

Molecular Psychiatry (2012), 1–15& 2012 Macmillan Publishers Limited All rights reserved 1359-4184/12

www.nature.com/mp

Page 2: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

cellular and/or network levels. For example, at thecellular level, the subtle synaptic defects that arebelieved to contribute to illness can be studied withcell-based models. Furthermore, while the cyclicalbehavioral swings of BD cannot be reproduced,patterns of spontaneous and stimulated neuronalnetwork activity can be measured in vitro. Usingcell-based models, one can study ASD, SCZD and BDby observing the abnormal development of neuronsand their circuitry in vitro.

In this review, we will discuss (1) post-mortem andanimal studies demonstrating that cellular pheno-types exist at the neuronal level in these disorders(Tables 1 and 2), (2) functional magnetic resonanceimaging (fMRI) and electrophysiological evidencefrom humans and rodents suggesting that networkdefects contribute to these disorders (Tables 1 and 2)and (3) the recent findings of novel in vitro models ofpsychiatric disorders (Table 3).

Evidence for cellular phenotypes in psychiatricdisorders

Aberrant neuronal connectivity, as assessed by den-dritic arborization and synaptic density, is a char-acteristic that appears to be shared between ASD,SCZD and BD. Perturbed neuronal migration has alsobeen linked to psychiatric disorders, althoughwhether this contributes to, or functions separatelyfrom, abnormal neuronal connectivity remains to bedemonstrated.

Altered dendritic arborizationAt onset, ASD is often characterized by excessive brainvolume; MRI studies observed increased cerebral whitematter volume in 2- to 4-year-old autistic children,10–12

which has been correlated with an excess number ofneurons in the prefrontal cortex.13 Over the lifetime ofthe ASD patient, however, the brain overgrowthphenotype is typically reversed; studies of adults withASD have observed cortical thinning14,15 and reducedfrontal lobe10,16,17 and corpus callosum18 volumes.While we found few anatomical post-mortem studies

of ASD, there are reports of reduced dendritic arboriza-tion in hippocampal neurons in two cases of ASD.19 InRett syndrome (RTT), a severe and rare ASD caused bythe mutation of the MECP2 gene,20,21 post-mortemstudies report reduced neuronal cell size and dendriticarborization throughout the cortex.22 Reports of fragileX syndrome (FX), another monogenetic form of ASD,have been less conclusive: while one study reporteddifferences in dendritic arborization following in vitrodifferentiation of neurospheres derived from post-mortem human FXS brain tissue,23 a second groupfailed to observe significant differences in a similarstudy.24

Decreased whole-brain volume is consistently ob-served in SCZD,25–27 particularly in the gray matter of thefrontal cortex, temporal lobe (particularly the hippo-campus and amygdala) and the basal ganglia,27–29 andlongitudinal studies report that progressive brainvolume declines for at least 20 years after the onsetof symptoms. Post-mortem studies of brains frompatients with SCZD have not found evidence ofneuronal loss; instead, they observe smaller neuronalsomas30,31 reduced dendritic arborizations31,32 andincreased neuronal density without changes in abso-lute cell number in the cortex and hippocampus.30,33

Decreased brain volumes in the limbic system,particularly the amygdala and hippocampus, and inthe frontal cortex are associated with BD.34,35 Itremains unclear, however, whether brain volumechanges are a preexisting factor contributing to thedevelopment of BD or a consequence of prolongedillness; one recent study suggests that brain volumechanges are more tightly correlated to active psycho-sis than BD.36 Despite observations of diminishedbrain volumes and reduced neuronal density in BDpatients,37,38 we found no report of altered dendriticarborization or synaptic density in post-mortemstudies of BD patient brains.

Mouse models of a number of psychiatric disordershave been developed. For the most part, these micehave reduced expression of rare, highly penetrantgenes implicated in ASD, SCZD or BD. RTT (Mecp2null) mouse brains show a reduction in neuronal size;39

Figure 1 Cell-based modeling of psychiatric disorders. Fibroblast cells obtained from patients can be used to generate livehuman neurons with a genetic background known to produce the disease state. Fibroblasts can be reprogrammed to human-derived induced pluripotent stem cells (hiPSCs) by transient expression of OCT4, SOX2, KLF4 and cMYC and thensubsequently differentiated into mature neurons. Alternately, fibroblasts can be directly converted into a neuronal fate bytransient expression of ASCL1, BRN2, MYT1L and NEUROD.

iPSC modeling of psychiatric disordersKJ Brennand et al

2

Molecular Psychiatry

Page 3: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

Table

1S

um

mary

of

pu

bli

shed

cell

ula

ran

dn

etw

ork

ph

en

oty

pes

inh

um

an

SC

ZD

pati

en

ts

Dis

ease

Stu

dy

Bra

inre

gio

n/c

ell

typ

eO

bse

rvati

on

Refe

ren

ce

AS

DM

RI,

post

-m

ort

em

Cere

bru

mIn

cre

ase

dcere

bra

lw

hit

em

att

er

volu

me

inch

ild

ren

,excess

nu

mber

of

neu

ron

sin

pre

fron

tal

cort

ex

inch

ild

ren

Carp

er

et

al.

,10

Cou

rch

esn

e,1

1H

azle

tet

al.

,12

Cou

rch

esn

eet

al.

13

AS

DM

RI

Cere

bru

m,

corp

us

call

osu

mR

ed

uced

fron

tal

an

dp

ari

eta

llo

be

gra

ym

att

er

volu

me

inold

er

ch

ild

ren

an

dad

ult

s,in

cre

ase

dven

tric

ula

rvolu

me,

red

uced

corp

us

call

osu

mvolu

me

Cou

rch

esn

eet

al.

,14

Sch

mit

zet

al.

,16

Bru

net

al.

,17

Wri

gh

tet

al.

,27

Fra

zie

ran

dH

ard

en

18

AS

DP

ost

-mort

em

Hip

pocam

pu

s,cort

ex

Red

uced

den

dri

tic

arb

ori

zati

on

inh

ipp

ocam

pu

s,gre

ate

rcort

ical

pyra

mid

al

spin

ed

en

sity

corr

ela

ted

wit

hd

ecre

ase

dcogn

itiv

efu

ncti

on

,lo

ssof

vert

ical

an

dh

ori

zon

tal

org

an

izati

on

of

cort

ical

layers

Raym

on

det

al.

,19

Hu

tsle

ran

dZ

han

g,4

8

Wegie

let

al.

72

AS

DfM

RI

Cort

ex

Incre

ase

dcon

necti

vit

ybetw

een

pro

xim

al:

post

eri

or

cin

gu

late

/para

hip

pocam

pal

gyru

s,d

ecre

ase

dcon

necti

vit

ybetw

een

dis

tal:

fron

tal

lobe-p

ari

eta

llo

be,

insu

lar

cort

ices-

som

ato

sen

sory

cort

ices

of

am

ygd

ala

,fr

on

tal

cort

ex-p

ost

eri

or

cin

gu

late

d,

decre

ase

din

terh

em

isp

heri

csy

nch

ron

izati

on

Mon

ket

al.

,80

Ken

ned

yan

dC

ou

rch

esn

e,8

1

Ebis

ch

et

al.

,82

Din

stein

et

al.

,83

Ken

ned

yan

dC

ou

rch

esn

e84

AS

D(R

TT

)P

ost

-mort

em

Cort

ex,

hip

pocam

pu

sR

ed

uced

cell

size

an

dd

en

dri

tic

arb

ori

zati

on

incort

ex,

red

uced

nu

mber

of

den

dri

tic

spin

es

inh

ipp

ocam

pu

s

Bau

man

et

al.

,22

Ch

ap

leau

et

al.

49

AS

D(F

X)

Invit

roP

ost

-mort

em

neu

rosp

here

cu

ltu

re,

feta

lcort

ical

NP

Ccu

ltu

re

Neu

ron

sw

ith

few

er

an

dsh

ort

er

neu

rite

s,sm

all

er

cell

bod

yvolu

me

an

dd

ecre

ase

dgli

al

dif

fere

nti

ati

on

;n

orm

al

neu

rogen

esi

sby

feta

lN

PC

s

Cast

ren

et

al.

,23

Bh

att

ach

ary

ya

et

al.

24

AS

D(F

X)

Post

-mort

em

Cort

ical

pyra

mid

al

cell

sL

on

ger,

more

slen

der

den

dri

tic

spin

esh

ap

eIr

win

et

al.

50

SC

ZD

MR

IG

ray

matt

er

fron

tal

cort

ex,

tem

pora

llo

be,

hip

pocam

pu

s,am

ygd

ala

,basa

lgan

gli

a

Decre

ase

dvolu

me

(wh

ole

bra

in)

inearl

yp

hase

SC

ZD

Vit

aet

al.

,25

Ste

en

et

al.

,26

Th

om

pso

net

al.

,28

Ell

ison

-Wri

gh

tet

al.

29

SC

ZD

Post

-mort

em

Cort

ex,

hip

pocam

pu

sR

ed

uced

den

dri

tic

arb

ori

zati

on

,re

du

ced

som

asi

ze,

incre

ase

dn

eu

ron

al

den

sity

,d

ecre

ase

dsy

nap

tic

den

sity

Rajk

ow

ska

et

al.

,30

Kolo

meets

et

al.

,31

Bla

ck

et

al.

,32

Sele

mon

et

al.

,33

Gare

yet

al.

,51

Gla

ntz

an

dL

ew

is52,

Kolo

meets

et

al.

53

SC

ZD

fMR

IF

ron

tal/

tem

pora

llo

be

Bra

inacti

vit

yabn

orm

ali

tyin

fron

tal

an

dte

mp

ora

llo

bes

Yu

rgelu

n-T

od

det

al.

,85

Yu

rgelu

n-T

od

det

al.

86

SC

ZD

fMR

IC

ort

ex

Cort

ical

hyp

er-

acti

vit

yan

dh

yp

er-

con

necti

vit

yin

pre

fron

tal

cort

ex

at

rest

,re

du

ced

acti

vati

on

of

med

ial

pre

fron

tal

du

rin

gw

ork

ing

mem

ory

task

sin

earl

yp

hase

SC

ZD

,in

cre

ase

dfu

ncti

on

al

con

necti

vit

ybetw

een

the

ven

tral

pre

fron

tal

cort

ex

an

dp

ost

eri

or

pari

eta

lcort

ex

gre

ate

r,d

ecre

ase

dfu

ncti

on

al

con

necti

vit

ybetw

een

the

dors

al

pre

fron

tal

cort

ex

an

dp

ost

eri

or

pari

eta

lcort

ex

gre

ate

r

Wh

itfi

eld

-Gabri

eli

et

al.

,87

Tan

et

al.

88

iPSC modeling of psychiatric disordersKJ Brennand et al

3

Molecular Psychiatry

Page 4: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

and abnormalities in dendritic arborization.39–41 Con-versely, dendritic arborization defects have not beenreported in FX (Fmr1 null) mice. Many SCZD mousemodels, including Disc1 and heterozygous-null Nrg1and Erbb4 mice, have reduced neurite outgrowthand reduced dendritic complexity.42–45 Although fewgenetic mouse models of BD have been reported,neurons from mice with a point mutation in thecircadian Clock gene display complex changes indendritic morphology,46 which can be ameliorated withlithium, a drug routinely used in the treatment of BD.46

Altered synaptic densityA number of genes implicated in ASD, SCZD and BDhave been associated with synaptic maturation andfunction.47 Post-mortem synaptic spine density has notbeen adequately explored in human patients withASD. One report found that relative to controls, spinedensities on cortical pyramidal cells were greater inASD subjects, and highest spine densities were mostcommonly found in ASD subjects with lower levels ofcognitive functioning.48 Conversely, the number ofspines in dendrites of neurons from post-mortem RTTbrains is reduced.49 In FX patients, post-mortemstudies have identified abnormalities in dendriticspine shape in cortical pyramidal cells, which tendto be both longer and more slender than controls.50

Post-mortem studies of SCZD patient brains foundreduced dendritic spine density in the cortex51,52 andhippocampus.31,53 What post-mortem analysis of ASDand SCZD has failed to resolve is whether diseaseprogression reflects developmental aberrations duringneuronal differentiation or activity-dependent atrophyof neuronal dendrites or synapses in mature neurons.33

Animal studies recapitulate these synaptic defects.Using mouse models of RTT, decreased Mecp2levels have been implicated in defects of synapticcontact formation and synaptic transmission.41,54–56

Comparably, inherited mutations of Shank3, whichalso model ASD, result in reduced dendritic synapticspine induction and maturation.57 Similar to post-mortem observations of FX patient brains, Fmr1 micehave abnormally thin and elongated dendritic spinemorphology and greater spine density.58 Fmr1, thegene affected in FX, regulates the translation ofmessages important for activity-dependent synapticmodulation.59 Although a number of animal models ofASD recapitulate defects in synaptic maturation, thedirection of the change varies depending uponthe gene under investigation, which is consistent withthe hypothesis that ASD is a spectrum of complexgenetic disorders involving impaired developmentalsynaptic maturation, stabilization, elimination orpruning. Studies of mouse models of SCZD have alsoobserved synaptic defects; there is reduced hippo-campal synaptic transmission in Disc1 mice,42,43

impaired synaptic maturation and function in Nrg1mice60–62 and fewer cortical neurons with slightlysmaller spines in mouse models of the human SCZDcopy number variant at 22q11.2.63–65 In mice withreduced Reelin (Reln) expression (putative models ofT

able

1C

on

tin

ued

Dis

ease

Stu

dy

Bra

inre

gio

n/c

ell

typ

eO

bse

rvati

on

Refe

ren

ce

SC

ZD

An

ato

mic

al

netw

ork

san

aly

sis

Cort

ex

Cort

ical

org

an

izati

on

alt

ere

d,

loss

of

netw

ork

hu

bs

infr

on

tal

cort

ex,em

erg

en

ce

of

hu

bs

ou

tsid

ecort

ex

Bass

ett

et

al.

89

SC

ZD

Post

-mort

em

,P

ET

Su

bst

an

ian

igra

,st

riatu

m,

cort

ex

Incre

ase

dD

Are

cep

tor

sen

siti

vit

y,in

cre

ase

dD

Are

cep

tor

levels

insu

bst

an

ian

igra

,corr

ela

tion

of

DA

recep

tor

exp

ress

ion

level

inte

mp

ora

lcort

ex

an

dst

riatu

mto

posi

tive

sym

pto

ms

of

SC

ZD

Ow

en

et

al.

,91

Kess

ler

et

al.

,90

SC

ZD

Ph

arm

acolo

gy

Not

dete

rmin

ed

NM

DA

an

tagon

ist

keta

min

ein

du

ces

SC

ZD

-lik

esy

mp

tom

s,M

GL

UR

2/3

agon

ists

am

eli

ora

teth

em

Kry

stal

et

al.

,92

Pati

let

al.

93

SC

ZD

Post

-mort

em

Hip

pocam

pu

s,cort

ex

Red

uced

GL

Ure

cep

tor

exp

ress

ion

Mead

or-

Wood

ruff

an

dH

ealy

94

BD

MR

IL

imbic

syst

em

(am

ygd

ala

/h

ipp

ocam

pu

s)R

ed

uced

bra

involu

me:

lim

bic

syst

em

(am

ygd

ala

,h

ipp

ocam

pu

s,fr

on

tal

cort

ex)

inad

ole

scen

tsK

arc

hem

skiy

et

al.

,34

Fra

zie

r,35

BD

Post

-mort

em

En

torh

inal

cort

ex

Decre

ase

dcell

nu

mber

an

dd

en

sity

of

GA

BA

neu

ron

sR

ajk

ow

ska

et

al.

,37

Pan

tazop

ou

los

et

al.

38

Abbre

via

tion

s:A

SD

,au

tism

spectr

um

dis

ord

er;

BD

,bip

ola

rd

isord

er;

DA

,d

op

am

ine;

GL

U,

glu

tam

ate

;F

X,

fragil

eX

syn

dro

me;

fMR

I,fu

ncti

on

al

magn

eti

cre

son

an

ce

imagin

g;

MR

I,m

agn

eti

cre

son

an

ce

imagin

g;

NP

C,

neu

ral

pro

gen

itor

cell

;R

TT

,R

ett

syn

dro

me;

SC

ZD

,sc

hiz

op

hre

nia

dis

ord

er.

iPSC modeling of psychiatric disordersKJ Brennand et al

4

Molecular Psychiatry

Page 5: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

Table

2S

um

mary

of

pu

bli

shed

cell

ula

ran

dn

etw

ork

ph

en

oty

pes

inro

den

tm

od

els

of

SC

ZD

Dis

ease

Gen

eB

rain

regio

n/c

ell

typ

eO

bse

rvati

on

Refe

ren

ce

AS

DC

ntn

ap

2C

ort

ex,

GA

BA

Imp

air

ed

mig

rati

on

of

cort

ical

pro

jecti

on

neu

ron

s,re

du

ced

GA

BA

erg

icn

eu

ron

s,d

ecre

ase

dn

eu

ral

syn

ch

ron

yP

en

agari

kan

oet

al.

100

AS

DS

han

k3

Hip

pocam

pal

neu

ral

cu

ltu

res

Red

uced

den

dri

tic

spin

ein

du

cti

on

/matu

rati

on

invit

roD

ura

nd

et

al.

57

AS

D(R

TT

)M

ecp

2H

ipp

ocam

pu

s,cort

ex,

cere

bell

um

,re

tin

ogen

icu

late

syn

ap

se,

NP

Cs

Red

ucti

on

inn

eu

ron

al

size,

den

dri

tic

arb

ori

zati

on

abn

orm

ali

ties,

thin

ner

cort

ical

layers

,re

du

ced

spin

ed

en

sity

,d

efe

cts

insy

nap

tic

matu

rati

on

an

dsy

nap

tic

tran

smis

sion

,im

pair

ed

exp

eri

en

ce

dep

en

den

tre

mod

eli

ng,

an

dalt

ere

dgen

eexp

ress

ion

Ch

en

et

al.

,39

Kis

hi

an

dM

ackli

s,40

Sm

rtet

al.

,41

Asa

ka

et

al.

,54

More

tti

et

al.

,55

Nels

on

et

al.

,56

Nou

tel

et

al.

,98

Dan

iet

al.

99

AS

D(F

X)

Fm

r1C

ort

ex,

neu

rosp

here

cu

ltu

reT

hin

an

delo

ngate

dd

en

dri

tic

spin

es

on

pyra

mid

al

neu

ron

s,in

cre

ase

dsp

ine

den

sity

alo

ng

ap

ical

den

dri

tes,

invit

rod

iffe

ren

tiate

dn

eu

ron

sh

ave

few

er

an

dsh

ort

er

neu

rite

san

da

small

er

cell

bod

yvolu

me

Com

ery

et

al.

,58

Cast

ren

et

al.

23

SC

ZD

Dis

c1

Cort

ex,

hip

pocam

pu

sR

ed

ucti

on

infe

tal

cort

ical

neu

ral

pro

gen

itor

pro

life

rati

on

an

dp

rem

atu

ren

eu

ral

dif

fere

nti

ati

on

,re

du

ced

cort

ical

mig

rati

on

,d

imin

ish

ed

resp

on

seto

cA

MP

-sen

siti

ve

rep

uls

ive

cu

es,

red

uced

neu

rite

ou

tgro

wth

,sy

nap

tic

tran

smis

sion

an

dalt

ere

dd

istr

ibu

tion

of

hip

pocam

pal

neu

ron

s

Mao

et

al.

,67

Kam

iya

et

al.

,74

Kvajo

et

al.

,42

Li

et

al.

43

SC

ZD

Dis

c1

kn

ockd

ow

nH

ipp

ocam

pal

ad

ult

-born

neu

ron

sA

du

ltn

ew

born

neu

ron

ssh

ow

accele

rate

dd

en

dri

tic

develo

pm

en

tan

dsy

nap

sefo

rmati

on

,d

efe

cts

inaxon

al

targ

eti

ng,

en

han

ced

excit

abil

ity

Fau

lker

et

al.

,70

Du

an

et

al.

69

SC

ZD

Nrg

1C

ort

ex,

peri

ph

era

ln

erv

es

Aberr

an

tta

ngen

tial

mig

rati

on

of

neu

ron

sd

eri

ved

from

the

ven

tral

tele

ncep

halo

n,

imp

air

ed

syn

ap

tic

matu

rati

on

an

dfu

ncti

on

,h

yp

om

yeli

nati

on

Lop

ez-B

en

dit

oet

al.

,44

Barr

os

et

al.

,60

Ch

en

et

al.

62

SC

ZD

Erb

B4

Hip

pocam

pu

s,cort

ex

Aberr

an

tn

eu

rite

ou

tgro

wth

an

dsy

nap

sem

atu

rati

on

,re

du

ced

lon

g-t

erm

pote

nti

ati

on

,su

pp

ress

ed

Src

-dep

en

den

ten

han

cem

en

tof

NM

DA

Rre

spon

ses

du

rin

gth

eta

-bu

rst

stim

ula

tion

,re

du

ced

excit

ato

ryin

pu

ton

toG

AB

Aerg

icn

eu

ron

s,P

PI

defi

cit

s

Kri

vosh

eya

et

al.

,45

Pit

ch

er

et

al.

,61

Pit

ch

er

et

al.

,101

Barr

os

et

al.

,60

Li

et

al.

,95

Ch

en

et

al.

62

SC

ZD

22q11.2

Cort

ex,

hip

pocam

pu

sF

ew

er

cort

ical

neu

ron

sw

ith

small

er

spin

es,

alt

ere

dsh

ort

-an

dlo

ng-

term

syn

ap

tic

pla

stic

ity

an

dcalc

ium

kin

eti

cs,

imp

air

ed

hip

pocam

pal-

pre

fron

tal

syn

ch

ron

y

Fen

elo

net

al.

63

Earl

set

al.

,64

Sig

urd

sson

et

al.

65

SC

ZD

Reln

Cort

ex,

hip

pocam

pu

sD

ecre

ase

dd

en

dri

tic

spin

em

atu

rati

on

,d

en

sity

an

dp

last

icit

yP

ap

pas

et

al.

66

SC

ZD

Dtn

bp

1G

AB

AD

ecre

ase

dP

PI,

red

uced

evoked

gam

ma

acti

vit

yC

arl

son

et

al.

104

BD

Clo

ck

Str

iatu

m(n

ucle

us

accu

mben

s)In

cre

ase

dle

ngth

an

dcom

ple

xit

yof

den

dri

tes,

norm

al

syn

ap

tic

den

sity

,d

ysf

un

cti

on

al

gam

ma

acti

vit

yacro

ssli

mbic

cir

cu

its,

imp

roved

by

lith

ium

treatm

en

t

Dzir

asa

et

al.

46

Abbre

via

tion

s:A

SD

,au

tism

spectr

um

dis

ord

er;

BD

,bip

ola

rd

isord

er;

FX

,fr

agil

eX

syn

dro

me;

PP

I,p

rep

uls

ein

hib

itio

n;

RT

T,

Rett

syn

dro

me;

SC

ZD

,sc

hiz

op

hre

nia

dis

ord

er.

iPSC modeling of psychiatric disordersKJ Brennand et al

5

Molecular Psychiatry

Page 6: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

Table

3S

um

mary

of

pu

bli

shed

rep

ort

sof

hiP

SC

-base

dm

od

els

of

AS

D,

SC

ZD

an

dB

D

Dis

ease

Refe

ren

ce

Gen

eti

cm

uta

tion

Neu

ron

al

ph

en

oty

pe

hiP

SC

meth

od

Sou

rce

of

cell

sP

ati

en

tse

x;

age

at

bio

psy

(years

);avail

able

ph

en

oty

pic

info

rmati

on

RT

TC

heu

ng

et

al.

122

MeC

P2

(D3–4,

T158

M,

R306C

)D

ecre

ase

dso

ma

size

Retr

ovir

us:

fou

rfa

cto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

)

Fib

robla

st:

pati

en

tbio

psy

(1)

an

dC

ori

ell

GM

11270

(2),

GM

17880

(3)

(1)

Fem

ale

;6

gro

wth

an

dd

evelo

pm

en

tal

dela

y,in

abil

ity

tow

alk

wit

hou

tass

ista

nce,

ata

xia

,n

on

verb

al,

has

no

han

du

sean

dcon

stan

tre

peti

tive

han

dm

oti

on

s,so

me

trem

or,

has

had

ep

ilep

tic

seiz

ure

san

dsi

gn

ific

an

tabn

orm

al

ele

ctr

oen

cep

halo

gra

m,

teeth

gri

nd

ing,

som

esl

eep

dif

ficu

ltie

s,an

dbre

ath

hold

ing

an

dh

yp

erv

en

tila

tion

(2)

Fem

ale

;8;

norm

al

lyso

som

al

en

zym

es,

cli

nic

all

yaff

ecte

d,

cla

ssic

al

sym

pto

ms

(3)

Fem

ale

;5;ass

ista

nce

requ

ired

for

walk

ing,d

ela

yin

gro

wth

an

dd

evelo

pm

en

t,sl

eep

pro

ble

ms,

abn

orm

al

EE

Gw

ith

no

sym

pto

ms

of

seiz

ure

s,gri

nd

ing

of

teeth

,bre

ath

hold

ing,

hyp

erv

en

tila

tion

,n

on

verb

al,

lack

of

han

du

sage,

rep

eti

tive

han

dm

oti

on

s,d

iffi

cu

lty

eati

ng

an

dsl

igh

tre

flu

xes,

slig

ht

trem

or,

small

feet

RT

TM

arc

hett

oet

al.

123

MeC

P2

(1155d

el3

,Q

244X

,T

158

M,

R306C

)

Red

uced

som

asi

ze

an

dsp

ine

den

sity

,fe

wer

syn

ap

ses,

alt

ere

dcalc

ium

sign

ali

ng,

ele

ctr

op

hysi

olo

gic

al

abn

orm

ali

ties

Retr

ovir

us:

fou

rfa

cto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

)

Fib

robla

st:

Cori

ell

GM

11272

(1),

GM

16548

(2),

GM

17880

(3),

GM

11270

(4)

(1)

Fem

ale

;3;

norm

al

lyso

som

al

en

zym

es,

cli

nic

all

yaff

ecte

d,

cla

ssic

al

sym

pto

ms

(2)

Fem

ale

;5;

cli

nic

all

yaff

ecte

d,

slig

htl

ycu

rved

spin

e,

am

bu

lato

ry,

slig

ht

rigid

ity

an

dsp

ast

icit

y,d

ecre

asi

ng

head

cir

cu

mfe

ren

ce,

aberr

an

tsl

eep

patt

ern

s,d

ecre

ase

dh

an

du

sage,

rep

eti

tive

han

dm

oti

on

s,bre

ath

hold

ing,

non

verb

al,

con

stip

ati

on

,d

ecre

ase

dh

an

dan

dfe

et

cir

cu

lati

on

,ra

rese

lf-

inju

riou

sbeh

avio

r,sl

igh

teati

ng

pro

ble

ms

an

dre

flu

zes,

teeth

gri

nd

ing,

slig

ht

EE

Gabn

orm

ali

ties,

trem

ors

(3)

Fem

ale

;5;ass

ista

nce

requ

ired

for

walk

ing,d

ela

yin

gro

wth

an

dd

evelo

pm

en

t,sl

eep

pro

ble

ms,

abn

orm

al

EE

Gw

ith

no

sym

pto

ms

of

seiz

ure

s,gri

nd

ing

of

teeth

,bre

ath

hold

ing,

hyp

erv

en

tila

tion

,n

on

verb

al,

lack

of

han

du

sage,

rep

eti

tive

han

dm

oti

on

s,d

iffi

cu

lty

eati

ng

an

dsl

igh

tre

flu

xes,

slig

ht

trem

or,

small

feet

(4)

Fem

ale

;8;

norm

al

lyso

som

al

en

zym

es,

cli

nic

all

yaff

ecte

d,

cla

ssic

al

sym

pto

ms

RT

TA

nan

iev

et

al.

124

MeC

P2

(T158

M,

V247X

,R

306C

)D

ecre

ase

inn

ucle

ar

an

dn

eu

ron

size

Len

tivir

us:

fou

rfa

cto

rs(O

CT

4,N

AN

OG

,S

OX

2,

LIN

28),

Retr

ovir

us:

4fa

cto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

)

Fib

robla

st:

Cori

ell

GM

17880

(1),

GM

07982

(2),

GM

11270

(3)

(1)

Fem

ale

;5;ass

ista

nce

requ

ired

for

walk

ing,d

ela

yin

gro

wth

an

dd

evelo

pm

en

t,sl

eep

pro

ble

ms,

abn

orm

al

EE

Gw

ith

no

sym

pto

ms

of

seiz

ure

s,gri

nd

ing

of

teeth

,bre

ath

hold

ing,

hyp

erv

en

tila

tion

,n

on

verb

al,

lack

of

han

du

sage,

rep

eti

tive

han

dm

oti

on

s,d

iffi

cu

lty

eati

ng

an

dsl

igh

tre

flu

xes,

slig

ht

trem

or,

small

feet

(2)

Fem

ale

;25;

cli

nic

all

yaff

ecte

d,

mic

rocep

haly

,se

vere

lyre

tard

ed

,h

an

dw

rin

gin

gst

art

ing

at

age

2,

scoli

osi

sat

age

12,

kyp

hosc

oli

osi

sat

age

25,

start

ed

tolo

sesk

ills

at

2years

old

,C

Tsc

an

at

25

show

ed

atr

op

hy,

slow

,abn

orm

al

EE

G,

no

sleep

pro

ble

ms

(3)

Fem

ale

;8;

norm

al

lyso

som

al

en

zym

es,

cli

nic

all

yaff

ecte

d,

cla

ssic

al

sym

pto

ms

iPSC modeling of psychiatric disordersKJ Brennand et al

6

Molecular Psychiatry

Page 7: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

Table

3C

on

tin

ued

Dis

ease

Refe

ren

ce

Gen

eti

cm

uta

tion

Neu

ron

al

ph

en

oty

pe

hiP

SC

meth

od

Sou

rce

of

cell

sP

ati

en

tse

x;

age

at

bio

psy

(years

);avail

able

ph

en

oty

pic

info

rmati

on

Tim

oth

ysy

nd

rom

eP

asc

aet

al.

129

CA

CN

A1C

Defe

cts

incalc

ium

sign

ali

ng,

decre

ase

dexp

ress

ion

of

cort

ical

gen

es,

incre

ase

dp

rod

ucti

on

of

nore

pin

ep

hri

ne

an

dd

op

am

ine

Retr

ovir

us:

fou

rfa

cto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

)

Fib

robla

st:

pati

en

tbio

psy

(1)

Fem

ale

;n

ot

state

d;

not

state

d(2

)N

ot

state

d;

not

state

d;

not

state

d

FX

SU

rbach

et

al.

127

FM

R1

No

neu

ron

sgen

era

ted

Retr

ovir

us:

fou

rfa

cto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

)

Fib

robla

st:

Cori

ell

GM

05848

(1),

GM

07072

(2),

GM

09497

(3)

(1)

Male

;4;in

cre

ase

dear

size,elo

ngate

dfa

ce,ap

pears

pro

gn

ath

ic,

men

tal

reta

rdati

on

,u

nd

efi

ned

con

necti

ve

tiss

ue

dysp

lasi

a(2

)M

ale

;22;

9/5

0cord

blo

od

lym

ph

ocyte

ssh

ow

ed

fra(X

),m

oth

er

isan

obli

gate

carr

ier

for

fra(x

)(3

)M

ale

;28;

aff

ecte

dbro

ther,

larg

eears

,m

en

tal

reta

rdati

on

,m

acro

-orc

hid

ism

,h

yp

era

cti

ve,

20%

of

PB

Lp

osi

tive

for

fra(x

)

FX

SS

heri

dan

et

al.

128

FM

R1

Few

er

an

dsh

ort

er

neu

ral

pro

cess

es,

incre

ase

dgli

al

cell

sw

ith

more

com

pact

morp

holo

gy

Retr

ovir

us:

fou

rfa

cto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

)

Fib

robla

st:

Cori

ell

GM

05848

(1),

GM

05131

(2),

GM

05185

(3)

(1)

Male

;4;in

cre

ase

dear

size,elo

ngate

dfa

ce,ap

pears

pro

gn

ath

ic,

men

tal

reta

rdati

on

,u

nd

efi

ned

con

necti

ve

tiss

ue

dysp

lasi

a(2

)M

ale

;3;

aff

ecte

dbro

ther

an

du

ncle

,(3

)M

ale

;26;

46,f

ra(X

),Y

pre

sen

tin

30–50%

of

PB

L

SC

ZD

Ch

ian

get

al.

131

DIS

C1

No

neu

ron

sgen

era

ted

Ep

isom

e:

fou

rfa

cto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

)

Fib

robla

st:

pati

en

tbio

psy

(1)

Male

;N

A;

dia

gn

ose

dw

ith

ch

ron

icu

nd

iffe

ren

tiate

dsc

hiz

op

hre

nia

,au

dit

ory

an

dvis

ual

hall

ucin

ati

on

s,m

ult

iple

delu

sion

san

dh

ad

form

al

thou

gh

td

isord

er

(2)

Fem

ale

;N

A;

dia

gn

ose

dch

ron

icp

ara

noid

sch

izop

hre

nia

,au

dit

ory

an

dvis

ual

hall

ucin

ati

on

s,m

ult

iple

delu

sion

san

dh

ad

form

al

thou

gh

td

isord

er

SC

ZD

Bre

nn

an

det

al.

132

Sp

ora

dic

Red

uced

neu

ron

al

con

necti

vit

y,fe

wer

neu

rite

s,d

ecre

ase

dP

SD

95

an

dglu

tam

ate

recep

tor

exp

ress

ion

levels

Tetr

acycli

ne-i

nd

ucib

lele

nti

vir

us:

five

facto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

,LIN

28)

Fib

robla

st:

Cori

ell

GM

02038

(1),

GM

01792

(2),

GM

01835

(3),

GM

02497

(4)

(1)

Male

;22;

on

set

at

age

6,

com

mit

ted

suic

ide

(2)

Male

;26;

recu

rren

ces

of

agit

ati

on

,d

elu

sion

sof

pers

ecu

tion

,fe

ar

of

ass

ass

inati

on

,fa

ther

an

dsi

ster

aff

ecte

d(3

)F

em

ale

;27;

sch

izoaff

ecti

ve

dis

ord

er,

pro

ble

ms

of

dru

gabu

se,

hosp

itali

zed

,fa

ther

aff

ecte

d(4

)M

ale

;23;

para

logic

al

thin

kin

g,

spli

ttin

gof

eff

ect

from

con

ten

t,su

spic

iou

sness

,aff

ecti

ve

shie

ldin

g,

on

set

at

age

15,

hosp

itali

zed

,p

osi

tive

fam

ily

his

tory

SC

ZD

Pau

lsen

et

al.

133

Sp

ora

dic

Ele

vate

dextr

a-

mit

och

on

dri

al

oxygen

con

sum

pti

on

,in

cre

ase

dle

vels

of

reacti

ve

oxygen

specie

s

Retr

ovir

us:

fou

rfa

cto

rs(O

CT

4,

SO

X2,

KLF

4,

c-M

YC

)

Fib

robla

st:

pati

en

tbio

psy

Fem

ale

;48;

clo

zap

ine-r

esi

stan

t

Abbre

via

tion

s:A

SD

,au

tism

spectr

um

dis

ord

er;

BD

,bip

ola

rd

isord

er;

EE

G,

ele

ctr

oen

cep

halo

gra

mte

st;

FX

,fr

agil

eX

syn

dro

me;

hiP

SC

,h

um

an

-deri

ved

ind

uced

plu

rip

ote

nt

stem

cell

;N

A,

not

ap

pli

cable

;P

BL

,p

eri

ph

era

lblo

od

lym

ph

ocyte

;R

TT

,R

ett

syn

dro

me;

SC

ZD

,sc

hiz

op

hre

nia

dis

ord

er.

iPSC modeling of psychiatric disordersKJ Brennand et al

7

Molecular Psychiatry

Page 8: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

SCZD and BD), there is decreased dendritic spinematuration and plasticity, leading to decreased spinedensity,66 whereas Clock mice, a model of mania inBD, appear to have normal synaptic density.46

Particularly with respect to SCZD, aberrations insynaptic activity have also been observed in adultneurogenesis in the hippocampus. Similar to corticalembryonic development, where downregulation ofDisc1 results in premature cell cycle exit of neuralprogenitor cells,67,68 adult-born neurons with reducedDisc1 have hastened neural development. Disc1knockdown results in accelerated dendritic develop-ment, soma hypertrophy, aberrant positioning andincreased neural excitability.69–71 It remains unknownif aberrant adult neurogenesis contributes to psychia-tric disease in humans.

Aberrant neuronal migrationIn ASD patients, defects of migration can lead to avariety of morphological outcomes, particularly hetero-topias and dysplastic changes. One recent pathologicalstudy identified a number of abnormalities andlesions in most of the ASD brains studied, includinga loss of vertical and horizontal organization of corticallayers in some patients.72 It has been hypothesizedthat altered expression of cytoskeletal proteins and lossof neuronal polarity contribute to these corticalmigration defects.73

Animal models also show phenotypes consistentwith abnormal neuronal migration: mice with re-duced Disc1 activity have reduced cortical migra-tion,74 Nrg1 mutant mice have reduced tangentialneural migration from the ventral telencephalon44,45

and Cntnap2-null mice have impaired migration ofcortical projection neurons.75 Disc1 mutant mice havealtered distribution of hippocampal mossy fiberterminals on CA3,42 and axons with Disc1 knockdownmiss CA3 altogether and project onto CA1.70 dnDISC1neurites have deficits in neurite repulsionin vitro.42 Although DISC1 is a rare SCZD allele, anunderstanding of the downstream targets or bindingpartners through which it mediates its cellulareffects may identify drug targets relevant to thebroader SCZD population. One putative downstreamtarget of DISC1 is Glycogen Synthase 3-beta (Gsk3b),67

Gsk3b functions within several central path-ways (including cAMP and Wnt) is a direct targetof lithium (a drug commonly used to treat BD)76,77

and mounting evidence indicates that Gsk3bmay be a central mediator of axon outgrowthdynamics.78 Cell-based assays will allow the studyof the effects of Gsk3b, cAMP and WNT levels onneurite outgrowths and axon migration of live humanneurons.

Evidence for network phenotypes in psychiatricdisorders

While comparable neuronal phenotypes, particularlyaberrant dendritic arborization, synaptic density andneuronal migration, are shared between ASD, SCZD

and BD, these cellular phenotypes likely result invastly different network effects in each disorder.Functional imaging facilitates the study of the abnor-mal neural circuitry behind cognitive dysfunction.

One hypothesis concerning ASD is that short-distance over-connectivity in the cortex leads to afailure of long-distance coupling.79 This hypothesispredicts that impaired long-distance connectivity inthe cortex impedes information integration acrossdiverse functional systems (emotional, sensory, auto-nomic, memory). Consistent with this prediction,fMRI studies of resting state brain activity haveobserved increased connectivity between proximalregions, such as the posterior cingulate and theparahippocampal gyrus,80 and decreased connectivitybetween the distal regions, such as the frontal cortexand the parietal lobe,81 the insular cortices and thesomatosensory cortices or amygdala,82 the frontalcortex and the posterior cingulate,80 as well asdecreased interhemispheric synchronization.83 Com-parable defects in long-distance connectivity werefound when ASD patients performed social andintrospective tasks.84 Among ASD patients, a negativecorrelation exists between functional connectivity inthese regions and severity of social and communica-tion impairment.80,82

Just as pathological studies of SCZD reporteddecreased frontal and temporal lobe volumes, earlyfMRI studies of SCZD patients revealed brain activityabnormalities in the frontal and temporal lobes.85,86

More recent studies have further shown that SCZDpatients exhibit cortical hyper-activity and hyper-connectivity of the prefrontal cortex at rest, butreduced activation of the medial prefrontal cortexduring working memory tasks.87 While functionalconnectivity of the parietal cortex to the ventralprefrontal cortex is greater in SCZD, it is reduced tothe dorsal prefrontal cortex.88 This is consistent withanatomical neuronal network maps, which reveal aloss of network ‘hubs’ in the frontal cortex, andincreased connection distance. These network aberra-tions are thought to result from neurodevelopmentalabnormalities impacting cortical organization.89

Although fMRI studies can reveal regions of thebrain with aberrant activity in the disease state, theycannot elucidate the specific neuronal cell typesaffected. Therefore, pharmacological and post-mor-tem studies have generated hypotheses concerningthe cell types affected by SCZD. Similar studies ofASD and BD have been less successful in identifyingthe specific cell types relevant to disease.

Good evidence now links aberrant neurotransmittersignaling to SCZD. Dopamine receptor antagonistsreduce the symptoms of SCZD and evidence nowlinks SCZD with increased dopamine receptor levelsand sensitivity.90,91 Comparably, glutamate-blockingdrugs such as ketamine produce symptoms generallyassociated with SCZD,92 whereas the glutamatereceptor2/3 agonist LY2140023 may ameliorate thesymptoms of SCZD.93 Post-mortem studies of SCZDbrains have found decreased glutamate receptor

iPSC modeling of psychiatric disordersKJ Brennand et al

8

Molecular Psychiatry

Page 9: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

expression,94 whereas among GABAergic interneur-ons, a decrease in GAD67 and calcium-bindingproteins was found. Changes in GABAergic neuronsare particularly relevant as they are thought toproduce gamma oscillations, which synchronizepyramidal neuron firing, an activity that is impairedin SCZD. Evidence in mice suggests that SCZD results,at least in part, from reduced excitatory glutamatergicinput onto GABAergic inhibitory neurons.60,95,96 Itremains unclear whether aberrant dopamine, gluta-mate or GABA signaling is the primary cause of SCZD,as aberrant activity of any neuronal cell type couldaffect neurotransmitter activity of the remaining celltypes in the disease state.

In model organisms from Drosophila to mice, genesassociated with ASD, SCZD and BD have been shownto regulate synaptic activity and plasticity. Forexample, a screen in Drosophila for genes critical inmaintaining homeostatic modulation of synaptictransmission identified the SCZD gene Dysbindin(DTNBP1). Dtnbp1 acts presynaptically, in a dose-dependent manner, to regulate adaptive neuralplasticity.97 In Mecp2 mice, although synapse forma-tion, elimination and strengthening are normal, theexperience-dependent phase of synapse remodelingis impaired98 and Mecp2 mice show altered activity-dependent neural gene expression.99 Cntnap2-nullmice, lacking a gene associated with ASD, havereduced GABAergic neurons and decreased neuronalsynchrony.100 Disc1 mice have reduced hippocampalsynaptic transmission.42,43 Nrg1 mice have impairedsynaptic maturation and function58,60–62,101 and22q11.2 mice show altered short- and long-termsynaptic plasticity as well as calcium kinetics inCA3 presynaptic terminals. Defects in synapticplasticity at the cellular level likely contributeto the network aberrations observed in psychiatricdisorders.

One characteristic network defect observed inSCZD is prepulse inhibition (PPI). PPI is a measureof sensory gating, in which a weaker prestimulus(prepulse) inhibits the reaction of an organism to asubsequent strong startling stimulus (pulse). Deficitsin PPI are observed in Nrg1 mice60,95 and are reversedby dopamine receptor antagonists.102,103 Dtnbp1 micedisplay not only decreased PPI but also reducedevoked g-activity, a second pattern seen in patientswith SCZD.104 In humans, polymorphisms in circa-dian genes such as CLOCK convey risk for BD; mutantClock mice also have dysfunctional g-activity acrosslimbic circuits, which can be improved by chroniclithium treatment.46

While PPI is attributed to glutamatergic activity,reduced g-activity indicates abnormal GABAergicneurotransmission. Therefore, although pharmacolo-gical evidence implicates dopaminergic and glutama-tergic neurons in SCZD, network analysis revealsdefects in both glutamatergic and GABAergic activityin SCZD and BD. Aberrations originating in any oneneuronal subtype would ultimately be expected toaffect activity in other types of neurons and in a

variety of brain regions. The ability to testsynaptic activity in defined populations of humanglutamatergic, GABAergic and dopaminergicneurons affected by ASD, SCZD or BD might help toelucidate the neuronal subtypes at the core of eachdisorder.

Introduction to hiPSCs and iNeurons

The transient expression of four factors (OCT3/4,KLF4, SOX2 and c-MYC) is sufficient to directlyreprogram adult somatic cells into an iPSC state.105–107

Because hiPSCs can be derived from adult patientsafter the development of disease, hiPSCs represent apotentially limitless source of human cells withwhich to study disease, even without knowing whichgenes are interacting to produce the disease state inan individual patient. Methods to efficiently differ-entiate pluripotent stem cells to neurons were devel-oped initially in studies using human embryonic stemcells.108 Through the addition of various morphogensto recapitulate the cues of embryonic development,ESCs and iPSCs can be directed to differentiate toregional identities including forebrain,109 midbrain/hindbrain110,111 and spinal cord.112,113 It is generallythought that every cell type present in vivo can bedifferentiated in vitro using hiPSCs, although meth-ods for many remain unexplored or inefficient.

An alternative approach for generating patient-specific neurons to study complex psychiatric dis-orders is now possible. Expression of four factors(ASCL1, BRN2, MYT1L and NEUROD) can convertfibroblasts into functional iNeurons in vitro.114,115 Theprocess is rapid, generating electrophysiologicallymature neurons with functional synapses within 14days, and it is efficient, yielding up to 8% neurons. Todate, methods exist to transform fibroblasts directly toglutamatergic115 and dopaminergic neurons,116 butmethods to generate GABAergic iNeurons have notyet been reported. The regional identity of eachneurotransmitter subtype remains unclear.

Both hiPSC neurons and iNeurons have thecapacity to generate vast numbers of live humanneurons for the study of psychiatric disorders.Because iNeuron generation bypasses neuronal differ-entiation and maturation, hiPSC neurons are likelythe best method by which to model developmentalfacets of disease. For example, if SCZD ultimatelyresults from abnormal synaptic maturation, it ispossible that direct reprogramming would bypassthe developmental window in which the SCZDcellular phenotype can be observed in vitro. Addi-tionally, as aberrant ASCL1, BRN2 and MYT1L haveall been linked to neurological disease,117–120 it is notunreasonable to predict that overexpression of one ormore of these key neuronal genes might affect theinitiation or progression of a psychiatric disorder invitro. Conversely, the rapid experimental timeframe ofiNeuron generation makes it an ideal system withwhich to study phenotypic effects in mature neurons.If ASD is indeed a disease of activity-dependent

iPSC modeling of psychiatric disordersKJ Brennand et al

9

Molecular Psychiatry

Page 10: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

synaptic modulation rather than synaptic maturation,iNeurons represent a more direct cell type with whichto assay network properties. As the efficiency ofiNeuron generation increases, and spontaneous neu-ronal networks result, this method may facilitaterobust and swift network analysis of ASD, SCZD andBD neurons. It is important to note that both strategiesfacilitate novel experiments of innate neuron-specificdeficits in psychiatric disease that are not confoundedby environmental factors, such as treatment history,drug and alcohol abuse or poverty, that typicallyplague clinical studies.

Psychiatric disorders result in hiPSC neuronalphenotypes in vitro

While no reported studies have yet characterizediNeurons from patients with psychiatric disorders, anumber of groups, including ours, have now pub-lished studies of hiPSC neurons derived from patientswith ASD and SCZD. During neuronal differentiationof hiPSCs, a number of neuronal genes alreadyimplicated in ASD, SCZD and BD, such as transcrip-tion factors and chromatin modifiers like POU3F2and ZNF804A and cell adhesion genes like NRXN1and NLGN1,121 are upregulated, permitting compar-isons of expression levels in diseased and healthy livehuman neurons.

Three groups have now generated hiPSCs from atotal of seven RTT patients, representing a number ofunique point mutations and deletions in the MeCP2gene (Q244X, 1155del 32, T158 M, R306C,D3-4,R294X, V247X).122–124 None of the groups observedaltered replication or differentiation of RTT hiPSCs orneural progenitor cells. Rather, consistent withanimal and post-mortem patient studies, all threegroups reported that neuronal soma size of RTThiPSC neurons is reduced by approximately 10–20%compared with controls.122–124 Furthermore, we ob-served that RTT hiPSC neurons have reduced spinedensity, decreased neuronal spontaneous calciumsignaling and decreased spontaneous excitatory andinhibitory postsynaptic currents. The reproducibilityof these findings across three independent reportsvalidates the use of hiPSC-based models. Further-more, by demonstrating the ability to test drugs torescue synaptic deficiency in RTT neurons, thesestudies hint at future uses of hiPSC neurons for high-throughout drug screening to identify new therapeu-tic drugs for psychiatric disorders.

FX is caused by the absence of expression of thefragile X mental retardation 1 (FMR1) gene,125 whichis believed to result from transcriptional silencingduring embryonic development, owing to a CGGtriplet-repeat expansion in the 50 untranslated regionof the gene.126 Although the somatic cells of three FXpatients were successfully reprogrammed to pluripo-tency, the FMR1 gene remained inactive in all FXhiPSC lines, unlike FX embryonic stem cell lines.Consequently, the authors of this first report of FXhiPSCs concluded that ‘FX-iPSCs do not model the

differentiation-dependent silencing of the FMR1gene,’ and therefore chose not to assess their FX-hiPSC neurons for phenotypic abormalities.127 Morerecently, a second group generated hiPSCs from threepatients (including one patient common to the firstgroup) via nearly identical methods. They noted thata number of hiPSC lines had FMR1 CGG-repeatlengths that were clearly different from the originalfibroblasts, and they also failed to detect FMR1 geneexpression in the original FX fibroblasts or their FXhiPSCs.128 Despite also not observing reactivatedFMR1 expression in FX hiPSCs, this group comparedneural differentiation of FX and control hiPSCs. Theyobserved that neural cultures generated from FXhiPSCs consisted of neurons with fewer and shorterprocesses, as well as a larger number of glial cellswith more compact morphology, suggesting thatdecreased FMR1 expression levels, rather than theslow silencing of FMR1 during neuronal differentia-tion, are sufficient to produce the disease state in FX.

Timothy syndrome is caused by a mutation in theL-type calcium channel Ca(v)1.2 and is associatedwith heart arrhythmias and ASD. From two patientswith Timothy syndrome, hiPSC-derived corticalneural progenitor cells (NPCs) and neurons weregenerated. Neurons from these individuals wereshown to have aberrant calcium signaling,129 whilean earlier publication by this same group demon-strated that hiPSC-derived cardiomyocytes from thesesame patients had irregular contraction, abnormalcalcium transients and irregular electrical activity.130

Timothy syndrome hiPSC neurons underwent abnor-mal cortical differentiation, showing decreased ex-pression of cortical genes and increased production ofnorepinephrine and DA. Notably, treatment withroscovitine, a cyclin-dependent kinase inhibitor andatypical L-type-channel blocker, was sufficient toameliorate many characteristics of Timothy syndromeneurons in vitro.129

DISC1 mutations cause a rare monogenic form ofSCZD. The generation of hiPSCs from SCZD patientswith a DISC1 mutation have now been reported,131

although neurons differentiated from these hiPSCshave not yet been characterized. We predict thatDISC1–hiPSC-derived neurons will ultimately beshown to recapitulate the cellular phenotypes ob-served in dnDISC mice, just as RTT and FX-hiPSCneurons have replicated findings from mouse studies.

We recently reported neuronal phenotypes ofhiPSC neurons from four patients with complexgenetic forms of SCZD. When assayed by retrogradetransmission of rabies virus neuronal labeling, SCZD-hiPSC neurons showed reduced neuronal connectiv-ity and altered gene expression profiles.132 Whilenearly 25% of genes with altered expression had beenpreviously implicated in SCZD, we also identified anumber of new pathways that may contribute toSCZD. A second group has now reported an oxygenmetabolism phenotype associated with SCZD;133 theyobserved a twofold increase in extra-mitochondrialoxygen consumption as well as elevated levels of

iPSC modeling of psychiatric disordersKJ Brennand et al

10

Molecular Psychiatry

Page 11: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

reactive oxygen species in neural progenitor cellsderived from hiPSCs from one SCZD patient relativeto controls. Although a small study, this observationis consistent with animal studies134,135 and deservesattention. Oxygen metabolism defects have not beenwell demonstrated in human neurons, owing to a lackof live human cells for study. This is an excellentexample of the type of hiPSC study that can inve-stigate hypotheses not testable in human patients.

Limitations of hiPSC-based modeling

A number of major limitations currently restricthiPSC-based studies, particularly concerning thescalability of hiPSC generation, neural differentiationand phenotypic characterization of derived neuronsand neural networks. These technical limitationshave made it hard to accurately address the inherentvariability of cell-based studies, which exist in threemajor forms: (1) neuron-to-neuron (intra-patient), (2)hiPSC-to-hiPSC (intra-patient) and (3) patient-to-patient (inter-patient). To produce meaningful data,each cell-based experiment should ideally comparemultiple neuronal differentiations from multipleindependent hiPSC lines from multiple patients.Owing to cost and time constraints, such largeexperiments have not yet been completed. Conse-quently, the hiPSC studies reported to date mayultimately prove to be proof-of-concept demonstra-tions until methods to compare derived neurons fromhundred or thousands of patients and controls arerefined.

Intra-patient variability results from differencesbetween neurons and iPSCs generated from a singlepatient; it is the major constraint on signal to noise incell-based experiments. Differences between indivi-dual hiPSC neurons derived from a single patientproduce neuron-to-neuron variability. To some extent,this variability may be unavoidable, although it iscurrently exacerbated by the heterogeneity of cellularsubtypes in hiPSC neural populations; none of thereports described in this review compare pureneuronal populations of a specific subtype. At theexperimental level, neural subtype heterogeneityresults because current neuronal differentiation pro-tocols are not 100% efficient and, in contrast to thehematopoietic system, cell surface markers by whichspecific subtypes of neurons might be purified havenot been developed. It is well established thatindividual hiPSC lines vary genetically, epigeneti-cally and in terms of neural differentiation propen-sities to produce hiPSC-to-hiPSC variability. Geneticdifferences include the location and number of viralintegrations produced during the reprogrammingprocess and spontaneous mutations that have beenobserved during hiPSC generation and expansion.136

Epigenetic differences reflect the somatic cell typeused for reprogramming and the completeness of itschromatin remodeling.137 Differences in developmen-tal potential exist among human embryonic stem celllines138 and between individual hiPSC lines.139

Inter-patient variability reflects the heterogeneity inclinical outcomes between patients with ASD, SCZDor BD. Consequently, given the small sample size(typically 1–4 patients) of the current hiPSC-basedstudies discussed in this review, a major concern iswhether their findings are representative of the largerpatient population. In the short term, this has beenaddressed by recruiting patients with well-definedclinical or genetic characteristics as well as matchedhealthy controls. Ultimately, methods will have to bedeveloped to permit comparisons of thousands ofpatients.

Future directions of cell-based studies

Whole-brain disorders should be studied at the levelof component aberrations of cells and neural net-works. Neuroimaging, post-mortem anatomical andpharmacological studies of patients may be measuringconsequences of the disease state, rather than itsorigin. Cell-based studies will lead to the discernmentand characterization of the molecular causes of ASD,SCZD and BD and facilitate studies of the cellular andnetwork phenotypes that serve as neuronal predis-positions to disease. Furthermore, these studiesconfer the ability to test various neuron non-cell-autonomous effects, such as inflammation, oxidativestress, activity-dependent modulations and the influ-ence of stress hormones in psychiatric disorders.High-throughput screening of new classes of com-pounds capable of pharmacological amelioration ofneuronal and/or network phenotypes for treatment ofthese disorders is possible.

Small defects at the cellular level could ultimatelymanifest as complex psychiatric disorders with anarray of symptoms in patients. For example, ifneurons derived from psychiatric patients show adecrease in the absolute number of connectionsbetween cells, and if this phenotype is restricted toa specific subtype of neurons, this finding might hintat the central cell type relevant to the disease state.Because synaptic strength is highly modulated bysynaptic activity, a decrease in the strength ofindividual connections between neurons in psychia-tric patients could indicate aberrant synaptic activityor plasticity in patient brains. Finally, perturbedneuronal migration or axon targeting in vitro mightsuggest that mis-targeted neuronal connections,rather than decreased neuronal connectivity, iscentral to disease. Cellular phenotypes hint at theneuronal predispositions contributing to psychiatricdisorders and may help to unlock the complexities ofpsychiatric illness.

While overlapping genetic susceptibilities mightproduce a common cellular phenotype, or predis-position, to psychiatric illness, clinical outcome maybe determined by activity at the network level.Synaptic pruning (either whole brain or in specificregions) is an activity-dependent process that couldgenerate the clinical differences distinguishing ASD,SCZD and BD. Cell-based studies of neuronal and

iPSC modeling of psychiatric disordersKJ Brennand et al

11

Molecular Psychiatry

Page 12: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

network aberrations in psychiatric disorders may leadto predictions of activity-dependent environmentalinfluences that contribute to disease progression.

Human iPSC- and iNeuron-based methods have thepotential to simplify whole-brain disorders like ASD,SCZD and BD to their cellular and network compo-nents, contributing to our understanding of theseconditions. Although many technical issues, particu-larly, concerning the scalability of hiPSC generation,neuronal differentiation and neural assays remain, webelieve that studies of neuronal networks constructedfrom defined neuronal populations are feasible. Byrecapitulating and monitoring healthy and diseasenetworks in a dish, it is likely that new methods ofin vitro modeling of psychiatric disorders willresult in new insights into the mechanism of diseaseinitiation, progression and, ultimately, treatment.

Conflict of interest

The authors declare no conflict of interest.

Acknowledgments

The Gage Laboratory is partially funded by CIRMGrant RL1-00649-1, The Lookout and Mathers Foun-dation, the Helmsley Foundation as well as Sanofi-Aventis. We thank J Simon for illustrations and MLGage for editorial comments.

References

1 Association AP. Diagnostic and statistical manual of mentaldisorders: DSM-IV. 3rd ed., rev. edn, vol. 4th ed, AmericanPsychiatric Press: Washington, DC, 1994; 886, pp 69–70, 263,350–359.

2 Di Giorgio FP, Boulting GL, Bobrowicz S, Eggan KC. Humanembryonic stem cell-derived motor neurons are sensitive to thetoxic effect of glial cells carrying an ALS-causing mutation. CellStem Cell 2008; 3: 637–648.

3 Marchetto MC, Muotri AR, Mu Y, Smith AM, Cezar GG, Gage FH.Non-cell-autonomous effect of human SOD1 G37R astrocytes onmotor neurons derived from human embryonic stem cells. CellStem Cell 2008; 3: 649–657.

4 Ritvo ER, Freeman BJ, Mason-Brothers A, Mo A, Ritvo AM.Concordance for the syndrome of autism in 40 pairs of afflictedtwins. Am J Psychiatry 1985; 142: 74–77.

5 Sullivan PF, Kendler KS, Neale MC. Schizophrenia as a complextrait: evidence from a meta-analysis of twin studies. Arch GenPsychiatry 2003; 60: 1187–1192.

6 Tsuang MT, Stone WS, Faraone SV. Genes, environment andschizophrenia. Br J Psychiatry Suppl 2001; 40: s18–s24.

7 Bottenstein JE, Sato GH. Growth of a rat neuroblastoma cell linein serum-free supplemented medium. Proc Natl Acad Sci USA1979; 76: 514–517.

8 Benitez-King G, Riquelme A, Ortiz-Lopez L, Berlanga C, Rodri-guez-Verdugo MS, Romo F et al. A non-invasive method to isolatethe neuronal linage from the nasal epithelium from schizophre-nic and bipolar diseases. J Neurosci Methods 2011; 201: 35–45.

9 Matigian N, Abrahamsen G, Sutharsan R, Cook AL, Vitale AM,Nouwens A et al. Disease-specific, neurosphere-derived cells asmodels for brain disorders. Dis Model Mech 2010; 3: 785–798.

10 Carper RA, Moses P, Tigue ZD, Courchesne E. Cerebral lobes inautism: early hyperplasia and abnormal age effects. Neuroimage2002; 16: 1038–1051.

11 Courchesne E, Karns CM, Davis HR, Ziccardi R, Carper RA, TigueZD et al. Unusual brain growth patterns in early life in patients

with autistic disorder: an MRI study. Neurology 2001; 57:245–254.

12 Hazlett HC, Poe M, Gerig G, Smith RG, Provenzale J, Ross A et al.Magnetic resonance imaging and head circumference study ofbrain size in autism: birth through age 2 years. Arch GenPsychiatry 2005; 62: 1366–1376.

13 Courchesne E, Mouton P, Calhoun M, Semendeferi K, Ahrens-Barbeau C, Hallet M et al. Neuron number and size in prefrontalcortex of children with autism. JAMA 2011; 206: 2001–2010.

14 Courchesne E, Press GA, Yeung-Courchesne R. Parietal lobeabnormalities detected with MR in patients with infantile autism.AJR Am J Roentgenol 1993; 160: 387–393.

15 Hadjikhani N, Joseph RM, Snyder J, Tager-Flusberg H. Anatomi-cal differences in the mirror neuron system and social cognitionnetwork in autism. Cereb Cortex 2006; 16: 1276–1282.

16 Schmitz N, Daly E, Murphy D. Frontal anatomy and reaction timein Autism. Neurosci Lett 2007; 412: 12–17.

17 Brun CC, Nicolson R, Lepore N, Chou YY, Vidal CN, DeVito TJet al. Mapping brain abnormalities in boys with autism. HumBrain Mapp 2009; 30: 3887–3900.

18 Frazier TW, Hardan AY. A meta-analysis of the corpus callosumin autism. Biol Psychiatry 2009; 66: 935–941.

19 Raymond GV, Bauman ML, Kemper TL. Hippocampus in autism:a Golgi analysis. Acta Neuropathol 1996; 91: 117–119.

20 Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, ZoghbiHY. Rett syndrome is caused by mutations in X-linked MECP2,encoding methyl-CpG-binding protein 2. Nat Genet 1999; 23:185–188.

21 Van den Veyver IB, Zoghbi HY. Methyl-CpG-binding protein 2mutations in Rett syndrome. Curr Opin Genet Dev 2000; 10:275–279.

22 Bauman ML, Kemper TL, Arin DM. Pervasive neuroanatomicabnormalities of the brain in three cases of Rett’s syndrome.Neurology 1995; 45: 1581–1586.

23 Castren M, Tervonen T, Karkkainen V, Heinonen S, Castren E,Larsson K et al. Altered differentiation of neural stem cellsin fragile X syndrome. Proc Natl Acad Sci USA 2005; 102: 17834–17839.

24 Bhattacharyya A, McMillan E, Wallace K, Tubon Jr TC, CapowskiEE, Svendsen CN. Normal neurogenesis but abnormal gene

expression in human Fragile X cortical progenitor cells. Stem

Cells Dev 2008; 17: 107–117.25 Vita A, De Peri L, Silenzi C, Dieci M. Brain morphology in first-

episode schizophrenia: a meta-analysis of quantitative magnetic

resonance imaging studies. Schizophr Res 2006; 82: 75–88.26 Steen RG, Mull C, McClure R, Hamer RM, Lieberman JA. Brain

volume in first-episode schizophrenia: systematic review and

meta-analysis of magnetic resonance imaging studies. Br

J Psychiatry 2006; 188: 510–518.27 Wright IC, Rabe-Hesketh S, Woodruff PW, David AS, Murray RM,

Bullmore ET. Meta-analysis of regional brain volumes in schizo-

phrenia. Am J Psychiatry 2000; 157: 16–25.28 Thompson PM, Vidal C, Giedd JN, Gochman P, Blumenthal J,

Nicolson R et al. Mapping adolescent brain change

reveals dynamic wave of accelerated gray matter loss in very

early-onset schizophrenia. Proc Natl Acad Sci USA 2001; 98:

11650–11655.29 Ellison-Wright I, Glahn DC, Laird AR, Thelen SM, Bullmore E.

The anatomy of first-episode and chronic schizophrenia: an

anatomical likelihood estimation meta-analysis. Am J Psychiatry

2008; 165: 1015–1023.30 Rajkowska G, Selemon LD, Goldman-Rakic PS. Neuronal and

glial somal size in the prefrontal cortex: a postmortem morpho-

metric study of schizophrenia and Huntington disease. Arch Gen

Psychiatry 1998; 55: 215–224.31 Kolomeets NS, Orlovskaya DD, Rachmanova VI, Uranova NA.

Ultrastructural alterations in hippocampal mossy fiber synapses

in schizophrenia: a postmortem morphometric study. Synapse

2005; 57: 47–55.32 Black JE, Kodish IM, Grossman AW, Klintsova AY, Orlovskaya D,

Vostrikov V et al. Pathology of layer V pyramidal neurons in theprefrontal cortex of patients with schizophrenia. Am J Psychiatry2004; 161: 742–744.

iPSC modeling of psychiatric disordersKJ Brennand et al

12

Molecular Psychiatry

Page 13: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

33 Selemon LD, Goldman-Rakic PS. The reduced neuropil hypoth-esis: a circuit based model of schizophrenia. Biol Psychiatry1999; 45: 17–25.

34 Karchemskiy A, Garrett A, Howe M, Adleman N, Simeonova DI,Alegria D et al. Amygdalar, hippocampal, and thalamic volumesin youth at high risk for development of bipolar disorder.Psychiatry Res 2011; 194: 319–325.

35 Frazier JA, Chiu S, Breeze JL, Makris N, Lange N, Kennedy DNet al. Structural brain magnetic resonance imaging of limbic andthalamic volumes in pediatric bipolar disorder. Am J Psychiatry2005; 162: 1256–1265.

36 Edmiston EE, Wang F, Kalmar JH, Womer FY, Chepenik LG,Pittman B et al. Lateral ventricle volume and psychotic featuresin adolescents and adults with bipolar disorder. Psychiatry Res2011; 194: 400–402.

37 Rajkowska G, Halaris A, Selemon LD. Reductions in neuronaland glial density characterize the dorsolateral prefrontal cortex inbipolar disorder. Biol Psychiatry 2001; 49: 741–752.

38 Pantazopoulos H, Lange N, Baldessarini RJ, Berretta S. Parvalbu-min neurons in the entorhinal cortex of subjects diagnosedwith bipolar disorder or schizophrenia. Biol Psychiatry 2007; 61:640–652.

39 Chen RZ, Akbarian S, Tudor M, Jaenisch R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-likephenotype in mice. Nat Genet 2001; 27: 327–331.

40 Kishi N, Macklis JD. MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation ratherthan cell fate decisions. Mol Cell Neurosci 2004; 27:306–321.

41 Smrt RD, Eaves-Egenes J, Barkho BZ, Santistevan NJ, Zhao C,Aimone JB et al. Mecp2 deficiency leads to delayed maturationand altered gene expression in hippocampal neurons. NeurobiolDis 2007; 27: 77–89.

42 Kvajo M, McKellar H, Arguello PA, Drew LJ, Moore H,MacDermott AB et al. A mutation in mouse Disc1 that models aschizophrenia risk allele leads to specific alterations in neuronalarchitecture and cognition. Proc Natl Acad Sci USA 2008; 105:7076–7081.

43 Li W, Zhou Y, Jentsch JD, Brown RA, Tian X, Ehninger Det al. Specific developmental disruption of disrupted-in-schizophrenia-1 function results in schizophrenia-relatedphenotypes in mice. Proc Natl Acad Sci UnSA 2007; 104:18280–18285.

44 Lopez-Bendito G, Cautinat A, Sanchez JA, Bielle F, Flames N,Garratt AN et al. Tangential neuronal migration controls axonguidance: a role for neuregulin-1 in thalamocortical axonnavigation. Cell 2006; 125: 127–142.

45 Krivosheya D, Tapia L, Levinson JN, Huang K, Kang Y, Hines Ret al. ErbB4-neuregulin signaling modulates synapse develop-ment and dendritic arborization through distinct mechanisms.J Biol Chem 2008; 283: 32944–32956.

46 Dzirasa K, Coque L, Sidor MM, Kumar S, Dancy EA, Takahashi JSet al. Lithium ameliorates nucleus accumbens phase-signalingdysfunction in a genetic mouse model of mania. J Neurosci 2010;30: 16314–16323.

47 Sudhof TC. Neuroligins and neurexins link synaptic function tocognitive disease. Nature 2008; 455: 903–911.

48 Hutsler JJ, Zhang H. Increased dendritic spine densities oncortical projection neurons in autism spectrum disorders. BrainRes 2010; 1309: 83–94.

49 Chapleau CA, Calfa GD, Lane MC, Albertson AJ, Larimore JL,Kudo S et al. Dendritic spine pathologies in hippocampalpyramidal neurons from Rett syndrome brain and after expres-sion of Rett-associated MECP2 mutations. Neurobiol Dis 2009; 35:219–233.

50 Irwin SA, Patel B, Idupulapati M, Harris JB, Crisostomo RA,Larsen BP et al. Abnormal dendritic spine characteristics in thetemporal and visual cortices of patients with fragile-X syndrome:a quantitative examination. Am J Med Genet 2001; 98: 161–167.

51 Garey LJ, Ong WY, Patel TS, Kanani M, Davis A, Mortimer AMet al. Reduced dendritic spine density on cerebral corticalpyramidal neurons in schizophrenia. J Neurol Neurosurg Psy-chiatry 1998; 65: 446–453.

52 Glantz LA, Lewis DA. Decreased dendritic spine density onprefrontal cortical pyramidal neurons in schizophrenia. ArchGen Psychiatry 2000; 57: 65–73.

53 Kolomeets NS, Orlovskaya DD, Uranova NA. Decreased numer-ical density of CA3 hippocampal mossy fiber synapses inschizophrenia. Synapse 2007; 61: 615–621.

54 Asaka Y, Jugloff DG, Zhang L, Eubanks JH, Fitzsimonds RM.Hippocampal synaptic plasticity is impaired in the Mecp2-nullmouse model of Rett syndrome. Neurobiol Dis 2006; 21: 217–227.

55 Moretti P, Levenson JM, Battaglia F, Atkinson R, Teague R,Antalffy B et al. Learning and memory and synaptic plasticity areimpaired in a mouse model of Rett syndrome. J Neurosci 2006;26: 319–327.

56 Nelson ED, Kavalali ET, Monteggia LM. MeCP2-dependenttranscriptional repression regulates excitatory neurotransmis-sion. Curr Biol 2006; 16: 710–716.

57 Durand CM, Perroy J, Loll F, Perrais D, Fagni L, Bourgeron T et al.SHANK3 mutations identified in autism lead to modification ofdendritic spine morphology via an actin-dependent mechanism.Mol Psychiatry 2011; 17: 71–84.

58 Comery TA, Harris JB, Willems PJ, Oostra BA, Irwin SA, Weiler IJet al. Abnormal dendritic spines in fragile X knockout mice:maturation and pruning deficits. Proc Natl Acad Sci USA 1997;94: 5401–5404.

59 Weiler IJ, Spangler CC, Klintsova AY, Grossman AW, Kim SH,Bertaina-Anglade V et al. Fragile X mental retardation protein isnecessary for neurotransmitter-activated protein translation atsynapses. Proc Natl Acad Sci USA 2004; 101: 17504–17509.

60 Barros CS, Calabrese B, Chamero P, Roberts AJ, Korzus E, Lloyd Ket al. Impaired maturation of dendritic spines without disorga-nization of cortical cell layers in mice lacking NRG1/ErbBsignaling in the central nervous system. Proc Natl Acad SciUSA 2009; 106: 4507–4512.

61 Pitcher GM, Beggs S, Woo RS, Mei L, Salter MW. ErbB4 is asuppressor of long-term potentiation in the adult hippocampus.Neuroreport 2008; 19: 139–143.

62 Chen YJ, Zhang M, Yin DM, Wen L, Ting A, Wang P et al. ErbB4 inparvalbumin-positive interneurons is critical for neuregulin 1regulation of long-term potentiation. Proc Natl Acad Sci USA2010; 107: 21818–21823.

63 Fenelon K, Mukai J, Xu B, Hsu PK, Drew LJ, Karayiorgou M et al.Deficiency of Dgcr8, a gene disrupted by the 22q11.2 microdele-tion, results in altered short-term plasticity in the prefrontalcortex. Proc Natl Acad Sci USA 2011; 108: 4447–4452.

64 Earls LR, Bayazitov IT, Fricke RG, Berry RB, Illingworth E,Mittleman G et al. Dysregulation of presynaptic calcium andsynaptic plasticity in a mouse model of 22q11 deletionsyndrome. J Neurosci 2010; 30: 15843–15855.

65 Sigurdsson T, Stark KL, Karayiorgou M, Gogos JA, Gordon JA.Impaired hippocampal-prefrontal synchrony in a genetic mousemodel of schizophrenia. Nature 2010; 464: 763–767.

66 Pappas GD, Kriho V, Pesold C. Reelin in the extracellular matrixand dendritic spines of the cortex and hippocampus: acomparison between wild type and heterozygous reeler mice byimmunoelectron microscopy. J Neurocytol 2001; 30: 413–425.

67 Mao Y, Ge X, Frank CL, Madison JM, Koehler AN, Doud MK et al.Disrupted in schizophrenia 1 regulates neuronal progenitorproliferation via modulation of GSK3beta/beta-catenin signaling.Cell 2009; 136: 1017–1031.

68 Singh KK, Ge X, Mao Y, Drane L, Meletis K, Samuels BA et al.Dixdc1 is a critical regulator of DISC1 and embryonic corticaldevelopment. Neuron 2010; 67: 33–48.

69 Duan X, Chang JH, Ge S, Faulkner RL, Kim JY, Kitabatake Y et al.Disrupted-In-Schizophrenia 1 regulates integration of newlygenerated neurons in the adult brain. Cell 2007; 130: 1146–1158.

70 Faulkner RL, Jang MH, Liu XB, Duan X, Sailor KA, Kim JY et al.Development of hippocampal mossy fiber synaptic outputs bynew neurons in the adult brain. Proc Natl Acad Sci USA 2008;105: 14157–14162.

71 Kim JY, Duan X, Liu CY, Jang MH, Guo JU, Pow-anpongkul N etal. DISC1 regulates new neuron development in the adult brainvia modulation of AKT-mTOR signaling through KIAA1212.Neuron 2009; 63: 761–773.

iPSC modeling of psychiatric disordersKJ Brennand et al

13

Molecular Psychiatry

Page 14: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

72 Wegiel J, Kuchna I, Nowicki K, Imaki H, Marchi E, Ma SY et al.The neuropathology of autism: defects of neurogenesis andneuronal migration, and dysplastic changes. Acta Neuropathol2010; 119: 755–770.

73 Rorke LB. A perspective: the role of disordered genetic controlof neurogenesis in the pathogenesis of migration disorders.J Neuropathol Exp Neurol 1994; 53: 105–117.

74 Kamiya A, Kubo K, Tomoda T, Takaki M, Youn R, Ozeki Y et al. Aschizophrenia-associated mutation of DISC1 perturbs cerebralcortex development. Nat Cell Biol 2005; 7: 1167–1178.

75 Just MA, Cherkassky VL, Keller TA, Minshew NJ. Corticalactivation and synchronization during sentence comprehensionin high-functioning autism: evidence of underconnectivity. Brain2004; 127(Part 8): 1811–1821.

76 Ruiz i Altaba A, Melton DA. Involvement of the Xenopushomeobox gene Xhox3 in pattern formation along the anterior-posterior axis. Cell 1989; 57: 317–326.

77 O’Brien WT, Klein PS. Validating GSK3 as an in vivo targetof lithium action. Biochem Soc Trans 2009; 37(Part 5):1133–1138.

78 Kim WY, Zhou FQ, Zhou J, Yokota Y, Wang YM, Yoshimura Tet al. Essential roles for GSK-3s and GSK-3-primed substrates inneurotrophin-induced and hippocampal axon growth. Neuron2006; 52: 981–996.

79 Courchesne E, Redcay E, Morgan JT, Kennedy DP. Autism at thebeginning: microstructural and growth abnormalities underlyingthe cognitive and behavioral phenotype of autism. Dev Psycho-pathol 2005; 17: 577–597.

80 Monk CS, Peltier SJ, Wiggins JL, Weng SJ, Carrasco M, Risi S et al.Abnormalities of intrinsic functional connectivity in autismspectrum disorders. Neuroimage 2009; 47: 764–772.

81 Kennedy DP, Courchesne E. The intrinsic functional organizationof the brain is altered in autism. Neuroimage 2008; 39:1877–1885.

82 Ebisch SJ, Gallese V, Willems RM, Mantini D, Groen WB, RomaniGL et al. Altered intrinsic functional connectivity of anterior andposterior insula regions in high-functioning participantswith autism spectrum disorder. Hum Brain Mapp 2011; 32:1013–1028.

83 Dinstein I, Pierce K, Eyler L, Solso S, Malach R, Behrmann M etal. Disrupted neural synchronization in toddlers with autism.Neuron 2011; 70: 1218–1225.

84 Kennedy DP, Courchesne E. Functional abnormalities of thedefault network during self- and other-reflection in autism. SocCogn Affect Neurosci 2008; 3: 177–190.

85 Yurgelun-Todd DA, Renshaw PF, Gruber SA, Ed M, Waternaux C,Cohen BM. Proton magnetic resonance spectroscopy of thetemporal lobes in schizophrenics and normal controls. SchizophrRes 1996; 19: 55–59.

86 Yurgelun-Todd DA, Waternaux CM, Cohen BM, Gruber SA,English CD, Renshaw PF. Functional magnetic resonance imagingof schizophrenic patients and comparison subjects during wordproduction. Am J Psychiatry 1996; 153: 200–205.

87 Whitfield-Gabrieli S, Thermenos HW, Milanovic S, Tsuang MT,Faraone SV, McCarley RW et al. Hyperactivity and hyperconnec-tivity of the default network in schizophrenia and in first-degreerelatives of persons with schizophrenia. Proc Natl Acad Sci USA2009; 106: 1279–1284.

88 Tan HY, Sust S, Buckholtz JW, Mattay VS, Meyer-Lindenberg A,Egan MF et al. Dysfunctional prefrontal regional specializationand compensation in schizophrenia. Am J Psychiatry 2006; 163:1969–1977.

89 Bassett DS, Bullmore E, Verchinski BA, Mattay VS, WeinbergerDR, Meyer-Lindenberg A. Hierarchical organization of humancortical networks in health and schizophrenia. J Neurosci 2008;28: 9239–9248.

90 Kessler RM, Woodward ND, Riccardi P, Li R, Ansari MS,Anderson S et al. Dopamine D2 receptor levels in striatum,thalamus, substantia nigra, limbic regions, and cortex in schizo-phrenic subjects. Biol Psychiatry 2009; 65: 1024–1031.

91 Owen F, Cross AJ, Crow TJ, Longden A, Poulter M, Riley GJ.Increased dopamine-receptor sensitivity in schizophrenia.Lancet 1978; 2: 223–226.

92 Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, BremnerJD et al. Subanesthetic effects of the noncompetitive NMDAantagonist, ketamine, in humans. Psychotomimetic, perceptual,cognitive, and neuroendocrine responses. Arch Gen Psychiatry1994; 51: 199–214.

93 Patil ST, Zhang L, Martenyi F, Lowe SL, Jackson KA, Andreev BVet al. Activation of mGlu2/3 receptors as a new approach to treatschizophrenia: a randomized Phase 2 clinical trial. Nat Med2007; 13: 1102–1107.

94 Meador-Woodruff JH, Healy DJ. Glutamate receptor expressionin schizophrenic brain. Brain Res Brain Res Rev 2000; 31:288–294.

95 Li B, Woo RS, Mei L, Malinow R. The neuregulin-1 receptorerbB4 controls glutamatergic synapse maturation and plasticity.Neuron 2007; 54: 583–597.

96 Stefansson H, Sigurdsson E, Steinthorsdottir V, Bjornsdottir S,Sigmundsson T, Ghosh S et al. Neuregulin 1 and susceptibility toschizophrenia. Am J Hum Genet 2002; 71: 877–892.

97 Dickman DK, Davis GW. The schizophrenia susceptibility genedysbindin controls synaptic homeostasis. Science (New York,NY) 2009; 326: 1127–1130.

98 Noutel J, Hong YK, Leu B, Kang E, Chen C. Experience-dependentretinogeniculate synapse remodeling is abnormal in MeCP2-deficient mice. Neuron 2011; 70: 35–42.

99 Dani VS, Chang Q, Maffei A, Turrigiano GG, Jaenisch R,Nelson SB. Reduced cortical activity due to a shift in the balancebetween excitation and inhibition in a mouse model of Rettsyndrome. Proc Natl Acad Sci USA 2005; 102: 12560–12565.

100 Penagarikano O, Abrahams BS, Herman EI, Winden KD,Gdalyahu A, Dong H et al. Absence of CNTNAP2 leads toepilepsy, neuronal migration abnormalities, and core autism-related deficits. Cell 2011; 147: 235–246.

101 Pitcher GM, Kalia LV, Ng D, Goodfellow NM, Yee KT, Lambe EKet al. Schizophrenia susceptibility pathway neuregulin 1-ErbB4suppresses Src upregulation of NMDA receptors. Nat Med 2011;17: 470–478.

102 Geyer MA, Swerdlow NR, Mansbach RS, Braff DL. Startleresponse models of sensorimotor gating and habituation deficitsin schizophrenia. Brain Res Bull 1990; 25: 485–498.

103 Caine SB, Geyer MA, Swerdlow NR. Effects of D3/D2 dopaminereceptor agonists and antagonists on prepulse inhibition ofacoustic startle in the rat. Neuropsychopharmacology 1995; 12:139–145.

104 Carlson GC, Talbot K, Halene TB, Gandal MJ, Kazi HA, SchlosserL et al. From the cover: Dysbindin-1 mutant mice implicatereduced fast-phasic inhibition as a final common diseasemechanism in schizophrenia. Proc Natl Acad Sci USA 2011;108: E962–E970.

105 Takahashi K, Yamanaka S. Induction of pluripotent stem cellsfrom mouse embryonic and adult fibroblast cultures by definedfactors. Cell 2006; 126: 663–676.

106 Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, TomodaK et al. Induction of pluripotent stem cells from adult humanfibroblasts by defined factors. Cell 2007; 131: 861–872.

107 Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, FraneJL, Tian S et al. Induced pluripotent stem cell lines derivedfrom human somatic cells. Science (New York, NY) 2007; 318:1917–1920.

108 Tropepe V, Hitoshi S, Sirard C, Mak TW, Rossant J, van der KooyD. Direct neural fate specification from embryonic stem cells: aprimitive mammalian neural stem cell stage acquired through adefault mechanism. Neuron 2001; 30: 65–78.

109 Watanabe K, Kamiya D, Nishiyama A, Katayama T, Nozaki S,Kawasaki H et al. Directed differentiation of telencephalicprecursors from embryonic stem cells. Nat Neurosci 2005; 8:288–296.

110 Kawasaki H, Mizuseki K, Nishikawa S, Kaneko S, Kuwana Y,Nakanishi S et al. Induction of midbrain dopaminergic neuronsfrom ES cells by stromal cell-derived inducing activity. Neuron2000; 28: 31–40.

111 Perrier AL, Tabar V, Barberi T, Rubio ME, Bruses J, Topf N et al.Derivation of midbrain dopamine neurons from human embryo-nic stem cells. Proc Natl Acad Sci USA 2004; 101: 12543–12548.

iPSC modeling of psychiatric disordersKJ Brennand et al

14

Molecular Psychiatry

Page 15: Modeling psychiatric disorders at the cellular and network …...Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number

112 Li XJ, Du ZW, Zarnowska ED, Pankratz M, Hansen LO, Pearce RAet al. Specification of motoneurons from human embryonic stemcells. Nat Biotechnol 2005; 23: 215–221.

113 Wichterle H, Lieberam I, Porter JA, Jessell TM. Directeddifferentiation of embryonic stem cells into motor neurons. Cell2002; 110: 385–397.

114 Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Sudhof TC,Wernig M. Direct conversion of fibroblasts to functional neuronsby defined factors. Nature 2010; 463: 1035–1041.

115 Pang ZP, Yang N, Vierbuchen T, Ostermeier A, Fuentes DR, YangTQ et al. Induction of human neuronal cells by definedtranscription factors. Nature 2011; 476: 220–223.

116 Kim J, Su SC, Wang H, Cheng AW, Cassady JP, Lodato MA et al.Functional integration of dopaminergic neurons directly con-verted from mouse fibroblasts. Cell Stem Cell 2011; 9: 413–419.

117 Vrijenhoek T, Buizer-Voskamp JE, van der Stelt I, Strengman E,Sabatti C, Geurts van Kessel A et al. Recurrent CNVs disruptthree candidate genes in schizophrenia patients. Am J HumGenet 2008; 83: 504–510.

118 Riley B, Thiselton D, Maher BS, Bigdeli T, Wormley B,McMichael GO et al. Replication of association between schizo-phrenia and ZNF804A in the irish case-control study ofschizophrenia sample. Mol Psychiatry 2010; 15: 29–37.

119 Sun G, Tomita H, Shakkottai VG, Gargus JJ. Genomic organizationand promoter analysis of human KCNN3 gene. J Hum Genet 2001;46: 463–470.

120 Ide M, Yamada K, Toyota T, Iwayama Y, Ishitsuka Y, Minabe Yet al. Genetic association analyses of PHOX2B and ASCL1 inneuropsychiatric disorders: evidence for association of ASCL1with Parkinson’s disease. Hum Genet 2005; 117: 520–527.

121 Lin M, Pedrosa E, Shah A, Hrabovsky A, Maqbool S, Zheng Det al. RNA-Seq of human neurons derived from iPS cells revealscandidate long non-coding RNAs involved in neurogenesis andneuropsychiatric disorders. PLoS One 2011; 6: e23356.

122 Cheung AY, Horvath LM, Grafodatskaya D, Pasceri P, Weksberg R,Hotta A et al. Isolation of MECP2-null rett syndrome patient hiPScells and isogenic controls through X-chromosome inactivation.Hum Mol Genet 2011; 20: 2103–2115.

123 Marchetto MC, Carromeu C, Acab A, Yu D, Yeo GW, Mu Y et al.A model for neural development and treatment of rett syndromeusing human induced pluripotent stem cells. Cell 2010; 143:527–539.

124 Ananiev G, Williams EC, Li H, Chang Q. Isogenic pairs of wildtype and mutant induced pluripotent stem cell (iPSC) lines fromrett syndrome patients as in vitro disease model. PLoS One 2011;6: e25255.

125 O’Donnell WT, Warren ST. A decade of molecular studies offragile X syndrome. Annu Rev Neurosci 2002; 25: 315–338.

126 Nichol Edamura K, Pearson CE. DNA methylation and replica-tion: implications for the ‘deletion hotspot’ region of FMR1. HumGenet 2005; 118: 301–304.

127 Urbach A, Bar-Nur O, Daley GQ, Benvenisty N. Differentialmodeling of fragile X syndrome by human embryonic stemcells and induced pluripotent stem cells. Cell Stem Cell 2010; 6:407–411.

128 Sheridan SD, Theriault KM, Reis SA, Zhou F, Madison JM,Daheron L et al. Epigenetic characterization of the FMR1 gene andaberrant neurodevelopment in human induced pluripotent stemcell models of fragile x syndrome. PLoS One 2011; 6: e26203.

129 Pasca SP, Portmann T, Voineagu I, Yazawa M, Shcheglovitov A,Pasca AM et al. Using iPSC-derived neurons to uncover cellularphenotypes associated with Timothy syndrome. Nat Med 2011;17: 1657–1662.

130 Yazawa M, Hsueh B, Jia X, Pasca AM, Bernstein JA, Hallmayer Jet al. Using induced pluripotent stem cells to investigate cardiacphenotypes in Timothy syndrome. Nature 2011; 471: 230–234.

131 Chiang CH, Su Y, Wen Z, Yoritomo N, Ross CA, Margolis RL et al.Integration-free induced pluripotent stem cells derived fromschizophrenia patients with a DISC1 mutation. Mol Psychiatry2011; 16: 358–360.

132 Brennand KJ, Simone A, Jou J, Gelboin-Burkhart C, Tran N,Sangar S et al. Modelling schizophrenia using human inducedpluripotent stem cells. Nature 2011; 473: 221–225.

133 Paulsen BD, Maciel RD, Galina A, da Silveira MS, Souza CD,Drummond H et al. Altered oxygen metabolism associated toneurogenesis of induced pluripotent stem cells derived from aschizophrenic patient. Cell Transplant 2011 (e-pub ahead ofprint).

134 Meechan DW, Maynard TM, Tucker ES, LaMantia AS. Threephases of DiGeorge/22q11 deletion syndrome pathogenesisduring brain development: patterning, proliferation, and mito-chondrial functions of 22q11 genes. Int J Dev Neurosci 2011; 29:283–294.

135 Park YU, Jeong J, Lee H, Mun JY, Kim JH, Lee JS et al. Disrupted-in-schizophrenia 1 (DISC1) plays essential roles in mitochondriain collaboration with Mitofilin. Proc Natl Acad Sci USA 2010;107: 17785–17790.

136 Gore A, Li Z, Fung HL, Young JE, Agarwal S, Antosiewicz-Bourget J et al. Somatic coding mutations in human inducedpluripotent stem cells. Nature 2011; 471: 63–67.

137 Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery JR,Hon G et al. Hotspots of aberrant epigenomic reprogrammingin human induced pluripotent stem cells. Nature 2011; 471:68–73.

138 Osafune K, Caron L, Borowiak M, Martinez RJ, Fitz-Gerald CS,Sato Y et al. Marked differences in differentiation propensityamong human embryonic stem cell lines. Nat Biotechnol 2008;26: 313–315.

139 Hu BY, Weick JP, Yu J, Ma LX, Zhang XQ, Thomson JA et al.Neural differentiation of human induced pluripotent stem cellsfollows developmental principles but with variable potency. ProcNatl Acad Sci USA 2010; 107: 4335–4340.

iPSC modeling of psychiatric disordersKJ Brennand et al

15

Molecular Psychiatry