11
Cell Stem Cell Resource An Efficient and Reversible Transposable System for Gene Delivery and Lineage-Specific Differentiation in Human Embryonic Stem Cells Arnaud Lacoste, 1,2 Frada Berenshteyn, 1 and Ali H. Brivanlou 1, * 1 Laboratory of Molecular Embryology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA 2 Present address: Novartis Institutes for Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA *Correspondence: [email protected] DOI 10.1016/j.stem.2009.07.011 SUMMARY Unraveling the therapeutic potential of human embryonic stem cells (hESC) requires tools to modify their genome. We have engineered the PiggyBac transposable element to create an efficient system for gene delivery in hESCs. This redesigned system, named ‘‘ePiggyBac,’’ can deliver up to 18 Kb inserts, and transgene expression is observed in almost 90% of hES cells. ePiggyBac transposons can also carry insulators, inducible expression cassettes, and short hairpin RNAs for gain- and loss-of-function approaches. In hES cells, ePiggyBac’s efficiency is superior to that of viral vectors and previously described transposons, including other PiggyBac- based systems. In addition, ePiggyBac transgenes can be removed from the hESC genome without leaving any mutation. We used this system to direct hESC differentiation toward a neuronal phenotype. We then removed the transposons to obtain trans- gene-free neuronal precursors and neurons. The ability to create fully reversible genetic modifications represents an important step toward clinical applica- tions of hESCs. INTRODUCTION Human pluripotent stem cells hold remarkable potential for regenerative medicine and drug screens. They also provide a unique opportunity to improve our understanding of human development and diseases. However, a difficulty in generating transgenic hESC lines and use of reporter systems or gain- and loss-of-function approaches still limit our ability to fully unravel their potential. Currently, gene transfer into hESCs is mostly based on the use of viral vectors. These vectors can mediate transgene expres- sion in 20%–80% of human ES cells (Ben-Dor et al., 2006; Pfeifer et al., 2002), but their cargo size is restricted to about 5 Kb and their insert is often limited to one expression cassette. Nonviral systems that use the Sleeping Beauty transposon are also restricted to a cargo size of 5–6 Kb (Wilber et al., 2007). These characteristics limit the use of selectable markers, inducible cassettes, insulators, and large regulatory sequences often required to restrict transgene expression to specific cell types. Baculoviruses can deliver larger cargo to hESC (Zeng et al., 2007), but the long-term effect of adeno-associated rep protein expression could lead to undesirable effects (McCarty et al., 2004). In principle, plasmids and bacterial artificial chromo- somes (BACs) can be used when large promoters are required, but stable integration of these vectors in the hESC genome is relatively inefficient and plasmid-borne transgenes are subject to silencing (Braam et al., 2008). In addition, BACs usually carry several intact genes in addition to the engineered locus; therefore, their integration in the genome leads to gene multipli- cations. This may have important consequences when the supernumerary genes have regulatory functions. Regardless of efficiency, gene transfer technologies currently available in hESCs such as viral vectors, Sleeping Beauty trans- posons, or PhiC31 integrase (Thyagarajan et al., 2008) mediate irreversible genome modifications. This constitutes a major barrier to the clinic because the presence of exogenous inserts in certain loci may lead to higher predisposition toward tumori- genesis or uncontrolled cellular behavior. In the present study, we designed a gene transfer system for reversible integration of large multigene inserts in hESCs. We chose the PiggyBac transposon (Cary et al., 1989; Fraser et al., 1995, 1996) as a starting point for our design because this gene delivery system is nonviral and can carry cargo sizes of up to 14.3 Kb. It also exhibits higher activity than other transposons in mammalian models (Cadinanos and Bradley, 2007; Ding et al., 2005; Wilson et al., 2007). Importantly, PiggyBac does not leave mutations upon remobilization in HEK293 cells (Wilson et al., 2007), suggesting that it could be used for fully reversible transgene delivery in hESC. This property was recently used to generate transgene-free induced pluripotent cells in the mouse (Yusa et al., 2009; Kaji et al., 2009; Woltjen et al., 2009). RESULTS Engineering a PiggyBac Transposon for Efficient Gene Transfer Efficiency in hESCs The original 2.4 Kb PiggyBac element includes the PiggyBac transposase, which mediates gene transfer, and a transposon flanked by 5 0 - and 3 0 -terminal repeats necessary for transposi- tion (Cary et al., 1989; Fraser et al., 1995, 1996). For gene delivery experiments, the PiggyBac system can be divided into a donor plasmid that contains the transposon and a helper plasmid that expresses the transposase (Cary et al., 1989; Wilson et al., 332 Cell Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier Inc.

An Efficient and Reversible Transposable System for Gene Delivery and Lineage-Specific Differentiation in Human Embryonic Stem Cells

Embed Size (px)

Citation preview

Cell Stem Cell

Resource

An Efficient and Reversible Transposable Systemfor Gene Delivery and Lineage-SpecificDifferentiation in Human Embryonic Stem CellsArnaud Lacoste,1,2 Frada Berenshteyn,1 and Ali H. Brivanlou1,*1Laboratory of Molecular Embryology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA2Present address: Novartis Institutes for Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA

*Correspondence: [email protected] 10.1016/j.stem.2009.07.011

SUMMARY

Unraveling the therapeutic potential of humanembryonic stem cells (hESC) requires tools to modifytheir genome. We have engineered the PiggyBactransposable element to create an efficient systemfor gene delivery in hESCs. This redesigned system,named ‘‘ePiggyBac,’’ can deliver up to 18 Kb inserts,and transgene expression is observed in almost 90%of hES cells. ePiggyBac transposons can also carryinsulators, inducible expression cassettes, andshort hairpin RNAs for gain- and loss-of-functionapproaches. In hES cells, ePiggyBac’s efficiency issuperior to that of viral vectors and previouslydescribed transposons, including other PiggyBac-based systems. In addition, ePiggyBac transgenescan be removed from the hESC genome withoutleaving any mutation. We used this system to directhESC differentiation toward a neuronal phenotype.We then removed the transposons to obtain trans-gene-free neuronal precursors and neurons. Theability to create fully reversible genetic modificationsrepresents an important step toward clinical applica-tions of hESCs.

INTRODUCTION

Human pluripotent stem cells hold remarkable potential for

regenerative medicine and drug screens. They also provide a

unique opportunity to improve our understanding of human

development and diseases. However, a difficulty in generating

transgenic hESC lines and use of reporter systems or gain-

and loss-of-function approaches still limit our ability to fully

unravel their potential.

Currently, gene transfer into hESCs is mostly based on the use

of viral vectors. These vectors can mediate transgene expres-

sion in 20%–80% of human ES cells (Ben-Dor et al., 2006; Pfeifer

et al., 2002), but their cargo size is restricted to about 5 Kb and

their insert is often limited to one expression cassette. Nonviral

systems that use the Sleeping Beauty transposon are also

restricted to a cargo size of 5–6 Kb (Wilber et al., 2007). These

characteristics limit the use of selectable markers, inducible

cassettes, insulators, and large regulatory sequences often

332 Cell Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier In

required to restrict transgene expression to specific cell types.

Baculoviruses can deliver larger cargo to hESC (Zeng et al.,

2007), but the long-term effect of adeno-associated rep protein

expression could lead to undesirable effects (McCarty et al.,

2004). In principle, plasmids and bacterial artificial chromo-

somes (BACs) can be used when large promoters are required,

but stable integration of these vectors in the hESC genome is

relatively inefficient and plasmid-borne transgenes are subject

to silencing (Braam et al., 2008). In addition, BACs usually carry

several intact genes in addition to the engineered locus;

therefore, their integration in the genome leads to gene multipli-

cations. This may have important consequences when the

supernumerary genes have regulatory functions.

Regardless of efficiency, gene transfer technologies currently

available in hESCs such as viral vectors, Sleeping Beauty trans-

posons, or PhiC31 integrase (Thyagarajan et al., 2008) mediate

irreversible genome modifications. This constitutes a major

barrier to the clinic because the presence of exogenous inserts

in certain loci may lead to higher predisposition toward tumori-

genesis or uncontrolled cellular behavior.

In the present study, we designed a gene transfer system for

reversible integration of large multigene inserts in hESCs. We

chose the PiggyBac transposon (Cary et al., 1989; Fraser et al.,

1995, 1996) as a starting point for our design because this gene

delivery system is nonviral and can carry cargo sizes of up to

14.3 Kb. It also exhibits higher activity than other transposons

in mammalian models (Cadinanos and Bradley, 2007; Ding

et al., 2005; Wilson et al., 2007). Importantly, PiggyBac does

not leave mutations upon remobilization in HEK293 cells (Wilson

et al., 2007), suggesting that it could be used for fully reversible

transgene delivery in hESC. This property was recently used to

generate transgene-free induced pluripotent cells in the mouse

(Yusa et al., 2009; Kaji et al., 2009; Woltjen et al., 2009).

RESULTS

Engineering a PiggyBac Transposon for Efficient GeneTransfer Efficiency in hESCsThe original 2.4 Kb PiggyBac element includes the PiggyBac

transposase, which mediates gene transfer, and a transposon

flanked by 50- and 30-terminal repeats necessary for transposi-

tion (Cary et al., 1989; Fraser et al., 1995, 1996). For gene delivery

experiments, the PiggyBac system can be divided into a donor

plasmid that contains the transposon and a helper plasmid

that expresses the transposase (Cary et al., 1989; Wilson et al.,

c.

Cell Stem Cell

ePiggyBac

2007). To test the ability of PiggyBac to mediate gene transfer in

hESCs, we first designed basic EGFP- or RFP-expressing donor

plasmids (Figure 1A) that contain a 50-terminal repeat (50TR) of

313 bp and a 30-terminal repeat (30TR) of 235 bp. These two

minimal sequences are necessary and sufficient for transposi-

tion (Li et al., 2005). Our initial attempts used a helper plasmid

expressing the original insect PiggyBac transposase. Donor and

helper plasmids were cotransfected in the hESC line RUES2

and genome integration efficiency was assessed by counting

the number of EGFP-positive colonies 7 days after transfection.

In control experiments, the donor plasmid was transfected in

the absence of transposase in circular or linearized forms to

compare the efficiency of PiggyBac-mediated gene delivery

with that of random integration of plasmid DNA.

The original PiggyBac transposase is active in hESCs and

mediates higher genome integration efficiency than random inte-

gration of a linearized plasmid (Figure 1B), but transposition effi-

ciency remains low (10.4 ± 6.04 EGFP-positive colonies per 2000

transfected cells). This prompted us to engineer the PiggyBac

transposable element to optimize its activity in hESCs. Because

PiggyBac was originally isolated from an insect, we introduced

silent mutations to generate a codon usage similar to that of

humans. A piggyBac transposase that uses rodent codon usage

has already been described (Cadinanos and Bradley, 2007), but

human splice site prediction algorithms (Brunak et al., 1991)

detect the presence of cryptic splicing sites in that synthetic

transgene. This prompted us to design an improved transposase

cDNA for use in human embryonic stem cells (Figure S1 available

online). The codon-humanized PiggyBac transposase increased

genome integration efficiency by 10-fold: 5% of transfected cells

stably integrated the transgene and gave rise to green fluorescent

colonies (Figure 1B). This rate of transgenesis is similar to that of

other transposable systems for inserts of this size (Wilber et al.,

2007).

We next engineered the transposon’s 50- and 30-terminal

repeats to further improve gene delivery. The terminal repeats

were modified by random PCR-mediated mutagenesis. Twenty

different mutants were isolated for each terminal repeat and

used to replace their wild-type counterparts in the original donor

plasmid (Figure 1A). Mutated donor plasmids were then cotrans-

fected with a helper plasmid containing the codon-humanized

transposase cDNA. A 59% increase in transposition efficiency

(Figure 1C) was observed in a 50TR mutant carrying T53C and

C136T substitutions (Figure S2). The improved PiggyBac

system, comprising the codon-humanized transposase and

the T53C/C136T mutant 50TR, was named ePiggyBac for

‘‘enhanced PiggyBac.’’

To ensure that ePiggyBac could be widely used in hESC

research, we performed gene delivery experiments in hESC

and nonhuman primate ESC lines. ePiggyBac could efficiently

transfer EGFP- or RFP-expressing transgenes in different

hESC lines (Figure 1D). We also found that ePiggyBac-mediated

gene delivery does not alter the hESC karyotype (Figure S3) or

the ability of hESCs to express pluripotency markers, form

embryoid bodies, and differentiate into all three germ layers

(Figures S4 and S5). In addition, gene transfer efficiency re-

mained high in nonhuman primate ESCs (Figure 1D), indicating

that ePiggyBac can be used as a generic gene delivery system

in primate ESCs.

Ce

We also investigated whether silencing, which is known to

occasionally occur in plasmid- and viral-based transgenes in

hESCs, affects ePiggyBac-mediated transgenesis. We gener-

ated two single-transgenic RUES2 lines expressing either

EGFP or RFP and a double transgenic RUES2 line expressing

both EGFP and RFP. These lines were passaged 25 times without

selection and the proportion of cells that had lost transgene

expression was then quantified by FACS analysis. Results

showed that in these three independent clones, the proportion

of nonfluorescent events detected by flow cytometry was

between 0.1% and 0.2% (Figure 1E), indicating that silencing of

ePiggyBac transposons is rare, possibly more so than silencing

of plasmid- and virus-based transgenes.

One of the most important advantages of PiggyBac over other

gene delivery systems lies in its ability to carry large cargo size

(Ding et al., 2005). To investigate ePiggyBac’s ability to integrate

large DNA constructs in the hESC genome, we added inserts of

increasing size to our basic transposon (Figure 1F). We found

that, although transposition efficiency decreased significantly

for inserts larger than 12 Kb, ePiggyBac could deliver inserts of

up to 18 Kb in size to the hESC genome, which is larger than

the insert size delivered by the original PiggyBac system

(Figure 1F) or the maximum 14.3 Kb insert size previously

reported for mouse embryos (Fraser et al., 1995).

We next tested whether ePiggyBac could deliver large inserts

containing several components such as expression cassettes,

insulator sequences, and plasmid rescue systems. We created

a 12 Kb transposable insert containing an origin of replication

for plasmid rescue, a constitutively expressed neomycin-phos-

photransferase cassette and a reporter construct where EGFP

expression is driven by the human Oct4 promoter (Figure 1G;

Gerrard et al., 2005). We included a chicken HS4 insulator (Recil-

las-Targa et al., 2002) to prevent the constitutively expressed

neomycin cassette from interfering with the hOct4-EGFP

reporter, which should label only undifferentiated hESCs (Gerrard

et al., 2005). After gene delivery, 7 days of neomycin selection

were sufficient to select EGFP-positive colonies that could be

expanded to establish an hOct4-EGFP RUES2 cell line. To test

the specificity of EGFP labeling, we compared levels of fluores-

cence in cells cultured in nondifferentiating conditions to those

in cells maintained in differentiating conditions (Figure 1H).

Live-cell imaging showed that EGFP labeling is lost in cells main-

tained in differentiating conditions and in 18-day-old embryoid

bodies. In addition, coimmunostaining showed that the decrease

in EGFP expression is consistent with the decrease in endoge-

nous Oct4 protein expression (Figures 1I and 1J). These experi-

ments indicate that ePiggyBac can integrate large multicompo-

nent inserts into the hESC genome without altering their

functionality.

Finally, we systematically altered gene-delivery parameters

to define conditions that would further increase gene-delivery

efficiency and complement our molecular modifications to the

transposase and transposon. We found that expressing the

transposase from the CAG promoter, by means of 6 mg of DNA

per 1.5 3 105 cells, a transposase/transposon ratio of 1:2, and

transfection solution L instead of V (Siemen et al., 2005) led to

a dramatic increase in transgenesis efficiency. By using ePiggy-

Bac under these conditions, we commonly obtain transgene

expression in almost 90% (Figure 1K) of hESC colonies for a

ll Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier Inc. 333

Cell Stem Cell

ePiggyBac

# of

fluo

resc

ent c

olon

ies

% o

f flu

ores

cent

col

onie

s

12

*

1 2 3 4 5 6 7 8 9 10 11 13 14 15 16 17 18 19 20wt

200

150

100

50

00

30

60

90

120

neg. lin. wt co

*

Tran

spos

on c

opy

num

ber

μg ePiggyBac transposaseGene delivery protocol

RUES2 RUES1 H1 HUES10

BGN1 M. fascicularis M. nemestrina

# of

fluo

resc

ent c

olon

ies

M. mulatta

EGFPPGK pA

A

B C

D

RFPPGK pA

EGFPhOct4 pANeo Ins

F

H

G

I J

CM

d

ay

8

EGFP

Oct 4

No

C

M d

ay

8

EGFP

Oct 4

CM

No

C

M

day 0 day 2 day 4 day 6 day 8 EB day 18

Size (Kb)

0

50

100

150

200

1.5 3 4 6 8 10 12 14 16 18

Num

ber o

f col

onie

s

Merge

Sytox Sytox

Merge

K

RUES2 RUES2-EGFP

RUES2-RFP RUES2-EGFP/RFP

EGFP

RF

P

E

wild-typeePiggyBac

Basic Improved

wild-typeePiggyBac

0

50

100

0

4

8

12

0 0.5 1 1.5 2 2.5 3

L

Figure 1. An Engineered PiggyBac Transposable Element for Efficient Gene Delivery in Human and Nonhuman Primate ESCs

(A) Schematic representation of the basic EGFP- or RFP-expressing transposable transgenes. Purple triangles represent 50- and 30-terminal repeats required for

transposition.

(B) RUES2 cells were transfected with a circular plasmid containing the EGFP-expressing transposon (neg.), a linearized plasmid containing the EGFP-expressing

transposon (lin.), or a circular plasmid containing the transposon together with a helper plasmid expressing the original PiggyBac transposase (wt) or the codon-

humanized PiggyBac transposase (co). Two thousand cells were plated and cultured for 7 days before numbers of fluorescent colonies were determined. The

codon-humanized transposase mediated significantly higher gene transfer efficiency (p < 0.01 co versus wt, n = 3 independent repeats).

(C) To improve gene-transfer efficiency, 20 50TR mutants were generated by random PCR mutagenesis. Mutant number 16, which carries T53C and C136T

mutations, led to a significant (p < 0.01 versus control, n = 3 independent repeats) increase in gene delivery.

(D) The engineered PiggyBac system composed of the codon-humanized transposase and the T53C/C136T mutant 50TR mediates efficient gene delivery in hESC

lines RUES2, RUES1, H1, HUES10, and BGN1 as well as Macacca ESC lines.

(E) FACS analysis of three independent transgenic RUES2 clones (RUES2-EGFP, RUES2-RFP, RUES2-EGFP/RFP) after 25 passages without selection indicate

that ePiggyBac transposon silencing is minimal.

(F) Inserts of increasing size were added to the basic PGK-EGFP transposon and number of fluorescent colonies were counted 7 days after gene delivery

by ePiggyBac (solid bars) or original PiggyBac (open bars). ePiggyBac was able to deliver transgenes of up to 18 Kb with significantly higher efficiency

(p < 0.001 ePiggyBac versus original PiggyBac, n = 3 studies).

(G) Schematic representation of the hOct4-EGFP transposable element. The 12 Kb transgene includes a neomycin selection cassette (Neo), an HS4 chicken

insulator, and a 4 Kb human Oct4 promoter driving EGFP expression (pA, poly(A) signal).

(H) Time-lapse imaging of single neomycin-resistant colonies kept undifferentiated in conditioned medium (CM) or differentiated in nonconditioned medium (No

CM). Fluorescence levels remain high in undifferentiated cells while they decrease with time as differentiation progresses. 18-day embryoid bodies (EB) have lost

most of the hOct4-EGFP labeling.

334 Cell Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier Inc.

Cell Stem Cell

ePiggyBac

3 Kb transposon (72.1% ± 8.3% for an 18 Kb transposon), which

is higher than gene delivery efficiencies reported for viral vectors

(Ben-Dor et al., 2006; Pfeifer et al., 2002). We currently use

ePiggyBac routinely and regularly conduct karyotypic analyses

to verify that our ePiggyBac transgenic cell lines remain free of

chromosomal abnormalities. Analysis of more than 200 meta-

phases in various cell lines at passage numbers ranging from

15 to 55 has so far not revealed any chromosomal abnormalities.

This indicates that ePiggyBac transgenesis does not cause more

chromosome instability in hESCs than other gene-delivery

methods. SplinkTA PCR analyses of 70 samples indicated that

transgenic hESCs typically carried 1–12 copies when 2–3 mg of

the transposase-expressing plasmid is delivered to the cells

(Figure 1L). Interestingly, changing the amount of transposase-

expressing plasmid allows control over the number of transpo-

sons integrated in the hESC genome (Figure 1L).

Interestingly, we have noticed that in mouse ES cells, around

90% of colonies also exhibit stable transgene expression (not

shown). For comparison, Cadinanos and Bradley (2007) reported

that transfection of 107 AB2.2 mouse ES cells with a mouse

codon-optimized PiggyBac transposase gave rise to only about

1000 transgenic colonies. Therefore, it appears that the combi-

nation of transposase cDNA and terminal repeat modifications

that constitute ePiggyBac are not merely incremental improve-

ments. In ES cells, ePiggyBac’s efficiency is not only superior

to that of viral vectors, it is also largely superior to that of previ-

ously described transposons, including other PiggyBac-based

systems (Cadinanos and Bradley, 2007; Woltjen et al., 2009;

Kaji et al., 2009). In addition, the efficiency of ePiggyBac is

conserved between human, nonhuman primate, and rodent

ES cells.

ePiggyBac Retains Basic PiggyBac CharacteristicsTo determine whether gene transfer was the result of a transpo-

sition event, sites of integration were isolated via a plasmid

rescue strategy (Figure 2A), sequenced, and analyzed by BLAST

search of the Ensembl database. Analysis of 62 ePiggyBac inte-

gration sites revealed no obvious consensus sequence other

than the TTAA tetranucleotide sequence (Figure 2B) previously

shown to be required for PiggyBac transposition (Cadinanos

and Bradley, 2007; Cary et al., 1989; Ding et al., 2005; Fraser

et al., 1995, 1996; Li et al., 2005; Wilson et al., 2007). A nucleotide

frequency plot (Figure 2C) also revealed a preference for AT-rich

regions around the core TTAA sequence. Sequence analyses

showed that ePiggyBac integrations occurred in all chromo-

somes of the female hESC line RUES2 (Figure 2D). Interestingly,

72% of integrations occurred within 10 Kb of a known transcrip-

tion start site, and 96% of integrations that took place

downstream of the transcription start site occurred in introns

(Figure 2E) as described for the original PiggyBac transposable

Ce

element (Ding et al., 2005; Wilson et al., 2007). These data

indicate that mutations introduced in the PiggyBac system

increased gene delivery efficiency without altering its basic prop-

erties. It is noteworthy that preferential transgene integration in

introns is also a feature of viral-based gene-delivery methods.

This indicates that alterations in gene expression patterns in

ePiggyBac transgenics are theoretically possible, as with virus-

based or other gene-delivery methods.

ePiggyBac Transgenes Can Be Removedfrom the GenomeIn contrast to viral vectors and PhiC31 systems, transposons

can be remobilized and moved from one locus to another by

re-expression of the transposase (Elick et al., 1996; Wilson

et al., 2007). Theoretically, re-expression of the transposase

followed by random transgene segregation upon remobilization

could enable reversion from a transgenic to a wild-type geno-

type. This approach could therefore be used for reversible

transgenesis in hESCs. However, our initial attempts to remove

transgenes from transgenic hESCs by re-expression of the

ePiggyBac transposase were largely unsuccessful. It appeared

that extensive passaging and selection procedures would be

needed to select the few hESCs where transgenes have been

segregated out of the genome. As an alternative, we investi-

gated whether transposons could be transferred from the

genome to a recipient plasmid.

Transposons are commonly transferred from a donor plasmid

to a recipient plasmid or to genomic DNA. Transfer of ePiggyBac

transposons from the genome to a plasmid would enable rever-

sions from a transgenic to a completely intact genome because

PiggyBac does not leave any mutation upon excision. However,

transposition from the genome to a recipient plasmid is an

unlikely event because plasmids are million times smaller than

the human genome, and therefore, genomic DNA is a much

more likely target for integration. To increase the probability

that ePiggyBac transposons excised from the genome are

transferred to a plasmid rather than moved to another genomic

location, we first engineered a recipient plasmid carrying 14

‘‘upstream activating sequences’’ (UAS). Next, we generated

a helper plasmid where the Gal4 DNA binding domain is fused

to the N terminus of the ePiggyBac transposase. Upon cotrans-

fection of these two vectors, the Gal4-ePiggyBac chimeric

protein binds to the UAS-carrying recipient plasmid with high

affinity, which strongly increases chances that transposons are

transferred to the recipient plasmid without significantly affecting

transposase activity (Maragathavally et al., 2006). To test

whether this strategy enables the removal of transposons from

the hESC genome, we created an RFP-expressing transposon

where neomycin-phosphotransferase and thymidine kinase

cassettes are used for negative and positive selection,

(I and J) Simultaneous immunohistochemical labeling of both endogenous Oct4 and EGFP in undifferentiated (I) and differentiated (J) cells confirms that EGFP

labeling is consistent with endogenous Oct 4 expression. The hOct4-EGFP reporter transgene delivered by ePiggyBac is therefore functional.

(K) Final optimization of transposition parameters: compared to basic gene delivery protocols (basic) inspired by published data (Siemen et al., 2005), expressing

the transposase from the CAG promoter, with 6 mg of DNA per 1.5 3 105 cells, a transposase/transposon ratio of 1:2 and transfection solution L (improved) led to

a significant increase in transgenesis efficiency.

(L) Different amounts of ePiggyBac transposase per nucleofection reaction allows control over the number of transposons integrated in the hESC genome.

Inset in EB photo shows embryoid body EGFP fluorescence level.

Scale bars represent 100 mm (A–D, H), 200 mm (I, J). Asterisks indicate significant difference (p < 0.01) between clone 16 and wtPiggyBac.

ll Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier Inc. 335

Cell Stem Cell

ePiggyBac

0

25

50

75

100

Introns Exons 5' End 3' End

Freq

uenc

y (%

)

E

PGK pAAmpori EGFP

A

B

C

D

Figure 2. ePiggyBac Retains Basic Properties of the Original

PiggyBac System

(A) Schematic representation of the plasmid rescue system used to sequence

and map integration sites. In addition to the EGFP-expressing cassette, the

transposon includes a DNA replication origin and an ampicillin selection

cassette. 62 integration sites were sequenced.

(B) Consensus logo analysis revealed no obvious consensus sequence other

than the TTAA tetranucleotide sequence required for PiggyBac transposition.

(C) Frequency plots revealed a preference for AT-rich regions around the site of

integration.

(D) ePiggyBac exhibited no hot spot for integration and all chromosomes of the

XX ESC line RUES2 were hit.

(E) Distribution of insertions within known transcription units revealed a prefer-

ence for introns over exons, a 10 Kb window upstream of the transcription start

site, and a 10 Kb window downstream of the poly(A) site.

336 Cell Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier In

respectively (Figure 3A). This construct enabled us to test the

approach schematized in Figure 3B. For transgene integration,

the transposon is cotransfected with the ePiggyBac-expressing

vector. Transposon remobilization is expected to occur once or

several times until the helper plasmid is diluted by cell division

and transposase activity wears off. Once transposase activity

has disappeared, the transgene remains immobile on the

host genome. For transposon removal, binding of the Gal4-

ePiggyBac chimeric protein to the UAS-carrying recipient

plasmid increases the chances that the transposon is remobi-

lized from the host genome to the recipient plasmid (Maragatha-

vally et al., 2006). To test this strategy, we first used ePiggyBac

to create an RUES2 cell line containing a single copy of the RFP-

expressing transposon (Figure 3A) as determined by SplinkTA

PCR. This cell line was maintained under 200 mg/ml neomycin

selection for 5 passages to eliminate nontransgenic cells. It is

ganciclovir sensitive and exhibits uniform RFP expression

(Figure 3C).

Upon cotransfection of the Gal4-ePiggyBac helper and recip-

ient plasmid, 6.48% ± 1.04% colonies exhibited mosaic RFP

expression (Figure 3D), whereas mosaic RFP expression was

not observed in nontransfected cultures (not shown). DNA

extracted from these hESC was used to transform E. coli

bacteria. Bacterial transformants were then selected for both

ampicillin and kanamycin resistance conferred by the recipient

plasmid and transposon, respectively. Sequence analyses

revealed that the transposable element originally carried by the

transgenic hESC genome had been transferred to the recipient

plasmid (Figure 3E). In addition, presence of the typical TTAA

tetranucleotide at the junction between transposon and plasmid

sequences indicated that the transposon had been inserted in

the recipient plasmid by ePiggyBac transposition.

Ganciclovir selection was used to recover RFP-negative hESC

colonies. These colonies did not survive under neomycin

selection (not shown), indicating that the cells had lost all three

selectable markers: RFP, neomycin phosphotransferase, and

thymidine kinase. Importantly, untransfected RFP-hESCs or

controls where RFP-hESCs were transfected with either helper

or recipient plasmid alone did not give rise to ganciclovir-resis-

tant colonies (Figure 3F). This result indicates that re-expression

of the transposase alone is inefficient at removing transgenes

and that transgene removal is dependent on the Gal4 and UAS

components. This result also excludes the possibility that

ganciclovir resistance, neomycin sensitivity, and loss of RFP

expression are due to silencing of selection cassettes on the

transposon. Furthermore, SplinkTA PCR genotyping (Figure 3G)

indicated that ganciclovir-resistant/neomycin-sensitive/RFP-

negative colonies did not carry any transgene. Finally, we looked

for unequivocal evidence that transposon removal had occurred

without leaving footprint mutations in the genome by sequencing

transposon integration sites before and after excision. Compar-

isons of transposon integration site sequences before and after

excision revealed that transgene removal had occurred without

creation of footprint mutations and genomic sequences had

reverted to a wild-type state (Figure 3H). As of this writing,

mutation-free excisions have been performed in 32 independent

hESC clones, and although Wang et al. (2008) have reported that

only 95% excisions are mutation free, we have not yet observed

mutations at the site of excision in hESCs. It is possible that

c.

Cell Stem Cell

ePiggyBac

E

H

RFPTK

PGK

pA

ORI

Neo

genomic DNA

ePBac

RFPTK PGK pAORI Neo

genomic DNA

ePBac

UAS

G4

R6Kγ -UASG4-ePiggyBac

F

A

C

RFP

D

RFP

Sytox

Sytox

Merge

Merge

B

G

RU

ES2

RU

ES2-

RFP

Gan

R-1

Gan

R-2

RFPTK PGK pAORI Neo

RUES2 CAGGGAGTTTAACATCTGGAAGAGCCGGGAGTTCTTTGTGCTRUES2-RFP AAATTACATTAACATCTGGAAGAGCCGGGAGTTCTTTGTGCTRevertant CAGGGAGTTTAACATCTGGAAGAGCCGGGAGTTCTTTGTGCT

0

100

200

no.G

anR

colo

nies

Trangene integration Trangene removal

Figure 3. ePiggyBac Transposons Can Be Removed from the

Genome

(A) Schematic representation of the transgene integrated in hESC line RUES2.

The construct includes a pUC DNA replication origin for plasmid rescue,

neomycin/kanamycin phosphotransferase (Neo) and thymidine kinase (TK)

cassettes for negative and positive selections, respectively, and an RFP-

expressing insert using the PGK promoter and SV40 poly(A) (pA) signal.

Ce

ePiggyBac’s higher transposition efficiency in hESCs efficiency

allows for cleaner excision. In addition, karyotypic analysis of

20 metaphases indicated that transposon removal does not

compromise chromosomal integrity (Figure S6).

Overall, these data indicate that ePiggyBac can both deliver

large multicomponent transgenes to the hESC genome and

remove them without leaving any mutation.

ePiggyBac-Mediated Loss and Gain of Function DirectshESC Differentiation toward Specific Cell Types withoutGenome AlterationsWe next investigated whether the ability to deliver large multi-

component transgenes to hESC and remove them could be

used to drive hESC differentiation toward a specific cell type.

We first created a transposable and inducible short hairpin

RNA (shRNA)-expressing system in order to knock down pluripo-

tency gene expression. This transposon (Figure 4A) expresses

shRNAs modeled after human miR-30 (Silva et al., 2005).

Previous studies have shown that miR-30-based shRNAs inhibit

gene expression more efficiently than simple stem-loop shRNAs

and their expression can be driven by Pol II promoters. This

(B) Schematic representation of the integration/removal approach. For

transgene integration, the transposon is cotransfected with the ePiggyBac-

expressing vector. Transposon remobilization is expected to occur once or

several times until the helper plasmid is diluted by cell division and transpo-

sase activity wears off. Once transposase activity has disappeared, the

transgene remains immobile on the host genome. For transposon removal,

we created a fusion between the Gal4 DNA binding domain (G4) and the

ePiggyBac transposase (ePBac). The R6Kg-UAS recipient plasmid includes

an ampicillin resistance cassette (Amp), 14 repeats of the UAS sequence,

and an R6Kg DNA replication origin. Upon expression, the chimeric G4-ePBac

transposase binds to the UAS sequences, which is expected to increase

chances that transposons are transfered to the recipient plasmid. The R6Kg

DNA replication origin is active only in E. coli strains that provide the p protein

encoded by the pir gene. DH5a E. coli strains used in this study for plasmid

rescue are pir�; therefore upon expression of Gal4-ePiggyBac and DNA

extraction from hESCs, the recipient plasmid can give rise to ampicillin-

resistant DH5a colonies only if a transposon containing a pUC replication

origin has been inserted in the recipient plasmid. In addition, the modified

plasmid resulting from integration of the transposon into the recipient plasmid

only contains one functional replication origin in DH5a strains. This reduces

possible replication defects and decreases in plasmid rescue efficiency result-

ing from interference between two replication origins in the same plasmid.

(C) After neomycin selection, an RFP-positive line containing a single copy of

the transgene was established.

(D) Upon transfection of the recipient plasmid (R6Kg-UAS) and Gal4-

ePiggyBac, helper colonies exhibit mosaic RFP expression.

Scale bar represents 100 mm.

(E) Analyses of six plasmid sequences from ampicillin/kanamycin resistant

E. coli clones reveal that the transposon originally carried by the hESC genome

(black sequence) has been inserted in the recipient plasmid (blue sequence) at

a TTAA junction typical of ePiggyBac transposition.

(F) Ganciclovir-resistant colonies are observed only after cotransfection of

both the Gal4-ePiggyBac helper and R6Kg-UAS recipient plasmid.

(G) PCR genotyping reveals that ganciclovir-resistant hESC lines (GanR-1 and

GanR-2) have reverted from a transgenic (RUES2-RFP) to a wild-type (RUES2)

RUES2 genotype. The lower band on the gel indicates nonmutated allele.

(H) DNA sequences of a transposon integration locus in nontransgenic cells

(RUES2), transgenic cells before transposon removal (RUES2-RFP), and

transgenic cells after transposon removal (Revertant). Sequence analyses

show that transgene (blue) removal did not leave any mutation in the RUES2

genome (green). The chromatogram confirms the absence of mutations in the

revertant sequence.

ll Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier Inc. 337

Cell Stem Cell

ePiggyBac

shRNATet TRE pANeoA

B

Ins

c

- DOX + DOX

Oct4 CK18 Sytox

Oct4 CK18 Sytox

Oct4 CK18 Sytox

+ D

OX

- DO

X+

DO

X sh

EGFP

C

E

D

+ D

OX

- DO

X

shEG

FP+

DO

X

Oct4

Nanog

Sox2

GAPDH

Gata 6

Sox1

Brachyury

Cdx2

RT + RT -+

DO

X

- DO

X

shEG

FP+

DO

X

Figure 4. ePiggyBac Transposons Can Deliver Doxycyclin-Inducible

shRNA-Expressing Cassettes to Induce hESC Differentiation

(A) Schematic representation of the shRNA-expressing transposable element.

The transgene includes a TetON cassette, a neomycin selection cassette

(Neo), an HS4 chicken insulator, and Tet-responsive elements (TRE) driving

shRNA expression (pA, poly(A) signal).

(B) Quantitative RT-PCR analyses show that 4 days after the addition of

doxycyclin (+DOX), the expression of pluripotency markers Oct4, Sox2, and

Nanog decreases whereas levels of differentiation markers Gata6, Sox1,

Brachyury, and Cdx2 increase compared to samples grown in the absence

of doxycyclin (�DOX) or in cells expressing a control anti-EGFP shRNA

(shEGFP +DOX).

(C and D) Cells grown in the absence of doxycyclin, which formed tightly

packed colonies characteristic of undifferentiated hESCs (C), whereas cells

expressing the anti-Oct4 shRNA exhibited a flattened morphology reminiscent

of differentiated cells (D).

338 Cell Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier In

enabled the use of the TetON system and control gene knock-

down by addition of doxycyclin to the cell culture medium.

RUES2 cells were transfected with an shRNA vector targeting

Oct4 and grown in the presence of neomycin until 500- to

1000-cell colonies were obtained. Doxycyclin was then added

to the conditioned culture medium to induce anti-Oct4 shRNA

expression. RT-PCR analyses showed that 4 days after the addi-

tion of doxycyclin, the expression of pluripotency markers Oct4,

Sox2, and Nanog decreased whereas differentiation markers

Gata6, Sox1, Brachyury, and Cdx2 increased compared to

samples grown in the absence of doxycyclin or in cells express-

ing a control anti-EGFP shRNA (Figure 4B; Figure S7A). Concom-

itantly, clear morphological differences appeared between cells

grown in the absence of doxycyclin, which formed tightly packed

colonies (Figure 4C) characteristic of undifferentiated hESCs,

and cells expressing the anti-Oct4 shRNA, which exhibited a flat-

tened morphology reminiscent of differentiated cells (Figure 4D).

These morphological changes were accompanied by a strong

increase in immunoreactivity for the trophectoderm marker cyto-

keratin 18 in filamentous structures characteristic of epithelial

cell types (Figure 4E; Figure S7). These results indicate that

the ePiggyBac system can be used to deliver loss-of-function

vectors that trigger hESC differentiation in a doxycyclin-

inducible manner despite the presence of nondifferentiating

culture conditions.

We next investigated whether a combination of loss- and gain-

of-function systems could be used to drive differentiation toward

one particular cell type, namely a neural cell type. We added

a thymidine kinase cassette to our TetON system (Figure 4A)

and first transfected RUES2 cells with a gain-of-function

construct expressing human Sox1 cDNA to direct differentiation

toward neuroectoderm (not shown). We noticed that in condi-

tioned medium, overexpression of Sox1 alone does not direct

differentiation toward neuroectoderm (not shown), possibly

because it cannot counteract FGF and activin/nodal signaling

activated by conditioned medium to maintain hESC self-renewal.

Therefore, we cotransfected the Sox1 gain-of-function construct

together with loss-of-function constructs expressing shRNAs

against Oct4, Gata6, Sox 17, Brachyury, or Cdx2 (1 mg of each

construct per nucleofection reaction) to induce differentiation

but minimize the acquisition of endodermal, mesodermal, or

trophectodermal phenotypes. In preliminary experiments, we

tested that each individual shRNA was able to knock down its

target gene (Figure S8). Upon delivery of the transgene cocktail,

500- to 1000-cell transgenic colonies were obtained under

neomycin selection before doxycyclin was added to the condi-

tioned culture medium. After 10 days of doxycyclin treatment,

expression levels of pluripotency markers decreased signifi-

cantly and expression of endogenous neural-specific genes

Sox1 and NFH became preponderant over that of endodermal,

mesodermal, and trophectodermal markers (Figure 5A). Further-

more, hESCs differentiated into flattened cell types, and groups

of 10–50 round structures (Figure 5B) reminiscent of structures

defined by others as neural rosettes (Zhang et al., 2001) had

(E) These morphological changes were accompanied by a loss in Oct4 immu-

noreactivity and a strong increase in immunoreactivity for the trophectoderm

marker cytokeratin 18.

Scale bars represent 30 mm (C and D), 50 mm (E).

c.

Cell Stem Cell

ePiggyBac

0

5

10

Oct

4

Nan

og

Sox

2

Mix

l1

Sox

17

hCG

b

Sox

1

Pax

6

NFH

mR

NA

leve

ls

- Dox+ Dox

Cho

rdin

A

H

Endo Sox1shCdx2

shBrashSox17shGata6shOct4

Sox1

Neg

.

1 2 3 4 5 6 7 8 9 10

Nestin

Map2

B C

D E

F G

Nestin / Pax6

DCX / NFH

Figure 5. ePiggyBac Transposons Can Direct hESC Differentiation

toward a Neural Phenotype and Be Removed

(A) Quantitative RT-PCR analyses indicate that in hESCs transfected with

a cocktail of gain- and loss-of-function transposons, expression of pluripo-

tency markers significantly decrease upon doxycyclin treatment and the

expression of neuroectodermal marker Pax6 and neural-specific genes

Sox1 and NFH is preponderant over that of endodermal (Mixl1, Sox17), meso-

dermal (Chordin), and trophectodermal (hCG beta) markers.

Cel

appeared in 91% (n = 309) of flattened cell colonies. Immunoflu-

orescent staining for the neuroectodermal marker Pax6 and the

neural precursor marker nestin (Figure 5C) confirmed that the

round structures were neural rosettes (Zhang et al., 2001). These

results indicate that ePiggyBac can deliver cocktails of doxycy-

clin-inducible loss- and gain-of-function transgenes that drive

differentiation toward a neuronal cell type without the need for

a defined culture medium (other than the conditioned medium

commonly used to maintain hESCs), feeder cells, or embryoid

body formation (Zhang et al., 2001).

We finally asked whether transgenes could be removed

without loss of the neural phenotype. We isolated neural rosettes

by enzymatic treatment and let them form neural spheres (Zhang

et al., 2001) in conditioned medium supplemented with doxycy-

clin. Seven-day-old neural spheres were then triturated to obtain

5- to 10-cell clumps, nucleofected with the Gal4-ePiggyBac

helper and UAS-carrying recipient plasmid, and cultured in

suspension in N2 medium. 48 hr after transfection, ganciclovir

selection was applied and ganciclovir-resistant neural spheres

were obtained after an additional 7 days of culture (Figure 5D).

Neural spheres were then cut in halves. One half of the sphere

was plated on matrigel-coated plates and cultured for 4 days.

Upon attachment to the matrigel substrate, they formed neurites

expressing the neural precursor marker nestin and the neuronal

markers doublecortin, neurofilament heavy chain, and Map2

(Figures 5E–5G). PCR genotyping was performed on the other

half of the neural sphere to confirm that the transgenes had

been removed (Figure 5H). Interestingly, neural spheres origi-

nating from a clone containing all six transposons were devoid

of transgenes after ganciclovir selection. These results indicate

that ePiggyBac can deliver cocktails of transgenes that direct

differentiation toward a specific phenotype, which is conserved

upon transgene removal.

DISCUSSION

Most, if not all, gene delivery systems fall into two categories:

systems that mediate irreversible gene integrations (e.g., viral

vectors, PhiC31-based systems, plasmids, and BACs) and

systems that can be excised but leave mutations in the host

genome upon excision (e.g., Cre-, Flp-, or Sleeping beauty-

based systems). These irreversible genetic alterations could

potentially lead to higher predisposition toward uncontrolled

cellular behavior and they constitute a significant barrier to clin-

ical applications of hESCs. ePiggyBac minimizes these concerns

by enabling fully reversible gene delivery.

(B and C) Neural rosettes exhibiting Pax6 and Nestin immunoreactivity also

appear in doxycyclin-treated cultures.

(D) Upon transgene removal, ganciclovir-resistant neural spheres are selected.

(E–G) These neural spheres give rise to nestin- (E), doublecortin (DCX)-, NFH-

(F), and Map2-positive (G) neurons.

(H) PCR genotyping on ten different samples confirms that ganciclovir-resis-

tant neural spheres are devoid of transposons. Transposon-specific primers

that detect the inverted repeats of ePiggyBac were used to demonstrate

that regardless of number of insertions. all transgenes were removed. Primers

detecting endogenous Sox1 genomic sequences were used as positive

controls (Endo Sox1). Neg., indicates the genotype of the parent neural sphere

where transgenes have not been removed.

Scale bars represent 50 mm (B), 5 mm (C), 100 mm (D), 20 mm (E and G), 10 mm (F).

l Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier Inc. 339

Cell Stem Cell

ePiggyBac

The present data show that in ES cells, ePiggyBac’s efficiency

is not only superior to that of viral vectors, it is also superior to

that of previously described transposases, including other Pig-

gyBac-based systems (Cadinanos and Bradley, 2007; Yusa

et al., 2009; Woltjen et al., 2009; Kaji et al., 2009). In addition,

the efficiency of ePiggyBac is conserved between human,

nonhuman primate, and rodent ES cells. Our data show that

ePiggyBac can deliver inserts of up to 18 Kb, which enables the

use of large promoters for cell-specific transgene expression.

Transgenes need not be active in undifferentiated hESCs to

permit the selection of transgenic cells because in addition to

a reportersystem,ePiggyBactransposonscan includeaselection

cassette regulated by a separate promoter. Therefore, in vivo

labeling of specific cell types arising from transgenic hESC differ-

entiation is now straightforward. This should greatly facilitate the

isolation and purification of cell populations of medical interest.

The ability to use multiple expression cassettes also permits

the design of conditional loss- and gain-of-function strategies

to direct hESC differentiation toward cell types that can be

used for drug screens, toxicological assays, or basic research

on human diseases. In addition, the possibility to remove trans-

genes from the hESC genome without leaving any mutation at

the site of insertion eliminates risks associated with irreversible

gene delivery approaches. Differentiation via gain- and loss-of-

function cassettes also reduces the need for defined media

formulation. Ultimately, this should facilitate the generation of

clinical-grade hESC derivatives by reducing the need for

animal-derived media components and feeders.

EXPERIMENTAL PROCEDURES

DNA Constructs

The original PiggyBac transposase DNA was obtained from Dr. M. Fraser

(University of Notre Dame, IN). The ePiggyBac transposase cDNA was custom

synthesized by Integrated DNA Technologies. To generate helper plasmids,

transposase cDNAs were cloned in pBluescript (Stratagene) downstream of

a human phosphoglycerate kinase (PGK) or CAG promoter and upstream of

an SV40 polyadenylation signal sequence. Minimal 313 bp 50TR and 235 bp

30TR were custom synthesized and cloned upstream and downstream,

respectively, of expression cassettes containing the PGK promoter driving

EGFP (BD Biosciences) or tagRFP (Evrogen). The plasmid rescue system

also included an ampicillin resistance cassette and an origin of replication

from pBluescript cloned between the 50- and 30-terminal repeats. To improve

gene transfer efficiency, mutations in terminal repeats were introduced with

the GeneMorph II random PCR mutagenesis system (Stratagene). To generate

the Oct4-EGFP transposon, the human Oct4 promoter (Gerrard et al., 2005)

was cloned upstream of EGFP. The 250 bp chicken b-globin HS4 core insu-

lator (Recillas-Targa et al., 2002) was custom synthesized and inserted

between the hOct4-EGFP reporter and a neomycin/kanamycin cassette

subcloned from pEGFP-N1 (BD Biosciences). Finally, the hOct4-EGFP

reporter, HS4 insulator, and selection cassette were flanked by 50 and 30TR.

The helper plasmid for transgene removal was generated by cloning the

Gal4 DNA binding domain between the PGK promoter and the transposase

cDNA of the ePiggyBac helper plasmid to create a Gal4-ePiggyBac fusion.

Two consensus nuclear localization signals (MPKKKRKVDPKKKRKVD) were

incorporated at the N terminus of the Gal4 DNA binding domain, and a flexible

linker (GGSGGSGSS) was incorporated at its C terminus. The recipient

plasmid was generated by adding 14 UAS sequences to the pLD53 backbone

(Metcalf et al., 1996). This plasmid contains an R6Kg origin of replication,

which is inactive unless the p protein is provided by the bacterial host. It

was therefore maintained in pir-2 E. coli (Invitrogen). For nucleofection, all

plasmids were prepared with a Qiaprep Spin Miniprep kit (QIAGEN) and

purified by phenol:chloroform extraction and ethanol precipitation. For RNA

340 Cell Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier In

interference, shRNA were designed as described (Silva et al., 2005). shRNAs

used in this study are summarized in Table S1.

hESC Lines and Culture Conditions

hESC lines: RUES1 (James et al., 2006), RUES2, H1 (Thomson et al., 1998),

HUES10 (Cowan et al., 2004), and BGN1 (Mitalipova et al., 2003), as well as

Macacca fascicularis, M. nemestrina (both from the Reproductive Biology

Core, Washington National Primate Research Center), and M. mulatta line

R366.4 (Thomson et al., 1995). ESCs were grown on mitotically inactivated

MEF feeders (strain CF-1, Chemicon) in HUESM (DMEM supplemented with

20% KSR, 100 mM nonessential amino acids, 2 mM GlutaMAX, 100 mM b-mer-

captoethanol, 100 U/ml penicillin, 100 mg/ml streptomycin, and 13 B27

supplement without Vitamin A; all from Invitrogen) supplemented with 20 ng/

ml bFGF. All hESC lines were transferred to feeder-free conditions on Matri-

gel-coated dishes and cultured in conditioned medium (CM) for at least one

passage prior to gene delivery. For the preparation of CM, mitotically inacti-

vated MEFs were seeded at a density of 4 3 106 cell per 100 mm dish and

incubated in HUESM supplemented with 20 ng/ml bFGF for 24 hr. CM was

then collected, supplemented with 20 ng/ml bFGF, and used to feed human

and macacca ESCs. For expansion, hESC were enzymatically passaged

with 1 mg/ml dispase treatment (James et al., 2006). Embryoid bodies were

generated by incubation of hESC cultures in dispase until colonies detached

from the substrate. Individual aggregates were then transferred to low attach-

ment 96-well plates (Evergreen) and grown in HUESM for at least 2 weeks prior

to GFP or immunofluorescence imaging. To generate teratomas, 1–2 3 106

hESCs were injected into the rear leg muscle or subcutaneously into SCID/

Beige mice. Teratomas were allowed to develop for 6–8 weeks, excised, fixed

in neutral buffered formalin, and analyzed histologically by trained patholo-

gists. hESC lines were karyotyped by standard G-banding.

Gene Transfer

Prior to dissociation and nucleofection, hESC and Macacca ESC cultures or

dissociated neural spheres were treated for 1 hr with 10 mM ROCK inhibitor

Y-27632 which strongly diminishes dissociation-induced apoptosis and

increases cloning efficiency (Watanabe et al., 2007). ESCs were then dissoci-

ated in 0.25% (wt/vol) trypsin-EDTA at 37�C for 5 min, washed in CM with

ROCK inhibitor, and resuspended in nucleofection solution V or L (Amaxa).

Helper and transposon plasmids were added to the cell suspension, and

nucleofection was performed with program setting B-016 as described

(Siemen et al., 2005). ESCs were then plated in CM supplemented with

10 mM ROCK inhibitor at densities of 2 3 105 cells per 6 cm dish for the selec-

tion of clonal cell line hOct4-EGFP or 2 3 103 cells per 6 cm dish for fluorescent

colony counts. Culture medium was changed to CM without ROCK inhibitor

20–24 hr after nucleofection and colonies were allowed to form for 7 days

before numbers of fluorescent colonies were evaluated. EGFP-positive colony

count based on microscopic observation was preferred over single-cell counts

by flow cytometry. This constitutes a more stringent and more functionally

relevant way to assess the efficiency of a gene delivery system because trans-

gene silencing is frequent in hESCs and only a fraction of single transgenic

cells gives rise to an EGFP-labeled marked cell line that can be used for further

applications (Braam et al., 2008). In experiments where selection was needed,

exposure to neomycin (100 mg/ml) or ganciclovir (1 mM) was initiated 3 days

after nucleofection and maintained until large resistant colonies became

visible. Media were refreshed daily.

Plasmid Rescue and hESC Genotyping

Genomic DNA isolated from transgenic hESC clones was digested with

restriction enzymes BamHI, BglII, and NotI. DNA was self-ligated at low

concentration with T4 DNA ligase overnight at 16�C, precipitated with 100%

isopropanol, and washed with 70% ethanol before transformation in DH10B

Escherichia coli and selection on ampicillin. For transgene removal experi-

ments, undigested DNA was extracted from hESC cultures and used for

E. coli transformations. Bacterial transformants were then selected on both

ampicillin and kanamycin. Plasmid DNA from resistant E. coli clones was

sequenced with primers that read through the 50TR of the PiggyBac trans-

poson. Number of insertions were determined as described (Yin and Largaes-

pada, 2007) by SplinkTA PCR, which usually provides a higher resolution than

Southern blots. hESCs were genotyped before and after transposon removal

c.

Cell Stem Cell

ePiggyBac

with the following primers: 30TR-F 50-CTTAAGGAATTCGATAAAAG-30 and

30TR-R 50-GATACATTGATGAGTTTGG-30, which read through the 30TR of

the PiggyBac transposon, and 18-F 50-CGTGCTTACAGGCATTGAGC-30 and

18-R 50-CTGGAACCTCGACTTCTTGG-30, which flank the integration site of

the Neo-TK-RFP transposon (Figure 3A) on chromosome 18. Neural spheres

were genotyped with transposon-specific primers (forward 50-CTCTAGAT

CATAATCAGCC-30, reverse 50-ACGCCTTAAGATACATTGATGAG-30 ), and

primers detecting endogenous Sox1 genomic sequences (forward 50-GAG

ATTCATCTCAGGATTGAGATTC-30, reverse 50-CGAGCGCAATGGCAGATTT

AC-30) were used as positive controls.

mRNA Quantifications

RNA was isolated with RNA-Bee (Tel-Test Inc. Friendswood, TX) and first-

strand cDNA was generated with a SuperScript III cDNA First Strand Synthesis

kit (Invitrogen). Quantitative real-time PCR reactions were performed with

a LightCycler 480 SYBR Green I Master Kit (Roche). Primers used to measure

mRNA levels are summarized in Table S2.

Immunohistochemical Assays and Confocal Imaging

Undifferentiated hESCs or adherent embryoid bodies plated on Matrigel-

coated MatTek coverslip dishes were fixed in 4% paraformaldehyde (PFA),

washed in phosphate-buffered saline solution, and blocked in 0.1% Triton

X-100 and 3% donkey serum in PBS. Samples were exposed to primary anti-

bodies in blocking solution overnight at 4�C, washed 3 times in PBS with 0.1%

tween-20 at room temperature, and exposed to Alexa-conjugated secondary

antibodies (dilution 1:500). Primary antibodies included Oct3/4 (Signal Trans-

duction Labs), tagRFP (Evrogen), GATA6, Nestin, Cytokeratin 18, and double-

cortin (Santa Cruz), Muscle MHC/MF20 (Developmental Studies Hybridoma

Bank), Neurofilament Heavy Chain (SMI32, Sternberger), Map2 (Millipore)

Nanog, and Sox2 (R&D Systems). Alexa-conjugated secondary antibodies

and SytoxOrange nuclear counterstain were purchased from Molecular

Probes. All imaging was performed on a Zeiss LSM 500 Pascal confocal

microscope.

Flow Cytometry

Prior to dissociation and analysis, hESC were treated for 1 hr with 10 mM ROCK

inhibitor Y-27632 to minimize cell death. Cells were then dissociated in 0.25%

(wt/vol) trypsin-EDTA at 37�C for 5 min, washed in CM with ROCK inhibitor,

and resuspended in ice-cold PBS supplemented with 0.1% BSA and

0.5 mM EDTA. Live cell analyses were then performed immediately with a

BD LSRII-2 and DiVa 5.0.2 software (Becton Dickinson).

Statistical Analysis

The statistical significance (p values) in means of two-sample comparison was

determined with Student’s t test (Microsoft Excel). The statistical significance

in mean values among multiple sample groups was examined with two-way

ANOVA and Bonferroni’s post-hoc test. Values shown in graphs represent

the mean ± SD.

SUPPLEMENTAL DATA

Supplemental Data include ten figures and two tables and can be found

with this article online at http://www.cell.com/cell-stem-cell/supplemental/

S1934-5909(09)00347-6.

ACKNOWLEDGMENTS

We thank Dr. Scott A. Noggle for advice during the design of the experiments

and Shalini Jaiswal for outstanding technical assistance. This work was sup-

ported by private funding from The Rockefeller University, The Starr founda-

tion, NIH grant 5RO1 GM073379 (for work on hESC lines H1 and BGN1) to

A.H.B., and by a grant from the Emerald Foundation to A.L.

Received: January 12, 2009

Revised: May 4, 2009

Accepted: July 21, 2009

Published: September 3, 2009

Ce

REFERENCES

Ben-Dor, I., Itsykson, P., Goldenberg, D., Galun, E., and Reubinoff, B.E. (2006).

Lentiviral vectors harboring a dual-gene system allow high and homogeneous

transgene expression in selected polyclonal human embryonic stem cells.

Mol. Ther. 14, 255–267.

Braam, S.R., Denning, C., van den Brink, S., Kats, P., Hochstenbach, R., Pass-

ier, R., and Mummery, C.L. (2008). Improved genetic manipulation of human

embryonic stem cells. Nat. Methods 5, 389–392.

Brunak, S., Engelbrecht, J., and Knudsen, S. (1991). Prediction of human

mRNA donor and acceptor sites from the DNA sequence. J. Mol. Biol. 220,

49–65.

Cadinanos, J., and Bradley, A. (2007). Generation of an inducible and

optimized piggyBac transposon system. Nucleic Acids Res. 35, e87.

Cary, L.C., Goebel, M., Corsaro, B.G., Wang, H.G., Rosen, E., and Fraser, M.J.

(1989). Transposon mutagenesis of baculoviruses: Analysis of Trichoplusia ni

transposon IFP2 insertions within the FP-locus of nuclear polyhedrosis

viruses. Virology 172, 156–169.

Cowan, C.A., Klimanskaya, I., McMahon, J., Atienza, J., Witmyer, J., Zucker,

J.P., Wang, S., Morton, C.C., McMahon, A.P., Powers, D., et al. (2004). Deriva-

tion of embryonic stem-cell lines from human blastocysts. N. Engl. J. Med. 350,

1353–1356.

Ding, S., Wu, X., Li, G., Han, M., Zhuang, Y., and Xu, T. (2005). Efficient

transposition of the piggyBac (PB) transposon in mammalian cells and mice.

Cell 122, 473–483.

Elick, T.A., Bauser, C.A., and Fraser, M.J. (1996). Excision of the piggyBac

transposable element in vitro is a precise event that is enhanced by the expres-

sion of its encoded transposase. Genetica 98, 33–41.

Fraser, M.J., Cary, L., Boonvisudhi, K., and Wang, H.G. (1995). Assay for

movement of Lepidopteran transposon IFP2 in insect cells using a baculovirus

genome as a target DNA. Virology 211, 397–407.

Fraser, M.J., Ciszczon, T., Elick, T., and Bauser, C. (1996). Precise excision of

TTAA-specific lepidopteran transposons piggyBac (IFP2) and tagalong (TFP3)

from the baculovirus genome in cell lines from two species of Lepidoptera.

Insect Mol. Biol. 5, 141–151.

Gerrard, L., Zhao, D., Clark, A.J., and Cui, W. (2005). Stably transfected human

embryonic stem cell clones express OCT4-specific green fluorescent protein

and maintain self-renewal and pluripotency. Stem Cells 23, 124–133.

James, D., Noggle, S.A., Swigut, T., and Brivanlou, A.H. (2006). Contribution of

human embryonic stem cells to mouse blastocysts. Dev. Biol. 295, 90–102.

Kaji, K., Norrby, K., Paca, A., Mileikovsky, M., Mohseni, P., and Woltjen, K.

(2009). Virus-free induction of pluripotency and subsequent excision of

reprogramming factors. Nature 458, 771–775.

Li, X., Harrell, R.A., Handler, A.M., Beam, T., Hennessy, K., and Fraser, M.J., Jr.

(2005). piggyBac internal sequences are necessary for efficient transformation

of target genomes. Insect Mol. Biol. 14, 17–30.

Maragathavally, K.J., Kaminski, J.M., and Coates, C.J. (2006). Chimeric Mos1

and piggyBac transposases result in site-directed integration. FASEB J. 20,

1880–1882.

McCarty, D.M., Young, S.M., Jr., and Samulski, R.J. (2004). Integration of ad-

eno-associated virus (AAV) and recombinant AAV vectors. Annu. Rev. Genet.

38, 819–845.

Metcalf, W.W., Jiang, W., Daniels, L.L., Kim, S.K., Haldimann, A., and Wanner,

B.L. (1996). Conditionally replicative and conjugative plasmids carrying lacZ

alpha for cloning, mutagenesis, and allele replacement in bacteria. Plasmid

35, 1–13.

Mitalipova, M., Calhoun, J., Shin, S., Wininger, D., Schulz, T., Noggle, S.,

Venable, A., Lyons, I., Robins, A., and Stice, S. (2003). Human embryonic

stem cell lines derived from discarded embryos. Stem Cells 21, 521–526.

Pfeifer, A., Ikawa, M., Dayn, Y., and Verma, I.M. (2002). Transgenesis by lenti-

viral vectors: Lack of gene silencing in mammalian embryonic stem cells and

preimplantation embryos. Proc. Natl. Acad. Sci. USA 99, 2140–2145.

Recillas-Targa, F., Pikaart, M.J., Burgess-Beusse, B., Bell, A.C., Litt, M.D.,

West, A.G., Gaszner, M., and Felsenfeld, G. (2002). Position-effect protection

ll Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier Inc. 341

Cell Stem Cell

ePiggyBac

and enhancer blocking by the chicken beta-globin insulator are separable

activities. Proc. Natl. Acad. Sci. USA 99, 6883–6888.

Siemen, H., Nix, M., Endl, E., Koch, P., Itskovitz-Eldor, J., and Brustle, O.

(2005). Nucleofection of human embryonic stem cells. Stem Cells Dev. 14,

378–383.

Silva, J.M., Li, M.Z., Chang, K., Ge, W., Golding, M.C., Rickles, R.J., Siolas, D.,

Hu, G., Paddison, P.J., Schlabach, M.R., et al. (2005). Second-generation

shRNA libraries covering the mouse and human genomes. Nat. Genet. 37,

1281–1288.

Thomson, J.A., Kalishman, J., Golos, T.G., Durning, M., Harris, C.P., Becker,

R.A., and Hearn, J.P. (1995). Isolation of a primate embryonic stem cell line.

Proc. Natl. Acad. Sci. USA 92, 7844–7848.

Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S.S., Waknitz, M.A., Swiergiel, J.J.,

Marshall, V.S., and Jones, J.M. (1998). Embryonic stem cell lines derived from

human blastocysts. Science 282, 1145–1147.

Thyagarajan, B., Liu, Y., Shin, S., Lakshmipathy, U., Scheyhing, K., Xue, H.,

Ellerstrom, C., Strehl, R., Hyllner, J., Rao, M.S., et al. (2008). Creation of engi-

neered human embryonic stem cell lines using phiC31 integrase. Stem Cells

26, 119–126.

Watanabe, K., Ueno, M., Kamiya, D., Nishiyama, A., Matsumura, M., Wataya,

T., Takahashi, J.B., Nishikawa, S., Muguruma, K., and Sasai, Y. (2007). A

ROCK inhibitor permits survival of dissociated human embryonic stem cells.

Nat. Biotechnol. 25, 681–686.

342 Cell Stem Cell 5, 332–342, September 4, 2009 ª2009 Elsevier In

Wang, W., Lin, C., Lu, D., Ning, Z., Cox, T., Melvin, D., Wang, X., Bradley, A.,

and Liu, P. (2008). Chromosomal transposition of PiggyBac in mouse embry-

onic stem cells. Proc. Natl. Acad. Sci. USA 105, 9290–9295.

Wilber, A., Linehan, J.L., Tian, X., Woll, P.S., Morris, J.K., Belur, L.R., McIvor,

R.S., and Kaufman, D.S. (2007). Efficient and stable transgene expression in

human embryonic stem cells using transposon-mediated gene transfer.

Stem Cells 25, 2919–2927.

Wilson, M.H., Coates, C.J., and George, A.L., Jr. (2007). PiggyBac trans-

poson-mediated gene transfer in human cells. Mol. Ther. 15, 139–145.

Woltjen, K., Michael, I.P., Mohseni, P., Desai, R., Mileikovsky, M., Hamalainen,

R., Cowling, R., Wang, W., Liu, P., Gertsenstein, M., et al. (2009). piggyBac

transposition reprograms fibroblasts to induced pluripotent stem cells.

Nature 458, 766–770.

Yusa, K., Rad, R., Takeda, J., and Bradley, A. (2009). Generation of transgene-

free induced pluripotent mouse stem cells by the piggyBac transposon. Nat.

Methods 6, 363–369.

Yin, B., and Largaespada, D.A. (2007). PCR-based procedures to isolate

insertion sites of DNA elements. Biotechniques 43, 79–84.

Zeng, J., Du, J., Zhao, Y., Palanisamy, N., and Wang, S. (2007). Baculoviral

vector-mediated transient and stable transgene expression in human

embryonic stem cells. Stem Cells 25, 1055–1061.

Zhang, S.C., Wernig, M., Duncan, I.D., Brustle, O., and Thomson, J.A. (2001).

In vitro differentiation of transplantable neural precursors from human

embryonic stem cells. Nat. Biotechnol. 19, 1129–1133.

c.