Você está na página 1de 13

SCIENCE ADVANCES | RESEARCH ARTICLE

CELL BIOLOGY Copyright 2017


The Authors, some

Directed differentiation of human pluripotent stem cells rights reserved;


exclusive licensee
to blood-brain barrier endothelial cells American Association
for the Advancement
of Science. No claim to
Tongcheng Qian, Shaenah E. Maguire, Scott G. Canfield, Xiaoping Bao, William R. Olson, original U.S. Government
Eric V. Shusta,* Sean P. Palecek* Works. Distributed
under a Creative
The blood-brain barrier (BBB) is composed of specialized endothelial cells that are critical to neurological health. A Commons Attribution
key tool for understanding human BBB development and its role in neurological disease is a reliable and scalable NonCommercial
source of functional brain microvascular endothelial cells (BMECs). Human pluripotent stem cells (hPSCs) can the- License 4.0 (CC BY-NC).
oretically generate unlimited quantities of any cell lineage in vitro, including BMECs, for disease modeling, drug
screening, and cell-based therapies. We demonstrate a facile, chemically defined method to differentiate hPSCs
to BMECs in a developmentally relevant progression via small-molecule activation of key signaling pathways.
hPSCs are first induced to mesoderm commitment by activating canonical Wnt signaling. Next, these mesoderm
precursors progress to endothelial progenitors, and treatment with retinoic acid leads to acquisition of BBB-specific
markers and phenotypes. hPSC-derived BMECs generated via this protocol exhibit endothelial properties, includ-

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


ing tube formation and low-density lipoprotein uptake, as well as efflux transporter activities characteristic of
BMECs. Notably, these cells exhibit high transendothelial electrical resistance above 3000 ohmcm2. These hPSC-
derived BMECs serve as a robust human in vitro BBB model that can be used to study brain disease and inform
therapeutic development.

INTRODUCTION Human pluripotent stem cells (hPSCs) have the potential to generate
An intact blood-brain barrier (BBB) serves as a key interface between large quantities of specialized human cells for studying development
the blood circulation and the central nervous system (CNS). The pri- and modeling disease (2931). Previously, we reported the generation
mary anatomical component of the BBB is provided by brain micro- of pure populations of hPSC-derived BMECs via co-differentiation of
vascular endothelial cells (BMECs) (1, 2) that work in concert with hPSCs to neural and endothelial progenitors, followed by selective pu-
supporting cells, such as astrocytes, pericytes, and neurons, to form rification of the BMECs (32). In addition, we demonstrated that retinoic
the neurovascular unit (1, 3, 4). BMECs are connected by tight junc- acid (RA) addition during BMEC differentiation enhanced barrier
tions and display low levels of vesicular traffic, leading to extremely properties to physiologic levels (33). Presumably, the neural progenitors
low vascular permeability. BMECs also express molecular influx and in this co-differentiation platform induce the endothelial progenitors to
efflux transporters, which regulate the delivery of nutrients from the acquire BMEC-specific traits, which are then enhanced by RA treat-
blood to the brain and removal of compounds from the brain, respec- ment. However, the undefined nature of this co-differentiation platform
tively. A functional BBB prevents most of the small-molecule drugs complicates the investigation of mechanisms that specify BMEC fates in
and nearly all large-molecule biologics from entering the brain (5). the hPSC-derived ECs. In addition, this undefined protocol can result in
Thus, the BBB is a highly efficient barrier that protects the brain line-to-line and batch-to-batch variability in BMEC yield and pheno-
and limits CNS drug delivery (6). Moreover, BBB dysfunction has types (32, 3436), despite recent efforts that have explored the use of
been associated with many CNS disorders, including stroke (79), defined medium to accelerate portions of the differentiation process
Alzheimers disease (10, 11), multiple sclerosis (12), Parkinsons dis- (36). Other studies have also shown that human BMEC-like cells can
ease (13), traumatic brain injury (14, 15), and HIV (1618). be generated from alternative stem and progenitor cell sources, includ-
Although the BBB has been extensively studied in animal models ing hematopoietic stem cells (37), endothelial progenitors (38), and
(1921), in vitro models based on primary human BMECs (22, 23), hPSC-derived ECs cocultured with hPSC-derived pericytes, astrocytes,
and immortalized human brain EC lines (2, 24, 25), these models lack and neurons (39) or C6 glioma cells (40). Nevertheless, none of these
key attributes of the human BBB. Animal models cannot fully repre- previous studies report a chemically defined, robust process for gener-
sent the human BBB because of species differences, particularly in ating human BMECs exhibiting physiologic BBB phenotypes.
transporter expression and function (26). Human primary BMECs During embryonic development, mesoderm-derived ECs form a
are difficult to obtain in sufficient quantities for drug screening and vascular plexus covering the developing neural tube (41, 42). As
disease models and cannot be readily expanded in high-fidelity cultures. nascent blood vessels enter the developing CNS, canonical Wnt
Immortalized cell lines exhibit a loss of BMEC-specific properties, in- signaling is necessary to induce BMEC barrier properties (4345).
cluding loss of tight junctions yielding subphysiologic transendothelial RA has also been shown to regulate BMEC specification. During
electrical resistance (TEER) (27). These limitations have prevented BBB development, radial glial cells supply the CNS with RA (46),
our full understanding of human BBB development, function, and dis- and this RA signaling induces barrier formation and BBB-specific
ease (28). gene expression (33, 46, 47). In addition to Wnt regulation of BBB in-
duction in vivo, previous studies have demonstrated that activation of
canonical Wnt signaling can also direct hPSCs to mesodermal lineages
Department of Chemical and Biological Engineering, University of Wisconsin- in vitro (31, 4850). Thus, we hypothesized that appropriate differenti-
Madison, Madison, WI 53706, USA.
*Corresponding author. Email: sppalecek@wisc.edu (S.P.P.); eshusta@wisc.edu ation stagespecific modulation of canonical Wnt signaling would in-
(E.V.S.) duce mesodermal and endothelial commitment in hPSCs, and combine

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 1 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

with subsequent RA signaling to drive the acquisition of BMEC markers RESULTS


and phenotypes. hPSCs progress from intermediate mesoderm to BMECs
Here, we report a chemically defined method to differentiate hPSCs Given the roles of canonical Wnt signaling in both mesoderm specifi-
to BMECs via sequential Wnt and RA pathway activation. During this cation and BBB development, we first treated hPSCs with CHIR99021,
differentiation process, hPSCs progress through stages resembling a glycogen synthase kinase 3b (GSK-3b) inhibitor and canonical Wnt
primitive streak and intermediate mesoderm to vascular endothelial pathway agonist, to direct hPSCs to mesoderm-derived endothelial
growth factor receptor 2positive (VEGFR2+) endothelial progenitors progenitors. Before treatment, IMR90-4 induced pluripotent stem cells
that become virtually pure populations of CD31+ ECs displaying key (iPSCs) were seeded on a Matrigel-coated six-well plate at a density of
BMEC phenotypes including tight junctions, low passive permeability, 35 103 cells/cm2 and expanded in an undifferentiated state for 3 days
and polarized efflux transporters. The resultant, developmentally rele- in mTeSR1 (Fig. 1A). Previously, we showed that 6 mM CHIR99021
vant BMEC differentiation strategy is defined, robust, and facile. treatment induced hPSC differentiation to a primitive streaklike stage

A
35 103 cells/cm2 6 M
CHIR Replate A
B
D0 D1 D6 D8 D10

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


3D mTeSR1 1D DeSR1 5D DeSR2 2D hECSR1 hECSR2

Primitive Intermediate EC
hPSCs mesoderm progenitors BMECs
streak
D0 D1 D4 D5 D10
OCT4 Brachyury PAX2 VEGFR2 CD31
NANOG Claudin-5
Pgp
OCT4/DAPI Brachyury/DAPI
104
B E F 99.6 0.3%
103
Brachyury

102

101 IgG
100
0 200 400 600 800 1.0k

NANOG/DAPI PAX2/DAPI FSC


104
C G H 94.2 1.1%
103
PAX2

102

101 IgG
0
10
0 200 400 600 800 1.0k

TRA-1-60/DAPI VEGFR2/DAPI FSC


J M
4
10
D I 98.9 0.3%
103
VEGFR2

102

101 IgG
100
0 200 400 600 800 1.0k

FSC
Fig. 1. Schematic of BMEC differentiation protocol. (A) Singularized hPSCs are seeded on six-well plates coated with Matrigel, vitronectin, or Synthemax and
expanded for 3 days in mTeSR1. Differentiation to primitive streak is initiated by 24-hour treatment with 6 mM CHIR99021 in DeSR1. Cells progress to intermediate
mesoderm and endothelial progenitors during culture in serum-free defined DeSR2 medium. At day 6, BMEC specification is induced by culture in hESFM supplemented
with 2% B27, 10 mM RA, and bFGF (20 ng/ml) (hECSR1) for 2 days. After replating on Matrigel or fibronectin/collagen IV substrates, BMECs are obtained. (B to D) The
pluripotent state of expanded hPSCs was verified before differentiation by immunofluorescence for OCT4 (B), NANOG (C), and TRA-1-60 (D). DAPI, 4,6-diamidino-2-
phenylindole. (E and F) The expression of the primitive streak marker brachyury was assessed by immunofluorescence (E) and flow cytometry (F) 24 hours after
CHIR99021 treatment. IgG, immunoglobulin G; FSC, forward scatter. (G and H) On day 4 of differentiation, the expression of the intermediate mesoderm marker
PAX2 was quantified. (I and J) On day 5, the expression of the endothelial progenitor marker VEGFR2 was analyzed. Scale bars, 100 mm.

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 2 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

in a serum- and albumin-free medium (51). Hence, at day 0, 6 mM 25 CLDN genes, all 407 solute carrier (SLC) transporters, and all 53
CHIR99021 was added to DeSR1 [unconditioned medium (UM) adenosine 5-triphosphate (ATP)binding cassette (ABC) transport-
lacking KnockOut Serum Replacement: DMEM/F12, 1% minimum es- ers regardless of previous knowledge of BBB association (table S1).
sential medium (MEM)nonessential amino acids (NEAA), 0.5% Primary human BMECs expressed 234 of these genes. BMECs differ-
GlutaMAX, and 0.1 mM b-mercaptoethanol (32)] to initiate differenti- entiated from hPSCs via the defined method expressed many of these
ation. After 24 hours, the medium was removed and cells were transi- same genes [206 of 234 (88%)], as did BMECs differentiated via the
tioned to DeSR2 (DeSR1 plus B27 supplement) for another 5 days with UM method [208 of 234 (89%); Fig. 2K], indicating a close similarity
daily medium changes. At day 0, pluripotency was verified by OCT4, between hPSC-derived BMECs and primary human BMECs with re-
NANOG, and TRA-1-60 immunofluorescence (Fig. 1, B to D). After spect to transcripts having likely relevance to BBB function.
24 hours of CHIR99021 treatment, almost 100% of the cells expressed Initially, differentiation was performed on Matrigel, which has been
the primitive streak marker brachyury, as assessed by immunolabeling shown to support BMEC generation from hPSCs (32). However, to re-
(Fig. 1E) and flow cytometry (Fig. 1F). In concert with brachyury ex- move the lot-to-lot variability inherent to Matrigel and to fully define
pression, primitive streak genes T and MIXL1 (52) peaked at day 2 and the differentiation platform, we explored differentiation on Synthemax
then markedly decreased (fig. S1). At day 4, more than 90% of the cells and recombinant human vitronectin coatings. Undifferentiated
expressed the intermediate mesoderm marker PAX2 (Fig. 1, G and H), IMR90-4 iPSCs were expanded on either Synthemax- or vitronectin-
and PAX2 expression peaked at day 6 (fig. S1). Nearly 100% of the cells coated surfaces for 3 days and then subjected to the differentiation
expressed the endothelial progenitor marker VEGFR2 at day 5 (Fig. 1, process shown in Fig. 1A. Cells were replated onto a human placenta

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


I and J), whereas the expression level of the endothelial progenitor derived collagen IV/human plasmaderived fibronectin-coated sur-
marker CD34 gradually increased and then diminished after day 6 face at day 8. Immunofluorescence at day 10 demonstrated the
(fig. S1). expression of key BMEC proteins in cells differentiated on defined
At day 6, cells were switched to hECSR1 medium [human endo- matrices (fig. S3, A and B).
thelial serum-free medium (hESFM) supplemented with basic fibro-
blast growth factor (bFGF, 20 ng/ml), 10 mM RA, and B27] to induce hPSC-derived BMECs exhibit BBB phenotypes
RA signaling in the hPSC-derived endothelial progenitors in an at- In addition to the examination of BMEC gene and protein expression,
tempt to drive the specification to BMECs. Cells were maintained in we also evaluated endothelial and BMEC phenotypes. After 8 days of
this medium for 2 days. At day 8, cells were replated onto a Matrigel- differentiation, cells were replated onto a Matrigel-coated surface at
coated substrate in hECSR1, and at day 9, the medium was switched 1 million cells/cm2 and maintained in hECSR1 medium. At day 9,
to hECSR2 (hECSR1 lacking RA and bFGF). The expression of CDH5 culture medium was switched to hECSR2. Day 10 hPSC-derived
[vascular endothelial cadherin (VE-cadherin)] was substantially induced BMECs exhibited EC properties, including the expression of von
after RA treatment (fig. S1). The expression of the tight junctionrelated Willebrand factor (vWF) (Fig. 3A), formation of tube-like structures
genes TJP1, CLDN5, and OCLN and the efflux transporter ABCB1 also on Matrigel in the presence of VEGF (Fig. 3B), uptake of acetylated
increased during differentiation (fig. S1). The resultant day 10 BMEC- low-density lipoprotein (LDL) (Fig. 3C), and up-regulation of inter-
like cells were a pure population expressing endothelial markers (CD31 cellular adhesion molecule 1 (ICAM-1) expression after treatment
and VE-cadherin), BBB glucose transporter (GLUT-1), tight junction with tumor necrosis factora (TNF-a) (Fig. 3, D to F). BMEC efflux
proteins (ZO-1, claudin-5, and occludin), and efflux transporters (BCRP, transporter activities were also measured at day 10. Efflux transport-
MRP1, and Pgp) (Fig. 2, A to I). Thus, sequential treatment of hPSCs er accumulation assays were performed by quantifying intracellular
with CHIR99021 and RA directed hPSCs through endothelial pro- accumulation of fluorescent substrates, including the Pgp substrate
genitors to ECs that expressed BMEC markers. We next tested whether rhodamine 123, the MRP family substrate 2,7-dichlorofluorescein
the differentiation protocol illustrated in Fig. 1A generated cells ex- diacetate (DCFDA), and the BCRP family substrate Hoechst. In the
pressing BMEC markers in additional hPSC lines, including H9 hu- presence of the transporter-specific inhibitors cyclosporine A (CsA;
man embryonic stem cells (hESCs) and 19-9-11 iPSCs. These lines Pgp), MK571 (MRP), and Ko143 (BCRP), the intracellular accumu-
also produced cells expressing endothelial and BMEC markers, in- lation of fluorescent substrates increased between 150 and 220%, in-
cluding CD31, GLUT-1, ZO-1, claudin-5, occludin, MRP1, BCRP1, dicating the activity of each class of transporters in hPSC-derived
and Pgp, at day 10 (fig. S2). BMECs (Fig. 3, G to I). In addition, BMECs differentiated on defined
Next, RNA sequencing was used to compare global gene expres- vitronectin or Synthemax substrates also exhibited Pgp, MRP, and
sion profiles in the hPSC-derived BMECs differentiated, as shown BCRP transporter activities similar to those differentiated on Matrigel
in Fig. 1A, with BMECs generated from our previously reported co- (fig. S3C). Next, polarization of Pgp activity was demonstrated by
differentiation system [UM (32)] and primary human BMECs. As measuring rhodamine 123 flux across the BMEC monolayer in the pres-
expected, hPSC-derived BMECs from three independent differentia- ence and absence of the Pgp-specific inhibitor CsA and in both the ap-
tions clustered closely and were similar to those generated from the ical to basolateral (A-B) and basolateral to apical (B-A) directions. As
undefined UM platform. Moreover, the hPSC-derived BMECs clustered shown in Fig. 3J, CsA treatment increased rhodamine 123 transport
with primary human BMECs and were distinct from undifferentiated across the BMEC monolayer by 160% in the A-B direction. In con-
hPSCs and hPSC-derived ectoderm, endoderm, and mesoderm (Fig. 2J). trast, CsA inhibition resulted in a 23% decrease in rhodamine 123
The Pearson correlation coefficient between defined BMECs and crossing the barrier in the B-A direction (indicated in Fig. 1A), indi-
primary human BMECs was 0.77 (P < 0.001), which indicates a strong cating Pgp efflux function polarized in the B-A direction. Finally,
positive association between these two groups. We next analyzed the BMECs differentiated via the defined protocol exhibited Pgp accu-
expression of a subset of genes that regulate key BBB attributes, includ- mulation and transport similar to those differentiated via our previ-
ing tight junctions and molecular transporters. The gene set comprises ously reported undefined co-differentiation protocol (fig. S4, UM
20 tight junctionrelated genes (1, 5356) and an unbiased list of all protocol).

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 3 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

A CD31 VE-cadherin GLUT-1

ZO-1 Claudin-5 Occludin

BCRP MRP1 Pgp

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


4
B 10 99.2 0.8% C 10 4
99.9 0.1% D 10 4
99.9 0.3% E 104 99.8 0.1%
3
10 103 103 103

Claudin-5
GLUT-1
CD31

ZO-1

102 102 102 102


1 IgG IgG IgG IgG
10 10 1
101 101

100 0 200 400 600 800 1.0k 100 0 200 400 600 800 1.0k 100 0 200 400 600 800 1.0k 100 0 200 400 600 800 1.0k
FSC FSC FSC FSC
F 10 4
99.7 0.2% G 10 4
99.9 0.1% H 10 4
99.8 0.1% I 104
98.6 0.8%

103 103 103 103


Occludin

MRP1
BCRP

Pgp

102 102 102 102

10 1 IgG 10 1 IgG 10 1 IgG 101 IgG

100 0 200 400 600 800 1.0k 100 0 200 400 600 800 1.0k 100 0 200 400 600 800 1.0k 100 0 200 400 600 800 1.0k
FSC FSC FSC FSC

J K
Row max
HBMEC
UM-BMEC
D-BMEC2
D-BMEC3
D-BMEC1
Endo
hPSC
Mes
Ecto
Row min

Fig. 2. hPSC-derived BMECs express key BMEC proteins and have gene expression profiles similar to those of primary human BMECs. (A to I) At day 10, BMECs
differentiated as shown in Fig. 1A were characterized by immunocytofluorescence (A) and flow cytometry (B to I) for key endothelial and BMEC markers. Scale bars,
100 mm. (J) Hierarchical clustering of whole transcripts counted by RNA sequencing was plotted using GENE-E. Hierarchical clustering analysis was performed on log2-
transformed gene counts of RNA sequencing expression data of undifferentiated hPSCs (7678); hPSC-derived endoderm (Endo) (76), ectoderm (Ecto) (77), and mesoderm
(Mes) (78); BMECs differentiated under defined conditions as illustrated in Fig. 1A (D-BMEC1, D-BMEC2, and D-BMEC3: IMR90-4derived BMECs at day 10 generated from three
independent differentiations); BMECs differentiated in UM (UM-BMECs); and human primary BMECs (hBMECs). Distances were computed using one minus Pearson correlation
with average linkage. (K) A set of 506 tight junction and transporter genes (table S1) was used to investigate gene expression similarity between primary human BMECs, hPSC-
derived BMECs differentiated under defined conditions, and hPSC-derived BMECs differentiated in UM (32). The gene set included 20 tight junctionrelated genes (1, 5356), all
25 CLDN genes, all 407 solute carrier (SLC) transporters, and all 53 ATP-binding cassette (ABC) transporters. CLDN, SLC, and ABC genes were included without previous knowl-
edge of BBB association. A threshold of >1 fragment per kilobase million (FPKM) was used to define expressed versus nonexpressed transcripts.

Finally, previous studies have shown that coculturing BMECs, includ- tained as a monoculture or cocultured with primary human pericytes
ing those that are iPSC-derived, with neural progenitor cells, astrocytes, for the first 24 hours, followed by coculture with hPSC EZ sphere
and pericytes can enhance BBB properties such as TEER (33, 5760). derived astrocytes and neurons (1:3) (61) for additional 3 days. Max-
Day 8 iPSC-derived BMECs seeded on Transwells were either main- imum TEER for the monoculture system was ~3000 ohmcm2, and

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 4 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

A vWF/DAPI B C LDL/DIC

D ICAM-1/before E ICAM-1/after F 2.0k IgG


ICAM-1
1.5k

Count
62% increase
1.0k ICAM-1
after TNF-

500

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


0
100 101 102 103 104

Fluorescence
Rhodamine 123 accumulation Hoechst accumulation DCFDA accumulation Rhodamine 123 transport

G *** H ** I ** J **
A-B **
Percentage

B-A

*
**
**

Fig. 3. hPSC-derived BMECs exhibit key BBB phenotypes. hPSC-derived BMECs were differentiated as illustrated in Fig. 1A. (A) Immunofluorescence images of vWF
(red) and DAPI staining (blue) in hPSC-derived BMECs at day 10. (B) hPSC-derived BMECs were dissociated with Accutase and replated at 1 105 cells/cm2 on Matrigel.
Images were taken after 24 hours in hECSR2 with VEGF (50 ng/ml). (C) hPSC-derived BMECs at day 10 were analyzed with an LDL Uptake Assay kit. LDL is shown in red on a
merged bright-field image. (D to F) ICAM-1 induction in hPSC-derived BMECs. hPSC-derived BMECs at day 10 were treated with TNF-a (10 ng/ml) for 16 hours. Immuno-
fluorescence images for ICAM-1 were acquired before (D) and after (E) TNF-a treatment. (F) Cells were dissociated with Accutase, and ICAM-1 expression was quantified by
flow cytometry before and after TNF-a treatment. Efflux transporter activities were measured by the intracellular accumulation of (G) rhodamine 123, (H) Hoechst, and
(I) DCFDA, substrates for Pgp, BCRP, and MRP, respectively. CsA, Ko143, and MK571 were used as specific inhibitors of Pgp, BCRP, and MRP, respectively. (J) The polarization of
Pgp was measured by rhodamine 123 transport across the BMEC monolayer from the apical to basolateral side (A-B) and vice versa (B-A). Inhibitor-treated samples were
independently normalized to each respective noninhibitor-treated control sample. (K) TEER was measured in hPSC-derived BMECs cocultured with astrocytes, neurons, and
pericytes. Data were collected from three independent replicates and are means SEM. *P < 0.05, **P < 0.01, ***P < 0.001. Scale bars, 100 mm.

coculture further elevated TEER by 30% at day 2 and helped maintain addition to the optimal initial day 3 cell seeding density used above
an elevated TEER throughout the duration of the experiment com- (35 103 cells/cm2), we tested a range of seeding densities, from 8.8
pared to the monoculture control (Fig. 3K). 103 to 140 103 cells/cm2, to explore how density affects BMEC yield
and phenotype. As shown in Fig. 4A, TEER was a strong function of
Cell density is crucial for BMEC differentiation seeding density, with only 35 103 cells/cm2 yielding BMECs with
Cell density has been shown to be crucial for efficient hPSC differen- substantial barrier function at day 2 after transfer onto Transwells.
tiation to a variety of lineages, including BMECs (35, 6264). Thus, in The BMEC TEER peaked 2 days after replating and plateaued above

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 5 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

A B
***

D Claudin-5 E
C 800
Isotype 8.8k 35k

Geometric mean
140k 100
104 104 10104 4 100

Claudin-5 (%)
15.4 1.1% 99.2 0.3% 600 70k
Claudin-5

Count
103 103 10103 3
35k 80
400 17.5k
102 102 10102 2
50 60
8.8k
101 101 10101 1
200 Isotype 40

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


100 10100 0 20
100
0 200 400 600 800 1.0k 0 200 400 600 800 1.0k 00 200
200 400
400 600
600 800
800 1.0k
1.0k 0 0
100 101 102 103 104 8.8k 17.5k 35k 70k 140k
FSC FSC FSC Fluorescence Density (cells/cm2)
F
Claudin-5/DAPI 8.8k Claudin-5/DAPI 17.5k Claudin-5/DAPI 35k Claudin-5/DAPI 70k Claudin-5/DAPI 140k

H Occludin I
G Isotype 8.8k 35k 800
104 104

Geometric mean
10 4
100 120
23.5 1.3% 95.1 1.1% 140k
Occludin

Occludin (%)
103 103 600 80
103
70k 100
Count

102 102 102 35k 60


400
17.5k 40 80
101 101 101 8.8k
200
0 0 Isotype 20
10 10 100 60
0 200 400 600 800 1.0k 0 200 400 600 800 1.0k 0 200 400 600 800 1.0k
0 0
FSC 8.8k 17.5k 35k 70k 140k
FSC FSC 100 10 1
10 2
10 3
104

Density (cells/cm2)
Fluorescence
J
Occludin/DAPI 8.8k Occludin/DAPI 17.5k Occludin/DAPI 35k Occludin/DAPI 70k Occludin/DAPI 140k

Fig. 4. Initial seeding density is critical for BMEC differentiation. (A) hPSCs were seeded at the indicated densities (from 8800 to 140,000 cells/cm2) and differ-
entiated to BMECs, as illustrated in Fig. 1A. TEER was measured 2 days after replating on Transwell membranes at 106 cells/cm2. (B) TEERs of hPSC-derived BMECs were
measured daily for 7 days after replating on Transwell membranes. Data were collected from three independent replicates and are means SEM. ***P < 0.001. (C to E) The
percentage of claudin-5positive cells and expression levels of claudin-5 were quantified by flow cytometry at day 8 for cells differentiated at the indicated seeding density
(cells/cm2). (F) The localization of claudin-5 in cells differentiated at different seeding densities was investigated by immunofluorescence. White arrows indicate sample
areas lacking claudin-5 expression, and red arrows indicate discontinuous claudin-5. (G to I) The percentage of occludin-positive cells and expression levels of occludin
were quantified by flow cytometry at day 8 for cells differentiated at the indicated seeding density (cells/cm2). (J) The localization of occludin in cells differentiated at
different seeding densities was investigated by immunofluorescence. White arrows indicate sample areas lacking occludin expression, and red arrows indicate dis-
continuous occludin. Flow cytometry plots are representative of three independent experiments. Insets indicate the mean percentage of cells in the immunopositive
gated region SEM. Scale bars, 100 mm.

2000 ohmcm2 through day 7 (Fig. 4B). At non-optimum seeding den- cess. Cells differentiated at all densities tested yielded EC populations
sities, TEER gradually increased through 6 days after replating, reach- with nearly 100% VEGFR2+ cells at day 5 and more than 90% CD31+
ing approximately 1000 ohmcm2. We next assessed the expression of cells at day 10 (fig. S5). This suggested that deficits in barrier function
endothelial markers to investigate the endothelial specification pro- may be a result of poor BMEC specification. Thus, we assessed BMEC

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 6 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

marker expression in populations differentiated at different seeding tin or Synthemax required RA supplementation to achieve substantial
densities. Nearly 100% of cells expressed Pgp after the differentiation barrier properties (fig. S11). We then investigated the expression and
process at either day 8 or day 10 (fig. S6). However, only cells differ- localization of tight junction proteins. Both occludin and ZO-1 were
entiated at the optimal seeding density of 35 103 cells/cm2 yielded a expressed in nearly all cells at day 10 regardless of RA treatment; how-
pure claudin-5expressing population with maximal claudin-5 expres- ever, RA treatment significantly increased the expression levels of
sion (Fig. 4, C to E). In addition, a subpopulation of cells differentiated occludin and ZO-1 (Fig. 5, E and F). Although occludin and ZO-1 ex-
from non-optimal starting densities also lacked junctional claudin-5 pression was lower in the absence of RA, immunofluorescence results
(Fig. 4F, white arrows) or exhibited discontinuous claudin-5 localization indicate that nearly all the cells differentiated in the absence of RA still
at cell junctions (Fig. 4F, red arrows). Similarly, only cells differen- expressed occludin and ZO-1; however, the junctional distribution
tiated from seeding densities of at least 35 103 cells/cm2 yielded a was nonuniform (Fig. 5G, indicated with red arrows and compare
nearly pure population of occludin-expressing cells (Fig. 4, G to I). to Fig. 2A). In contrast to the results with occludin and ZO-1, in the
Immunofluorescence analysis of occludin also showed that a large absence of RA, only around 60% of the ECs expressed claudin-5
fraction of cells differentiated from initial densities less than 35 compared to 100% of RA-treated cells expressing claudin-5 (Fig. 5,
103 cells/cm2 lacked occludin expression (Fig. 4J, white arrows). Un- H and I). In addition, claudin-5 expression was also substantially
like claudin-5 and occludin, seeding density did not have a significant greater in RA-treated cells (Fig. 5I). Immunofluorescence indicated
effect on ZO-1 expression, but cells differentiated at initial densities of that in the absence of RA, many of the cells did not express claudin-5
8.8 103 cells/cm2 showed discontinuous ZO-1 localization (fig. S7, red (Fig. 5J, white arrows), and those that did exhibited nonuniform junc-

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


arrows). Immunofluorescence for additional BMEC markers also indi- tional distribution of claudin-5 similar to that observed with occludin
cated poor localization of CD31, MRP1, and BCRP in cells differen- and ZO-1 (Fig. 5J, red arrows). Together, these results suggest that RA
tiated at non-optimum initial cell densities (fig. S7, compare 35k to is not necessary for hPSC differentiation to ECs but enhances key
other densities). H9 hESCs and 19-9-11 iPSCs yielded BMECs exhibit- BMEC phenotypes in the hPSC-derived ECs, including the expression
ing TEER at or above 2000 ohmcm2 at an optimal initial seeding den- and localization of tight junction proteins that promote barrier func-
sity of 35 103 cells/cm2 (fig. S8, A and B). We also compared the tion as measured by TEER.
differentiation reproducibility with that of the previously reported
UM protocol (33). Although both methods produce BMECs capable
of substantial barrier formation from multiple hPSC lines, BMECs DISCUSSION
differentiated from H9 hESCs and 19-9-11 iPSCs using the defined Here, we demonstrate a robust and efficient process to differentiate
method exhibited higher TEERs and lower batch-to-batch variation hPSCs to BMECs in a defined manner. The cells progress as a homo-
(fig. S8A). Finally, 35 103 cells/cm2 was also found to be the optimal geneous population from a pluripotent state to primitive streaklike
seeding density for differentiation on Synthemax and vitronectin and intermediate mesodermlike stages, to endothelial progenitors,
substrates, with vitronectin substrates performing more closely to and eventually to ECs that express BMEC markers and exhibit BBB
Matrigel than Synthemax substrates in TEER assays (fig. S9). barrier and efflux transporter properties. This differentiation method
uses a completely defined platform, including culture medium and
RA enhances BMEC phenotypes substrates. Defined reagents exhibit less lot-to-lot variability, leading
Previously, we have shown that RA induces BBB properties in hPSC- to more robust and efficient differentiation and allowing differentia-
derived BMECs (33). Other studies have also demonstrated that RA tion results to be more reliable and reproducible. We have tested three
signaling regulates BBB formation and induces BBB phenotypes different hPSC lines with this differentiation protocol, and all of these
(46, 47). To determine the role of RA in specifying BMEC differen- lines were able to differentiate into pure populations of BMEC, with
tiation and enhancing the BMEC phenotypes described in Figs. 2 definitive BMEC properties at the same optimum cell density.
and 3, we compared differentiation in the presence and absence of In vivo, ECs that form the BBB originate from mesoderm progeni-
RA using the protocol illustrated in Fig. 1A. From day 6 to day 8, cells tors located outside the CNS (65). In contrast to previous BMEC dif-
were maintained either in hECSR1 or in hECSR1 lacking RA. Quan- ferentiation protocols (32, 37) that rely on coculture of endothelial
titative polymerase chain reaction (qPCR) showed that the expres- progenitors with pericytes, astrocytes, or differentiating neural cells,
sion of the tight junctionrelated genes TJP1, CLDN5, and OCLN this differentiation strategy instead relies on sequential Wnt and RA
and the efflux transporters ABCG2, ABCC1, and ABCB1 was greater signaling activation to first specify ECs and then enhance BMEC prop-
(3- to 20-fold) in cells exposed to RA (Fig. 5A). Nearly 100% of cells erties, respectively. First, activation of canonical Wnt signaling by
expressed CD31 at day 6, and this expression was preserved in the CHIR99021 addition directs hPSCs to brachyury-positive primitive
presence of RA induction at day 8 (Fig. 5B). Immunofluorescence for streaklike cells that then differentiate to PAX2-positive intermediate
CD31 and other BMEC markers for cells differentiated in the absence mesoderm and EC progenitors when differentiated at the appropriate
of RA, including VE-cadherin, GLUT-1, and MRP1, is shown in fig. density in DeSR2 medium. Next, RA treatment for 2 days drives these
S10A. Nearly 100% of cells differentiated in the absence and presence endothelial progenitor cells to express key BMEC markers and exhibit
of RA expressed Pgp, but RA-treated cells expressed more Pgp than BMEC-specific properties, including high TEER and efflux transporter
nontreated cells (Fig. 5C). To evaluate the barrier formation potential activity. Our experiments showed that although RA was not necessary
of the differentiated BMECs and to assess the effects of RA treatment, to obtain ECs, RA treatment significantly increased BBB properties such
we replated day 8 BMECs onto Transwells and measured TEER at day as TEER. The TEER enhancement correlated with increased expression
10. As shown in Fig. 5D, cells differentiated in the presence of RA ex- and improved localization of the tight junction proteins occludin and
hibited physiologically relevant TEER (~4000 ohmcm2), whereas claudin-5. These findings are similar to those results observed after RA
those differentiated in the absence of RA exhibited significantly re- treatment of hPSC-derived BMECs generated by co-differentiation with
duced barrier properties. Similarly, BMECs differentiated on vitronec- neural cells using our previously reported undefined protocol (33), in

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 7 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

A B C Pgp D
104 800 ***
NO RA-D10
** ** ** ** ** * D6 97.2 0.4% 99.6 0.3%
Isotype
103 D8 97.4 0.3% 600 RA-D10
99.8 0.1%

Count
79.2% increase

CD31
102 400

CD31
200
101

0
100 0 200 400 600 800 1.0k 100 101 102 103 104

FSC Fluorescence

E Occludin F ZO-1
NO RA-D10 1.2k NO RA-D10
1.2k
Isotype 95.1 0.6% Isotype 99.1 0.3%
RA-D10 RA-D10 G
99.1 0.5% 900 99.4 0.1%
900
191% increase 328% increase Occludin/DAPI ZO-1/DAPI
Count

Count

600 600

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


300 300

0 0
100 101 102 103 104 100 101 102 103 104

Fluorescence Fluorescence
Claudin-5
H 104 Isotype I 800 Isotype
NO RA-D10 60.0 2.8% NO RA-D10
RA-D10 99.8 0.1% RA-D10 J
103 600 436% increase Claudin-5/DAPI
Claudin-5

Count

2
10 400

101 200

100 0
0 200 400 600 800 1.0k 100 101 102 103 104
FSC Fluorescence
Fig. 5. RA induces acquisition of key BMEC phenotypes in EC progenitors. BMECs were differentiated as shown in Fig. 1A in the presence or absence of RA, as
indicated. (A) At day 8, the expression of tight junction and transporter genes was assessed by qPCR. (B) Flow cytometry for CD31 expression was performed at days 6 (D6)
and 8 (D8). (C) Pgp expression was quantified by flow cytometry at day 10. At day 6, the medium was switched to hESFM containing or lacking RA, as indicated. (D) At day 8,
cells were replated onto Matrigel-coated Transwell membranes at 106 cells/cm2 in the presence or absence of 10 mM Y-27632. TEER was measured at day 10, 2 days after
replating. (E to G) Occludin (E and G) and ZO-1 (F and G) expression and localization were assessed by flow cytometry and immunofluorescence at day 10. Red arrows indicate
discontinuous occludin or ZO-1 localization. (H and I) At day 10, the expression levels of claudin-5 in BMECs differentiated in the presence or absence of RA were assessed by
flow cytometry. (J) The localization and expression of claudin-5 of cells differentiated in the absence of RA were determined via immunofluorescence (white arrows indicate
cells lacking claudin-5, and red arrows indicate discontinuous claudin-5). Images, bar graphs, and flow cytometry plots are representative of three independent experiments.
Data from three independent replicates are means SEM. *P < 0.05, **P < 0.01, ***P < 0.001. Insets denote either percentage of positively labeled cells or percentage increase
in marker expression upon RA treatment. Scale bars, 100 mm.

addition to those studies that have explored the barrier-enhancing (6668). An initial cell seeding density of 35 103 cells/cm2 at day 3
effects of astrocyte or neuron coculture with hPSC-derived BMECs is necessary to yield homogeneous populations of BMECs with high
(33, 60). However, even in the absence of RA treatment, the previously expression and proper localization of key BBB proteins, in turn
reported undefined protocol still yielded pure populations of claudin-5 leading to optimal barrier properties. In addition, this optimum
expressing BMECs, whereas RA was critical in the defined protocol for seeding density translated to multiple hPSC lines and to differentia-
generating pure claudin-5expressing populations. Thus, the effects of tion on defined matrices. Cells differentiated at non-optimal seeding
defined differentiation do not exactly mimic those provided by cocul- densities expressed BMEC markers but exhibited a reduced TEER,
tured neural cells, yet the resultant BMECs were quite similar on a likely resulting from diminished claudin-5 and occludin expression
whole-transcriptome level to BMECs generated using the undefined co- and improper junctional localization. Thus, RA signaling and cell
culture approach and to primary human BMECs. density similarly regulate the capability for the endothelial progenitors
Previously, we have shown that cell seeding density can affect to acquire BMEC properties. Although we determined that seeding
BMEC differentiation from hPSCs using the neural co-differentiation density is a crucial regulator of hPSC differentiation to BMECs, we
protocol (35). Other studies have also demonstrated that cell seeding do not yet know the mechanism by which density affects differentia-
density is crucial for efficient hPSC differentiation to various lineages tion efficiency.

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 8 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

Coculturing hPSC-derived BMECs with pericytes, astrocytes, and Alexa Fluor 488; 1:200) for 1 hour at room temperature. Cells were
neurons further elevated TEER, consistent with previous studies that then washed with PBS three times, followed by treatment with DAPI
showed that coculturing BMECs with these neural cells can enhance Fluoromount-G (Southern Biotech), and were visualized. A detailed
BBB properties (57, 58, 6971). These data suggest that it will be possible list of antibodies is shown in table S2.
to integrate these defined hPSC-derived BMECs with other cells of the
neurovascular unit to create an isogenic patient-derived model that can Flow cytometry
be used to study the role of neurovascular unit in human neurological Cells were dissociated with Accutase, fixed in 1% PFA for 15 min at
diseases (60). In addition, this defined BMEC differentiation method room temperature, and then washed with 0.5% bovine serum albumin
has the potential to be a powerful and robust tool for preclinical studies (BSA) (Bio-Rad) plus 0.1% Triton X-100 three times. Cells were stained
of pharmaceutical transport through the BBB. with primary and secondary antibodies diluted in 0.5% BSA plus
0.1% Triton X-100, as previously described (31). Data were collected
on a FACSCalibur flow cytometer (Becton Dickinson) and analyzed
MATERIALS AND METHODS using FlowJo. Corresponding isotype antibodies were used as FACS
hPSC culture and differentiation (fluorescence-activated cell sorting) gating control. Details about anti-
Human iPSCs [iPS(IMR90)-4 (72), iPS-DF 19-9-11T (73), and hESCs body source and usage are provided in table S2.
(H9) (29)] were maintained on Matrigel (Corning)coated surfaces in
mTeSR1 (STEMCELL Technologies), as previously described (74). Quantitative real-time PCR

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


Before differentiation, hPSCs were singularized with Accutase (Inno- Total RNA was extracted with the RNeasy Mini Kit (Qiagen) and treated
vative Cell Technologies) and plated onto Matrigel-coated plates at a with deoxyribonuclease (Qiagen). One microgram of total RNA was
density between 25 103 and 50 103 cells/cm2 in mTeSR1 supple- reverse-transcribed into complementary DNA using an oligo(dT)
mented with 10 mM Rho-associated protein kinase (ROCK) inhibitor primer with SuperScript III Reverse Transcriptase (Invitrogen).
Y-27632 (Selleckchem). hPSCs were expanded in mTeSR1 for 3 days. Quantitative real-time PCR was done in triplicate with iQ SYBR Green
For differentiation on defined substrates, singularized hPSCs were SuperMix (Bio-Rad). Glyceraldehyde-3-phosphate dehydrogenase
plated onto the vitronectin-coated (Thermo Fisher Scientific) or (GAPDH) was used as an endogenous housekeeping control. Primer
Synthemax-coated (Corning) surface at a density between 25 103 sequences are provided in table S3.
and 50 103 cells/cm2 in mTeSR1 supplemented with 10 mM ROCK
inhibitor Y-27632 (Selleckchem). To initiate differentiation at day 0, LDL uptake assay
cells were treated with 6 mM CHIR99021 (Selleckchem) in DeSR1: Differentiated BMECs at day 10 were analyzed with the LDL Uptake
DMEM/Hams F12 (Thermo Fisher Scientific), 1 MEM-NEAA Assay Kit (Abcam). Culture medium was aspirated and replaced with
(Thermo Fisher Scientific), 0.5 GlutaMAX (Thermo Fisher Scientific), LDL-DyLight 550 working solution. Cells were then incubated for
and 0.1 mM b-mercaptoethanol (Sigma). After 24 hours, the medium 3 hours at 37C, followed by three washes with PBS, and then visualized
was changed to DeSR2: DeSR1 plus 1 B27 (Thermo Fisher Scientific) via fluorescent microscopy with excitation and emission wavelengths of
every day for another 5 days. At day 6, the medium was switched to 540 and 570 nm, respectively. After visualization, cells were fixed with a
hECSR1: hESFM (Thermo Fisher Scientific) supplemented with bFGF cell-based fixative solution for 10 min. Cells were then washed with tris-
(20 ng/ml), 10 mM RA, and 1 B27. After 2 days of culture in hECSR1 buffered saline plus 0.1% Triton X-100 for 5 min, followed by 30-min
medium, day 8 cells were dissociated with Accutase and plated at blocking with Cell-Based Assay Blocking Solution. Cells were then
1 106 cells/cm2 in hECSR1 onto 48-well tissue culture plates or stained with rabbit anti-LDL receptor primary antibody and DyLight
1.12-cm2 Transwell-Clear permeable inserts (0.4 mm pore size) coated 488conjugated secondary antibody. Images were taken with a fluores-
with Matrigel (100 mg/ml). For cells differentiated on vitronectin or cent microscope with excitation and emission wavelengths of 485 and
Synthemax substrates, cells were dissociated with Accutase and plated 535 nm, respectively.
at 1 106 cells/cm2 in hECSR1 onto 48-well tissue culture plates or
1.12-cm2 Transwell-Clear permeable inserts (0.4 mm pore size) coated Efflux transporter accumulation and transport assay
with human placentaderived collagen IV (400 mg/ml)/human plasma Pgp, BCRP, and MRP functionality was assessed by intracellular accu-
derived fibronectin (100 mg/ml). At day 9, the medium was changed to mulation of fluorescent transporter substrates and transport of fluo-
hECSR2 (hECSR1 without RA or bFGF) for longer-term mainte- rescent substrates across BMEC monolayers. Rhodamine 123 (10 mM;
nance. Y-27632 was added to increase the attachment (75) of cells dif- Sigma), Hoechst (20 mM; Thermo Fisher Scientific), and DCFDA
ferentiated in the absence of RA and was permitted to ensure confluent (10 mM; Life Technologies) were used as the specific substrates for
monolayer formation and confirm that the reduction in TEER was not Pgp, BCRP1, and MRP1, respectively. BMECs at day 10 were pretreated
only a result of incomplete monolayer formation. for 1 hour with or without specific transporter inhibitors [10 mM CsA
(Pgp inhibitor), 10 mM Ko143 (BCRP inhibitor) (Sigma), and 1 mM
Immunochemistry MK571 (MRP inhibitor) (Sigma) in Hanks balanced salt solution
Cells were rinsed with ice-cold phosphate-buffered saline (PBS) once (HBSS)]. Cells were then treated with transporter substrates in HBSS
and fixed in either ice-cold methanol or 4% paraformaldehyde (PFA) and incubated for 1 hour at 37C on an orbital shaker. Cells were
for 15 min. Cells were then blocked with 10% goat serum in PBS washed with PBS three times and then lysed with radioimmunoprecip-
containing 0.3% Triton X-100 for 30 min (10% PBSGT). Primary anti- itation assay buffer (Pierce Biotechnology). Fluorescence intensity was
bodies were diluted in 10% PBSGT, and cells were incubated in the measured on a plate reader (485-nm excitation and 530-nm emission
antibody solutions at 4C overnight or at room temperature for 2 hours. for rhodamine 123 and DCFDA and 360-nm excitation and 497-nm
After three PBS washes, cells were incubated with secondary antibodies emission for Hoechst). Fluorescence intensity was subsequently nor-
in 10% PBSGT (goat anti-rabbit Alexa Fluor 594 and goat anti-mouse malized to cell number determined using a hemocytometer.

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 9 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

Endothelial cell tube formation fig. S4. BMECs differentiated from hPSCs in defined and undefined protocols exhibit similar
Each well of a 24-well tissue culture plate was coated with 300 ml of Pgp activities.
fig. S5. BMECs differentiated at different seeding densities express VEGFR2 and CD31.
Matrigel (10 mg/liter). BMECs at day 10 were dissociated with Ac- fig. S6. BMECs differentiated at different seeding densities express Pgp.
cutase and plated in hECSM1 plus VEGF (50 ng/ml) without RA or fig. S7. BMECs differentiated at different densities express BMEC proteins but can exhibit
bFGF at 2 105 cells per well. Phase-contrast images were acquired aberrant protein localization.
after 24 hours. fig. S8. TEER of BMECs differentiated from different hPSC lines.
fig. S9. TEER in BMECs differentiated from hPSCs at different seeding densities.
fig. S10. BMECs differentiated in the absence of RA exhibit low expression and mislocalization
RNA sequencing and data analysis of EC and BMEC proteins.
Total RNA of day 10 IMR90-4 iPSCderived BMECs with either fig. S11. RA enhances TEER for cells differentiated on Matrigel-, vitronectin-, and Synthemax-
defined protocol or UM protocol and primary human brain micro- coated surfaces.
table S1. Expression of tight junction and transporter genes in iPSC-derived and primary
vascular ECs (ACBRI 376, Cell Systems) were prepared with the
human BMECs.
Direct-zol RNA MiniPrep Plus Kit (Zymo Research) according to table S2. Antibodies used in this study.
the manufacturers instructions. Samples were sequenced on Illumina table S3. qPCR primers used in this study.
HiSeq 2500 at the University of Wisconsin-Madison Biotechnology
Center. The resulting sequence reads were mapped to the human ge-
nome (hg19) using HISAT49, and the RefSeq transcript levels (FPKMs)
REFERENCES AND NOTES
were quantified using the Python script rpkmforgenes.py50. A hierar- 1. B. Obermeier, R. Daneman, R. M. Ransohoff, Development, maintenance and disruption

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


chical clustering of whole transcripts was performed using GENE-E of the blood-brain barrier. Nat. Med. 19, 15841596 (2013).
on the log2-transformed gene counts. Distances were computed using 2. B. B. Weksler, E. A. Subileau, N. Perrire, P. Charneau, K. Holloway, M. Leveque,
one minus Pearson correlation with average linkage. FASTQ files of H. Tricoire-Leignel, A. Nicotra, S. Bourdoulous, P. Turowski, D. K. Male, F. Roux,
J. Greenwood, I. A. Romero, P. O. Couraud, Blood-brain barrier-specific properties of a
hPSCs (7678) and hPSC-derived ectoderm (77), endoderm (76), and human adult brain endothelial cell line. FASEB J. 19, 18721874 (2005).
mesoderm (78) were downloaded from the Gene Expression Omnibus 3. N. J. Abbott, L. Rnnbck, E. Hansson, Astrocyteendothelial interactions at the
(GEO) or ArrayExpress (www.ebi.ac.uk/arrayexpress/) database. The bloodbrain barrier. Nat. Rev. Neurosci. 7, 4153 (2006).
expression of a subset of genes that regulate key BBB attributes, in- 4. P. Ballabh, A. Braun, M. Nedergaard, The bloodbrain barrier: An overview: structure,
regulation, and clinical implications. Neurobiol. Dis. 16, 113 (2004).
cluding tight junctions and molecular transporters, was analyzed. The
5. W. M. Pardridge, The blood-brain barrier: Bottleneck in brain drug development. NeuroRx
gene set comprises 20 tight junctionrelated genes (1, 5356) and an 2, 314 (2005).
unbiased list of all 25 CLDN genes, all 407 solute carrier (SLC) trans- 6. Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier.
porters, and all 53 ATP-binding cassette (ABC) transporters regardless Adv. Drug Deliv. Rev. 64, 640665 (2012).
of previous knowledge of BBB association (table S1). Transcript levels 7. K. E. Sandoval, K. A. Witt, Blood-brain barrier tight junction permeability and ischemic
stroke. Neurobiol. Dis. 32, 200219 (2008).
(FPKMs) were set at a threshold of >1 FPKM, which indicates moderate 8. D. Fernndez-Lpez, J. Faustino, R. Daneman, L. Zhou, S. Y. Lee, N. Derugin,
expression (79). Primary human BMECs were used to screen out the M. F. Wendland, Z. S. Vexler, Bloodbrain barrier permeability is increased after acute
BBB-related genes from the gene list with the threshold of >1 FPKM. adult stroke but not neonatal stroke in the rat. J. Neurosci. 32, 95889600 (2012).
9. Y. Yang, G. A. Rosenberg, Bloodbrain barrier breakdown in acute and chronic
cerebrovascular disease. Stroke 42, 33233328 (2011).
Coculture of hPSC-derived BMECs with pericytes, neurons,
10. J. R. Cirrito, R. Deane, A. M. Fagan, M. L. Spinner, M. Parsadanian, M. B. Finn, H. Jiang,
and astrocytes J. L. Prior, A. Sagare, K. R. Bales, S. M. Paul, B. V. Zlokovic, D. Piwnica-Worms,
hPSC-derived BMECs at day 8 were replated onto 12-well Transwell D. M. Holtzman, P-glycoprotein deficiency at the blood-brain barrier increases amyloid-b
inserts at a density of 1 106 cells/cm2 and cocultured with primary deposition in an Alzheimer disease mouse model. J. Clin. Invest. 115, 32853290 (2005).
human pericytes residing on the bottom of the 12-well plates (1 105 cells 11. G. L. Bowman, J. A. Kaye, M. Moore, D. Waichunas, N. E. Carlson, J. F. Quinn, Bloodbrain
barrier impairment in Alzheimer disease: Stability and functional significance.
per well) for 24 hours in hECSR1 medium. Following coculture with peri- Neurology 68, 18091814 (2007).
cytes, BMECs were cocultured with EZ spherederived neurons and as- 12. A. Minagar, J. S. Alexander, Blood-brain barrier disruption in multiple sclerosis.
trocytes (a total of 1 105 cells per well; 1:3) in hECSR2 for the remainder Mult. Scler. 9, 540549 (2003).
of the experiment. TEER was measured as a function of time following 13. R. Kortekaas, K. L. Leenders, J. C. H. van Oostrom, W. Vaalburg, J. Bart, A. T. M. Willemsen,
N. H. Hendrikse, Bloodbrain barrier dysfunction in parkinsonian midbrain in vivo.
initiation of coculture.
Ann. Neurol. 57, 176179 (2005).
14. D. Shlosberg, M. Benifla, D. Kaufer, A. Friedman, Bloodbrain barrier breakdown as a
Statistics therapeutic target in traumatic brain injury. Nat. Rev. Neurol. 6, 393403 (2010).
Data are presented as means SEM. Statistical significance was de- 15. A. Beaumont, A. Marmarou, K. Hayasaki, P. Barzo, P. Fatouros, F. Corwin, C. Marmarou,
termined by Students t test (two-tailed) between two groups. Three J. Dunbar, in Brain Edema XI (Springer, 2000), pp. 125129.
16. J. D. Huber, R. D. Egleton, T. P. Davis, Molecular physiology and pathophysiology of tight
or more groups were analyzed by one-way analysis of variance junctions in the bloodbrain barrier. Trends Neurosci. 24, 719725 (2001).
(ANOVA) followed by Tukeys post hoc tests. P < 0.05 was considered 17. Y. Persidsky, M. Stins, D. Way, M. H. Witte, M. Weinand, K. S. Kim, P. Bock, H. E. Gendelman,
statistically significant. M. Fiala, A model for monocyte migration through the blood-brain barrier during
HIV-1 encephalitis. J. Immunol. 158, 34993510 (1997).
18. P. Annunziata, Blood-brain barrier changes during invasion of the central nervous system
by HIV-1. J. Neurol. 250, 901906 (2003).
SUPPLEMENTARY MATERIALS 19. M. L. Brines, P. Ghezzi, S. Keenan, D. Agnello, N. C. de Lanerolle, C. Cerami, L. M. Itri,
Supplementary material for this article is available at http://advances.sciencemag.org/cgi/ A. Cerami, Erythropoietin crosses the bloodbrain barrier to protect against experimental
content/full/3/11/e1701679/DC1 brain injury. Proc. Natl. Acad. Sci. U.S.A. 97, 1052610531 (2000).
fig. S1. Gene expression during hPSC differentiation to BMECs. 20. L. Belayev, R. Busto, W. Zhao, M. D. Ginsberg, Quantitative evaluation of blood-brain
fig. S2. BMECs differentiated from H9 hESCs and 19-9-11 iPSCs express EC- and BMEC-related barrier permeability following middle cerebral artery occlusion in rats. Brain Res. 739,
proteins. 8896 (1996).
fig. S3. BMECs differentiated on Synthemax and vitronectin express EC- and BMEC-related 21. M. Asahi, X. Wang, T. Mori, T. Sumii, J.-C. Jung, M. A. Moskowitz, M. E. Fini, E. H. Lo, Effects
proteins and have efflux transporter activities. of matrix metalloproteinase-9 gene knock-out on the proteolysis of bloodbrain

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 10 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

barrier and white matter components after cerebral ischemia. J. Neurosci. 21, 77247732 of immortalized brain endothelial cells as a tool for generating a standardized model of
(2001). the blood brain barrier in vitro. PLOS ONE 8, e70233 (2013).
22. M. F. Stins, F. Gilles, K. S. Kim, Selective expression of adhesion molecules on human brain 45. J. M. Stenman, J. Rajagopal, T. J. Carroll, M. Ishibashi, J. McMahon, A. P. McMahon,
microvascular endothelial cells. J. Neuroimmunol. 76, 8190 (1997). Canonical Wnt signaling regulates organ-specific assembly and differentiation of CNS
23. D. Wong, K. Dorovini-Zis, Upregulation of intercellular adhesion molecule-1 (ICAM-1) vasculature. Science 322, 12471250 (2008).
expression in primary cultures of human brain microvessel endothelial cells by cytokines 46. M. R. Mizee, D. Wooldrik, K. A. M. Lakeman, B. van het Hof, J. A. R. Drexhage, D. Geerts,
and lipopolysaccharide. J. Neuroimmunol. 39, 1121 (1992). M. Bugiani, E. Aronica, R. E. Mebius, A. Prat, H. E. de Vries, A. Reijerkerk, Retinoic acid
24. Y. Sano, F. Shimizu, M. Abe, T. Maeda, Y. Kashiwamura, S. Ohtsuki, T. Terasaki, M. Obinata, induces bloodbrain barrier development. J. Neurosci. 33, 16601671 (2013).
K. Kajiwara, M. Fujii, M. Suzuki, T. Kanda, Establishment of a new conditionally 47. M. R. Mizee, P. G. Nijland, S. M. A. van der Pol, J. A. R. Drexhage, B. van het Hof, R. Mebius,
immortalized human brain microvascular endothelial cell line retaining an in vivo P. van der Valk, J. van Horssen, A. Reijerkerk, H. E. de Vries, Astrocyte-derived retinoic
bloodbrain barrier function. J. Cell. Physiol. 225, 519528 (2010). acid: A novel regulator of bloodbrain barrier function in multiple sclerosis.
25. B. Weksler, I. A. Romero, P.-O. Couraud, The hCMEC/D3 cell line as a model of the human Acta Neuropathol. 128, 691703 (2014).
blood brain barrier. Fluids Barriers CNS 10, 16 (2013). 48. R. C. Lindsley, J. G. Gill, M. Kyba, T. L. Murphy, K. M. Murphy, Canonical Wnt signaling is
26. S. Syvnen, . Lindhe, M. Palner, B. R. Kornum, O. Rahman, B. Lngstrm, G. M. Knudsen, required for development of embryonic stem cell-derived mesoderm. Development 133,
M. Hammarlund-Udenaes, Species differences in blood-brain barrier transport of 37873796 (2006).
three positron emission tomography radioligands with emphasis on P-glycoprotein 49. X. Lian, X. Bao, A. Al-Ahmad, J. Liu, Y. Wu, W. Dong, K. K. Dunn, E. V. Shusta, S. P. Palecek,
transport. Drug Metab. Dispos. 37, 635643 (2009). Efficient differentiation of human pluripotent stem cells to endothelial progenitors via
27. M. A. Deli, Bloodbrain barrier models, in Handbook of Neurochemistry and Molecular small-molecule activation of WNT signaling. Stem Cell Reports 3, 804816 (2014).
Neurobiology: Neural Membranes and Transport (Springer, 2007), pp. 2955. 50. A. Q. Lam, B. S. Freedman, R. Morizane, P. H. Lerou, M. T. Valerius, J. V. Bonventre, Rapid
28. P. Naik, L. Cucullo, In vitro bloodbrain barrier models: Current and perspective and efficient differentiation of human pluripotent stem cells into intermediate
technologies. J. Pharm. Sci. 101, 13371354 (2012). mesoderm that forms tubules expressing kidney proximal tubular markers. J. Am. Soc.

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


29. J. A. Thomson, J. Itskovitz-Eldor, S. S. Shapiro, M. A. Waknitz, J. J. Swiergiel, V. S. Marshall, Nephrol. 25, 12111225 (2014).
J. M. Jones, Embryonic stem cell lines derived from human blastocysts. Science 282, 51. X. Lian, X. Bao, M. Zilberter, M. Westman, A. Fisahn, C. Hsiao, L. B. Hazeltine, K. K. Dunn,
11451147 (1998). T. J. Kamp, S. P. Palecek, Chemically defined, albumin-free human cardiomyocyte
30. S.-C. Zhang, M. Wernig, I. D. Duncan, O. Brstle, J. A. Thomson, In vitro differentiation generation. Nat. Methods 12, 595596 (2015).
of transplantable neural precursors from human embryonic stem cells. Nat. Biotechnol. 52. E. S. Ng, L. Azzola, K. Sourris, L. Robb, E. G. Stanley, A. G. Elefanty, The primitive streak
19, 11291133 (2001). gene Mixl1 is required for efficient haematopoiesis and BMP4-induced ventral mesoderm
31. X. Lian, C. Hsiao, G. Wilson, K. Zhu, L. B. Hazeltine, S. M. Azarin, K. K. Raval, J. Zhang, patterning in differentiating ES cells. Development 132, 873884 (2005).
T. J. Kamp, S. P. Palecek, Robust cardiomyocyte differentiation from human pluripotent 53. H.-C. Bauer, I. A. Krizbai, H. Bauer, A. Traweger, You Shall Not PassTight junctions of
stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl. Acad. the blood brain barrier. Front. Neurosci. 8, 392 (2014).
Sci. U.S.A. 109, E1848E1857 (2012). 54. E. G. Geier, E. C. Chen, A. Webb, A. C. Papp, S. W. Yee, W. Sadee, K. M. Giacomini, Profiling
32. E. S. Lippmann, S. M. Azarin, J. E. Kay, R. A. Nessler, H. K. Wilson, A. Al-Ahmad, S. P. Palecek, solute carrier transporters in the human bloodbrain barrier. Clin. Pharmacol. Ther.
E. V. Shusta, Derivation of blood-brain barrier endothelial cells from human pluripotent 94, 636639 (2013).
stem cells. Nat. Biotechnol. 30, 783791 (2012). 55. M. A. Huntley, N. Bien-Ly, R. Daneman, R. J. Watts, Dissecting gene expression at the
33. E. S. Lippmann, A. Al-Ahmad, S. M. Azarin, S. P. Palecek, E. V. Shusta, A retinoic acid-enhanced, blood-brain barrier. Front. Neurosci. 8, 355 (2014).
multicellular human blood-brain barrier model derived from stem cell sources. Sci. Rep. 56. S. Liebner, C. J. Czupalla, H. Wolburg, Current concepts of blood-brain barrier
4, 4160 (2014). development. Int. J. Dev. Biol. 55, 467476 (2011).
34. E. S. Lippmann, A. Al-Ahmad, S. P. Palecek, E. V. Shusta, Modeling the bloodbrain barrier 57. C. Weidenfeller, C. N. Svendsen, E. V. Shusta, Differentiating embryonic neural progenitor
using stem cell sources. Fluids Barriers CNS 10, 2 (2013). cells induce bloodbrain barrier properties. J. Neurochem. 101, 555565 (2007).
35. H. K. Wilson, S. G. Canfield, M. K. Hjortness, S. P. Palecek, E. V. Shusta, Exploring the effects 58. E. S. Lippmann, C. Weidenfeller, C. N. Svendsen, E. V. Shusta, Bloodbrain barrier
of cell seeding density on the differentiation of human pluripotent stem cells to brain modeling with cocultured neural progenitor cellderived astrocytes and neurons.
microvascular endothelial cells. Fluids Barriers CNS 12, 13 (2015). J. Neurochem. 119, 507520 (2011).
36. E. K. Hollmann, A. K. Bailey, A. V. Potharazu, M. D. Neely, A. B. Bowman, E. S. Lippmann, 59. C.-H. Lai, K.-H. Kuo, The critical component to establish in vitro BBB model: Pericyte.
Accelerated differentiation of human induced pluripotent stem cells to bloodbrain Brain Res. Rev. 50, 258265 (2005).
barrier endothelial cells. Fluids Barriers CNS 14, 9 (2017). 60. S. G. Canfield, M. J. Stebbins, B. S. Morales, S. W. Asai, G. D. Vatine, C. N. Svendsen,
37. R. Cecchelli, S. Aday, E. Sevin, C. Almeida, M. Culot, L. Dehouck, C. Coisne, B. Engelhardt, S. P. Palecek, E. V. Shusta, An isogenic bloodbrain barrier model comprising brain
M.-P. Dehouck, L. Ferreira, A stable and reproducible human blood-brain barrier model endothelial cells, astrocytes and neurons derived from human induced pluripotent stem
derived from hematopoietic stem cells. PLOS ONE 9, e99733 (2014). cells. J. Neurochem. 140, 874888 (2017).
38. J. Boyer-Di Ponio, F. El-Ayoubi, F. Glacial, K. Ganeshamoorthy, C. Driancourt, M. Godet, 61. A. D. Ebert, B. C. Shelley, A. M. Hurley, M. Onorati, V. Castiglioni, T. N. Patitucci,
N. Perrire, O. Guillevic, P. O. Couraud, G. Uzan, Instruction of circulating endothelial S. P. Svendsen, V. B. Mattis, J. V. McGivern, A. J. Schwab, D. Sareen, H. W. Kim, E. Cattaneo,
progenitors in vitro towards specialized blood-brain barrier and arterial phenotypes. C. N. Svendsen, EZ spheres: A stable and expandable culture system for the generation
PLOS ONE 9, e84179 (2014). of pre-rosette multipotent stem cells from human ESCs and iPSCs. Stem Cell Res. 10,
39. K. Yamamizu, M. Iwasaki, H. Takakubo, T. Sakamoto, T. Ikuno, M. Miyoshi, T. Kondo, 417427 (2013).
Y. Nakao, M. Nakagawa, H. Inoue, J. K. Yamashita, In vitro modeling of blood-brain barrier 62. B. K. Gage, T. D. Webber, T. J. Kieffer, Initial cell seeding density influences pancreatic
with human iPSC-derived endothelial cells, pericytes, neurons, and astrocytes via endocrine development during in vitro differentiation of human embryonic stem cells.
notch signaling. Stem Cell Reports 8, 634647 (2017). PLOS ONE 8, e82076 (2013).
40. H. Minami, K. Tashiro, A. Okada, N. Hirata, T. Yamaguchi, K. Takayama, H. Mizuguchi, 63. H. Lu, L. Guo, M. J. Wozniak, N. Kawazoe, T. Tateishi, X. Zhang, G. Chen, Effect of cell
K. Kawabata, Generation of brain microvascular endothelial-like cells from human density on adipogenic differentiation of mesenchymal stem cells. Biochem. Biophys. Res.
induced pluripotent stem cells by co-culture with C6 glioma cells. PLOS ONE 10, Commun. 381, 322327 (2009).
e0128890 (2015). 64. J. J. Otero, W. Fu, L. Kan, A. E. Cuadra, J. A. Kessler, b-Catenin signaling is required for
41. W. Risau, H. Wolburg, Development of the blood-brain barrier. Trends Neurosci. 13, neural differentiation of embryonic stem cells. Development 131, 35453557 (2004).
174178 (1990). 65. H. Kurz, Cell lineages and early patterns of embryonic CNS vascularization. Cell Adh. Migr.
42. T. Era, N. Izumi, M. Hayashi, S. Tada, S. Nishikawa, S.-I. Nishikawa, Multiple mesoderm 3, 205210 (2009).
subsets give rise to endothelial cells, whereas hematopoietic cells are differentiated only 66. S. M. Chambers, C. A. Fasano, E. P. Papapetrou, M. Tomishima, M. Sadelain, L. Studer,
from a restricted subset in embryonic stem cell differentiation culture. Stem Cells 26, Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD
401411 (2008). signaling. Nat. Biotechnol. 27, 275280 (2009).
43. S. Liebner, M. Corada, T. Bangsow, J. Babbage, A. Taddei, C. J. Czupalla, M. Reis, A. Felici, 67. J. A. Selekman, N. J. Grundl, J. M. Kolz, S. P. Palecek, Efficient generation of functional
H. Wolburg, M. Fruttiger, M. M. Taketo, H. von Melchner, K. H. Plate, H. Gerhardt, epithelial and epidermal cells from human pluripotent stem cells under defined
E. Dejana, Wnt/b-catenin signaling controls development of the bloodbrain barrier. conditions. Tissue Eng. Part C Methods 19, 949960 (2013).
J. Cell Biol. 183, 409417 (2008). 68. E. S. Lippmann, M. C. EstevezSilva, R. S. Ashton, Defined human pluripotent stem cell
44. R. Paolinelli, M. Corada, L. Ferrarini, K. Devraj, C. Artus, C. J. Czupalla, N. Rudini, culture enables highly efficient neuroepithelium derivation without small molecule
L. Maddaluno, E. Papa, B. Engelhardt, P. O. Couraud, S. Liebner, E. Dejana, Wnt activation inhibitors. Stem Cells 32, 10321042 (2014).

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 11 of 12


SCIENCE ADVANCES | RESEARCH ARTICLE

69. A. Frey, B. Meckelein, H. Weiler-Gttler, B. Mckel, R. Flach, H. G. Gassen, Pericytes of M. P. Murphy, S. Rafii, H. E. Broxmeyer, M. C. Yoder, Differentiation of human pluripotent
the brain microvasculature express gglutamyl transpeptidase. Eur. J. Biochem. 202, 421429 stem cells to cells similar to cord-blood endothelial colonyforming cells. Nat. Biotechnol.
(1991). 32, 11511157 (2014).
70. S. Nakagawa, M. A. Deli, H. Kawaguchi, T. Shimizudani, T. Shimono, . Kittel, K. Tanaka, 79. M. H. Schulz, D. R. Zerbino, M. Vingron, E. Birney, Oases: Robust de novo RNA-seq
M. Niwa, A new bloodbrain barrier model using primary rat brain endothelial cells, assembly across the dynamic range of expression levels. Bioinformatics 28, 10861092
pericytes and astrocytes. Neurochem. Int. 54, 253263 (2009). (2012).
71. S. Nakagawa, M. A. Deli, S. Nakao, M. Honda, K. Hayashi, R. Nakaoke, Y. Kataoka, M. Niwa,
Pericytes from brain microvessels strengthen the barrier integrity in primary cultures Acknowledgments: We acknowledge the use of the Illumina RNA Seq Services of the
of rat brain endothelial cells. Cell. Mol. Neurobiol. 27, 687694 (2007). University of Wisconsin-Madison Biotechnology Center. Funding: This work was supported
72. J. Yu, M. A. Vodyanik, K. Smuga-Otto, J. Antosiewicz-Bourget, J. L. Frane, S. Tian, J. Nie,
by the NIH (grants R21 NS085351, R01 NS083688, and R01 EB007534) and the Takeda
G. A. Jonsdottir, V. Ruotti, R. Stewart, I. I. Slukvin, J. A. Thomson, Induced pluripotent stem
Pharmaceuticals New Frontier Science Program. Author contributions: The project was
cell lines derived from human somatic cells. Science 318, 19171920 (2007).
conceived by T.Q., E.V.S., and S.P.P. The experiments were designed by T.Q., E.V.S., and S.P.P.
73. J. Yu, K. Hu, K. Smuga-Otto, S. Tian, R. Stewart, I. I. Slukvin, J. A. Thomson, Human induced
and were carried out by T.Q., S.E.M., S.G.C., X.B., and W.R.O. The manuscript was written
pluripotent stem cells free of vector and transgene sequences. Science 324, 797801
by T.Q., E.V.S., and S.P.P. Competing interests: T.Q., E.V.S., and S.P.P. are authors on a patent
(2009).
application related to this work (U.S. patent application no. 15/478,463, filed 4 April 2017,
74. T. E. Ludwig, V. Bergendahl, M. E. Levenstein, J. Yu, M. D. Probasco, J. A. Thomson,
published 5 October 2017). All other authors declare that they have no competing interests.
Feeder-independent culture of human embryonic stem cells. Nat. Methods 3, 637646
Data and materials availability: The final analyzed data and raw FASTQ files were
(2006).
submitted to GEO with the accession number GSE97575 (www.ncbi.nlm.nih.gov/geo/query/
75. A. Pipparelli, Y. Arsenijevic, G. Thuret, P. Gain, M. Nicolas, F. Majo, ROCK inhibitor
acc.cgi?acc=GSE97575). All data needed to evaluate the conclusions in the paper are present
enhances adhesion and wound healing of human corneal endothelial cells.
in the paper and/or the Supplementary Materials. Additional data related to this paper
PLOS ONE 8, e62095 (2013).
may be requested from the authors.
76. B. R. Dye, D. R. Hill, M. A. H. Ferguson, Y.-H. Tsai, M. S. Nagy, R. Dyal, J. M. Wells,

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


C. N. Mayhew, R. Nattiv, O. D. Klein, E. S. White, G. H. Deutsch, J. R. Spence, In vitro
generation of human pluripotent stem cell derived lung organoids. eLife 4, e05098 Submitted 19 May 2017
(2015). Accepted 11 October 2017
77. A. M. B. Tadeu, S. Lin, L. Hou, L. Chung, M. Zhong, H. Zhao, V. Horsley, Transcriptional Published 8 November 2017
profiling of ectoderm specification to keratinocyte fate in human embryonic stem cells. 10.1126/sciadv.1701679
PLOS ONE 10, e0122493 (2015).
78. N. Prasain, M. R. Lee, S. Vemula, J. L. Meador, M. Yoshimoto, M. J. Ferkowicz, A. Fett, Citation: T. Qian, S. E. Maguire, S. G. Canfield, X. Bao, W. R. Olson, E. V. Shusta, S. P. Palecek,
M. Gupta, B. M. Rapp, M. R. Saadatzadeh, M. Ginsberg, O. Elemento, Y. Lee, Directed differentiation of human pluripotent stem cells to blood-brain barrier endothelial
S. L. Voytik-Harbin, H. M. Chung, K. S. Hong, E. Reid, C. L. ONeill, R. J. Medina, A. W. Stitt, cells. Sci. Adv. 3, e1701679 (2017).

Qian et al., Sci. Adv. 2017; 3 : e1701679 8 November 2017 12 of 12


Directed differentiation of human pluripotent stem cells to blood-brain barrier endothelial cells
Tongcheng Qian, Shaenah E. Maguire, Scott G. Canfield, Xiaoping Bao, William R. Olson, Eric V. Shusta and Sean P.
Palecek

Sci Adv 3 (11), e1701679.


DOI: 10.1126/sciadv.1701679

Downloaded from http://advances.sciencemag.org/ on November 12, 2017


ARTICLE TOOLS http://advances.sciencemag.org/content/3/11/e1701679

SUPPLEMENTARY http://advances.sciencemag.org/content/suppl/2017/11/06/3.11.e1701679.DC1
MATERIALS

REFERENCES This article cites 77 articles, 19 of which you can access for free
http://advances.sciencemag.org/content/3/11/e1701679#BIBL

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Use of this article is subject to the Terms of Service

Science Advances (ISSN 2375-2548) is published by the American Association for the Advancement of Science, 1200 New
York Avenue NW, Washington, DC 20005. 2017 The Authors, some rights reserved; exclusive licensee American
Association for the Advancement of Science. No claim to original U.S. Government Works. The title Science Advances is a
registered trademark of AAAS.

Você também pode gostar