Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

EMBRYONIC STEM CELLS/INDUCED

PLURIPOTENT STEM CELLS


iPSC Transplantation Increases Regeneration and
Functional Recovery After Ischemic Stroke in
Neonatal Rats
MONICA J. CHAU,a TODD C. DEVEAU,a MINGKE SONG,a XIAOHUAN GU,a DONGDONG CHEN,a
LING WEIa,b
Key Words. iPSCs • Neuronal differentiation • Ischemic stroke • Trophic factors

a ABSTRACT
Department of
Anesthesiology; bDepartment Limited treatments are available for perinatal/neonatal stroke. Induced pluripotent stem cells
of Neurology, Emory
(iPSCs) hold therapeutic promise for stroke treatment, but the benefits of iPSC transplantation
University School of
in neonates are relatively unknown. We hypothesized that transplanted iPSC-derived neural pro-
Medicine, Atlanta, Georgia,
genitor cells (iPSC-NPCs) would increase regeneration after stroke. Mouse pluripotent iPSCs
USA.
were differentiated into neural progenitors using a retinoic acid protocol. Differentiated neural
Correspondence: Ling Wei, MD, cells were characterized by using multiple criteria and assessments. Ischemic stroke was induced
Department of Anesthesiology, in postnatal day 7 (P7) rats by occluding the right middle cerebral artery and right common
101 Woodruff Cir, Suite 617, carotid artery. iPSC-NPCs (400,000 in 4 ml) were transplanted into the penumbra via intracranial
Emory University School of injection 7 days after stroke. Trophic factor expression in the peri-infarct tissue was measured
Medicine, Atlanta, Georgia using Western blot analysis. Animals received daily bromodeoxyuridine (BrdU) injections and
30322, USA. Telephone: 404- were sacrificed 21 days after stroke for immunohistochemistry. The vibrissae-elicited forelimb
718-8661; Fax. 404-712-1351;
e-mail: lwei7@emory.edu
placement test was used to evaluate functional recovery. Differentiated iPSCs expressed mature
neuronal markers, functional sodium and potassium channels, and fired action potentials. Sev-
Received December 18, 2013; eral angiogenic and neurogenic trophic factors were identified in iPSC-NPCs. Animals that
accepted for publication July 23, received iPSC-NPC transplantation had greater expression of stromal cell-derived factor 1-a
2014; first published online in (SDF-1a) and vascular endothelial growth factor (VEGF) in the peri-infarct region. iPSC-NPCs
STEM CELLS EXPRESS August 5, stained positive for neuronal nuclei (NeuN) or glial fibrillary acidic protein (GFAP) 14 days after
2014. transplantation. iPSC-NPC-transplanted animals showed greater numbers of BrdU/NeuN and
C AlphaMed Press
V
BrdU/Collagen IV colabeled cells in the peri-infarct area compared with stroke controls and per-
1066-5099/2014/$30.00/0 formed better in a sensorimotor functional test after stroke. iPSC-NPC therapy may play multi-
ple therapeutic roles after stroke by providing trophic factors, increasing angiogenesis and
http://dx.doi.org/ neurogenesis, and providing new cells for tissue repair. STEM CELLS 2014;32:3075–3087
10.1002/stem.1802

INTRODUCTION such as cognitive impairment, spasticity, or


death [4]. Children who suffer a perinatal stroke
Ischemic neonatal/perinatal stroke is a devastat- also are at risk of delayed effects such as seiz-
ing disease. In the United States, perinatal ures. Over 25% of perinatal stroke survivors
stroke occurs at a rate of 1 of 3500 live births develop a seizure within 3 years [6]. The medical
each year [1, 2]. The most common cause of
cost of perinatal stroke is a financial strain to
arterial ischemic stroke in children ages 0–15
patients and society [7].
years is cerebral arteriopathy, which comprises
The postnatal brain exhibits endogenous
more than half of all cases [1, 3]. Among the regeneration including neurogenesis and
causes of neonatal ischemic stroke are angiopa- angiogenesis after ischemic stroke [8–11]. Neu-
thies and thromboembolism from an intracranial ral progenitors are generated in the subven-
or extracranial vessel. There are few treatments tricular zone (SVZ) [12, 13]. As a physiological
available for ischemic stroke. Tissue plasminogen process, SVZ cells migrate to the olfactory
activator (tPA), a thrombolytic agent, is the only bulb via the rostral migratory stream to differ-
Food and Drug Administration-approved drug entiate into granule interneurons [14]. After
for the treatment of ischemic stroke in adults ischemic stroke, chemoattractants such as SDF-
[4]. The efficacy and safety of tPA in children is 1a in the injury are upregulated and divert
unknown. Currently, children with thrombosis neural progenitors from the rostral migratory
are treated with anticoagulation using unfractio- stream toward the ischemic infarct [13, 15,
nated or low-molecular-weight heparin [5]. Peri- 16]. Cerebral ischemia dramatically increases
natal stroke can result in irreversible outcomes SVZ neural progenitor cell proliferation,

STEM CELLS 2014;32:3075–3087 www.StemCells.com C AlphaMed Press 2014


V
3076 Regeneration and Functional Recovery by iPSCs in Rats

Figure 1. Experimental timeline. This experimental timeline delineates the in vitro and in vivo studies. Induced pluripotent stem cells
(iPSCs) were differentiated in suspension culture with the retinoic acid protocol. Neural progenitors were harvested and were either
plated for terminal differentiation or transplanted into the neonatal stroke rats. Abbreviation: RA, retinoic acid.

migration to the infarct, and differentiation into neurons [9, In contrast to multipotent bone marrow mesenchymal stem
17–21]. However, a large proportion of these progenitors and cells (BMSCs), iPSCs can be differentiated into cells of any of
new neurons do not survive upon reaching the injury site due the three primary germ layers [29] including neurons and glia
to the inflammatory and cytotoxic microenvironment sur- [30–32]. Although iPSCs have great therapeutic potential, a pro-
rounding the stroke [22]. Thus, the endogenous regenerative file of the major trophic factors expressed by iPSC-neural pro-
activities and their abilities in tissue repair are limited. genitor cells (NPCs) has not been delineated. Furthermore, their
In the field of regenerative medicine, transplanting exoge- regenerative abilities after transplantation into a neonatal ische-
nous cells such as neural progenitors derived from iPSCs has mic brain have not been investigated. This study is the first to
exhibited promise for enhanced tissue repair and functional examine the regenerative properties of neural progenitors
recovery after ischemic stroke [23–25]. iPSCs are pluripotent derived from iPSCs in a neonatal ischemic stroke model.
stem cells generated from adult somatic cells [26, 27]. They
were first created via the upregulation of four transcription
factors, Oct3/4, Sox2, c-Myc, and Klf4 [27, 28]. The use of MATERIALS AND METHODS
iPSCs circumvents ethical issues surrounding the use of
human embryonic stem cells (ESCs). iPSCs also have great iPSC Culture and Differentiation
potential for personalized medicine in which pluripotent stem The experimental timeline is illustrated in Figure 1, showing
cells can be generated using one’s own somatic cells such as neural differentiation of iPSCs in vitro relative to the timing of
skin fibroblasts. Autologous transplantation of these cells in vivo transplantation. Pluripotent mouse primary iPSCs (WP5
would avoid immune rejection after transplantation. line, Stemgent Cambridge, MA, https://www.stemgent.com/)

C AlphaMed Press 2014


V STEM CELLS
Chau, Deveau, Song et al. 3077

were maintained in their pluripotent state in culture medium ized with a 0.2% Triton-X 100 solution, and blocked with 1%
consisting of 15% ESC fetal bovine serum (ESC FBS, Gibco Life cold fish gelatin (Sigma, St. Louis, MO, http://www.sigmaal-
Technologies, Grand Island, NY, http://www.lifetechnologies. drich.com/united-states.html). Cells were incubated overnight
com/us/en/home/brands/gibco.html) in Dulbecco’s modified at 4 C with primary antibodies for Oct3/4 (sc-5279, 1:200;
Eagle Medium (Corning Cellgro, Manassas, VA, http://www. Santa Cruz Biotechnology, Santa Cruz, CA, http://www.scbt.
cellgro.com/). This media was supplemented with 1% nones- com/), Sox2 (sc-17320, 1:200; Santa Cruz Biotechnology), dou-
sential amino acids, 1% penicillin–streptomycin, 0.1% beta- blecortin (DCX, sc-8066, 1:100; Santa Cruz Biotechnology,
mercaptoethanol, and 1:10,000 leukemia inhibitory factor http://www.scbt.com/), NeuN (MAB377 1:200; Millipore, Bill-
(LIF). Cells were either passaged daily with trypsin-EDTA when erica, MA http://www.emdmillipore.com/US/en), b-III Tubulin
cells were approximately 80% confluent or media was (Tuj-1, MMS-435P, 1:200; Covance, http://www.covance.com/,
changed if the cells were not confluent enough for passage. Princeton, NJ), synaptosomal-associated protein 25 (SNAP-25,
Pluripotent cell cultures were maintained on mouse embry- AB5871P, 1:100; Millipore, Billerica, MA http://www.emdmilli-
onic fibroblast (MEF) feeder layers (Millipore, Billerica, MA, pore.com/US/en), synapsin 1 (51–5200, 1:200; Life Technolo-
http://www.emdmillipore.com/US/en) for about six passages gies, Grand Island, NY https://www.lifetechnologies.com/us/
after the initial thawing. The cultures were maintained at later en/home.html), neurofilament (AB9568, 1:100; Millipore Bill-
passages in 0.15% gelatin-coated flasks. iPSCs were differenti- erica, MA http://www.emdmillipore.com/US/en), and mCherry
ated in suspension culture with a “42/41” (4 days without/4 (AB167453, 1:100; Abcam, Cambridge, MA, http://www.
days with) retinoic acid (RA) (all trans-RA, Sigma, St. Louis, abcam.com/). To analyze trophic factors, iPSC-NPCs were incu-
MO, http://www.sigmaaldrich.com/united-states.html) proto- bated overnight with primary antibodies at 1:100 for fibro-
col in LIF-free media [33, 34]. Cells were dissociated from blast growth factor (FGF) (sc-7375; Santa Cruz Biotechnology,
flasks with trypsin without EDTA. iPSCs were plated in petri Dallas, TX http://www.scbt.com/), brain-derived neurotrophic
dishes in growth media without LIF in suspension culture. factor (BDNF) (sc-546; Santa Cruz Biotechnology, Dallas, TX
Within the first day, the cells formed embryoid bodies in sus- http://www.scbt.com/), erythropoietin (EPO) (sc-1310; Santa
pension culture. In the last 4 days, 5 3 1027 M of all-trans Cruz Biotechnology, Dallas, TX http://www.scbt.com/), stromal
RA was added to the media. Neural progenitors were har- cell-derived factor 1-a (SDF-1a) (MAB350; R&D systems, Min-
vested by dissociating cells with trypsin-EDTA for 10 minutes, neapolis, MN, http://www.rndsystems.com/), and glial cell-
inactivated with serum media, and run through a cheesecloth dervied neurotrophic factor (GDNF) (sc-328; Santa Cruz Bio-
filter to separate the cells. Cells were centrifuged and resus- technology, Dallas, TX http://www.scbt.com/). After incuba-
pended to a concentration of 100,000 cells/ll and were either tion, cells were washed with PBS three times for 5 minutes
transplanted immediately into the rats after stroke or plated each. Secondary antibodies were applied at 1:100 for 1 hour
in modified SATO media [35] on Poly-D-Lysine (PDL)/Laminin- at room temperature for Oct3/4 and SDF-1a, neuronal nuclei
coated dishes and allowed to terminally differentiate for at (NeuN), Tuj-1 (donkey anti-mouse Cy3, Jackson ImmunoRe-
least 5 days (Fig. 1). In vitro studies were performed in paral- search, West Grove, PA, http://www.jacksonimmuno.com/),
lel with transplantation to monitor the quality and differentia- doublecortin (DCX) (donkey anti-goat Cy3, Jackson ImmunoRe-
tion of the transplanted cells. search, West Grove, PA, http://www.jacksonimmuno.com/),
In studies visualizing iPSC-NPC transplantation and differen- GDNF (donkey anti-rabbit Cy5, Jackson ImmunoResearch, West
tiation in vivo, we used iPSCs that were genetically modified to Grove, PA http://www.jacksonimmuno.com/), sex determining
express the fluorescent protein, mCherry. Briefly, the mCherry region Y-box 2 (Sox2), FGF, EPO, synaptosomal-associated
expression vector was created by subcloning the mCherry coding protein 25 (SNAP-25) (AlexaFluor 488, donkey anti-goat; Invi-
sequence (Addgene Cambridge, MA http://www.addgene.org trogen Life Technologies, Grand Island, NY, https://www.life-
plasmid #20943, kind gift of Dr. Karl Deisseroth) [35] in place of technologies.com/us/en/home.html), and BDNF,
the Nanog coding sequencing of a backbone used previously in neurofilament, synapsin 1, and mCherry (donkey anti-rabbit
ESC studies (Addgene Cambridge, MA http://www.addgene.org Cy3, Jackson ImmunoResearch, West Grove, PA http://www.
plasmid #13838, kind gift of Dr. Shinya Yamanaka) [36]. A stable jacksonimmuno.com/). Dishes were washed with PBS and
mCherry iPSC line was created by transfecting the mCherry stained with Hoechst 33342 (1:20,000), washed, and cover-
expression vector into pluripotent iPSCs using Lipofectamine slipped with Vectashield mounting media (Vector Laboratories,
2000 (Life Technologies, Grand Island, NY, https://www.lifetech- Burlingame, CA, https://www.vectorlabs.com/).
nologies.com/us/en/home.html). Forty-eight hours after trans-
fection, puromycin (0.5–1 mg/ml, Sigma) was used to select Electrophysiological Characterization
stable clones expressing mCherry that were pooled and main- Whole-cell patch clamp recording was performed on mouse
tained on MEFs. We observed mCherry expression through at iPSC-derived neurons 10 days after neural progenitor harvest
least 30 passages as well as after cell freezing and recovery. using an EPC9 amplifier (HEKA, Elektronik, Lambrecht, Germany,
These cells underwent the same maintenance, differentiation, http://www.heka.com/) at room temperature. The recording
and transplantation protocol as cells without the mCherry external solution contained (mM) 135 NaCl, 5 KCl, 1 MgCl2, 2
marker. CaCl2, 10 HEPES, and 10 Glucose at a pH of 7.4. Recording elec-
trodes were pulled from borosilicate glass pipettes (Sutter
Immunocytochemistry Instrument, Novato, CA http://www.sutter.com/) and had a tip
Cells were fixed with 4% paraformaldehyde for 15 minutes resistance between 5 and 8 MX when filled with the internal
then washed three times for 5 minutes each with phosphate- solution (mM): 140 KCl, 2 MgCl2, 1 CaCl2, 2 Na2ATP, 10 EGTA,
buffered saline (PBS) after each protocol step. Cells were and 10 HEPES at a pH of 7.2. Series resistance was compensated
treated with 220 C ethanol/acetic acid solution, permeabil- by 60–80%. Linear leak and residual capacitance currents were

www.StemCells.com C AlphaMed Press 2014


V
3078 Regeneration and Functional Recovery by iPSCs in Rats

subtracted on-line using a P/6 protocol. Action potentials (APs) gery commenced when the rat showed no response to
were recorded (HEKA, Elektronik, Lambrecht, Germany http:// noxious stimuli. The right common carotid artery and the dis-
www.heka.com/) under current-clamp mode using Pulse soft- tal branches of right middle cerebral artery were permanently
ware (HEKA Elektronik Lambrecht, Germany http://www.heka. occluded via cauterization to induce ischemia in the right
com/). Tetrodotoxin (TTX, 1lM) was used to block voltage- whisker barrel cortex. Sham animals received skin incisions,
gated inward sodium currents. Data were filtered at 3 KHz and without arterial occlusion. Starting 3 days after stroke, animals
digitized at sampling rates of 20 KHz. AP amplitude was deter- received daily intraperitoneal (i.p.) injection of bromodeoxyur-
mined by measurement from the initial threshold to the peak idine (BrdU; Sigma St. Louis, MO http://www.sigmaaldrich.
of AP upstroke. com/united-states.html) to label any proliferating cells in the
brain. Pups were given 1/3 the adult BrdU dosage (16.7 mg/
Reverse Transcription PCR on iPSC-NPCs kg) until their body weight reached 30 g (usually P15) when
On the final day of the differentiation protocol (day 8) when they received the adult dose of 50 mg/kg.
iPSCs had differentiated into neural progenitors, total mRNA Seven days after stroke induction, animals received iPSC-
was extracted from the cells with Trizol reagent (Invitrogen NPC transplantation. Under 4% chloral hydrate anesthesia, the
Life Technologies Grand Island, NY https://www.lifetechnolo- skull over the penumbra was thinned by a dental drill. Ische-
gies.com/us/en/home/brands/invitrogen.html). For each dish, mic tissue was visually identified through the skull. 400,000
250 ll of Trizol was used to lyse the cells. Choloroform (50 iPSC-NPCs were delivered through 4 3 1 ll injections at four
ll) was added and samples were centrifuged. The RNA was sites in the peri-infarct region using a Hamilton syringe (Reno,
collected from the upper aqueous phase and precipitated NV http://www.hamiltoncompany.com/Syringes/). Control ani-
with 125 ll of isopropyl alcohol. RNA was centrifuged into a mals received 4 3 1 ll injections of SATO media at four sites
pellet and washed two times with 75% ethanol. RNA pellets as a vehicle control. The incision was closed with surgical
were air-dried and resuspended in DEPC-treated water and adhesive. To kill the animals, they were anesthetized and
RNA concentration was measured. The high capacity RNA-to- decapitated. The brain was dissected from the cranium, a 5-
cDNA kit (Life Technologies, Grand Island, NY) was used to mm coronal section that included the injury or a flattened
create cDNA from RNA samples. Polymerase chain reaction cortex of the injured side was flash frozen on dry ice in OCT
(PCR) analysis has a mixture of Taq buffer (New England Biol- mounting media.
abs, Ipswich, MA, https://www.neb.com/), forward primer,
reverse primer, dNTP (10 mM), Taq polymerase, water, and Western Blot on iPSC-NPCs and Peri-Infarct Brain
cDNA. Primer pairs for trophic factors (mouse) were designed Tissue
from the mRNA sequence found in the NCBI nucleotide bank. To examine trophic factors and receptors expressed by neural
The primer pairs are listed below. DNA samples were run out progenitors in culture, neural progenitors were collected after
in a 1.8% agarose gel and band intensity was quantified using RA differentiation. To analyze trophic factors in the peri-infarct
ImageJ software (NIH, Bethesda, MD, http://rsbweb.nih.gov/ij/ tissue after cell transplantation, tissue was dissected from the
download.html). peri-infarct area immediately after killing and total protein was
extracted using lysis buffer. Cells or tissues were homogenized
Primer Pairs. 18S: GACTCAACACGGGAAACCTC (forward), with lysis buffer containing protease inhibitor (1:100). The bicin-
ATGCCAGAGTCTCGTTCGTT (reverse) choninic acid assay was used to measure and normalize total
BDNF: CGACATCACTGGCTGACACT (forward), ATGTTTGCGGC protein concentrations for each sample. Protein samples for gel
ATCCAGGTA (reverse) CXCR4: GCCATGGCTGACTGGTACTT (for- electrophoresis were created by combining the original protein
ward), CACCCACATAGACGGCCTTT (reverse) solution with water and 5X loading buffer. Samples were boiled
EPO: ACCACCCCACCTGCTCCACTC (forward), GTTCGTCGGTC for 10 minutes at 95 C. The samples were run on a 7.5–20%
CACCACGGT (reverse) gradient acrylamide gel. For culture dishes, we used antibodies
FGF1: GTGGATGGGACAAGGGACAG (forward), GGTGTCTG for vascular endothelial growth factor (VEGF) (05–443; Millipore,
CGAGCCGTATAA (reverse) Billerica, MA, www.emdmillipore.com/US/en), VEGFR-2 (SC315;
GDNF: CGCTGACCAGTGACTCCAAT (forward), CTCTGCGACC Santa Cruz, Dallas, Texas, http://www.scbt.com/), VEGFR-3
TTTCCCTCTG (reverse) (AB1875; Millipore, Billerica, MA, http://www.emdmillipore.com/
SDF-1a: GCTCTGCATCAGTGACGGTA (forward), CCAGGTACT US/en), BDNF (SC546; Santa Cruz, Dallas, Texas, http://www.
CTTGGATCCAC (reverse) scbt.com/), Angiopoietin-3 (Ang3, PC671; Oncogene Research
VEGF: CTCACCAAAGCCAGCACATA (forward), AAATGCTTTCT Products, Cambridge, MA, http://www.oncogene.com/), and
CCGCTCTGA (reverse) SDF-1a (MAB350; R&D Systems, Minneapolis, MN, http://www.
rndsystems.com/). For tissue samples, antibodies probed for
Focal Ischemia Surgery and Cell Transplantation VEGF (05–443, 1:1000; Millipore, Billerica, MA, http://www.emd-
Wistar rat pups and dam were housed in a climate-controlled millipore.com/US/en), SDF-1a (MAB350, 1:500; R&D Systems,
room with a 12-hour light–dark cycle. Animals had free access Minneapolis, MN, http://www.rndsystems.com/), b-actin (A5441
to water and food. Pups received experimental treatment 1:5000; Sigma, St. Louis, MO, http://www.sigmaaldrich.com/
from ages P7 to P28 and were sacrificed by 4% chloral united-states.html), and tubulin (Cell signaling, Boston, MA,
hydrate (10 ml/kg) before decapitation. All animal procedures http://www.cellsignal.com/). For cultures, blots were incubated
were approved by the Emory University Institutional Animal in AP-conjugated secondary antibody anti-mouse, anti-rabbit, or
Care and Use Committee (IACUC). anti-goat (1:1000; Promega, Madison, WI, http://www.promega.
To induce focal cerebral ischemia, P7 rat pups were anes- com/). Tissue SDF-1a and VEGF blots were incubated in AP-
thetized with hypothermia as described previously [36]. Sur- conjugated secondary antibody anti-mouse (1:1000; Promega,

C AlphaMed Press 2014


V STEM CELLS
Chau, Deveau, Song et al. 3079

Madison, WI http://www.promega.com/) at room temperature, the injury have been stimulated. Animals were trained two
washed and developed with NBT/BCIP solution, and then times on each side before testing. Each animal was tested for
washed with water to stop the reaction. All blots were quanti- 6–10 times on each side and the percentage of successful
fied with Image J (NIH). Values for each band were normalized reaches was calculated.
against a b-actin or tubulin loading control.
Statistical Analysis
Statistical analyses were performed using Prism Graphpad
Immunohistochemistry (GraphPad Software, Inc., La Jolla, CA, http://www.graphpad.
In our in vivo analyses, fresh frozen brains were sectioned at com/scientific-software/prism/). Single comparisons were per-
20 lm on a cryostat microtome at 220 C using design-based formed using Student’s t-test. One- and two-way analysis of var-
stereology in which every 10th section was collected such iance (ANOVA) followed by Bonferroni’s post hoc analysis was
that two adjacent tissues were more than 200 lm apart. This used for multiple comparisons. Significance was identified by a
method avoided double-counting a cell in adjacent sections. p < .05. SEM was reported with the means.
Sectioned tissues were fixed with 10% buffered formalin and
treated with a 220 C ethanol/acetic acid solution. Tissue was
permeabilized with a 0.2% Triton-X 100 solution, and blocked RESULTS
for nonspecific binding with 1% fish gel. Tissues and cells
were washed three times with PBS between each step. Tis- iPSCs Differentiation into Functional Neurons In Vitro
sues were incubated overnight at 4 C with primary antibodies iPSCs in the expansion stage stained positive for pluripotent
for BrdU (OBT0030G, 1:400; AbD Serotec, Hercules, CA http:// markers, Sox2 and Oct3/4 (Fig. 2A–2F). iPSCs were maintained
www.abdserotec.com/), NeuN (MAB377 1:200; Millipore, Bill- on MEF feeder layers (Fig. 2G), expanded in feeder free condi-
erica, MA http://www.emdmillipore.com/US/en), Collagen IV tions on 0.2% gelatin-coated flasks (diluted from 2% gelatin
(AB769, 1:200; Millipore, Billerica, MA http://www.emdmilli- solution, Sigma-Aldrich, St. Louis MO, http://www.sigmaal-
pore.com/US/en), and glial fibrillary acidic protein (GFAP) drich.com), and were differentiated in suspension culture into
(MAB360, 1:200; Millipore Billerica, MA http://www.emdmilli- neural progenitors by a 42/41 RA differentiation protocol
pore.com/US/en). After washing with PBS, secondary antibod- [33]. The pluripotent cells in suspension formed embryoid
ies conjugated to fluorophores were used to visualize 5- bodies 1 day after differentiation and remained in aggregates
bromo-20 -deoxyuridine (BrdU) (donkey anti-rat Cy3, 1:300; throughout the differentiation process until harvest at day 8
Jackson ImmunoResearch, West Grove, PA http://www.jackso- (Fig. 2H). After harvesting aggregates, cells were plated on
nimmuno.com/), NeuN (donkey anti-mouse Cy5, 1:200), Colla- poly-D-lysine PDL/laminin-coated dishes. Cell counts at 1 day
gen IV (donkey anti-goat AlexaFluor 488, 1:200; Invitrogen Life after harvest indicated that 96.9% of cells expressed the neu-
Technologies Grand Island, NY http://www.lifetechnologies. ral progenitor marker, DCX (DCX-positive cells/total Hoechst
com/us/en/home.html), and GFAP (donkey anti-mouse Cy3, cells) (Fig. 2I, 2J). Five days after harvest, cells exhibited neu-
1:200; Jackson ImmunoResearch, West Grove, PA http://www. ronal morphology including processes projecting from the cell
jacksonimmuno.com/home/contact.asp). The secondary anti- body (Fig. 2K, 2L).
bodies were applied and incubated on the tissue for 1 hour To verify the formation of mature neurons and neuronal
at room temperature and washed three times with PBS. activity, we performed electrophysiological recordings on
Stained tissues and cells were analyzed under fluorescent these cells (Fig. 3A). Whole-cell patch clamp recordings were
microscopy. Pictures from six areas in the peri-infarct area performed 10 days after harvest. Differentiated cells exhibited
(based on the border between live and dead NeuN-positive neuronal functionality with action potentials triggered by a
cells adjacent to the ischemic core) were taken at 40X. Six tis- 30-lA current injection (average amplitude 5 53.31612.77
sue sections from each animal were quantified. NeuN/BrdU mV, n 5 7 cells; Fig. 3A-1). Inward Na1 current and outward
colabeled and Collagen IV/BrdU colabeled cells were counted K1 current were elicited by step voltage changes from a hold-
per animal. ing potential of 270 mV to depolarized levels (240 to 150
mV in 10 mV decrement; Fig. 3A). The inward Na1 current
Vibrissae-Elicited Forelimb Placement Behavior Test was highly sensitive to the specific channel blocker, tetrodo-
The vibrissae-elicited forelimb placement test is a test to toxin (TTX, 1 lM; Fig. 3A). To confirm differentiation into neu-
directly assay whisker somatosensation and forepaw motor ronal cells, we stained the cells for the mature neuronal
function [37, 38]. It assesses the ability for the whiskers of markers, Tuj-1, NeuN, and neurofilament (Fig. 3B–3D). At 5
the animal to sense the tactile stimulus of the table edge. If days after harvest, 87.5% of the cells expressed NeuN (NeuN-
the tactile stimulus is felt, animals will stereotypically place positive cells/total Hoechst-labeled cells) (Fig. 3C). The cells
their forelimb on the table. Without impeding the forelimb also expressed synapsin 1 and SNAP-25 (Fig. 3E, 3F).
movement, the rats were scruffed loosely to brush either To confirm that the genetically labeled mCherry iPSCs
right or left whiskers on a table edge. Animals were held in a were able to differentiate into functional neurons in vitro, we
horizontal position and passed from above the table edge to recorded action potentials from the differentiated mCherry
below the table edge once. Upon stimulation of the whiskers, iPSCs (Fig. 3G). Differentiated mCherry iPSCs also stained posi-
normal animals with no brain injury will stereotypically reach tive for NeuN and Tuj-1 (Fig. 3H, 3I).
out to the ledge with their forepaw. Animals with no deficit
will reach out approximately equally for both sides of whisker Trophic Factor Expression Profile of iPSC-NPCs In Vitro
stimulation. Animals with post-stroke deficits will reach out a Using immunocytochemistry, we detected expression of (SDF-
fewer number of times when the whiskers contralateral to 1a), fibroblast growth factor (FGF), glial cell-derived neutrophic

www.StemCells.com C AlphaMed Press 2014


V
3080 Regeneration and Functional Recovery by iPSCs in Rats

Figure 2. Induced pluripotent stem cells (iPSCs) differentiated down the neural lineage. (A–F): iPSCs expressed Sox2 and Oct3/4. (G):
iPSCs were maintained on a feeder layer. (H): Cells in suspension formed embryoid bodies during differentiation. (I, J): The cells
expressed doublecortin (DCX) 1 day after harvest. (K, L): Cells exhibited neuronal morphology including projected processes 5 days after
harvest. Abbreviation: DCX, doublecortin.
factor (GDNF), brain-derived neurotrophic factor (BDNF), and SDF-1a and VEGF in the peri-infarct tissue after ischemic
erythropoitin (EPO) in iPSC-NPCs (Fig. 4A–4G). We verified stroke. Tissue from the peri-infarct area was collected 2 days
trophic factor expression and expression of vascular endothelial after iPSC-NPCs were transplanted. Western blot analysis dem-
growth factor (VEGF) and C-X-C motif receptor 4 (CXCR4) with onstrated that SDF-1a expression in the peri-infarct area
PCR analysis of neural progenitors (Fig. 4H). In addition, the pro- increased after ischemic stroke. Animals with cell transplanta-
tein expression of several angiogenic trophic factors including tion showed a significant increase in SDF-1a expression com-
vascular endothelial growth factor (VEGF), brain-derived neuro- pared with the sham group (Fig. 5A, n 5 4; one-way ANOVA,
trophic factor (BDNF), angiopoietin-3 (Ang3), SDF-1a, and the F(2,9) 5 10.69, p 5 .0042; pairwise comparisons with Bonfer-
receptors for VEGF including vascular endothelial growth factor roni post hoc test showed a significant difference between
receptor-2 (VEGFR-2) and vascular endothelial growth factor sham and stroke1 iPSC, p < .005). Similarly, VEGF expression
receptor-3 (VEGFR-3) were detected using Western blot (Fig. 4I). in the iPSC-NPC transplantation group was significantly greater
than stroke and sham groups (Fig. 5B, n 5 3–4, one-way
Increased Trophic Factor Expression in the Ischemic ANOVA, F(2,8) 5 13.34, p 5 .0028; pairwise comparisons with
Brain After Transplantation Bonferroni post hoc test showed a significant difference
We tested if transplanting iPSC-NPCs into the peri-infarct area between sham and stroke1 iPSC, p < .005, stroke and stro-
would increase the levels of two important trophic factors, ke 1 iPSC, p < .05).

C AlphaMed Press 2014


V STEM CELLS
Chau, Deveau, Song et al. 3081

Figure 3. Induced pluripotent stem cells (iPSCs) differentiated into functional neurons. (A): iPSC-derived neurons exhibited (1) action
potentials, (2) Na1 and K1 currents, and (3) when tetrodotoxin was applied, INa was completely blocked. (B–D): Cells expressed imma-
ture and mature neuronal markers, Tuj-1, NeuN, and neurofilament (E, F) and neuronal synaptic markers and synaptosomal-associated
protein 25 (SNAP-25) (G–I). Differentiated mCherry iPSCs exhibited action potentials and neuronal markers, NeuN and Tuj-1. Abbrevia-
tions: NeuN, Neuronal Nuclei; SNAP-25, synaptosomal-associated protein 25; Tuj, tubulin.

Transplanted iPSC-NPCs Differentiate into Neural Line- (NeuN-positive) and proliferating vessels (Collagen IV-positive
age Cells cells labeling the vessel basement membrane). In stroke only
Animals received mCherry iPSC-NPC transplantation 7 days controls (Fig. 7A–7D) and stroke 1 iPSC-NPC transplanted brains
after stroke. We chose a delayed 7-day time point to avoid (Fig. 7E–7H), we compared the number of colabeled NeuN/
the cytotoxic milieu found in acute and subacute phases of BrdU cells (Fig. 7I–7K) and colabeled Collagen IV/BrdU cells in
ischemia [25, 39]. Much of the inflammatory response and the peri-infarct area (Fig. 7L–7N). There were significantly more
edema subsides by 7 days after stroke [40, 41], which pre- NeuN/BrdU (Fig. 7K, n 5 7–9; Student’s t-test t(14) 5 2.799,
sumably allows for a more hospitable environment for exoge- p 5 .0142) and Collagen IV/BrdU (Fig. 7N, n 5 7–9; Student’s t-
nous cell transplants [41, 42]. We observed that transplanted test t(14) 5 2.258, p 5 .0404) colabeled cells in the iPSC-NPC
mCherry iPSCs differentiated into neural lineage cells at 7 transplantation group compared with the stroke only group.
days after transplantation as indicated by the positive costain-
ing of mCherry with the mature neuronal markers, NeuN and Transplantation of iPSCs Increases Functional Recovery
neurofilament (Fig. 6A–6C). Also, a portion of transplanted
mCherry iPSCs differentiated into GFAP-positive cells as indi- Our stroke model produced a consistent injury in the sensorimotor
cated by the GFAP staining colabeled with mCherry (Fig. 6D). cortex that included the whisker barrel cortex in the neonatal rats
[43]. We analyzed the somatosensory function of sham, stroke,
Transplantation of iPSC-NPCs Increased Endogenous and stroke with iPSC-NPC transplantation animals using the
Neurogenesis and Angiogenesis vibrissae-elicited forelimb placement test 14 days after treatment
Dissected brain tissues were analyzed for neurogenesis and (21 days after the onset of stroke) [38]. Sham animals had a simi-
angiogenesis 21 days after stroke when active endogenous neu- lar (and not significantly different) percentage of successful reaches
ral progenitor proliferation and migration activity peaks post- with their left and right paws. Animals with stroke exhibited a sig-
ischemia [13, 42]. Animals were injected with BrdU (i.p.) start- nificantly lower percentage of successful reaches on the left/
ing 3 days after stroke to label newly formed neuronal cells impaired side compared with shams (Fig. 7O, n 5 7–9; repeated

www.StemCells.com C AlphaMed Press 2014


V
3082 Regeneration and Functional Recovery by iPSCs in Rats

Figure 4. Induced pluripotent stem cells (iPSC)–neural progenitor cells (NPCs) express trophic factors in vitro. (A–G): iPSC-NPCs
expressed SDF-1a, fibroblast growth factor (FGF), GDNF, EPO, and BDNF with immunohistochemistry. (H): Expression of CXCR4, SDF-1a,
vascular endothelial growth factor (VEGF), EPO, BDNF, FGF, and GDNF with polymerase chain reaction analysis of iPSC-NPCs. (I): Western
blot of iPSC-NPCs for VEGF, VEGFR-2, VEGFR-3, BDNF, Ang3, and SDF-1a. Abbreviations: BDNF, Brain-derived Neurotrophic Factor; EPO,
Erythropoietin; FGF, fibroblast growth factor; GDNF, Glial Cell Line-derived Neurotrophic Factor; SDF, Stromal Cell-Derived Factor 1-a;
VEGF, vascular endothelial growth factor.

measures two-way ANOVA shows a significant interaction tissue after transplantation, demonstrated the ability of trans-
F(2,22) 5 3.60, p 5 .0444 and a significant effect of paw side planted iPSC-NPCs to differentiate into neurons in vitro and in
F(1,22) 5 14.54, p 5 .001; pairwise comparisons with the Bonfer- vivo, and demonstrated increased levels of angiogenesis, neu-
roni post hoc test showed a significant difference in the left paw rogenesis and functional recovery in neonatal rats with stroke
when comparing sham and stroke group, p < .05). Animals that and iPSC-NPC transplantation. The benefits of stem cell trans-
had received iPSC-NPC transplantation showed no significant differ- plantation are multifold, involving generation of new neuronal
ence from sham animals (pairwise comparisons with the Bonfer- cells and trophic factor delivery to the injury region.
roni post hoc test showed no significant difference in the left paw To support our hypothesis that iPSCs can differentiate into
neurons to replace lost cells, we demonstrated that Oct3/4 and
when comparing sham and stroke1 iPSC-NPC groups, p > .05).
Sox2-positive pluripotent iPSCs could be differentiated into
DCX-positive neural progenitors with a 96.9% differentiation
rate. Furthermore, harvested iPSC-NPCs could differentiate into
DISCUSSION
neurons expressing several neuronal markers including Tuj-1,
Previous studies have shown that iPSC transplantation into an neurofilament, and NeuN. We confirmed that these neurons
adult ischemic stroke model increased regeneration and func- had functional electrophysiological activity. At 10 days after
tional recovery [24, 25, 39, 44], however the trophic factor harvest, the cells exhibited action potentials, and inward Na1
expression of iPSCs and their regenerative capabilities have and outward K1 currents. The SNAP-25 and synapsin 1 expres-
not been evaluated in neonatal stroke. In the current investi- sion of the differentiated cells suggested that they also have
gation, we characterized the stages of neuronal differentiation the machinery to build synapses and potentially regulate syn-
of iPSCs, profiled major regenerative trophic factors expressed aptic activity within the host tissue. Furthermore, transplanted
by iPSC-NPCs, evaluated the trophic factor levels in the brain iPSC-NPCs were able to differentiate into mature neurons and

C AlphaMed Press 2014


V STEM CELLS
Chau, Deveau, Song et al. 3083

Figure 5. Increased trophic factor expression after transplantation. (A): Tissues were dissected from the peri-infarct area 2 days after
induced pluripotent stem cells (iPSC)–neural progenitor cell (NPC) transplantation. Animals with transplantation showed a significant
increase in SDF-1a expression compared with sham animals (n 5 4, F(2,9) 5 10.69, p 5 .0042). (B): VEGF expression in the iPSC-NPC
transplantation group was significantly greater than stroke only and sham groups (n 5 3–4, F(2,8) 5 13.34, p 5 .0028). Abbreviations:
iPSC, induced pluripotent stem cell; SDF, stromal cell-derived factor 1-a; VEGF, vascular endothelial growth factor.

glial cells in vivo suggesting that these cells can provide iPSC-NPCs. Trophic factors found in several types of stem cells
rebuilding materials for tissue repair in the ischemic brain. have pleiotropic effects in that transplanting them after stroke
To support our hypothesis that trophic factors from iPSCs enhances endogenous neurogenesis and angiogenesis [13, 45,
increase regeneration, we analyzed major trophic factors in 46]. For example, mesenchymal stem cells (MSCs) have been

Figure 6. Transplanted induced pluripotent stem cell (iPSC)–neural progenitor cells (NPCs) differentiate into neural lineage cells. (A, B):
Transplanted mCherry iPSC-NPCs differentiated into NeuN-positive cells, (C) neurofilament-positive cells, and (D) glial fibrillary acidic
protein-positive cells. Abbreviations: GFAP, glial fibrillary acidic protein; NeuN, neuronal nuclei.

www.StemCells.com C AlphaMed Press 2014


V
3084 Regeneration and Functional Recovery by iPSCs in Rats

Figure 7. Transplantation of induced pluripotent stem cell (iPSC)–neural progenitor cells (NPCs) increased endogenous regeneration
and functional recovery. (A–D): Stroke only control. (E–H): Stroke 1 iPSC-NPC transplantation. White arrows show colabeling. (I–K):
Increased colabeled NeuN/bromodeoxyuridine (BrdU) cells in transplanted animals (n 5 7–9; t(14) 5 2.799, p 5 .0142). (L–N): Collagen
IV/BrdU cells in the peri-infarct area (n 5 7–9; t(14) 5 2.258, p 5 .0404). (O): Vibrissae-elicited forelimb placement test showed a signifi-
cantly lowered percentage of successful reaches on the left side of stroke animals compared with shams (n 5 7–9; F(2,22) 5 3.60,
p 5 .0444). No significant difference between sham and stroke 1 iPSC-NPC animals. Abbreviations: BrdU, bromodeoxyuridine; iPSC,
induced pluripotent stem cells; NeuN, Neuronal nuclei; NPC, neural progenitor cells.

shown to release a wide range of adaptive factors such as [47]. To illustrate their paracrine effects, MSC-conditioned
factors that are cytoprotective (endothelin), angiogenic media increased neurite outgrowth and branch number when
(VEGF), and play a role in cell migration (LRP-1 and LRP-6) applied to neurons [48]. Similarly, iPSCs have recently been

C AlphaMed Press 2014


V STEM CELLS
Chau, Deveau, Song et al. 3085

shown to express trophic factors including VEGF in of the circulatory system to maintain metabolic homeostasis
neuroepithelial-like stem cells derived from human iPSCs [44]. such as ionic concentrations. This functional unit is severely
We profiled the proregenerative trophic factors expressed by compromised after an ischemic event [63]. Previous work
iPSC-NPCs via reverse-transcriptase polymerase chain reaction demonstrated that angiogenesis and neurogenesis are causally
(RT-PCR) and found that the iPSC-NPCs express SDF-1a, FGF, linked to regenerate the neurovascular unit [11]. For example,
GDNF, EPO, and BDNF, VEGF, Ang3, and CXCR4, a receptor of when angiogenesis was blocked with Endostatin after stroke,
SDF-1a. These are all major angiogenic and neurotrophic fac- there was a correlating 10-fold drop of new neurons in the
tors that contribute to regeneration after stroke [49–54]. peri-infarct cortex [11]. When endothelial progenitor cells
Western blot analysis of the iPSC-NPCs further confirmed were transplanted, there was an increase of angiogenesis and
expression of VEGF, BDNF, Ang3, SDF-1a, VEGFR-2, and neurogenesis improving overall stroke recovery and behavioral
VEGFR-3. Since SDF-1a and VEGF are highly upregulated at deficits [45]. These findings support our observations in the
the stroke site [50, 55], SDF-1a and VEGF receptors expressed present investigation that increased neurogenesis and angio-
by iPSC-NPCs may help to localize the transplanted cells to genesis after transplantation could be synergistically contribut-
the injury area after transplantation to encourage local ing to the rebuilding of the neurovascular unit.
regeneration. Pluripotent stem cell transplantation has the potential to
After confirming the trophic factor expression of iPSC-NPCs be tumorigenic, a major concern in the field [64]. To evaluate
in vitro, we transplanted these iPSC-NPCs into the peri-infarct whether or not these cells will lead to tumorigenesis, further
area and found that animals with transplantation had elevated studies staining for markers such as Ki-67, proliferating cell
levels SDF-1a and VEGF in the peri-infarct tissue suggesting nuclear antigen (PCNA), prominin-1 (CD133), and p53 are
that iPSC-NPCs-derived trophic factors may contribute to the needed [65–67]. We transplanted progenitors, but not pluripo-
regenerative microenvironment. iPSC-NPCs were transplanted tent cells to avoid tumorigenesis. This agrees with the current
during the delayed phase of stroke and supplemented the ris- view that transplantation of undifferentiated stem cells includ-
ing levels of SDF-1a and VEGF in the infarct encouraging repair ing pluripotent iPSCs may lead to tumorigenesis [68, 69]. Thus
after stroke [13, 50]. Certain trophic factors also enhance neu- lineage-restricted stem cells or progenitors are less tumori-
rogenesis and progenitor migration. Chemoattractive trophic genic and better candidates for transplantation [70, 71].
factors like SDF-1a play an important role in directing endoge- To assess the functional recovery after iPSC-NPC transplan-
nous neural progenitors to the stroke lesion via the receptor, tation, whisker somatosensation was tested with the vibrissae-
CXCR4 [13, 50, 56]. By blocking the SDF-1a signaling with a elicited forelimb placement test. Animals with iPSC-NPC trans-
neutralizing antibody against CXCR4, migration was attenuated plantation showed an increase in the percentage of reaches
[50]. SDF-1a is pleiotropic and also contributes to angiogenesis not significantly different than shams. Previous studies have
after stroke by attracting endothelial cells for revascularization shown that stem cell transplantation increases functional
[57, 58]. SDF-1a and VEGF both play roles in endogenous neu- recovery when transplanted into adult [41, 72] and neonatal
ral progenitor migration as well as endothelial progenitor cell stroke animals [73], however we are the first to explore iPSCs
recruitment [50, 51] since endogenous neural and endothelial in neonatal stroke. It has been suggested that transplanted
cell progenitors express CXCR4 [59, 60]. cells may provide several benefits: a favorable trophic factor
Trophic factors such as VEGF enhance angiogenesis. Previ- environment that attenuates cell death, increasing neurogene-
ous studies in vitro have shown that VEGF stimulates the sis and angiogenesis after stroke, and the integration of trans-
tubule formation of endothelial cells (human umbilical vein planted cells into the existing circuitry to improve functional
endothelial cells [HUVEC]) resulting in angiogenesis [61]. With recovery. Thus, both exogenously transplanted iPSC-NPCs and
the addition of VEGF antibody, this effect was attenuated endogenous neural and endothelial progenitors contribute to
[61]. Similarly, transplanted neural stem cells that secreted tissue repair after neonatal ischemic stroke. This study is the
VEGF showed an increase in neovascularization in the peri- first to profile the many trophic factors expressed by iPSC-
infarct area demonstrating a greater blood vessel density in NPCs and show that transplantation of iPSC-NPCs increases
animals with transplantation compared with controls [62]. We angiogenesis and neurogenesis in neonatal stroke.
tested the endogenous regeneration after iPSC-NPC transplan-
tation and observed significant increases in neurogenesis and
angiogenesis in the peri-infarct area of animals with iPSC-NPC CONCLUSION
transplantation. An increase in the migration of neural pro-
Our results indicate iPS-NPC transplantation increases trophic
genitors and angiogenesis could be an effect of trophic factors
support (through increased trophic factor expression) which can
from transplanted iPCS-NPCs. The NeuN/BrdU cells originated
lead to increased endogenous regeneration in the peri-infarct
as endogenous neural progenitors that had migrated and dif-
area and functional recovery following focal ischemic stroke.
ferentiated into NeuN-positive cells before or after arriving at
the peri-infarct area. The source of the NeuN/BrdU cells is
likely endogenous and not exogenous since iPSC-NPCs show
AUTHOR CONTRIBUTIONS
low levels of proliferation after engraftment into the brain tis-
sue and in culture after harvest [39]. M.C.: conception and experimental design, data collection
The endogenous angiogenesis and neurogenesis observed and analysis, manuscript writing; T.D.: created mCherry stem
in this study reflect a remodeling of the damaged neurovascu- cell line and data collection; M.S.: performed patch clamp
lar unit [11]. The neurovascular unit is a functional entity in experiments and data analysis; X.G.: performed stroke sur-
the brain parenchyma where the CNS, comprising of neuronal gery and animal care; D.C.: stem cell cultures, data collection
cells, astrocytes, and pericytes interfaces with the vasculature and analysis; L.W.: conception and experimental design, data

www.StemCells.com C AlphaMed Press 2014


V
3086 Regeneration and Functional Recovery by iPSCs in Rats

analysis, manuscript revision, financial and administrative cloning help and expertise. This work was supported by NIH
support. grants NS075338, NS0458710, AHA EIA Award, and AHA Pre-
doctoral Fellowship.
ACKNOWLEDGMENTS
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
We thank Dr. Oskar Laur of the Custom Cloning Core Facility,
Emory University School of Medicine, Atlanta, GA, for his sub- The authors indicate no potential conflicts of interest.

REFERENCES rostral migratory stream. Dev Dyn 1998;213: 30 Dimos JT, Rodolfa KT, Niakan KK et al.
220–227. Induced pluripotent stem cells generated
1 Go AS, Mozaffarian D, Roger VL et al. 17 Zhang RL, Zhang ZG, Zhang L et al. Pro- from patients with ALS can be differentiated
Heart disease and stroke statistics—2013 liferation and differentiation of progenitor into motor neurons. Science 2008;321:1218–
update: A report from the American Heart cells in the cortex and the subventricular 1221.
Association. Circulation 2013;127:e6–e245. zone in the adult rat after focal cerebral 31 Chambers SM, Fasano CA, Papapetrou
2 Agrawal N, Johnston SC, Wu YW et al. ischemia. Neuroscience 2001;105:33–41. EP et al. Highly efficient neural conversion of
Imaging data reveal a higher pediatric stroke 18 Jin K, Sun Y, Xie L et al. Directed migra- human ES and iPS cells by dual inhibition of
incidence than prior US estimates. Stroke tion of neuronal precursors into the ischemic SMAD signaling. Nat Biotechnol 2009;27:
2009;40:3415–3421. cerebral cortex and striatum. Mol Cell Neuro- 275–280.
3 Ganesan V, Prengler M, McShane MA sci 2003;24:171–189. 32 Karumbayaram S, Novitch BG, Patterson
et al. Investigation of risk factors in children 19 Tonchev AB, Yamashima T, Sawamoto K M et al. Directed differentiation of human-
with arterial ischemic stroke. Ann Neurol et al. Enhanced proliferation of progenitor induced pluripotent stem cells generates
2003;53:167–173. cells in the subventricular zone and limited active motor neurons. STEM CELLS 2009;27:
4 Lynch JK, Hirtz DG, DeVeber G et al. neuronal production in the striatum and neo- 806–811.
Report of the National Institute of Neurologi- cortex of adult macaque monkeys after 33 Bain G, Kitchens D, Yao M et al. Embry-
cal Disorders and Stroke workshop on perina- global cerebral ischemia. J Neurosci Res onic stem cells express neuronal properties
tal and childhood stroke. Pediatrics 2002; 2005;81:776–788. in vitro. Dev Biol 1995;168:342–357.
109:116–123. 20 Thored P, Arvidsson A, Cacci E et al. Per- 34 Bain G, Ray WJ, Yao M et al. Retinoic
5 Monagle P, Adams M, Mahoney M et al. sistent production of neurons from adult acid promotes neural and represses mesoder-
Outcome of pediatric thromboembolic dis- brain stem cells during recovery after stroke. mal gene expression in mouse embryonic
ease: A report from the Canadian Childhood STEM CELLS 2006;24:739–747. stem cells in culture. Biochem Biophys Res
Thrombophilia Registry. Pediatr Res 2000;47: 21 Parent JM, Vexler ZS, Gong C et al. Rat Commun 1996;223:691–694.
763–766. forebrain neurogenesis and striatal neuron 35 Bottenstein JE, Sato GH. Growth of a rat
6 Wusthoff CJ, Kessler SK, Vossough A replacement after focal stroke. Ann Neurol neuroblastoma cell line in serum-free supple-
et al. Risk of later seizure after perinatal 2002;52:802–813. mented medium. Proc Natl Acad Sci USA
arterial ischemic stroke: A prospective cohort 22 Kokaia Z, Thored P, Arvidsson A et al. 1979;76:514–517.
study. Pediatrics 2011;127:e1550–1557. Regulation of stroke-induced neurogenesis in 36 Wei L, Han BH, Li Y et al. Cell death
7 Gardner MA, Hills NK, Sidney S et al. adult brain—Recent scientific progress. Cere- mechanism and protective effect of erythro-
The 5-year direct medical cost of neonatal bral Cortex 2006;16:I162–I167. poietin after focal ischemia in the whisker-
and childhood stroke in a population-based 23 Yuan T, Liao W, Feng NH et al. Human barrel cortex of neonatal rats. J Pharmacol
cohort. Neurology 2010;74:372–378. induced pluripotent stem cell-derived neural Exp Ther 2006;317:109–116.
8 Greenberg DA. Angiogenesis and stroke. stem cells survive, migrate, differentiate, and 37 Schaar KL, Brenneman MM, Savitz SI.
Drug News Perspect 1998;11:265–270. improve neurological function in a rat model Functional assessments in the rodent stroke
9 Arvidsson A, Collin T, Kirik D et al. Neu- of middle cerebral artery occlusion. Stem Cell model. Exp TransStroke Med 2010;2:13.
ronal replacement from endogenous precur- Res Ther 2013;4:73. 38 Hua Y, Schallert T, Keep RF et al. Behav-
sors in the adult brain after stroke. Nat Med 24 Chen SJ, Chang CM, Tsai SK et al. Func- ioral tests after intracerebral hemorrhage in
2002;8:963–970. tional improvement of focal cerebral ische- the rat. Stroke 2002;33:2478–2484.
10 Ohab JJ, Carmichael ST. Poststroke neu- mia injury by subdural transplantation of 39 Drury-Stewart D, Song M, Mohamad O
rogenesis: Emerging principles of migration induced pluripotent stem cells with fibrin et al. Highly efficient differentiation of neural
and localization of immature neurons. Neuro- glue. Stem Cells Dev 2010;19:1757–I767. precursors from human embryonic stem cells
scientist 2008;14:369–380. 25 Mohamad O, Drury-Stewart D, Song M and benefits of transplantation after ischemic
11 Ohab JJ, Fleming S, Blesch A et al. A et al. Vector-free and transgene-free human stroke in mice. Stem Cell Res Ther 2013;4:93.
neurovascular niche for neurogenesis after iPS cells differentiate into functional neurons 40 Li Y, Lu ZY, Ogle M et al. Erythropoietin
stroke. J Neurosci 2006;26:13007–13016. and enhance functional recovery after ische- prevents blood brain barrier damage induced
12 Li Y, Yu S, Mohamad O et al. Sublethal mic stroke in mice. PLoS One 2013;8:e64160. by focal cerebral ischemia in mice. Neuro-
transient global ischemia stimulates migra- 26 Takahashi K, Tanabe K, Ohnuki M et al. chem Res 2007;32:2132–2141.
tion of neuroblasts and neurogenesis in Induction of pluripotent stem cells from 41 Wei L, Cui L, Snider BJ et al. Transplanta-
mice. Trans Stroke Res 2010;1:184–196. adult human fibroblasts by defined factors. tion of embryonic stem cells overexpressing
13 Li WL, Yu SP, Ogle ME et al. Enhanced Cell 2007;131:861–872. Bcl-2 promotes functional recovery after
neurogenesis and cell migration following 27 Takahashi K, Yamanaka S. Induction of transient cerebral ischemia. Neurobiol Dis
focal ischemia and peripheral stimulation in pluripotent stem cells from mouse embryonic 2005;19:183–193.
mice. Dev Neurobiol 2008;68:1474–1486. and adult fibroblast cultures by defined fac- 42 Kuge A, Takemura S, Kokubo Y et al.
14 Kornack DR, Rakic P. The generation, tors. Cell 2006;126:663–676. Temporal profile of neurogenesis in the sub-
migration, and differentiation of olfactory 28 Takahashi K, Okita K, Nakagawa M et al. ventricular zone, dentate gyrus and cerebral
neurons in the adult primate brain. Proc Natl Induction of pluripotent stem cells from cortex following transient focal cerebral
Acad Sci U S A 2001;98:4752–4757. fibroblast cultures. Nat Protoc 2007;2:3081– ischemia. Neurol Res 2009;31:969–976.
15 Menezes JR, Smith CM, Nelson KC et al. 3089. 43 Wei L, Rovainen CM, Woolsey TA. Minis-
The division of neuronal progenitor cells dur- 29 Yu J, Vodyanik MA, Smuga-Otto K et al. trokes in rat barrel cortex. Stroke 1995;26:
ing migration in the neonatal mammalian Induced pluripotent stem cell lines derived 1459–1462.
forebrain. Mol Cell Neurosci 1995;6:496–508. from human somatic cells. Science 2007;318: 44 Oki K, Tatarishvili J, Wood J et al.
16 Smith CM, Luskin MB. Cell cycle length 1917–1920. Human-induced pluripotent stem cells form
of olfactory bulb neuronal progenitors in the functional neurons and improve recovery

C AlphaMed Press 2014


V STEM CELLS
Chau, Deveau, Song et al. 3087

after grafting in stroke-damaged brain. STEM 54 Kobayashi T, Ahlenius H, Thored P et al. injury. J Physiol Pharmacol 2009;60 Suppl 4:
CELLS 2012;30:1120–1133. Intracerebral infusion of glial cell line-derived 95–104.
45 Ishikawa H, Tajiri N, Shinozuka K et al. neurotrophic factor promotes striatal neuro- 64 Dressel R, Schindehutte J, Kuhlmann T
Vasculogenesis in experimental stroke after genesis after stroke in adult rats. Stroke et al. The tumorigenicity of mouse embryonic
human cerebral endothelial cell transplanta- 2006;37:2361–2367. stem cells and in vitro differentiated neuro-
tion. Stroke 2013;44:3473–3481. 55 Jin K, Zhu Y, Sun Y et al. Vascular endo- nal cells is controlled by the recipients’
46 Chau M, McCullough I, Yu SP et al. Neu- thelial growth factor (VEGF) stimulates neu- immune response. PLoS One 2008;3:e2622.
rogenesis and neural progenitor migration rogenesis in vitro and in vivo. Proc Natl Acad 65 Moore N, Lyle S. Quiescent, slow-cycling
after stroke. In: Girard A, Moreau L, eds. Sci U S A 2002;99:11946–11950. stem cell populations in cancer: A review of
Neuronal Migration: Disorders, Genetic Fac- 56 Imitola J, Raddassi K, Park KI et al. the evidence and discussion of significance.
tors, and Treatment Options. New York, NY: Directed migration of neural stem cells to J Oncol 2011;2011, pii: 396076.
Nova Science Publishers, Inc.; 2012;115–143. sites of CNS injury by the stromal cell- 66 Sathi GA, Tamamura R, Tsujigiwa H et al.
47 Nguyen BK, Maltais S, Perrault LP et al. derived factor 1alpha/CXC chemokine recep- Analysis of immunoexpression of common
Improved function and myocardial repair of tor 4 pathway. Proc Natl Acad Sci U S A cancer stem cell markers in ameloblastoma.
infarcted heart by intracoronary injection of 2004;101:18117–18122. Exp Ther Med 2012;3:397–402.
mesenchymal stem cell-derived growth 57 Gupta SK, Lysko PG, Pillarisetti K et al. 67 Zhang M, Song T, Yang L et al. Nestin and
factors. J Cardiovasc Trans Res 2010;3:547– Chemokine receptors in human endothelial CD133: Valuable stem cell-specific markers for
558. cells. Functional expression of CXCR4 and its determining clinical outcome of glioma
48 Oh JS, Liu ML, Jin HL et al. Effect of transcriptional regulation by inflammatory patients. J Exp Clin Cancer Res 2008;27:85.
bone marrow derived mesenchymal stem cell cytokines. J Biol Chem 1998;273:4282–4287. 68 Kawai H, Yamashita T, Ohta Y et al. Trider-
conditioned media for neurite outgrowth in 58 Volin MV, Joseph L, Shockley MS et al. mal tumorigenesis of induced pluripotent stem
human Ntera-2 neurons. Tissue Eng Regen- Chemokine receptor CXCR4 expression in cells transplanted in ischemic brain. J Cereb
erat Med 2009;6:562–567. endothelium. Biochem Biophys Res Commun Blood Flow Metab 2010; 30:1487–1493.
49 Frank J, Carroll CM, Aaranson K et al. 1998;242:46–53. 69 Nishimori M, Yakushiji H, Mori M et al.
Ischemia increases the angiogenic potency of 59 Salcedo R, Wasserman K, Young HA Tumorigenesis in cells derived from induced
basic fibroblast growth factor (FGF-2). Micro- et al. Vascular endothelial growth factor and pluripotent stem cells. Human Cell 2014;27:
surgery 1996;17:452–456. basic fibroblast growth factor induce expres- 29–35.
50 Robin AM, Zhang ZG, Wang L et al. Stro- sion of CXCR4 on human endothelial cells: In 70 Xu T, Zhang M, Laurent T et al. Concise
mal cell-derived factor 1alpha mediates neu- vivo neovascularization induced by stromal- review: Chemical approaches for modulating
ral progenitor cell motility after focal derived factor-1alpha. Am J Pathol 1999;154: lineage-specific stem cells and progenitors.
cerebral ischemia. J Cereb Blood Flow Metab 1125–1135. Stem Cells Trans Med 2013;2:355–361.
2006;26:125–134. 60 Ji JF, He BP, Dheen ST et al. Expression 71 Liao MC, Diaconu M, Monecke S et al.
51 Yamaguchi J, Kusano KF, Masuo O et al. of chemokine receptors CXCR4, CCR2, CCR5 Embryonic stem cell-derived neural progeni-
Stromal cell-derived factor-1 effects on ex and CX3CR1 in neural progenitor cells iso- tors as non-tumorigenic source for dopami-
vivo expanded endothelial progenitor cell lated from the subventricular zone of the nergic neurons. World J Stem Cells 2014;6:
recruitment for ischemic neovascularization. adult rat brain. Neurosci Lett 2004;355:236– 248–255.
Circulation 2003;107:1322–1328. 240. 72 Theus MH, Wei L, Cui L et al. In vitro
52 Chen J, Zhang C, Jiang H et al. Atorvas- 61 Bishop ET, Bell GT, Bloor S et al. An in hypoxic preconditioning of embryonic stem
tatin induction of VEGF and BDNF promotes vitro model of angiogenesis: Basic features. cells as a strategy of promoting cell survival
brain plasticity after stroke in mice. J Cereb Angiogenesis 1999;3:335–344. and functional benefits after transplantation
Blood Flow Metab 2005;25:281–290. 62 Horie N, Pereira MP, Niizuma K et al. into the ischemic rat brain. Exp Neurol 2008;
53 Wang L, Chopp M, Gregg SR et al. Neu- Transplanted stem cell-secreted VEGF effects 210:656–670.
ral progenitor cells treated with EPO induce post-stroke recovery, inflammation, and vas- 73 van Velthoven CT, Sheldon RA, Kavelaars
angiogenesis through the production of cular repair. STEM CELLS 2011; 29:274–85. A et al. Mesenchymal stem cell transplanta-
VEGF. J Cereb Blood Flow Metab 2008;28: 63 Madri JA. Modeling the neurovascular tion attenuates brain injury after neonatal
1361–1368. niche: Implications for recovery from CNS stroke. Stroke 2013;44:1426–1432.

www.StemCells.com C AlphaMed Press 2014


V

You might also like