You are on page 1of 8

Human Neural Stem Cell Grafts Modify Microglial

Response and Enhance Axonal Sprouting in Neonatal


Hypoxic–Ischemic Brain Injury
Marcel M. Daadi, PhD; Alexis S. Davis, MD; Ahmet Arac, MD; Zongjin Li, MD, PhD;
Anne-Lise Maag, BS; Rishi Bhatnagar, BS; Kewen Jiang, MD, PhD; Guohua Sun, MD;
Joseph C. Wu, MD, PhD; Gary K. Steinberg, MD, PhD

Background and Purpose—Hypoxic–ischemic (HI) brain injury in newborn infants represents a major cause of cerebral
palsy, development delay, and epilepsy. Stem cell-based therapy has the potential to rescue and replace the ischemic
tissue caused by HI and to restore function. However, the mechanisms by which stem cell transplants induce functional
recovery are yet to be elucidated. In the present study, we sought to investigate the efficacy of human neural stem cells
derived from human embryonic stem cells in a rat model of neonatal HI and the mechanisms enhancing brain repair.
Methods—The human neural stem cells were genetically engineered for in vivo molecular imaging and for postmortem
histological tracking. Twenty-four hours after the induction of HI, animals were grafted with human neural stem cells into the
forebrain. Motor behavioral tests were performed the fourth week after transplantation. We used immunocytochemistry and
neuroanatomical tracing to analyze neural differentiation, axonal sprouting, and microglia response. Treatment-induced
changes in gene expression were investigated by microarray and quantitative polymerase chain reaction.
Results—Bioluminescence imaging permitted real time longitudinal tracking of grafted human neural stem cells. HI
transplanted animals significantly improved in their use of the contralateral impeded forelimb and in the Rotorod test. The
grafts showed good survival, dispersion, and differentiation. We observed an increase of uniformly distributed microglia cells
in the grafted side. Anterograde neuroanatomical tracing demonstrated significant contralesional sprouting. Microarray
analysis revealed upregulation of genes involved in neurogenesis, gliogenesis, and neurotrophic support.
Conclusions—These results suggest that human neural stem cell transplants enhance endogenous brain repair through
Downloaded from http://ahajournals.org by on January 15, 2020

multiple modalities in response to HI. (Stroke. 2010;41:516-523.)


Key Words: axonal sprouting 䡲 cell therapy 䡲 hypoxia ischemia 䡲 microglia 䡲 neural stem cells

H ypoxic–ischemic (HI) brain injury causes brain damage


in the fetus and newborn infants and represents a major
cause of cerebral palsy, cognitive impairment, learning dis-
and synaptic changes; inflammatory and immune adaptation;
neurogenesis; gliogenesis; and angiogenesis.12 Axonal
sprouting and reorganization manifest by changes in dendritic
ability, and epilepsy.1 Although mild body hypothermia has arborization, spine remodeling, branching into the denervated
been shown to improve the outcome after neonatal HI areas, and de novo formation of novel projections.13–17
encephalopathy when initiated within 6 hours of birth,2,3 there Although limited, this spontaneous plasticity-mediated recov-
are no other effective interventions to improve the chronic ery is a promising target for drug and cell therapeutic
sequelae of perinatal asphyxia. Neural stem cell-based therapy interventions.17–21
offers the prospect to rescue damaged tissue, to replace lost cells, In parallel to endogenous central nervous system plasticity,
and to restore neurological function after cerebral HI. inflammation and immune components are markedly acti-
Early imaging studies in patients with stroke4 –7 and micro- vated de novo in the neonatal brain and peripheral organs
stimulation in experimental models of stroke8 –10 reported that after HI.22,23 Noteworthy, this inflammatory response to
in response to ischemic injury, the brain undergoes limited ischemic injury may exert neuroprotective and regenerative
compensatory changes in an effort to recover from structural effects on the central nervous system.24,25
and functional loss.11 These changes or neuroplasticity are In the present study, we evaluated functional recovery after
most prominent in early weeks and include axonal, dendritic, transplantation of multipotent human embryonic stem cell-

Received November 20, 2009; accepted December 1, 2009.


From the Department of Neurosurgery and Stanford Stroke Center (M.M.D., A.S.D., A.A., A.-L.M., R.B., K.J., G.S., G.K.S.), Stanford University
School of Medicine, Stanford, Calif; and Department of Radiology and Molecular Imaging Program (Z.L., J.C.W.), Stanford University School of
Medicine, Stanford, Calif.
Present address for K.J.: Department of Neurology, Children’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
Correspondence to Marcel M. Daadi, PhD, Stanford University, Department of Neurosurgery, MSLS P304, 1201 Welch Road, Stanford, CA
94305-5487. E-mail mdaadi@stanford.edu
© 2010 American Heart Association, Inc.
Stroke is available at http://stroke.ahajournals.org DOI: 10.1161/STROKEAHA.109.573691

516
Daadi et al Neural Stem Cells in Neonatal Hypoxia Ischemia 517

derived human neural stem cells (hNSCs) into the rat model of
neonatal HI. We also investigated modalities by which grafted
hNSCs provide therapeutic benefits to the HI-damaged brain.

Materials and Methods


Derivation of Multipotent hNSCs
Human neural stem cells were derived from human embryonic stem
cells and perpetuated as previously described.26,27

Induction of HI and Cell Transplantation


All animal experimentations were conducted according to the Na-
tional Institutes of Health guidelines and approved by the Institu-
tional Animal Care and Use Committee. Seven-day-old Sprague-
Dawley rats (Charles River Laboratories; Wilmington, Mass) were
subjected to permanent ligation of the left carotid artery followed by
90 minutes in a hypoxic chamber (8% O2 and 92% N2, at 37°C).28
The newborns were divided into HI vehicle (total n⫽12) and HI
transplant (total n⫽12) groups. Twenty-four hours after the induc-
tion of HI, animals were placed in a stereotaxic apparatus with
neonatal rat adaptor (Kopf Instruments). The skull bregma was
determined and a single cell suspension (50 000 cell/␮L) of hNSCs
(passages P9 to P15) was transplanted, using a Hamilton syringe,
into 3 sites (2 ␮L/site) in the left ischemic hemisphere at the
following stereotaxic coordinates in millimeters: anteroposterior: ⫹0.0,
mediolateral: ⫹3.0, dorsoventral: ⫺4.0; anteroposterior: ⫹0.5, me-
diolateral: ⫹2.5, dorsoventral: ⫺3.5; anteroposterior: ⫺1.0, medio-
lateral: ⫹2.0, dorsoventral: ⫺4.0. The injection rate was 1 ␮L/min,
and the cannula was left in place for 5 minutes before retraction.

Bioluminescence Imaging of Grafted hNSCs


In Vivo
Bioluminescence imaging was performed using the Xenogen in vivo
imaging system. Due to space limitation, we refer the reader to our
Downloaded from http://ahajournals.org by on January 15, 2020

recent publication27 where we described in detail the biolumines-


cence imaging technique.

Behavioral Tests
The animals were evaluated 4 weeks after transplantation for their
sensorimotor skills in the cylinder and in the Rotorod tests, as we
previously reported.26,29

Transcriptome Analysis
Total RNA was isolated from the ipsilateral hemisphere of normal, HI,
and HI transplant groups (n⫽3) using the RNAeasy kit (Qiagen)
according to the manufacturer’s instructions. The Oligo GEArray
neurogenesis and neural stem cell microarrays were purchased from
SuperArray (SA Biosciences Corporation). The protocol for the microarray
analyses followed the manufacturer’s recommendations. Array spot density
and differential probe expression were calculated using SuperArray’s
GEArray Expression Analysis Suite software. Spot density was normalized
to select positive and negative controls spotted onto each array.

Quantitative Reverse Transcriptase–Polymerase


Chain Reaction
Total RNA was extracted from tissue dissected from the ipsilateral
hemisphere of vehicle and hNSC-treated animals (n⫽3). Aliquots (1
␮g) of total RNA from the cells were reverse transcribed as Figure 1. Imaging of human embryonic stem cell-derived
previously described.30 Real-time polymerase chain reaction was hNSCs in HI neonates. A, Neural stem cells were isolated
performed using Maxima SYBR Green qPCR Master Mix (Fermen- through a multistep process previously described.26,27 Flow
tas) in the Stratagene thermocycler MX3000P (Stratagene) according cytometry analysis demonstrated the purity of the eGFP-
to the supplier’s instructions. Data were expressed as mean⫾SEM. expressing hNSCs. B, In vitro imaging analysis of genetically
The ⌬Ct for each candidate was calculated as ⌬Ct of [Ct (gene of engineered hNSCs show increasing fLuc activity with cell den-
sity and a linear correlation (R2⫽0.98). C, Data are representa-
interest)⫺Ct (18S)] and the ⌬⌬Ct was the difference between the Ct
tive of 3 independent experiments performed in triplicate. Rep-
of the treated sample (transplanted animals) and the Ct of the control
resentative bioluminescence imaging of a neonate rat
vehicle sample. The relative expression was calculated as the 2⌬⌬Ct
ˆ
transplanted with the hNSCs and monitored for 4 weeks. D,
according to the methods31 and plotted as relative levels of gene Color scale bar is in photon/s/cm2/sr.
expression. The rat primers were designed using the Primer3
software and are available on request.
518 Stroke March 2010

Figure 2. Characterization of hNSC grafts.


A, Example of triple labeling of trans-
planted animal with the human nuclear
specific marker hNuc (purple) and the
neuronal markers doublecortin (red) and
TuJ1 (green). B, Higher magnification
shows the dispersion of the grafted
hNuc⫹ (purple) hNSCs in the striatum.
Photomicrographs show coexpression of
hNuc (purple) and the neuronal markers:
TuJ1 (green; C), doublecortin (green; D),
and NeuN (green; E). F, Colocalization of
hNuc and the astroglial marker glial fibril-
lary acidic protein, and (G) example of
grafted cell expressing the GABAergic
neurotransmitter marker GAD. Bars: (A–B)
100 ␮m; (B, far right, E) 20 ␮m; (C, D, F,
G) 10 ␮m.
Downloaded from http://ahajournals.org by on January 15, 2020

Biotinylated Dextran Amine Injections tein, antigalactocerebrocide, anti-Nestin (Chemicon), anti-TuJ1 (Co-
The last week before euthanasia of the animals, 3 randomly selected vance), anti-CNPase (Aves Labs), and antidoublecortin (SantaCruz
animals from both animal groups were anesthetized and placed in the Biotechnology). Secondary antibodies raised in the appropriate hosts
stereotaxic apparatus. After craniotomy, 0.5 ␮L of biotinylated and conjugated to fluorescein isothiocyanate, RITC, AMCA, CY3,
dextran amine (BDA; 10 000 molecular weight; Molecular Probes; or CY5 chromogenes (Jackson ImmunoResearch) were used. Cells
10% wt/vol solution in sterile phosphate-buffered saline) was in- and sections were counterstained with the nuclear marker 4⬘,6-
jected stereotaxically into the sensorimotor cortex opposite to the HI diamidine-2⬘-phenylindole dihydrochloride. Fluorescence was de-
lesion site at the stereotaxic coordinates: anteroposterior: ⫹0.5 m, tected, analyzed, and photographed with a Zeiss LSM550 laser
mediolateral: 2.5 mm, and DV: ⫺1.5 mm. The scalp was then closed scanning confocal photomicroscope (Carl Zeiss).
and the animal returned to its cage. For the rat, anti-Iba-1 (ionized calcium binding adapter molecule;
Wako Bioproducts) and BDA immunohistochemistry was carried
Histopathology, Immunocytochemistry, and out, as we previously described.29 For each animal, quantitative
Microscopical Analysis estimates of the number of Iba-1⫹ cells were stereologically
Immunocytochemistry was performed as we previously reported.26 determined using the optical fractionator procedure,32 as we previ-
The following primary antibodies were used: antihuman Nuclei ously described in detail.29 The quantitative analysis of BDA-labeled
(hNuc), anti-NeuN, anti-GAD65/67, antiglial fibrillary acidic pro- fibers was performed, as we previously reported.29 The infarct
Daadi et al Neural Stem Cells in Neonatal Hypoxia Ischemia 519
Downloaded from http://ahajournals.org by on January 15, 2020

Figure 3. Transplanted hNSCs enhanced axonal sprouting. Confocal image analysis and quantification of BDA-labeled axons and axon
terminals in the sensorimotor cortex (A), corpus callosum (B), striatum (C), and in the thalamus (D) show significant increase on the ipsi-
lateral HI-lesioned and hNSC-transplanted side (n⫽3 per group). Data are expressed as mean⫾SEM, *P⬍0.05 versus vehicle group.

volume was estimated by summing up the infarcted areas in all variance was used to compare groups. Differences between the
animals of each group. To eliminate the effects of edema, infarct size groups were determined using Bonferroni post hoc test. A probabil-
was calculated as the contralateral hemisphere⫺ipsilateral nonische- ity value of ⬍0.05 was considered statistically significant.
mic hemisphere/contralateral hemisphere and expressed as percent-
age (⫻100%) of infarcted hemisphere.
Results
Statistics Real-Time Imaging of hNSC Transplants
Outcome measurement for each experiment was reported as
mean⫾SEM. Data were analyzed using SPSS 11 for Mac OS X The hNSCs grew as an adherent monolayer culture and all
(SPSS Inc). Significance of intergroup differences was performed by progeny expressed double fusion construct carrying fLuc and
applying Student t test where appropriate. One-way analysis of eGFP reporter genes (Figure 1A).
520 Stroke March 2010

Image analysis (Figure 1B) demonstrated a linear correla-


tion between plating density and the fLuc activity (Figure
1C). This stable and efficient expression of fLuc gene enabled
us to noninvasively image, in real time, the survival of the
grafted hNSCs at different time points (Figure 1D).

Characterization of the HI Infarct Size and the


hNSC Graft
Microscopic examination of cresyl violet-stained sections
showed significant loss of brain tissue in HI animals in the
neocortex, the striatum, and extending caudally to the hip-
pocampus. This atrophy was accompanied sometimes by the
formation of a porencephalic cyst. The estimation of the
infracted region revealed 17.8%⫾8.6% of the hemisphere
(n⫽6) for the transplanted group and 20.66%⫾12.5% (n⫽6)
for the vehicle group. Although hNSC-grafted animals
showed smaller infarct volume, the difference from the
vehicle group was not significant (n⫽6). Grafted hNSCs
identified with hNuc demonstrated good survival dispersion
and integration into the HI-damaged tissue (Figure 2A–B).
Transplanted cells expressed the neuroepithelial stem cell
marker nestin in 12.8%⫾3.4% and differentiated into neurons
expressing NeuN, TuJ1⫹ (42.9%⫾3.5%), doublecortin
(4.8%⫾1.1%; Figure 2C–E), and the GABAergic marker
GAD (47.6%⫾4.3%; Figure 2G). Grafted cells also differen-
tiated into glial fibrillary acidic protein-expressing astrocytes
(Figure 2F).
Figure 4. Microarray analysis and quantitative real-time reverse
hNSC Grafts Enhance Axonal Sprouting transcribed–polymerase chain reaction measurement of selected
To investigate whether the transplanted hNSCs influenced the genes upregulated in transplanted animals. Genes from the
Downloaded from http://ahajournals.org by on January 15, 2020

rewiring of the HI-damaged tissue, we used BDA to antero- expression profile with probability values ⬍0.01 were selected
for analysis. A, Fold of increase of genes involved in neurogen-
grade label the sensorimotor cortical projection originating esis, gliogenesis, and neurotrophic support in HI and HI trans-
from the contralateral side. We measured the density of plant groups relative to the naïve group. B, Quantitative reverse
BDA⫹ crossing fibers in the corpus callosum and of BDA⫹ transcribed–polymerase chain reaction of the selected genes
terminals in the ipsilateral sensorimotor cortex, the striatum, confirming the expression patterns seen in the microarray data.
and thalamic nuclei. The quantitative analysis of BDA-
labeled fibers and terminals, normalized to the total number throughout the brain in both vehicle and hNSC-grafted
of labeled somas at the injections site,29 revealed an increase animals without adverse infiltration or reaction against the
in both the number of fibers crossing toward the ipsilateral grafts (Figure 5A–B). Stereological quantitative analysis of
hemisphere (Figure 3B) and in the number of terminals in the the Iba1-positive cells demonstrated a significant increase in
ipsilateral sensorimotor cortex (Figure 3A), the striatum the transplanted striatum (Figure 5D).
(Figure 3C), and thalamic nuclei (Figure 3D).
Improvement of Motor Behavior in Rats That
Transcriptome Analysis Received the hNSC Grafts
Gene microarray analysis revealed, in comparison to the To determine the ability of hNSCs to functionally engraft, 1
vehicle group, a significant increase in central nervous system
month after transplantation, the sensorimotor skills of animals
rat endogenous genes involved: (1) in neurogenesis and cell
were evaluated using 2 neurobehavioral tests. Our results
migration (doublecortin, chemokine [C-X-C motif] receptor 4
showed that during the fourth week, HI transplanted animals
[CXCR4], oligodendrocyte lineage transcription factor 2, and
significantly improved in their use of the contralateral impeded
FGF2); (2) in myelination (myelin basic protein); and (3) in
forelimb (Figure 6A–B). The hNSC grafts significantly amelio-
genes involved in cell survival and neurite outgrowth (glial
derived neurotrophic factor, Neurturin, and insulin growth rated the locomotor deficits in the Rotorod test (Figure 6C).
factor-1; Figure 4A).33 Subsequently, we confirmed the mi-
croarray expression patterns of selected genes using real-time Discussion
quantitative reverse transcribed–polymerase chain reaction Genetically engineered hNSCs were efficiently and noninva-
(Figure 4B). sively imaged in real time after transplantation into the HI
model of newborn rats and up to 5 weeks of age. We report that
Grafts of hNSCs Modulate Microglial Presence in hNSCs engrafted into the ischemic brain enhanced axonal
the HI-Damaged Area sprouting and the expression of genes involved in neurogenesis,
Immunostaining with the pan-microglial marker Iba1 demon- gliogenesis, and neurotrophic support; modulated microglial
strated that microglia were homogenously distributed response; and improved motor function of the animals.
Daadi et al Neural Stem Cells in Neonatal Hypoxia Ischemia 521

Figure 5. Stereological analysis of Iba1-expressing microglia in vehicle and transplanted groups. Brain sections from vehicle (A) and
hNSC-grafted (B) animals were processed for the pan-microglial marker Iba1 immunohistochemistry. Stereological quantification of the
Iba1⫹ cells in the cortex (C) and striatum (D) of grafted animals. Tx indicates transplant. Bars: (A, B) 20 ␮m.
Downloaded from http://ahajournals.org by on January 15, 2020

It is generally believed that transplanted nonneural cells provide cell replacement and neurotrophic support.29,35,36
such as those derived from bone marrow or cord blood exert This neurotrophic support may be responsible for the signif-
neurotrophic effects on ischemia-injured tissue and may not icant axonal sprouting that we measured in the cortex, the
survive long term,34 whereas neural stem cells are thought to striatum, and the thalamus. The number of BDA-labeled axon

Figure 6. Transplantation of hNSCs improves sen-


sorimotor function. The independent use of the
impaired contralateral forelimb significantly
increased in the transplant group (B; n⫽3) on Days
28 and 30 posttransplantation (*P⬍0.05 versus
vehicle group. A, Bars represent percentages⫾SEM of
steps taken by the ipsilateral, contralateral, and both
forelimbs simultaneously. On Days 29 and 30, the
transplant group (n⫽9) showed significant improvement
(*P⬍0.05) on the Rotorod test.
522 Stroke March 2010

terminals may vary with the size of the injection site and the In conclusion, we provide evidence that growth factor-
number of BDA⫹ cell bodies. The BDA infusion led to small isolated and perpetuated hNSCs from human embryonic stem
(3 to 5 mm2) and circumscribed injection sites in the cells are amenable to genetic modification for real time in
sensorimotor cortex. There was no significant difference in vivo imaging and potentially for other therapeutic genes. We
the size of the injection site and the total number of labeled showed that these hNSCs are able to modify the host
cells between vehicle and transplanted groups. In addition, microenvironment and enhance neuroanatomical plasticity
the data were normalized to the total number of labeled cells. after HI in neonates and improve sensorimotor skills.
Thus, it is unlikely that the increase of axonal sprouting we
observed is due to differences in the size or location of the Acknowledgments
injection site. We thank Beth Hoyte for preparation of the figures.
After ischemia, regions of the contralateral hemispheres
become activated during the early phase of partial regain of
Sources of Funding
This work was supported in part by Russell and Elizabeth Siegelman,
function or spontaneous recovery in experimental models37 Bernard and Ronni Lacroute, the William Randolph Hearst Founda-
and in patients with stroke.38 – 40 In neonate HI rats, contrale- tion, CIRM RS1-00322, the Edward G. Hills Fund, and National
sional sprouting could give rise to alternative motor descend- Institutes of Health National Institute of Neurological Diseases and
ing pathways from the motor cortex relaying in subcortical Stroke grants RO1 NS27292, P01 NS37520, and R01 NS058784.
structures such as the red nucleus and the pontine formation
or direct corticospinal projections may be formed to compen-
Disclosures
None.
sate for functional loss.41 The newly formed pathways could
be generated by multiple mechanisms, including sprouting References
from the surviving neurons, unmasking of existing pathways 1. Vannucci RC. Hypoxic–ischemic encephalopathy. Am J Perinatol. 2000;
that are functionally inactive, or the compensatory descend- 17:113–120.
2. Gluckman PD, Wyatt JS, Azzopardi D, Ballard R, Edwards AD, Ferriero
ing control channels through alternative functionally active
DM, Polin RA, Robertson CM, Thoresen M, Whitelaw A, Gunn AJ.
but redundant pathways.42,43 Selective head cooling with mild systemic hypothermia after neonatal
The intracerebral or intravenous delivery of multipotent encephalopathy: multicentre randomised trial. Lancet. 2005;365:
adult progenitor cells provided protection to HI-damaged 663– 670.
3. Shankaran S, Laptook AR, Ehrenkranz RA, Tyson JE, McDonald SA,
tissue in neonates and improved motor and neurological Donovan EF, Fanaroff AA, Poole WK, Wright LL, Higgins RD, Finer
scores compared with the vehicle group.44 Using retrograde NN, Carlo WA, Duara S, Oh W, Cotten CM, Stevenson DK, Stoll BJ,
and anterograde neuroanatomical tracing, Park et al36 dem- Lemons JA, Guillet R, Jobe AH. Whole-body hypothermia for neonates
Downloaded from http://ahajournals.org by on January 15, 2020

with hypoxic–ischemic encephalopathy. N Engl J Med. 2005;353:


onstrated that neural stem cells grafted into HI-lesioned 1574 –1584.
neonates, along with a polyglycolic acid-based biodegradable 4. Chollet F, DiPiero V, Wise RJ, Brooks DJ, Dolan RJ, Frackowiak RS.
polymer scaffold, re-established long-distance cross-callosal The functional anatomy of motor recovery after stroke in humans: a study
with positron emission tomography. Ann Neurol. 1991;29:63–71.
neuronal connections. Recent genomic analysis studies dem-
5. Fridman EA, Hanakawa T, Chung M, Hummel F, Leiguarda RC, Cohen
onstrated that in addition to these growth factors, axonal LG. Reorganization of the human ipsilesional premotor cortex after
sprouting is activity-dependent in constraint-induced move- stroke. Brain. 2004;127:747–758.
ment therapy.45 Together, these data suggest that grafted 6. Ward NS. Functional reorganization of the cerebral motor system after
stroke. Curr Opin Neurol. 2004;17:725–730.
hNSCs could exert neurotrophic and/or activity-mediated 7. Rossini PM, Altamura C, Ferreri F, Melgari JM, Tecchio F, Tombini M,
effects on a HI-damaged local network and enhance axonal Pasqualetti P, Vernieri F. Neuroimaging experimental studies on brain
sprouting. Based on these observations, it seems reasonable plasticity in recovery from stroke. Eura Medicophys. 2007;43:241–254.
8. Nudo RJ, Wise BM, SiFuentes F, Milliken GW. Neural substrates for the
to propose that the newly innervated and recruited area of the effects of rehabilitative training on motor recovery after ischemic infarct.
contralesional hemisphere is becoming part of a reorganized Science. 1996;272:1791–1794.
network promoting motor recovery. 9. Brown CE, Li P, Boyd JD, Delaney KR, Murphy TH. Extensive turnover
of dendritic spines and vascular remodeling in cortical tissues recovering
Our data are in line with previous studies that show an from stroke. J Neurosci. 2007;27:4101– 4109.
increase in the resident microglia/macrophage cells (CD11b) 10. Kleim JA, Bruneau R, VandenBerg P, MacDonald E, Mulrooney R,
in ischemic animals that received neurosphere-derived cells.46 Pocock D. Motor cortex stimulation enhances motor recovery and reduces
Capone et al suggest that microglia activation is required for peri-infarct dysfunction following ischemic insult. Neurol Res. 2003;25:
789 –793.
neurosphere graft neuroprotective action through secretion of 11. Cramer SC, Chopp M. Recovery recapitulates ontogeny. Trends
growth factors, including insulin growth factor-1, vascular Neurosci. 2000;23:265–271.
endothelial growth factor-A, transforming growth factor-␤1, 12. Daadi MM, Steinberg GK. Manufacturing neurons from human
embryonic stem cells: biological and regulatory aspects to develop a safe
and brain-derived neurotrophic factor.46 Indeed, brain- cellular product for stroke cell therapy. Regen Med. 2009;4:251–263.
derived neurotrophic factor treatment of HI neonates im- 13. Carmichael ST, Wei L, Rovainen CM, Woolsey TA. New patterns of
proves spatial memory47 and selective ablation of microglia intracortical projections after focal cortical stroke. Neurobiol Dis. 2001;
8:910 –922.
through the mutant thymidine kinase gene driven by myeloid- 14. Dancause N, Barbay S, Frost SB, Plautz EJ, Chen D, Zoubina EV, Stowe
specific CD11b promoter exacerbated ischemic injury.48 AM, Nudo RJ. Extensive cortical rewiring after brain injury. J Neurosci.
Thus, these findings support the notion that microglia play a 2005;25:10167–10179.
dual role, proinflammatory or anti-inflammatory/neurotro- 15. Xiao J, Nan Z, Motooka Y, Low WC. Transplantation of a novel cell line
population of umbilical cord blood stem cells ameliorates neurological
phic, depending on their state of activation and functional deficits associated with ischemic brain injury. Stem Cells Dev. 2005;14:
phenotype.24 722–733.
Daadi et al Neural Stem Cells in Neonatal Hypoxia Ischemia 523

16. Napieralski JA, Butler AK, Chesselet MF. Anatomical and functional 32. West MJ, Slomianka L, Gundersen HJ. Unbiased stereological estimation
evidence for lesion-specific sprouting of corticostriatal input in the adult of the total number of neurons in the subdivisions of the rat hippocampus
rat. J Comp Neurol. 1996;373:484 – 497. using the optical fractionator. Anat Rec. 1991;231:482– 497.
17. Liu Z, Li Y, Zhang X, Savant-Bhonsale S, Chopp M. Contralesional 33. Lin L, Doherty D, Lile J, Bektesh S, Collins F. GDNF: a glial cell
axonal remodeling of the corticospinal system in adult rats after stroke line-derived neurotrophic factor for midbrain dopaminergic neurons.
and bone marrow stromal cell treatment. Stroke. 2008;39:2571–2577. Science. 1993;260:1130 –1132.
18. Kawamata T, Dietrich WD, Schallert T, Gotts JE, Cocke RR, Benowitz 34. Chopp M, Li Y, Zhang ZG. Mechanisms underlying improved recovery
LI, Finklestein SP. Intracisternal basic fibroblast growth factor enhances of neurological function after stroke in the rodent after treatment with
functional recovery and up-regulates the expression of a molecular neurorestorative cell-based therapies. Stroke. 2009;40(suppl):S143–145.
marker of neuronal sprouting following focal cerebral infarction. Proc 35. Buhnemann C, Scholz A, Bernreuther C, Malik CY, Braun H, Schachner
Natl Acad Sci U S A. 1997;94:8179 – 8184. M, Reymann KG, Dihne M. Neuronal differentiation of transplanted
embryonic stem cell-derived precursors in stroke lesions of adult rats.
19. Chen P, Goldberg DE, Kolb B, Lanser M, Benowitz LI. Inosine induces
Brain. 2006;129:3238 –3248.
axonal rewiring and improves behavioral outcome after stroke. Proc Natl
36. Park KI, Teng YD, Snyder EY. The injured brain interacts reciprocally
Acad Sci U S A. 2002;99:9031–9036.
with neural stem cells supported by scaffolds to reconstitute lost tissue.
20. Lee JK, Kim JE, Sivula M, Strittmatter SM. NOGO receptor antagonism
Nat Biotechnol. 2002;20:1111–1117.
promotes stroke recovery by enhancing axonal plasticity. J Neurosci. 37. Dijkhuizen RM, Singhal AB, Mandeville JB, Wu O, Halpern EF, Fin-
2004;24:6209 – 6217. klestein SP, Rosen BR, Lo EH. Correlation between brain reorganization,
21. Wiessner C, Bareyre FM, Allegrini PR, Mir AK, Frentzel S, Zurini M, ischemic damage, and neurologic status after transient focal cerebral
Schnell L, Oertle T, Schwab ME. Anti-NOGO-A antibody infusion 24 ischemia in rats: a functional magnetic resonance imaging study.
hours after experimental stroke improved behavioral outcome and corti- J Neurosci. 2003;23:510 –517.
cospinal plasticity in normotensive and spontaneously hypertensive rats. 38. Gerloff C, Bushara K, Sailer A, Wassermann EM, Chen R, Matsuoka T,
J Cereb Blood Flow Metab. 2003;23:154 –165. Waldvogel D, Wittenberg GF, Ishii K, Cohen LG, Hallett M. Multimodal
22. Offner H, Subramanian S, Parker SM, Afentoulis ME, Vandenbark AA, imaging of brain reorganization in motor areas of the contralesional
Hurn PD. Experimental stroke induces massive, rapid activation of the hemisphere of well recovered patients after capsular stroke. Brain. 2006;
peripheral immune system. J Cereb Blood Flow Metab. 2006;26: 129:791– 808.
654 – 665. 39. Butefisch CM, Netz J, Wessling M, Seitz RJ, Homberg V. Remote
23. Bona E, Andersson AL, Blomgren K, Gilland E, Puka-Sundvall M, changes in cortical excitability after stroke. Brain. 2003;126:470 – 481.
Gustafson K, Hagberg H. Chemokine and inflammatory cell response to 40. Cramer SC. Repairing the human brain after stroke: I. Mechanisms of
hypoxia–ischemia in immature rats. Pediatr Res. 1999;45:500 –509. spontaneous recovery. Ann Neurol. 2008;63:272–287.
24. Schwartz M. Macrophages and microglia in central nervous system 41. Z’Graggen WJ, Fouad K, Raineteau O, Metz GA, Schwab ME, Kartje
injury: are they helpful or harmful? J Cereb Blood Flow Metab. 2003; GL. Compensatory sprouting and impulse rerouting after unilateral py-
23:385–394. ramidal tract lesion in neonatal rats. J Neurosci. 2000;20:6561– 6569.
25. Wang X, Feuerstein GZ. The janus face of inflammation in ischemic 42. Lee RG, van Donkelaar P. Mechanisms underlying functional recovery
brain injury. Acta Neurochir Suppl. 2004;89:49 –54. following stroke. Can J Neurol Sci. 1995;22:257–263.
26. Daadi MM, Maag AL, Steinberg GK. Adherent self-renewable human 43. Rossini PM, Dal Forno G. Integrated technology for evaluation of brain
embryonic stem cell-derived neural stem cell line: functional engraftment function and neural plasticity. Phys Med Rehabil Clin N Am. 2004;15:
263–306.
Downloaded from http://ahajournals.org by on January 15, 2020

in experimental stroke model. PLoS ONE. 2008;3:e1644.


27. Daadi MM, Li Z, Arac A, Grueter BA, Sofilos M, Malenka RC, Wu JC, 44. Yasuhara T, Hara K, Maki M, Mays RW, Deans RJ, Hess DC, Carroll JE,
Borlongan CV. Intravenous grafts recapitulate the neurorestoration
Steinberg GK. Molecular and magnetic resonance imaging of human
afforded by intracerebrally delivered multipotent adult progenitor cells in
embryonic stem cell-derived neural stem cell grafts in ischemic rat brain.
neonatal hypoxic–ischemic rats. J Cereb Blood Flow Metab. 2008;28:
Mol Ther. 2009;17:1282–1291.
1804 –1810.
28. Vannucci RC, Connor JR, Mauger DT, Palmer C, Smith MB, Towfighi J,
45. Maier IC, Baumann K, Thallmair M, Weinmann O, Scholl J, Schwab ME.
Vannucci SJ. Rat model of perinatal hypoxic–ischemic brain damage. Constraint-induced movement therapy in the adult rat after unilateral
J Neurosci Res. 1999;55:158 –163. corticospinal tract injury. J Neurosci. 2008;28:9386 –9403.
29. Daadi MM, Lee SH, Arac A, Grueter BA, Bhatnagar R, Maag AL, Schaar 46. Capone C, Frigerio S, Fumagalli S, Gelati M, Principato MC, Storini C,
B, Malenka RC, Palmer TD, Steinberg GK. Functional engraftment of the Montinaro M, Kraftsik R, De Curtis M, Parati E, De Simoni MG.
medial ganglionic eminence cells in experimental stroke model. Cell Neurosphere-derived cells exert a neuroprotective action by changing the
Transplant. 2009;18:815– 826. ischemic microenvironment. PLoS ONE. 2007;2:e373.
30. Daadi MM, Weiss S. Generation of tyrosine hydroxylase-producing 47. Almli CR, Levy TJ, Han BH, Shah AR, Gidday JM, Holtzman DM.
neurons from precursors of the embryonic and adult forebrain. BDNF protects against spatial memory deficits following neonatal hyp-
J Neurosci. 1999;19:4484 – 4497. oxia–ischemia. Exp Neurol. 2000;166:99 –114.
31. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using 48. Lalancette-Hebert M, Gowing G, Simard A, Weng YC, Kriz J. Selective
real-time quantitative PCR and the 2(⫺Delta Delta C[T]) method. ablation of proliferating microglial cells exacerbates ischemic injury in
Methods. 2001;25:402– 408. the brain. J Neurosci. 2007;27:2596 –2605.

You might also like