You are on page 1of 14

doi:10.

1093/brain/awm306 Brain (2008), 131, 616 ^ 629

Anti-inflammatory mechanism of intravascular neural


stem cell transplantation in haemorrhagic stroke
Soon-Tae Lee,1,2,3,* Kon Chu,1,2,* Keun-Hwa Jung,1,2,4 Se-Jeong Kim,1 Dong-Hyun Kim,1 Kyung-Mook Kang,1
Nan Hyung Hong,1 Jin-Hee Kim,1,2 Jae-Joon Ban,1 Hee-Kwon Park,1,2 Seung U. Kim,5,6 Chung-Gyu Park,7
Sang Kun Lee,1,2 Manho Kim1,2 and Jae-Kyu Roh1,2
1
Stroke & Stem Cell Laboratory, Clinical Research Institute, Stem Cell Research Center, Department of Neurology, Seoul
National University Hospital, 2Program in Neuroscience, Neuroscience Research Institute of SNUMRC, Seoul National
University, 3Program in Public Health Service, Seoul National Hospital, 4Department of Epidemic Intelligence Service, Korea
Center for Disease Control and Prevention, Seoul, 5Institute for Regenerative Medicine, Gachon Medical University, Inchon,
South Korea, 6Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver,
Canada and 7Department of Microbiology and Immunology, Xenotransplantation Research Center, Transplantation Research
Institute, Tumor Immunity Medical Research Center, Seoul National University, Seoul, South Korea

*These authors contributed equally to this work.

Correspondence to: Jae-Kyu Roh, MD, PhD, Department of Neurology, Seoul National University Hospital, 28, Yongon-Dong,
Chongro-Gu, Seoul, 110 -744, Republic of Korea

Downloaded from by guest on November 6, 2014


E-mail: rohjk@snu.ac.kr

Neural stem cell (NSC) transplantation has been investigated as a means to reconstitute the damaged brain
after stroke. In this study, however, we investigated the effect on acute cerebral and peripheral inflammation
after intracerebral haemorrhage (ICH). NSCs (H1 clone) from fetal human brain were injected intravenously
(NSCs-iv, 5 million cells) or intracerebrally (NSCs-ic, 1 million cells) at 2 or 24 h after collagenase-induced ICH
in a rat model. Only NSCs-iv-2 h resulted in fewer initial neurologic deteriorations and reduced brain oedema
formation, inflammatory infiltrations (OX- 42, myeloperoxidase) and apoptosis (activated caspase-3, TUNEL)
compared to the vehicle-injected control animals. Rat neurosphere-iv-2 h, but not human fibroblast-iv-2 h, also
reduced the brain oedema and the initial neurologic deficits. Human NSCs-iv-2 h also attenuated both cerebral
and splenic activations of tumour necrosis factor-alpha (TNF-a), interleukin- 6 (IL- 6), and nuclear factor-kappa B
(NF-kB). However, we observed only a few stem cells in brain sections of the NSCs-iv-2 h group; in the main,
they were detected in marginal zone of spleens. To investigate whether NSCs interact with spleen to reduce
cerebral inflammation, we performed a splenectomy prior to ICH induction, which eliminated the effect of
NSCs-iv-2 h transplantation on brain water content and inflammatory infiltrations. NSCs also inhibited in vitro
macrophage activations after lipopolysaccharide stimulation in a cell-to-cell contact dependent manner. In sum-
mary, early intravenous NSC injection displayed anti-inflammatory functionality that promoted neuroprotec-
tion, mainly by interrupting splenic inflammatory responses after ICH.

Keywords: neural stem cell; spleen; cerebral inflammation; intracerebral haemorrhage; macrophage

Abbreviations: bFGF = basic fibroblast growth factor; ChAT = choline acetyltransferase; DMEM = Dulbecco’s Modified
Eagle’s Medium; EAE = experimental autoimmune encephalitis; EGF = epidermal growth factor; GFAP = glial fibrillary acidic
protein; HPF = high power field; ICH = intracerebral haemorrhage; IL = interleukin; LPS = lipopolysaccharide;
MPO = myeloperoxidase; MLPT = modified limb placing test; NF-kB = nuclear factor-kappaB; NSCs = neural stem cells;
SDF-1a = stromal cell derived factor-1alpha; TGF = tumour growth factor; TNF-a = tumour necrosis factor-alpha;
TUNEL = terminal deoxynucleotidyl transferase biotin-dUTP nick end labelling
Received June 25, 2007. Revised November 15, 2007. Accepted November 26, 2007. Advance Access publication December 21, 2007

ß The Author (2007). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: journals.permissions@oxfordjournals.org
Neural stem cells and spleen Brain (2008), 131, 616 ^ 629 617

Introduction cerebral and peripheral inflammatory response in acute


Neural stem cell (NSC) transplantation has been studied as stroke has important therapeutic relevance that should be
a promising tool for inducing regeneration of damaged clarified before developing NSC transplantation strategies.
brain tissue in many neurological disorders (Miller, 2006). In this study, we examined the influence of systemic NSC
We have shown that the intravenous transplantation of transplantation on brain and spleen inflammatory reactions
human NSCs promotes neurological recovery with cell during the acute period of ICH, and we observed that NSCs
replacement in intracerebral haemorrhage (ICH) (Jeong have an important ‘bystander’ anti-inflammatory effect on
et al., 2003). However, recent evidence indicates that NSC the spleen-macrophage system.
transplantation may protect the CNS from inflammatory
damage via a ‘bystander’ mechanism rather than by direct Material and Methods
cell replacement (Martino and Pluchino, 2006). NSCs
Human neural stem cell culture
appear to rescue degenerating neurons by modulating
All experimental procedures were approved by the Care of
the host environment by adopting a chaperone-like role Experimental Animals Committee at Seoul National University
(Ourednik et al., 2002; Hagan et al., 2003). In experimental Hospital and human cell use was approved by our institutional
autoimmune encephalomyelitis (EAE) models, NSCs exert review board. Primary dissociated neural stem cells were prepared
immune-like functions and induce the apoptosis of blood- from the ventricular zone (VZ) of fetal human brain (15 weeks
borne encephalitogenic T cells (Pluchino et al., 2003, 2005), gestation) and maintained. Detailed characteristics of this human
as well as decrease CNS inflammation via peripheral NSC line (H1 clone) have been described elsewhere (Ourednik
suppression of the adaptive immune response (Einstein et al., 2002; Jeong et al., 2003; Chu et al., 2004b,c,d; Imitola et al.,
2004; Chu et al., 2005; Lee et al., 2005; Parker et al., 2005; Lee
et al., 2007). This indicates the feasibility of using NSCs to
et al., 2006a). In brief, suspensions of dissociated NSCs
reduce cerebral inflammation and secondary brain damage
(5  105 cells per ml), isolated from the telencephalic VZ of a
after stroke. However, no previous study has been per- 15-week human fetal cadaver, were cultured in 98% DMEM

Downloaded from by guest on November 6, 2014


formed on the possible anti-inflammatory potential of (Dulbecco’s Modified Eagle’s Medium)/F12 (GIBCO, Invitrogen,
NSCs on innate inflammatory response after acute stroke. Carlsbad, CA, USA), 1% N2 supplement (GIBCO), 1% penicillin/
Intracerebral haemorrhage (ICH) represents at least streptomycin (GIBCO), 8 mg/ml heparin (Sigma), 10 ng/ml
15–20% of all strokes in the Caucasian population leukemia inhibiting factor (Chemicon-Millipore), 20 ng/ml basic
(Qureshi et al., 2001), and occurs with increasing frequency fibroblast growth factor (bFGF, Calbiochem) in uncoated 25 cm2
as a complication of the thrombolytic treatment of flasks (FalconTM, BD Bioscience) at 37 C in 5% CO2. NSCs were
cultured in poly-L-lysine-coated culture dishes or flasks as single
ischaemic stroke (Lapchak, 2002). ICH induces neurological
cells or large clusters, which can be subcultured and passaged
damage due to local tissue deformation and the subsequent
weekly over a period of 6 months. This cell line can give rise to
developments of excitotoxicity, apoptosis, and inflamma- multiple neural cell types throughout the neural axis during
tion (Aronowski and Hall, 2005). Of these, ICH-induced development and has the ability to reconstitute these regions when
inflammation appears to be a key factor of secondary brain perturbed (Snyder et al., 1997; Ourednik et al., 2001; Parker et al.,
damage, which suggests that anti-inflammatory approaches 2005). In addition, NSC clones generated using the same method
may lessen the outcome of haemorrhagic stroke (Jung et al., expresses all commonly accepted NSC markers (nestin, musashi,
2004; Chu et al., 2004a; Aronowski and Hall, 2005; Wang vimentin, etc.) and can form neurospheres (Kitchens et al., 1994;
and Dore, 2007; Sinn et al., 2007). Parker et al., 2005). When injected into rats for histologic analysis,
Both innate and adaptive inflammatory mechanisms NSCs were pre-labelled with VybrantÕ DiO cell-labelling solution
(Invitrogen) before transplantation, according to the manufac-
have been clearly shown to participate in the progression
turer’s instructions.
of cellular injury after stroke (Hallenbeck et al., 2005).
A prominent inflammatory response, which includes the Rat neurosphere and human fibroblast culture
activation of resident brain microglia and inflammatory We cultured neurospheres using the published method (Singec
infiltration into the brain, is initiated by neutrophils et al., 2006), on postnatal days 5–7 from Sprague-Dawley rats
followed by macrophages (Del Bigio et al., 1996). This (Orient Bio, South Korea). We used microdissected sections
process is accompanied by the massive, rapid activation of containing the subventricular zone (SVZ) for the culture, and
the peripheral immune system and the dynamic and wide- used DMEM/F12 medium (GIBCO) supplemented with B27
spread activation of inflammatory cytokines and chemo- (Invitrogen), 20 ng/ml bFGF (PeproTech, Rocky Hill, NJ, USA),
kines in spleen (Offner et al., 2006). In particular, the and 20 ng/ml epidermal growth factor (EGF, PeproTech) for the
spleen is the key organ that activates tumour necrosis culture medium. For passaging, we collected neurospheres with
37 C warmed 0.05% trypsin, centrifuged them, and mechanically
factor-alpha (TNF-a) and nuclear factor-kappa B (NF-kB)
triturated them, and we plated single cells again after counting
in systemic inflammatory response (Huston et al., 2006; cells. For the transplantation, neurospheres (at three passages)
Tracey, 2007). TNF-a is one of the major inflammatory were dissociated into single cells, and suspensions containing
mediators in stroke (Zheng and Yenari, 2004), and is largely 5  106 cells in 0.5 ml PBS were used. Primary human fibroblasts
produced by infiltrating macrophages (Gregersen et al., were cultured from a normal skin biopsy specimen with the
2000). Thus, the issue as to whether NSCs can modulate patient’s consent using the published method (Lee et al., 2006c).
618 Brain (2008), 131, 616 ^ 629 S.-T. Lee et al.

Cell suspensions containing 5  106 fibroblasts in 0.5 ml PBS were group) (Choudhri et al., 1997; Asahi et al., 2000; Lee et al., 2006b).
used for transplantation. In brief, hemispheric brain tissue was acquired from normal rats
who had been subjected to complete transcardial perfusion for the
removal of intravascular blood. Incremental volumes of homol-
Induction of ICH and human NSC ogous blood (0–200 ml) were added to each hemispheric sample,
transplantation along with PBS, to a total volume of 3 ml, followed by 30 s of
One hundred and seventy nine male Sprague-Dawley rats (Orient homogenization, 1 min of sonication on ice, and 30 min of
Bio), each weighing 210–240 g, were utilized. Experimental ICH centrifugation at 12 000 r.p.m. Drabkin’s reagent (1.6 ml; Sigma)
was induced via the stereotaxic intrastriatal administration of was then added to 0.4 ml aliquots, and allowed to stand for 15 min
bacterial collagenase type VII (0.23 IU dissolved in 1 ml saline, at room temperature. Optical density (OD) was then measured
Sigma), as described previously (Jeong et al., 2003; Chu et al., and recorded at 540 nm with a spectrophotometer (Molecular
2004a; Jung et al., 2004; Lee et al., 2006b). Sham ICH was induced Devices). These procedures showed a linear relationship between
with a stereotaxic needle insertion and an injection of equal the haemoglobin concentrations in the perfused rat brain and the
volume (1 ml) of saline instead of collagenase. Two or 24 h after volume of added blood. Measurements from the perfused ICH
ICH induction, human NSCs were administered intravenously brains were then compared with this standard curve, allowing us
(via tail vein, 5 million cells in 0.5 ml PBS/rat) or intracerebrally to obtain data regarding haemorrhage volume (in ml).
[1 million cells in 2 ml PBS/rat, divided at three co-ordinates
(anterior–posterior, medial–lateral, dorsal–ventral): (+1 mm,
+1.4 mm, 3.0 mm), (+0.2 mm, +2.0 mm, 3.0 mm), (0.4 mm,
Measurements of brain water content
+2.2 mm, 3.0 mm), respectively], as previously described (Jeong Water contents were measured at 3 days after ICH (n = 6 for each
et al., 2003; Kelly et al., 2004), namely intravenous transplantation group), as previously described (Jeong et al., 2003; Chu et al.,
at 2 h (the NSCs-iv-2 h group), intravenous transplantation at 2004a; Jung et al., 2004; Lee et al., 2006b). In brief, the rats were
24 h (the NSCs-iv-24 h group), intracerebral transplantation at 2 h anesthetized and sacrificed via decapitation. The rats’ brains were
(the NSCs-ic-2 h group), or intracerebral transplantation at 24 h removed immediately, and divided into two hemispheres along the
(the NSCs-ic-24 h group). Vehicle was administered to the ICH- midline, after which the cerebellum was removed from each brain.

Downloaded from by guest on November 6, 2014


vehicle group by injecting 0.5 ml of PBS via tail vein at 2 h after The brain samples were then immediately weighed on an
the ICH induction. electronic analytical balance to obtain wet weights, then dried in
a gravity oven at 100 C for 24 h, in order to obtain dry weights.
Water contents were expressed as a percentage of wet weight: the
Splenectomy formula used to calculate the water contents was as follows: (wet
Rats were anesthetized with ketamine and xylazine (Sigma). weight  dry weight)/(wet weight).
Spleens were identified by midline laparotomy and removed after
appropriate blood vessel ligation. Tissue preparation and immunohistochemistry
At 3 days or 35 days after ICH, rats (n = 4 in each of the groups)
Behavioural testing were re-anesthetized and perfused through the heart with 50 ml of
Behavioural testing (n = 10 for each group) was conducted weekly cold saline and 50 ml of 4% paraformaldehyde, in 0.1 mol/l PBS.
for 5 weeks after ICH using modified limb placing tests (MLPT), as Sections (30 mm thickness) of brains were immunostained
previously described (Jeong et al., 2003; Chu et al., 2004a; Lee et al., with antibodies against myeloperoxidase (MPO; DAKO), Ox-42
2006b). This test consists of two limb-placing tasks, which assess the (Chemicon), or activated caspase-3 (BD biosciences), and sections
sensorimotor integration of the forelimb and the hindlimb, by of spleens were stained with antibodies against ED1 (Serotec,
monitoring the subject’s responses to tactile and proprioceptive Oxford, UK), CD11b (Chemicon), choline acetyltransferase
stimulation. First, the rat is suspended 10 cm above a table, and the (ChAT, Serotec), and human nuclear antigen (HuN, Chemicon),
stretch of the forelimbs towards the table is observed and evaluated: as previously described (Jeong et al., 2003; Chu et al., 2004a; Jung
a normal stretch is scored as 0 points; abnormal flexion is scored et al., 2004; Lee et al., 2006b). The terminal deoxynucleotidyl
as 1 point. Next, the rat is positioned along the edge of the table, transferase biotin-dUTP nick end labelling (TUNEL) was used to
with its forelimbs suspended over the edge, and is then allowed to detect in situ DNA fragmentation using a commercial kit (R&D
move freely. Each forelimb (forelimb-second task, hindlimb-third systems). For fluorescent double staining of TUNEL and cell
task) is gently pulled down, and retrieval and placement is evaluated. markers, we used anti-NeuN (Chemicon), anti-glial fibrillary
Finally, the rat is placed near the table edge, in order to assess the acidic protein (GFAP, Chemicon) antibodies, and a fluorescent
lateral placement of the forelimb. The three tasks are scored in the TUNEL assay kit (Chemicon). Fluoro-Jade C staining was
following manner: normal performance is scored as 0 points; delayed performed according to the published protocol to detect neurons
(at least 2 s) and/or incomplete performance is scored as 1 point; no undergoing degeneration (Schmued et al., 2005). To confirm any
performance is scored as 2 points. A total score 7 points indicates cell-to-cell contact or co-labelled cell markers, sections were
maximal neurological deficit, and a score of 0 points denotes normal analyzed with a laser-scanning confocal microscopic (LSM 510,
performance. Carl Zeiss microimaging, Thornwood, NY, USA) imaging system.
One coronal brain section taken through centers of needle tracts
and two adjacent sections (1 mm width) were analysed by
Spectrophotometric assay of haemorrhage counting marker-specific cells throughout entire sections (three
volume sections per antibody staining). Total counts were converted to
Haemorrhage volumes were quantified at 3 days post-ICH using a cell densities in order to facilitate quantification. We investigated
spectrophotometric assay, as described previously (n = 6 for each NSC distributions in brain, spleen, lungs, livers and mesenteric
Neural stem cells and spleen Brain (2008), 131, 616 ^ 629 619

lymph nodes of the NSCs-iv-2 h groups with DAPI staining at 3 cells for each sample were analyzed on a FACS II flow cytometer
or 35 days after ICH. ED1+-activated macrophages in the spleen (BD Bioscience).
were counted in three marginal zone areas immediately adjacent
to the white pulp in at least three sections per animal using a Co-culture of rat macrophages with NSCs
magnification of 400 (high power field, HPF) over a microscopic
Peritoneal macrophages were isolated from Sprague-Dawley rats,
field. For determining hemispheric atrophy, three sections through
as previously described (Falcone and Ferenc, 1998). Briefly, rats
the needle entry site were Nissl stained at 35 days after ICH
were injected i.p. with 3 ml of 4% thioglycollate. Four days later
(n = 6 per group). The total hemispheric areas of each section were
cells were harvested by lavage with cold PBS. Peritoneal cells were
traced and measured using image analyzer (Image Pro-Plus,
recovered by centrifugation and resuspended in 24-well plates in
Media Cybernetics, Bethesda, MD, USA) as described previously
500 ml of RPMI-10% FBS containing penicillin (200 U/ml) and
(Matsushita et al. 2000; Chu et al. 2004a; Jung et al. 2004). The
streptomycin (0.2 mg/ml). Cells were allowed to adhere for 2 h and
hemispheric atrophy was expressed as a percentage of contralateral
then washed free of non-adherent cells. Adherent cells were then
hemispheric area.
re-seeded (106 cells per well) in 500 ml of RPMI. Two hours before
or after lipopolysaccharide (LPS) stimulation, NSCs (106 cells per
Western blotting and RT-PCR well) were added to these macrophages as mixed culture. For the
Twenty four hours after ICH induction, rats were sacrificed by double chamber system, equal numbers of NSCs were plated into
decapitation, and brains, spleens and mesenteric lymph nodes transwell membrane inserts (Costar) with a 0.4 mm pore size to
were immediately extracted (n = 4 in each group). Homogenates avoid direct cell-to-cell interactions and to allow the diffusion of
of ipsilateral (haemorrhagic) hemispheres and spleens were soluble factors between two cell populations (Shen et al., 2004).
serially processed for western blotting, as described previously LPS was added to the cultures at a final concentration of
(Jung et al., 2004; Lee et al., 2006d), using anti-NF-kB (1 : 200; 100 ng/ml. Supernatants were collected 4 or 24 h after adding LPS
BD Biosciences) and anti-b-actin antibody (Santa Cruz Biotech- and prepared for TNF-a and IL-6 ELISA using ELISA kits (R&D
nology). Relative optical densities were calculated versus measured Systems).
values of b-actin. We also conducted RT-PCR for TNF-a,

Downloaded from by guest on November 6, 2014


interleukin (IL)-1b, IL-4, IL-6, tumour growth factor (TGF)-b1, Statistical analysis
Fas and Fas ligand (FasL) at 24 h after ICH induction (n = 3 in
All data in this study are presented as means  SD. Error bars in
each group), as previously described (Lee et al., 2006d,). In brief,
the figures mean SD except Fig 1, in which the error bars are SE.
total RNA was isolated from homogenates (50 mg) of haemor-
The Mann–Whitney U-test was used for inter-group comparisons
rhagic hemispheres, spleens, or mesenteric lymph nodes using TRI
and we did not specify the test in these cases. For a nonparametric
reagent (Sigma). RT-PCRs were conducted using First strand
comparison among three and more unpaired groups, we used
cDNA Synthesis kits (Roche, Basel, Switzerland) over 25 cycles of
Kruskall-Wallis analysis of variances (ANOVA) and specified
95 C, 58 C and 72 C for 40 s each using the following primer sets:
the test. When P-values from Kruskall–Wallis was 50.05, Mann–
TNF-a: 50 -TAC TGA ACT TCG GGG TGA TTG GTC C-30 (sense)
Whitney U-test was further used for post-hoc inter-group
and 50 -CAG CCT TGT CCC TTG AAG AGA ACC-30 (antisense);
comparisons. A two-tailed probability value of 50.05 was
IL-1b: 50 -ATG GCA ACT GTC CCT GAA CT (sense) and 50 -GTC
considered to be significant.
ATC ATC CCA CGA GTC AC (antisense); IL-4: 50 -CTG CTT
TCT CAT ATG TAC CGGG (sense) and 50 -TTT CAG TGT TGT
GAG CGT GG (antisense); IL-6: 50 -CTT GGG ACT GAT GTT Results
GTT GAC-30 (sense) and 50 -CTC TGA ATG ACT CTG GCT TTG-
NSCs-iv-2 h reduced initial neurologic
30 (antisense); TGF-b1: 50 -CTA ATG GTG GAC CGC AAC AAC-
30 (sense) and 50 -CGG TTC ATG TCA TGG ATG GTG-30 deterioration
(antisense); Fas: 50 -CAA GGG ACT GAT AGC ATC TTT GAG We previously reported favourable neurologic recovery in
G-30 (sense) and 50 -TCC AGA TTC AGG GTC ACA GGT TG-30 the intravenous NSCs transplantation at 24 h after the ICH
(antisense); Fas-L: 50 -CAG CCC CTG AAT TAC CCA TGT C-30 induction (NSCs-iv-24 h group) compared to the vehicle-
(sense) and 50 -CAC TCC AGA GAT CAA AGC AGT TC-30 treated ICH group (ICH-vehicle) (Jeong et al., 2003). Thus,
(antisense). Transcript levels were quantified by analyzing the we first investigated whether an earlier NSCs transplanta-
scanned photographs of gels using appropriate imaging software
tion at 2 h after ICH (NSCs-iv-2 h) can induce more
(Molecular AnalystÕ , Bio-Rad). Relative optical densities were
favourable results. Although the ICH-vehicle group scored
determined by comparing measured values with GAPDH mean
values. over six points on the MLPT at 1 day after ICH
(6.85  0.38), the NSCs-iv-2 h group exhibited less pro-
found initial deficits (4.66  0.98, P50.01, Fig. 1). The
FACS analysis NSCs-iv-2 h group then continued to recover, and the
At 3 days after the ICH induction, single cell suspensions (5  105
difference between the two groups was statistically sig-
cells) were prepared from spleens in phosphate buffered saline
(PBS) supplemented with 2% fetal calf serum (FCS) and 0.05%
nificant until at least 5 weeks after ICH (all P50.01).
sodium azide. To remove contaminating erythrocytes, the cell Although the MLPT score of NSCs-iv-24 h group started to
suspensions were subjected to osmotic lysis using a hypotonic differ from that of the ICH-vehicle group at 4 weeks, the
ammonium chloride solution. With using FITC anti-rat CD11b NSCs-iv-2 h group showed less neurologic deficits than the
(2 mg/ml, BD Biosciences) and Phycoerythrin (PE) anti-rat TNF-a NSCs-iv-24 h group from day 1 to day 35 (all P50.01). We
(2 mg/ml, eBioscience, San Diego, CA, USA) antibodies, 5  104 also investigated the neurologic deficits of the NSCs-ic-2 h
620 Brain (2008), 131, 616 ^ 629 S.-T. Lee et al.

and NSCs-ic-24 h groups for comparison. Although the were not different to those of the ICH-vehicle group
MLPT scores of these two groups started to differ from that (Fig. 1). In summary, only the NSCs-iv-2 h group showed
of the ICH-vehicle group at 21 or 28 days after the ICH, less initial neurologic deficits compared to the other groups.
the neurologic deficits during the early periods (day 1–14)
NSCs-iv-2 h reduced brain water content
Because the lower levels of neurologic deficits observed in
the NSCs-iv-2 h group at 1 day after ICH could not be
explained by cell replacement or regeneration, we hypothe-
sized that some form of initial neuroprotection or anti-
inflammation had been initiated in the NSCs-iv-2 h group.
Thus, we measured brain water contents at 3 days after
ICH, because water content represents brain oedema which
is one of the most important surrogate markers of
perihematomal inflammation and tissue damage and
peaks at 3–4 days after ICH (Xi et al., 2006). ANOVA
suggested an intergroup difference among the ipsilateral
brain water contents of the five groups (p50.05, Kruskall–
Wallis ANOVA, Fig. 2A). Mean brain water content in
ipsilateral (haemorrhagic) hemispheres was 81.63  0.55%
in the ICH-vehicle group, and 80.10  0.27% in the
NSCs-iv-2 h group (P50.01), whereas in contralateral
(non-haemorrhagic) hemispheres these were 79.63  0.36%

Downloaded from by guest on November 6, 2014


Fig. 1 Modified limb placing test (MLPT). At 24 h after ICH, the in the ICH-vehicle group and 79.30  0.25% in the NSCs-
NSCs-iv-2 h group exhibited less profound deficits than the ICH- iv-2 h group (P = 0.08), demonstrating that NSCs-iv-2 h
vehicle group, and then progressively recovered. The MLPT score
of NSCs-iv-24 h group started to differ from that of the ICH- significantly reduced brain oedema in haemorrhagic hemi-
vehicle group at 4 weeks after ICH. NSCs-iv-2 h group showed less spheres. However, the brain water contents in the other
neurologic deficits than NSCs-iv-24 h group from day 1 to day 35. groups, i.e. the NSCs-iv-24 h group [81.18  0.45% (ipsi-
The MLPT scores of the NSCs-ic-2 h and the NSCs-ic-24 h groups lateral); 79.58  0.26% (contralateral)], the NSCs-ic-2 h
started to differ from that of the ICH-vehicle group at 21 or 28 group [80.95  0.35% (ipsilateral); 79.82  0.31% (contra-
days respectively, and the neurologic deficits during the early
periods (day 1^14) were not different to those of the ICH-vehicle lateral)], and the NSCs-ic-24 h group [81.25  0.45%
group. P50.05, P50.01 versus ICH-vehicle. yP50.01 versus (ipsilateral); 79.93  0.36% (contralateral)], were not dif-
NSCs-iv-24 h. ôP50.05, ‰P50.01 versus NSCs-ic-2 h. ferent from those of the ICH-vehicle group. These results

Fig. 2 Brain water content and haemorrhage volume. Brain water contents were significantly lower in the ipsilateral (haemorrhagic)
hemispheres of the NSCs-iv-2 h group than in all other groups at 3 days after ICH (A). Water contents of the other groups were not
different from those of the ICH-vehicle group. NSC transplantations either intravenously or intracerebrally at 2 or 24 h did not influence
haemorrhagic volume (B). Intravenous injection of rat neurosphere cells at 2 h (ICH-rat neurosphere-iv-2 h group) also reduced the mean
brain water content in the ipsilateral hemisphere, while the human fibroblast injection (ICH-human fibroblast-iv-2 h group) showed no
significant effect on the brain water content compared to the vehicle injection (C). P50.01 versus all other groups.
Neural stem cells and spleen Brain (2008), 131, 616 ^ 629 621

Downloaded from by guest on November 6, 2014


Fig. 3 Histological analysis of inflammation and apoptosis. As representative photographs show (A), the NSC-iv-2 h group showed lower
numbers of OX 42+ and MPO+ inflammatory cells and lower levels of activated caspase-3+ cells and TUNEL+ apoptotic cells in perihe-
matomal regions at 3 days after ICH than the other four groups, as representative photographs show (B). The NSCs-iv-24 h group showed
reductions of MPO+ and caspase-3+ cells versus the ICH-vehicle group. The # signs indicate hematoma areas. Bar = 100 mm. P50.05
versus all other four groups. yP50.01 versus the ICH-vehicle group.

indicate that intravenous transplantation at 24 h, or Accordingly, NSCs attenuated the brain oedema formation
intracerebral. transplantation at 2 h or 24 h failed to and initial neurologic deficits regardless of their allogenic or
reduce brain oedema. Only early intravenous transplanta- xenogenic transplantation conditions.
tion effectively reduced the brain oedema.
According to the results of spectrophotometric assays of NSCs-iv-2 h reduced brain inflammatory
haemorrhage volumes at 3 days after ICH, NSC transplan- infiltration and apoptotic cell numbers
tation by either method (intravenous, intracerebral, 2 h and Histologic quantifications of inflammatory infiltrations
24 h) exerted no influence on haemorrhage volumes and apoptotic cells were undertaken in perihematomal
(P40.05 in Kruskall–Wallis ANOVA; all P-values 40.05 areas to confirm the anti-inflammatory effects of NSCs-
versus ICH-vehicle, Fig. 2B), suggesting that equal burdens iv-2 h at 3 days after ICH (Fig. 3A). Initially, we observed
of haemorrhage were initiated. intergroup differences among the five treatment groups in
To investigate whether allogenic neural progenitor cells the inflammatory/apoptotic cell counts including OX42,
can also reduce the brain oedema formation, we measured MPO, Caspase-3 and TUNEL (P50.05, Kruskall–Wallis
brain water contents in two more experimental groups made ANOVA), and further analyzed the values by intergroup
by intravenous injections of rat neurosphere cells (ICH-rat comparison. It was found that the NSCs-iv-2 h group
neurosphere-iv-2 h) or human fibroblasts (ICH-human possessed significantly lower numbers of OX42+ microglia
fibroblast-iv-2 h) at 2 h after the ICH. The ICH-rat neuros- and macrophages than the ICH-vehicle group (P50.05),
phere-iv-2 h group showed the reduced brain water content whereas the other groups showed no difference versus the
in the ipsilateral hemisphere (80.05  0.36%, P50.01 versus ICH-vehicle group (ICH-vehicle: 105.0  20.8 cells/mm2,
ICH-vehicle). However, the ipsilateral brain water contents NSCs-iv-2 h: 24.0  6.32 cells/mm2, NSCs-iv-24 h:
of the ICH-human fibroblast-iv-2 h group (81.01  0.36%) 79.8  13.8 cells/mm2, NSCs-ic-2 h: 103.8  14.4 cells/mm2,
was not significantly different to that of the ICH-vehicle NSCs-ic-24 h: 113.5  12.6 cells/mm2) (Fig. 3B).
group (P = 0.078). In addition, the ICH-rat neurosphere- MPO+ neutrophil counts in perihematomal areas
iv-2 h group, but not ICH-human fibroblast-iv-2 h group, revealed the most significant reduction in the NSCs-iv-2 h
showed the reduced initial neurologic deficits (MLPT score) group (ICH-vehicle: 212.5  29.9 cells/mm2, NSCs-iv-2 h:
compared to the ICH-vehicle group (ICH-rat neurosphere- 44.3  9.2 cells/mm2, NSCs-iv-24 h: 111.3  19.7 cells/mm2,
iv-2 h: 4.00  0.82, P50.01 versus ICH-vehicle; ICH-human NSCs-ic-2 h: 190.3  20.3 cells/mm2, NSCs-ic-24 h:
fibroblast-iv-2 h: 6.2  0.84, P = 0.104 versus ICH-vehicle). 148.9  66.6 cells/mm2, P50.05 between NSCs-iv-2 h and
622 Brain (2008), 131, 616 ^ 629 S.-T. Lee et al.

ICH-vehicle) (Fig. 3). The NSCs-iv-24 h group also showed haemorrhagic brains and spleens compared to the normal
a reduction in MPO+ count versus the ICH-vehicle group samples, with using RT-PCR supported by optical density
(P50.05), but this was less pronounced than of the NSCs- analysis (all P50.05, n = 4 per group) (Fig. 4A and B). In
iv-2 h group (P50.05 versus NSCs-iv-24 h). However, the lymph nodes, only IL-4 was upregulated (P50.05) and
NSCs-ic-2 h and NSCs-ic-24 h groups showed no reduction TNF-a, IL-1b, and IL-6 were unchanged after ICH. The
in MPO+ neutrophil infiltration. levels of TGF-b were not affected by the ICH induction in
An analysis of active caspase-3+ apoptotic cell numbers all the three kinds of tissue.
showed that the NSCs-iv-2 h and NSCs-iv-24 h groups had In brain samples, the NSCs-iv-2 h group showed
lower counts than the ICH-vehicle group (all P50.05), and decreases of 79% in TNF-a, 62% in IL-4, 47% in IL-6
the NSCs-iv-2 h group had lowest counts than NSCs-iv-24 h mRNA levels versus the ICH-vehicle group (all P50.05,
(P50.05) (ICH-vehicle: 72.0  10.6 cells/mm2, NSCs-iv-2 h: Fig. 4A and B). The NSCs-iv-2 h group also showed
17.8  3.1 cells/mm2, NSCs-iv-24 h: 38.0  6.7 cells/mm2, decreases in Fas and FasL mRNA levels compared to the
NSCs-ic-2 h: 76.5  9.4 cells/mm2, NSCs-ic-24 h: ICH-vehicle group (by 35% and 66%, respectively, P50.05,
63.5  11.0 cells/mm2) (Fig. 3). Supplementary fig. 2A). In spleens, the NSCs-iv-2 h group
TUNEL staining revealed a high density of positively- showed decreases of 85% in TNF-a and 58% in IL-6
stained cells within and at the peripheries of haemorrhagic mRNA levels versus the ICH-vehicle group (all P50.05,
lesions (Fig. 3A). By quantitative analysis, the NSCs-iv-2 h Fig. 4A and B). In the lymph nodes, the NSCs-iv-2 h
group exhibited a significantly lower number of TUNEL+ injection attenuated IL-4 levels by 69% (P50.05,
cells (82.5  43.7 cells/mm2) than the ICH-vehicle group Supplementary Fig. 2B). In addition, western blotting for
(314.5  50.8 cells/mm2; P50.05, Fig. 3B). However, NF-kB expressions in the brain and spleen samples showed
the NSCs-iv-24 h (254  44.6 cells/mm2), NSCs-ic-2 h that levels were significantly attenuated in the NSCs-iv-2 h
(349.5  63.6 cells/mm2), and NSCs-ic-24 h group compared to the ICH-vehicle group (36% decrease in
(325.5  32.7 cells/mm2) groups were similar to the ICH-

Downloaded from by guest on November 6, 2014


the brains and 81% in the spleens, all P50.05 versus ICH-
vehicle group. The perihematomal TUNEL+ cells were vehicle, Fig. 4C).
mostly positive for the neuronal marker (NeuN) To investigate whether NSCs-iv-2 h changed TNF-a
(Supplementary Fig. 1A). Nevertheless, overall immunor- expressions in the spleen macrophages, we performed
eactivity against NeuN was diminished in the perihemato- flow cytometric analysis of splenocytes using anti-CD11b
mal area because of the loss of neuronal nuclear and/or and anti-TNF-a antibodies. In the ICH-vehicle spleen,
cytoskeletal structure (Matsushita et al., 2000), and it 24.2% of CD11b+ macrophages expressed TNF-a, while
seemed to be inaccurate to quantify TUNEL+NeuN+ cells. 17.6% of CD11b+ macrophages expressed TNF-a in the
Thus, we used Fluoro-Jade C staining to detect normal spleen (Fig. 4D). In the NSCs-iv-2 h group,
degenerating neurons in the perihematomal area (Wang however, only 19.1% of CD11b+ macrophages expressed
and Tsirka, 2005). The ICH-NSCs-iv-2 h group had a lower TNF-a. In addition, the spleens from the NSCs-iv-2 h
number of Fluoro-Jade C stained neurons than the ICH- group showed reduced numbers of ED1+ activated macro-
vehicle group at 3 days after ICH (ICH-vehicle: phages in the marginal zone compared to those from the
176  33 cells/mm2, ICH-NSCs-iv-2 h: 49  18 cells/mm2, ICH-vehicle group (ICH-vehicle: 63  9 cells/HPF, NSCs-iv-
P50.05, Supplementary Fig. 1B). To confirm the sustained 2 h: 30  8 cells/HPF, p50.05, Supplementary Fig. 2C). In
tissue protection, we also measured the hemisphere summary, these findings demonstrated that NSCs-iv-2 h
atrophies of the ICH-vehicle and the NSCs-iv-2 h groups reduced the levels of the important inflammatory mediators
at 35 days after ICH. The NSCs-iv-2 h group exhibited in both the brain and the spleen.
lesser degree of hemispheric atrophies than did the ICH-
vehicle group (ICH-vehicle: 24.5  3.5%, NSCs-iv-2 h:
16.0  8.2%, P50.05). Accordingly, the NSCs-iv-2 h group
Distributions of injected NSCs
demonstrated the most prominent and reproducible anti- We previously observed that a number of NSCs migrated
inflammatory and neuroprotective effects. to perihematomal areas and differentiated into NeuN+,
Neurofilament+, or GFAP+ cells in NSCs-iv-24 h group
when we analyzed brains at 6 weeks after the transplanta-
NSCs-iv-2 h showed attenuated levels of tion (Jeong et al., 2003). However, at 1 day after the
cerebral and splenic inflammatory mediators transplantation, only occasional NSCs were observed in a
We then analysed changes in the levels of inflammatory cerebral ischaemia model (Jin et al., 2005), although robust
mediators induced by NSCs-iv-2 h in brains, spleens and in situ proliferation was observed from 7 days after
lymph nodes at 1 day after ICH. We have previously shown transplantation (Chu et al., 2004b). In the present study,
that ICH increases the expressions of TNF-a, IL-6, Fas and we analyzed the distribution of injected NSCs in the NSCs-
FasL in the brain samples at 1 day after ICH (Sinn et al., iv-2 h group at 3 days after ICH when their anti-
2007). In the present study, we could confirm the inflammatory effects were apparent. However, only small
upregulations of TNF-a, IL-1b, IL-4 and IL-6 in the numbers were observed in brain [41 cell per high-power
Neural stem cells and spleen Brain (2008), 131, 616 ^ 629 623

Downloaded from by guest on November 6, 2014


Fig. 4 Changes in the expressions of cerebral and splenic inflammatory mediators. RT-PCR (A) and the optical density measure
(B) revealed the upregulations of TNF-a, IL-1b, IL- 4 and IL- 6 in the haemorrhagic brains and spleens compared to the normal samples. In
lymph nodes, only IL- 4 was upregulated. The NSCs-iv-2 h group showed decreases of TNF-a, IL- 4 and IL- 6 mRNA levels in brain samples,
as well as decreases of TNF-a and IL- 6 mRNA levels in spleen samples (B). In the lymph nodes, the NSCs-iv-2 h injection attenuated the
IL- 4 upregulation. Western blotting for NF-kB expressions in the brain and spleen samples showed that the levels were significantly atte-
nuated in the NSCs-iv-2 h group compared to the ICH-vehicle group (C). In the flow cytometric analysis of splenocytes, 24.2% of CD11b+
macrophages expressed TNF-a in the ICH-vehicle group, while 17.6% of CD11b+ macrophages expressed TNF-a in the normal spleen
(D, from each single representative sample). In the NSCs-iv-2 h group, however, only 19.1% of CD11b+ macrophages expressed TNF-a.

P50.05 versus normal. yP50.05 versus ICH-vehicle.

field (HPF)], lung (2–3 cells per HPF), and liver (about 1 for human nuclear antigen, confirming that DiO -labelled
cell per HPF) (Fig. 5). This distribution is in accord with cells were transplanted human cells (Supplementary
previous observations in an EAE model, where more NSCs Fig. 3F). About 80% of the NSCs observed in the spleen
were found to be distributed to the systemic organs rather were positive for the cholinergic neuronal marker (ChAT)
than to the CNS just after transplantation (Pluchino et al., (Supplementary Fig. 3G and H). When we analyzed the
2005). However, we observe few NSCs in mesenteric lymph distribution of NSCs in the NSCs-ic-2 h and NSCs-ic-24 h
nodes. In an EAE model, many transplanted NSCs were groups at 3 days after ICH, we observed abundant NSCs in
found in lymph nodes during the early stage, although they the perihematomal area as well as transplantation foci of
disappeared over several days (Einstein et al., 2007). the brain, but not in the systemic organs (Supplementary
However, in the present study, large numbers of NSCs Fig. 3I).
were detected in spleen, especially in the marginal zone area
(20–30 cells per HPF). Because this splenic zone contains
many macrophages (Mebius and Kraal, 2005), we immu-
nostained the spleen macrophages with anti-CD11b Ab. The Splenectomy eliminated the effect of
results obtained confirmed that NSCs were located in the NSCs-iv-2 h transplantation
vicinity of marginal zone macrophages (Fig. 5F). In addition, Because of the splenic bias shown by NSCs and the
we found that a number of NSCs were in cell-to-cell contact observed attenuations of inflammatory mediators in spleen,
with CD11b+ spleen macrophages under the confocal we hypothesized that this organ may facilitate inflammatory
microscopic views (Fig. 5G). response modulation in our stroke model. To prove this,
At 35 days after the transplantation, those NSCs-iv-2 h we performed splenectomy 3 days prior to the ICH
cells were still detected in lungs, livers and spleens induction, and then measured brain water contents at 3
(Supplementary Fig. 3A–E). However, we could observe days after ICH (n = 6 per each group). Mean brain water
few NSCs in the brains. The DiO-labelled cells were positive content was found to be 81.63  0.55% in the ICH-vehicle
624 Brain (2008), 131, 616 ^ 629 S.-T. Lee et al.

Downloaded from by guest on November 6, 2014


Fig. 5 Distributions of NSCs in vivo. NSCs were pre-labelled with DiO fluorescent dye before transplantation. In the NSCs-iv-2 h group,
only a few NSCs were detected in brain (A), lung (B) and liver (C) at 3 days after the transplantation (arrows). No NSCs were observed in
mesenteric lymph nodes (D). However, a number of NSCs are detected in spleen, especially in the marginal zone (MZ) (E). Immunostaining
for CD11b demonstrated that NSCs in the spleen are in the vicinity of the marginal zone macrophages (F). We could observe that a number
of NSCs were in cell-to-cell contact with CD11b+ spleen macrophages under the confocal microscopy (G, arrow heads, 1 mm step-size
optical sections along the z-axis). WP = white pulp. Dotted line indicates the WP border. Bar = 100 mm in A^F, and 10 mm in G.

group, and 80.10  0.27% in the NSCs-iv-2 h group (MLPT) at 1 day between the two groups (splenectomy-
(P50.01, Fig. 6). Interestingly, the mean brain water ICH-vehicle: 3.62  0.52, splenectomy-ICH-NSCs-iv-2 h:
content of splenectomized ICH rats (splenectomy- 3.33  0.52, P = 0.269), while the splenectomy-ICH-vehicle
ICH-vehicle) was 80.8  0.26% and was much lower than group showed less deficits compared to the ICH-vehicle
that of the ICH-vehicle group (P50.01), which suggests group (P50.01). In summary, NSCs-iv-2 h transplantation
that the spleen is involved in cerebral oedema formation failed to reduce brain oedema, perihematomal inflammatory
after ICH. Moreover, NSCs-iv-2 h transplantation in cells and initial neurologic deficits in splenectomized rats,
splenectomized ICH rats (splenectomy-ICH-NSCs-iv-2 h) which indicated that the spleen plays a central role in the
resulted in a mean brain water content of 80.82  0.25%, attenuation of ICH-associated brain injuries by intravenously
which was similar to that of the splenectomized ICH rats administered NSCs.
(splenectomy-ICH-vehicle) (P = 0.986). The mean brain
water content of non-ICH normal control rats was
78.86  0.30, and that of splenectomized non-ICH rats NSCs inhibited in vitro macrophage
was 78.91  0.13 (P = 0.714; no difference). activations via contact-dependency
In histologic quantifications of perihematomal inflam- TNF-a levels are one of the most important predictors of
matory cells, the splenectomy-ICH-vehicle group showed cerebral inflammation and oedema formation after ICH
less OX-42+ or MPO+ cells compared to the ICH-vehicle (Castillo et al., 2002). Because transplanted NSCs were
group (all P50.05, Fig. 6B). However, NSCs-iv-2 h trans- found to localize in macrophage-rich splenic areas, we
plantation in the splenectomy-ICH rats (splenectomy-ICH- further focused on the interaction between NSCs and
NSCs-iv-2 h) failed to further reduce the OX-42+ or MPO+ macrophages. Thioglycollate-elicited macrophages stimu-
inflammatory cell numbers (all P40.05). In addition, lated with lipopolysaccharide (LPS) (namely macrophage +
there was also no difference in the initial neurologic scores LPS) robustly secreted TNF-a in vitro (referred to as
Neural stem cells and spleen Brain (2008), 131, 616 ^ 629 625

Fig. 6 Splenectomy eliminated the effects of early systemic NSCs transplantation. Splenectomy before ICH (Third bar, splenectomy-
ICH-vehicle) reduced ipsilateral hemisphere water contents as compared with the ICH-vehicle group (First bar) (A). NSCs-iv-2 h
transplantation in splenectomized ICH rats (Fourth bar, splenectomy-ICH-NSCs-iv-2 h) had similar levels of brain water content as
splenectomy-ICH-vehicle rats (Third bar), which suggested that splenectomy eliminated the effects of NSCs. In histologic counts of
perihematomal inflammatory infiltrations (B), the splenectomy-ICH-vehicle group showed less OX- 42+ or MPO+ cells compared to the
ICH-vehicle group, and NSCs-iv-2 h transplantation in the splenectomy-ICH rats (splenectomy-ICH-NSCs-iv-2 h) failed to further reduce
the inflammatory cell numbers compared to the splenectomy-ICH-vehicle group. P50.05, P50.01. ns = not significant.

Downloaded from by guest on November 6, 2014


Fig. 7 NSCs inhibited in vitro macrophage activation via cell-to-cell contact. Macrophages stimulated with LPS robustly secreted TNF-a
(second bar), and macrophage/NSC mixed-cultures showed lower TNF-a levels in culture medium (third bar) at 4 h after the LPS stimula-
tion (A). However, this effect was eliminated when NSCs were cultured in transwell membrane inserts which prevented cell-to-cell
contact and permitted only the diffusion of soluble factors (fourth bar). NSCs alone showed no response to LPS stimulation (fifth bar).
When we added NSCs to the cultured macrophages at 2 h after the LPS stimulation and measured cytokine levels at 24 h after the LPS
stimulation, NSCs still attenuated the TNF-a secretion of macrophages (B). IL- 6 secretion were not inhibited by NSCs. P50.05, P50.01.
ns = not significant.

100  1.5%, at 4 h after the LPS stimulation, Fig. 7). we added NSCs to the cultured macrophages at 2 h after
However, when macrophages are co-cultured with NSCs the LPS stimulation and measured cytokine levels at 24 h
before LPS stimulation, TNF-a secretion induced by LPS after the LPS stimulation. In this experiment, NSCs again
stimulation significantly reduced to 70.9  6.9% (P50.01 attenuated the TNF-a secretion of macrophages by 56%
versus macrophage + LPS, n = 6 wells per group). However, (P50.05, n = 3 wells per group, Fig. 7B). However,
when NSCs were cultured in transwell membrane inserts the levels of IL-6 secretions were not changed by NSCs
that prevented their contacting macrophages and allowed co-culture (P = 0.369).
the diffusion of soluble factors, mean TNF-a secretion was
97.9  1.8%, i.e., no different to the macrophage + LPS
situation (P = 0.149). These results suggest that NSCs can Discussion
inhibit in vitro macrophage activations via a contact- In this study, we undertook to characterize the anti-
dependent mechanism. inflammatory effects of NSCs, and the mechanisms
As an adjuvant experiment to simulate the in vivo involved. Intravenous NSC transplantation during the
condition of NSCs-iv-2 h transplantation after ICH, hyperacute stage (at 2 h after ICH) was found to reduce
626 Brain (2008), 131, 616 ^ 629 S.-T. Lee et al.

initial neurologic deterioration, and to exhibit anti- white pulp. Moreover, based on spleen structure, most
inflammatory and anti-apoptotic properties with attenuat- blood flows through the marginal zone and directly along
ing brain oedema and the expressions of inflammatory the white pulp, which leads to efficient monitoring of the
mediators in brain and spleen. Unexpectedly, we also blood by the peripheral immune system (Mebius and Kraal,
observed that the spleen participates in cerebral inflamma- 2005). Macrophages represent a major component of the
tion as splenectomy reduced cerebral oedema and inflam- marginal zone and express pattern-recognition receptors
matory cell counts, and that NSCs modulate the splenic (e.g. toll-like receptors) that are utilized for pathogen and
inflammatory pathway to reduce the cerebral inflammation. antigen clearance (Mebius and Kraal, 2005). Accordingly,
In addition, NSCs inhibited in vitro macrophage activation early splenic inflammatory activation after stroke (Offner
via a contact-derived mechanism. et al., 2006) and the localization of systemically injected
Our results suggest that earlier intravenous NSC admin- NSCs in the splenic marginal zone well correspond.
istration can attenuate systemic inflammatory response after TNF-a activation can be detected a few hours after
haemorrhagic stroke and protect the brain via a bystander stroke; even before significant neuronal death has occurred
mechanism rather than via any direct cell replacement. To (Allan and Rothwell, 2001), and the spleen secretes robust
date, NSCs have been transplanted to regenerate damaged amounts of TNF-a into circulating blood at this early stage
brain tissue, and previously, we showed that human NSCs (Offner et al., 2006). TNF-a promotes early-stage inflam-
administered intravenously at 24 h after ICH promoted mation by increasing the expressions of chemotactic factors
neurological recovery and replaced lost neuronal cells and adhesion molecules by vascular endothelium, which
(Jeong et al., 2003). Moreover, suppressions of T-lymphocyte leads to the early infiltration of monocytes/macrophages,
activity by mesenchymal stem cells or neural precursors have neutrophils, and T cells into sites of injury (Barone and
been demonstrated in EAE models (Pluchino et al., 2003, Feuerstein, 1999; Stevens et al., 2002). Early modulation of
2005; Zappia et al., 2005; Einstein et al., 2007; Gerdoni et al., inflammatory cell response by administering an antibody
2007), and it has been suggested that the immunomodulatory blocking TNF-a at 2 h after stroke reduced brain damage

Downloaded from by guest on November 6, 2014


properties are a characteristics of stem cells rather than and improved neurologic outcomes in an experimental
neural cells (Miller and Bai, 2007). However, the present model (Hosomi et al., 2005). Accordingly, interventions
study is the first to report that intravenous NSCs adminis- targeting splenic TNF-a activation require a prompt
tered during the hyperacute stage in stroke can modulate approach. Our result that NSC transplantation at 24 h
innate cerebral inflammatory responses with interacting with after ICH had no attenuating effect on brain oedema/
peripheral inflammatory systems. inflammation confirm this idea, because TNF-a has
As demonstrated by this study, the spleen is important probably been activated systemically and centrally by this
both as an inducer of cerebral inflammation and as a target time, as shown by the present study and others (Offner
for NSC-based intervention. The link between brain and et al., 2006; Sinn et al., 2007). Thus, the observed
spleen inflammation can be referred to as ‘brain-spleen differences between NSC-iv-2 h and NSC-iv-24 h probably
inflammatory coupling’. Humoral pathways facilitate brain/ depend on the pathogenic time course.
systemic immunity communication. Cytokines like TNF-a, The homing of NSCs to damaged brain is dependent on
IL-1b and IL-6 secreted by cells in different tissues and organs stromal cell derived factor-1a (SDF-1a)-CXC chemokine
link their inflammatory responses (Chamorro et al., 2007). receptor 4 signalling, or on adhesion molecule-receptor
In addition, neural and hormonal mechanisms like the signalling (Imitola et al., 2004; Mueller et al., 2006).
cholinergic anti-inflammatory pathway, the adrenergic path- Although the expressions of SDF-1a and other adhesion
way and the hypothalamic pituitary axis (HPA) link the brain molecules in brain might be significant at later times, brain
and peripheral immune organs (Prass et al., 2003; Meisel homing signals might not be significant at 2 h after ICH
et al., 2005; Tracey, 2007). Thus, changes in the peripheral when NSCs-iv-2 h cells were injected in the present study.
immune system can occur within hours of brain injury, and In addition, blood-brain barrier might be less penetrable for
may later cause stroke-induced immune depression syn- NSCs at this time window. This might answer why NSCs-
drome (SIDS) or increase the risk of infection (Dirnagl et al., iv-2 h cells failed to migrate to the brain. However,
2007). In the present study, reductions in splenic inflamma- migration of NSCs to spleen might not be dependent on
tion by NSCs without significant migration to the brain were inducible factors. The H1 cell line used in this study highly
coupled with a reduction in brain inflammation and a expresses CD44+ (Chu et al., 2004b), which is also
favourable neurologic outcome. In addition, splenectomy expressed on inflammatory cells and mediates their
prior to ICH reduced the cerebral oedema formation and the infiltration via CD44/hyaluronan interaction (Pure and
number of perihematomal inflammatory cells per se. Cuff, 2001). In an EAE model, intravenously administered
Accordingly, modulations of spleen-mediated inflammation neurosphere cells were predominantly detected in lymph
may provide new solutions for cerebral anti-inflammatory nodes at 1 day after transplantation, and were not detected
treatment and brain protection. in brain, spinal cord, or even in spleen (Einstein et al.,
The splenic marginal zone is an important transit area 2007). These two different models, i.e. the haemorrhagic
for cells that are leaving the bloodstream and entering the stroke and the EAE models, with predominantly innate and
Neural stem cells and spleen Brain (2008), 131, 616 ^ 629 627

adaptive immune responses, respectively, may affect the Acknowledgements


biodistribution patterns and immunologic effects of neural This research was supported by a grant (SC3060) from
stem cells. Although the NSCs-iv-2 h treatment also reduced Stem Cell Research Center of the 21st Century Frontier
IL-4 levels in ICH brains and lymph nodes in the present Research Program funded by the Ministry of Science and
study, IL-4 is an anti-inflammatory molecule, mainly Technology, Republic of Korea.
secreted by a negative feedback mechanism responsive to
the other inflammatory cytokines (Tedgui and Mallat,
2001), and is not associated with the clinical outcome of References
stroke (Vila et al., 2003). Accordingly, further studies are Allan SM, Rothwell NJ. Cytokines and acute neurodegeneration. Nat Rev
Neurosci 2001; 2: 734–44.
warranted to reveal how NSCs migrate to immune organs
Aronowski J, Hall CE. New horizons for primary intracerebral hemorrhage
and how they interact with inflammatory cells, although the treatment: experience from preclinical studies. Neurol Res 2005; 27:
present study demonstrates initial evidences that the 268–79.
mechanism of macrophage inactivation by NSCs involves Asahi M, Asahi K, Jung JC, del Zoppo GJ, Fini ME, Lo EH. Role for
a contact-dependent interaction. matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene
The systemic transplantation of NSCs is probably the knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab
2000; 20: 1681–9.
least invasive method of cell administration. Intracerebral Barker RA, Widner H. Immune problems in central nervous system cell
direct neural transplantation requires invasive surgery and therapy. NeuroRx 2004; 1: 472–81.
leaves needle tracks and mass effects. In human, the local Barone FC, Feuerstein GZ. Inflammatory mediators and stroke: new
stereotaxic infusion of NSCs may be severely limited opportunities for novel therapeutics. J Cereb Blood Flow Metab 1999;
because infused cells must migrate substantial distances 19: 819–34.
Castillo J, Davalos A, Alvarez-Sabin J, Pumar JM, Leira R, Silva Y, et al.
and the time taken to embrace entire disease sites may be Molecular signatures of brain injury after intracerebral hemorrhage.
unacceptable. Thus, to overcome these obstacles, NSCs Neurology 2002; 58: 624–9.

Downloaded from by guest on November 6, 2014


must be administered at numerous sites. In contrast, an Chamorro A, Urra X, Planas AM. Infection after acute ischemic stroke: a
intravenous injection can disperse NSCs according to manifestation of brain-induced immunodepression. Stroke 2007; 38:
natural cues and allow them to migrate along the host’s 1097–103.
Choudhri TF, Hoh BL, Solomon RA, Connolly ES, Jr., Pinsky DJ. Use of a
‘chemotactic gradients’ (Pluchino et al., 2003; Chu et al.,
spectrophotometric hemoglobin assay to objectively quantify intracer-
2004b; Martino and Pluchino, 2006). A similar systemic ebral hemorrhage in mice. Stroke 1997; 28: 2296–302.
approach has been studied using hematopoietic stem cells Chu K, Jeong SW, Jung KH, Han SY, Lee ST, Kim M, et al. Celecoxib
in stroke models (Taguchi et al., 2004; Nomura et al., 2005; induces functional recovery after intracerebral hemorrhage with
Honma et al., 2006). Moreover, the transplantation of reduction of brain edema and perihematomal cell death. J Cereb
Blood Flow Metab 2004a; 24: 926–33.
NSCs into circulating blood may be easier and more
Chu K, Kim M, Chae SH, Jeong SW, Kang KS, Jung KH, et al.
effective in multifocal and large-lesioned CNS disorders Distribution and in situ proliferation patterns of intravenously injected
(Jeong et al., 2003; Pluchino et al., 2003; Chu et al., 2004d; immortalized human neural stem-like cells in rats with focal cerebral
Fujiwara et al., 2004; Pluchino et al., 2005). In addition, ischemia. Neurosci Res 2004b; 50: 459–65.
based on the present study, the migration of NSCs to the Chu K, Kim M, Jung KH, Jeon D, Lee ST, Kim J, et al. Human neural
spleen can be therapeutically beneficial and be another stem cell transplantation reduces spontaneous recurrent seizures
following pilocarpine-induced status epilepticus in adult rats. Brain
major contribution in stroke models. Nevertheless, the Res 2004c; 1023: 213–21.
immune responses in the brain and other systemic organs Chu K, Park KI, Lee ST, Jung KH, Ko SY, Kang L, et al. Combined
are complex, and cannot be ignored when it comes to treatment of vascular endothelial growth factor and human neural stem
repair strategies involving cellular transplants (Barker and cells in experimental focal cerebral ischemia. Neurosci Res 2005; 53:
Widner, 2004). Accordingly, further studies are warranted 384–90.
Chu K, Kim M, Park KI, Jeong SW, Park HK, Jung KH, et al. Human
in this issue. neural stem cells improve sensorimotor deficits in the adult rat brain
Collectively, our results suggest that systemic injections with experimental focal ischemia. Brain Res 2004d; 1016: 145–53.
of human NSCs during hyper-acute stage ICH have Del Bigio MR, Yan HJ, Buist R, Peeling J. Experimental intracerebral
potential therapeutic relevance, because they were found hemorrhage in rats. Magnetic resonance imaging and histopathological
to display strong anti-inflammatory functions that promote correlates. Stroke 1996; 27: 2312–20.
Dirnagl U, Klehmet J, Braun JS, Harms H, Meisel C, Ziemssen T, et al.
secondary neuroprotection by interrupting splenic inflam-
Stroke-induced immunodepression: experimental evidence and clinical
matory response. The present study demonstrates that relevance. Stroke 2007; 38: 770–3.
the spleen is centrally involved in innate immune response Einstein O, Fainstein N, Vaknin I, Mizrachi-Kol R, Reihartz E,
after ICH, and thus, represents an important target for the Grigoriadis N, et al. Neural precursors attenuate autoimmune
therapeutic development of stem cell therapies in acute encephalomyelitis by peripheral immunosuppression. Ann Neurol
2007; 61: 209–18.
stroke.
Falcone DJ, Ferenc MJ. Acetyl-LDL stimulates macrophage-dependent
plasminogen activation and degradation of extracellular matrix. J Cell
Physiol 1998; 135: 387–96.
Supplementary material Fujiwara Y, Tanaka N, Ishida O, Fujimoto Y, Murakami T, Kajihara H,
Supplementary material is available at Brain online. et al. Intravenously injected neural progenitor cells of transgenic rats can
628 Brain (2008), 131, 616 ^ 629 S.-T. Lee et al.

migrate to the injured spinal cord and differentiate into neurons, Lee ST, Chu K, Sinn DI, Jung KH, Kim EH, Kim SJ, et al. Erythropoietin
astrocytes and oligodendrocytes. Neurosci Lett 2004; 366: 287–91. reduces perihematomal inflammation and cell death with eNOS
Gerdoni E, Gallo B, Casazza S, Musio S, Bonanni I, Pedemonte E, et al. and STAT3 activations in experimental intracerebral hemorrhage.
Mesenchymal stem cells effectively modulate pathogenic immune J Neurochem 2006d; 96: 1728–39.
response in experimental autoimmune encephalomyelitis. Ann Neurol Martino G, Pluchino S. The therapeutic potential of neural stem cells. Nat
2007; 61: 219–27. Rev Neurosci 2006; 7: 395–406.
Gregersen R, Lambertsen K, Finsen B. Microglia and macrophages are Matsushita K, Meng W, Wang X, Asahi M, Asahi K, Moskowitz MA, et al.
the major source of tumor necrosis factor in permanent middle Evidence for apoptosis after intercerebral hemorrhage in rat striatum.
cerebral artery occlusion in mice. J Cereb Blood Flow Metab 2000; 20: J Cereb Blood Flow Metab 2000; 20: 396–404.
53–65. Mebius RE, Kraal G. Structure and function of the spleen. Nat Rev
Hagan M, Wennersten A, Meijer X, Holmin S, Wahlberg L, Mathiesen T. Immunol 2005; 5: 606–16.
Neuroprotection by human neural progenitor cells after experimental Meisel C, Schwab JM, Prass K, Meisel A, Dirnagl U. Central nervous
contusion in rats. Neurosci Lett 2003; 351: 149–52. system injury-induced immune deficiency syndrome. Nat Rev Neurosci
Hallenbeck JM, Hansson GK, Becker KJ. Immunology of ischemic vascular 2005; 6: 775–86.
disease: plaque to attack. Trends Immunol 2005; 26: 550–6. Miller RH, Bai L. The expanding influence of stem cells in neural repair.
Honma T, Honmou O, Iihoshi S, Harada K, Houkin K, Hamada H, et al. Ann Neurol 2007; 61: 187–8.
Intravenous infusion of immortalized human mesenchymal stem cells Miller RH. The promise of stem cells for neural repair. Brain Res 2006;
protects against injury in a cerebral ischemia model in adult rat. Exp 1091: 258–64.
Neurol 2006; 199: 56–66. Mueller FJ, Serobyan N, Schraufstatter IU, DiScipio R, Wakeman D,
Hosomi N, Ban CR, Naya T, Takahashi T, Guo P, Song XY, et al. Loring JF, et al. Adhesive interactions between human neural stem cells
Tumor necrosis factor-alpha neutralization reduced cerebral edema and inflamed human vascular endothelium are mediated by integrins.
through inhibition of matrix metalloproteinase production after Stem Cells 2006; 24: 2367–72.
transient focal cerebral ischemia. J Cereb Blood Flow Metab 2005; 25: Nomura T, Honmou O, Harada K, Houkin K, Hamada H, Kocsis JD. I.V.
959–67. infusion of brain-derived neurotrophic factor gene-modified human
Huston JM, Ochani M, Rosas-Ballina M, Liao H, Ochani K, Pavlov VA, mesenchymal stem cells protects against injury in a cerebral ischemia
et al. Splenectomy inactivates the cholinergic antiinflammatory pathway model in adult rat. Neuroscience 2005; 136: 161–9.
during lethal endotoxemia and polymicrobial sepsis. J Exp Med 2006; Offner H, Subramanian S, Parker SM, Afentoulis ME, Vandenbark AA,

Downloaded from by guest on November 6, 2014


Hurn PD. Experimental stroke induces massive, rapid activation of the
203: 1623–8.
peripheral immune system. J Cereb Blood Flow Metab 2006; 26: 654–65.
Imitola J, Raddassi K, Park KI, Mueller FJ, Nieto M, Teng YD, et al.
Ourednik J, Ourednik V, Lynch WP, Schachner M, Snyder EY. Neural
Directed migration of neural stem cells to sites of CNS injury by the
stem cells display an inherent mechanism for rescuing dysfunctional
stromal cell-derived factor 1alpha/CXC chemokine receptor 4 pathway.
neurons. Nat Biotechnol 2002; 20: 1103–10.
Proc Natl Acad Sci USA 2004; 101: 18117–22.
Ourednik V, Ourednik J, Flax JD, Zawada WM, Hutt C, Yang C, et al.
Jeong SW, Chu K, Jung KH, Kim SU, Kim M, Roh JK. Human neural
Segregation of human neural stem cells in the developing primate
stem cell transplantation promotes functional recovery in rats with
forebrain. Science 2001; 293: 1820–4.
experimental intracerebral hemorrhage. Stroke 2003; 34: 2258–63.
Parker MA, Anderson JK, Corliss DA, Abraria VE, Sidman RL, Park KI,
Jin K, Sun Y, Xie L, Mao XO, Childs J, Peel A, et al. Comparison of
et al. Expression profile of an operationally-defined neural stem cell
ischemia-directed migration of neural precursor cells after intrastriatal,
clone. Exp Neurol 2005; 194: 320–32.
intraventricular, or intravenous transplantation in the rat. Neurobiol Dis
Pluchino S, Zanotti L, Rossi B, Brambilla E, Ottoboni L, Salani G, et al.
2005; 18: 366–74.
Neurosphere-derived multipotent precursors promote neuroprotection
Jung KH, Chu K, Jeong SW, Han SY, Lee ST, Kim JY, et al. HMG-CoA
by an immunomodulatory mechanism. Nature 2005; 436: 266–71.
reductase inhibitor, atorvastatin, promotes sensorimotor recovery, Pluchino S, Quattrini A, Brambilla E, Gritti A, Salani G, Dina G, et al.
suppressing acute inflammatory reaction after experimental intracerebral Injection of adult neurospheres induces recovery in a chronic model of
hemorrhage. Stroke 2004; 35: 1744–9. multiple sclerosis. Nature 2003; 422: 688–94.
Kelly S, Bliss TM, Shah AK, Sun GH, Ma M, Foo WC, et al. Transplanted Prass K, Meisel C, Hoflich C, Braun J, Halle E, Wolf T, et al. Stroke-
human fetal neural stem cells survive, migrate, and differentiate in induced immunodeficiency promotes spontaneous bacterial infections
ischemic rat cerebral cortex. Proc Natl Acad Sci USA 2004; 101: and is mediated by sympathetic activation reversal by poststroke T
11839–44. helper cell type 1-like immunostimulation. J Exp Med 2003; 198:
Kitchens DL, Snyder EY, Gottlieb DI. FGF and EGF are mitogens for 725–36.
immortalized neural progenitors. J Neurobiol 1994; 25: 797–807. Pure E, Cuff CA. A crucial role for CD44 in inflammation. Trends Mol
Lapchak PA. Hemorrhagic transformation following ischemic stroke: Med 2001; 7: 213–21.
significance, causes, and relationship to therapy and treatment. Curr Qureshi AI, Tuhrim S, Broderick JP, Batjer HH, Hondo H, Hanley DF.
Neurol Neurosci Rep 2002; 2: 38–43. Spontaneous intracerebral hemorrhage. N Engl J Med 2001; 344:
Lee ST, Chu K, Park JE, Lee K, Kang L, Kim SU, et al. Intravenous 1450–60.
administration of human neural stem cells induces functional recovery Schmued LC, Stowers CC, Scallet AC, Xu L. Fluoro-Jade C results in ultra
in Huntington’s disease rat model. Neurosci Res 2005; 52: 243–9. high resolution and contrast labeling of degenerating neurons. Brain Res
Lee ST, Park JE, Lee K, Kang L, Chu K, Kim SU, et al. Noninvasive 2005; 1035: 24–31.
method of immortalized neural stem-like cell transplantation in an Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, et al.
experimental model of Huntington’s disease. J Neurosci Methods 2006a; Endothelial cells stimulate self-renewal and expand neurogenesis of
152: 250–4. neural stem cells. Science 2004; 304: 1338–40.
Lee ST, Chu K, Jung KH, Kim J, Kim EH, Kim SJ, et al. Memantine Singec I, Knoth R, Meyer RP, Maciaczyk J, Volk B, Nikkhah G, et al.
reduces hematoma expansion in experimental intracerebral hemorrhage, Defining the actual sensitivity and specificity of the neurosphere assay in
resulting in functional improvement. J Cereb Blood Flow Metab 2006b; stem cell biology. Nat Methods 2006; 3: 801–6.
26: 536–44. Sinn DI, Kim SJ, Chu K, Jung KH, Lee ST, Song EC, et al. Valproic acid-
Lee ST, Chu K, Kim EH, Jung KH, Lee KB, Sinn DI, et al. Quantification mediated neuroprotection in intracerebral hemorrhage via histone
of human neural stem cell engraftments in rat brains using ERV-3 real- deacetylase inhibition and transcriptional activation. Neurobiol Dis
time PCR. J Neurosci Methods 2006c; 157: 225–9. 2007; 26: 464–72.
Neural stem cells and spleen Brain (2008), 131, 616 ^ 629 629

Snyder EY, Yoon C, Flax JD, Macklis JD. Multipotent neural precursors Vila N, Castillo J, Dávalos A, Esteve A, Planas AM, Chamorro A. Levels of
can differentiate toward replacement of neurons undergoing targeted anti-inflammatory cytokines and neurological worsening in acute
apoptotic degeneration in adult mouse neocortex. Proc Natl Acad Sci ischemic stroke. Stroke 2003; 34: 671–5.
USA 1997; 94: 11663–8. Wang J, Dore S. Inflammation after intracerebral hemorrhage. J Cereb
Stevens SL, Bao J, Hollis J, Lessov NS, Clark WM, Stenzel-Poore MP. The use Blood Flow Metab 2007; 27: 894–908.
of flow cytometry to evaluate temporal changes in inflammatory cells Wang J, Tsirka SE. Tuftsin fragment 1-3 is beneficial when delivered after
following focal cerebral ischemia in mice. Brain Res 2002; 932: 110–9. the induction of intracerebral hemorrhage. Stroke 2005; 36: 613–8.
Taguchi A, Soma T, Tanaka H, Kanda T, Nishimura H, Yoshikawa H, Xi G, Keep RF, Hoff JT. Mechanisms of brain injury after intracerebral
et al. Administration of CD34+ cells after stroke enhances neurogenesis haemorrhage. Lancet Neurol 2006; 5: 53–63.
via angiogenesis in a mouse model. J Clin Invest 2004; 114: 330–8. Zappia E, Casazza S, Pedemonte E, Benvenuto F, Bonanni I, Gerdoni E,
Tedgui A, Mallat Z. Anti-inflammatory mechanisms in the vascular wall. et al. Mesenchymal stem cells ameliorate experimental autoimmune
Circ Res 2001; 88: 877–87. encephalomyelitis inducing T-cell anergy. Blood 2005; 106: 1755–61.
Tracey KJ. Physiology and immunology of the cholinergic antiinflamma- Zheng Z, Yenari MA. Post-ischemic inflammation: molecular mechanisms
tory pathway. J Clin Invest 2007; 117: 289–96. and therapeutic implications. Neurol Res 2004; 26: 884–92.

Downloaded from by guest on November 6, 2014

You might also like