Transduction of Neural Precursor Cells With TAT-Heat Shock

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

TRANSLATIONAL AND CLINICAL RESEARCH

Transduction of Neural Precursor Cells with TAT-Heat Shock Protein


70 Chaperone: Therapeutic Potential Against Ischemic Stroke After
Intrastriatal and Systemic Transplantation
THORSTEN R. DOEPPNER,a,b TOBIAS A. S. EWERT,c LARS T€oNGES,b JOSEPHINE HERZ,a ANIL ZECHARIAH,a
AYMAN ELALI,a ANNA-KRISTIN LUDWIG,d BERND GIEBEL,d FLORIAN NAGEL,e GUNNAR P. H. DIETZ,b,f,g
JENS WEISE,h DIRK M. HERMANN,a MATHIAS B€aHRb,g
a
Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany; bDepartment of
Neurology, University of Goettingen Medical School, Goettingen, Germany; cDepartment of Neurology,
University Medical Center Hamburg-Eppendorf, Hamburg, Germany; dInstitute for Transfusion Medicine,
University of Duisburg-Essen Medical School, Essen, Germany; eCenter for Molecular Neurobiology, Hamburg,
Germany; fDepartment 851, Neurodegeneration II, H. Lundbeck A/S, Valby, Denmark; gDFG Research Center for
the Molecular Physiology of the Brain (CMPB), Germany; hDepartment of Neurology, University of Jena Medical
School, Jena, Germany

Key Words. Adult stem cells • Cell transplantation • Cellular therapy • Nervous system • Neural stem cell • Progenitor cells

ABSTRACT
Novel therapeutic concepts against cerebral ischemia focus prevented secondary neuronal degeneration after intracere-
on cell-based therapies in order to overcome some of the bral delivery that was associated with enhanced functional
side effects of thrombolytic therapy. However, cell-based outcome. Furthermore, intracerebral transplantation of
therapies are hampered because of restricted understanding TAT-Hsp70-transduced NPCs enhanced postischemic neu-
regarding optimal cell transplantation routes and due to rogenesis and induced sustained high levels of brain-derived
low survival rates of grafted cells. We therefore trans- neurotrophic factor, glial cell line-derived neurotrophic fac-
planted adult green fluorescence protein positive neural tor, and vascular endothelial growth factor in vivo. Neuro-
precursor cells (NPCs) either intravenously (systemic) or protection after intracerebral cell delivery correlated with
intrastriatally (intracerebrally) 6 hours after stroke in the amount of surviving NPCs. On the contrary, systemic
mice. To enhance survival of NPCs, cells were in vitro pro- delivery of NPCs mediated acute neuroprotection via stabi-
tein-transduced with TAT-heat shock protein 70 (Hsp70) lization of the blood-brain-barrier, concomitant with
before transplantation followed by a systematic analysis of reduced activation of matrix metalloprotease 9 and
brain injury and underlying mechanisms depending on cell decreased formation of reactive oxygen species. Our
delivery routes. Transduction of NPCs with TAT-Hsp70 findings imply two different mechanisms of action of intra-
resulted in increased intracerebral numbers of grafted cerebrally and systemically transplanted NPCs, indicating
NPCs after intracerebral but not after systemic transplan- that systemic NPC delivery might be more feasible for
tation. Whereas systemic delivery of either native or trans- translational stroke concepts, lacking a need of in vitro
duced NPCs yielded sustained neuroprotection and induced manipulation of NPCs to induce long-term neuroprotection.
neurological recovery, only TAT-Hsp70-transduced NPCs STEM CELLS 2012;30:1297–1310
Disclosure of potential conflicts of interest is found at the end of this article.

cellular mediators into the extracellular space that promote


INTRODUCTION neurological recovery [7, 8]. Unfortunately, survival and dif-
ferentiation rates of new-born cells are low [4–6, 9, 10].
Neurogenesis persists in the adult rodent brain within the Therefore, exogenous NPC transplantation strategies have
subventricular zone (SVZ) [1–3]. Cerebral ischemia stimu- been used to overcome limitations of endogenous neurogene-
lates neurogenesis [4–6], and newly formed neurons release sis [7, 11, 12].

Author contributions: T.R.D.: conception and design, collection and/or assembly of data, data analysis and interpretation, manuscript
writing, and final approval of manuscript; T.A.E.: collection and/or assembly of data, provision of study material or patients, and final
approval of manuscript; L.T. and F.N.: provision of study material or patients and final approval of manuscript; J.H., A.Z., A.E., and A.-
K.L.: collection and/or assembly of data and final approval of manuscript; B.G. and G.P.D.: manuscript writing and final approval of
manuscript; J.W.: conception and design, financial support, and final approval of manuscript; D.M.H.: administrative support, manuscript
writing, and final approval of manuscript; M.B.: financial support, administrative support, and final approval of manuscript.
Correspondence: Thorsten R. Doeppner, M.Sc., M.D., Department of Neurology, University of Duisburg-Essen Medical School,
Hufelandstr. 55, 45147 Essen, Germany. Telephone: þ49-201-723-83133; Fax: þ49-201-723-1660; e-mail: thorsten.doeppner@uk-essen.
de Received October 6, 2011; accepted for publication March 16, 2012; first published online in STEM CELLS EXPRESS March 30, 2012.
V
C AlphaMed Press 1066-5099/2012/$30.00/0 doi: 10.1002/stem.1098

STEM CELLS 2012;30:1297–1310 www.StemCells.com


1298 Transplantation of TAT-Hsp70-Transduced NPCs

Table 1. groups and numbers of animals used for statistical analysis


Systemic delivery Intrastriatal delivery
PBS NPCs NPCs TAT-HA NPCs TAT-Hsp70 PBS NPCs NPCs TAT-HA NPCs TAT-Hsp70
Day 1 14 14 14 14 14 14 14 14
Day 4 15 14 14 14 14 14 14 14
Day 14 7 7 7 7 7 7 7 7
Day 28 8 9 9 9 8 8 9 8
Day 56 11 11 11 11 11 11 11 11
Animals were assigned to four treatment groups for both systemic and intrastriatal delivery of adult NPCs or PBS, respectively. All
injections were performed 6 hours after stroke onset. Five survival periods were defined for each group. Time points given refer to induction
of stroke, which was assigned as day 0. The number of animals given reflects the amount of surviving mice used for final analysis of
experimental results.
Abbreviations: Hsp70, heat shock protein 70; NPCs, neural precursor cells; PBS, phosphate buffered saline; TAT-HA, TAT-hemagglutinin.

Transplantation of NPCs ameliorates ischemic brain injury tations and the in vivo experimental paradigm is given in Sup-
in rodents [7, 8, 13–17], but its therapeutic benefit is hampered porting Information (Supporting Information Fig. S1). Survival
by low survival rates of grafted cells [18, 19]. Since cell trans- rates were 100% for all animals surviving 1, 4, 14, or 28 days for
plantation is usually done after stroke [20], grafted cells are not each experimental condition, and the amount of animals used per
exposed to ischemic insults directly. Rather, cells are trans- experimental group as described in Materials and Methods. The
planted into a nonfavorable tissue environment, in which they survival rate of animals receiving systemic or intracerebral injec-
are exposed to oxidative stress and inflammation [21, 22], tions of phosphate buffered saline (PBS) was 78.6% (11/14) on
which may cause protein misfolding and cell death [23, 24]. day 56 for both groups. Survival rate after intracerebral transplan-
tation of native NPCs, TAT-HA-transduced, or TAT-Hsp70-trans-
Regarding clinical translation, the most appropriate route of
duced NPCs was 84.6% (11/13) for each group. For animals that
NPC transplantation is an important but unresolved question.
had received systemic delivery of TAT-Hsp70-transduced NPCs
Among different transplantation routes, intravenous cell delivery survival was 91.6% (11/12), whereas for the remaining experi-
has been used due to its simplicity and clinical practicability [8, mental groups with a 56 days survival, the survival rate was
25, 26], although achieving low intracerebral cell numbers [15, 100%. Assessment of cell proliferation was performed on animals
27]. Therefore, local intracerebral injections are widely used, surviving either 28 days or 56 days. These mice received 11 con-
which can enhance the number of transplanted intracerebral secutive intraperitoneal (i.p.) injections of 5-bromo-2-desoxyuri-
cells but lack clinical utility due to invasive delivery procedures dine (BrdU; 50 mg/kg b.wt.) on days 8–18.
[28–32]. Until recently, only two studies systematically com-
pared the effect of different NPC transplantation routes on post- Preparation of Recombinant Proteins
ischemic outcome. However, these studies are limited to a maxi- TAT-Hsp70 and TAT-hemagglutinin (TAT-HA), which is also
mal observation period of 1 week and lack an analysis of included in the TAT-Hsp70 construct serving as a negative con-
underlying cellular mechanisms [33, 34]. trol, were prepared under native conditions as described [36].
To elucidate consequences of NPC delivery strategies, we Briefly, the recombinant genes were expressed in Escherichia coli
herein submitted mice to cerebral ischemia and transplanted strain BL21 (DE3) pLysS (Novagen, Madison, WI, www.
NPCs 6 hours later either intravenously (systemically) or novagen.com) and proteins were isolated in 10 mM Tris, pH 10,
intrastriatally (intracerebrally), followed by an analysis of 20% glycerol, 274 mM NaCl, 0.1% Pluronic, and 0.02% Tween
brain injury and remodeling for up to 8 weeks. In order to 80. Bacterial debris was removed by centrifugation and the cell
promote survival of grafted cells, NPCs were protein-trans- extracts were purified by affinity chromatography using Ni-tris-
duced with the chaperone Hsp70, which reduces apoptosis carboxymethyl-ethylene-diamine [37]. Protein was eluted by step-
and inflammation after hypoxic-ischemic injury [23, 24]. wise addition of binding buffer containing increasing concentra-
Since cellular entry of Hsp70 is poor, transduction was tions of imidazole. The column eluate was purified from imidaz-
achieved in vitro using the membrane-permeable and neuro- ole by gel filtration (Sephadex G-25 M, GE Healthcare Bio-
protective TAT-Hsp70 fusion protein before transplantation Sciences AB, Munich, Germany, www.gehealthcare.com).
[10, 35]. This study has explicit meaning for future cell-based According to this procedure, TAT-Hsp70 is highly stable after
stroke therapies, since it highlights the importance of cell cell transduction as no protein degradation could be detected 24
transplantation routes, implying different mechanisms by hours after protein application, suggesting that the intracellular
half-life time is at least a few days [36]. For transplantation of
which transplanted NPCs induce poststroke neuroprotection.
NPCs, in vitro incubation period with either TAT-Hsp70 (250
nM) or TAT-HA (250 nM) was 4 hours.

Preparation and Cultivation of Adult SVZ-Derived


MATERIALS AND METHODS NPCs
NPCs were isolated from the SVZ of 19 6–8 weeks old male
Animals and Experimental Groups nontransgenic C57BL/6 mice for in vitro experiments [19]. For in
Experimental procedures were in accordance with the National vivo experiments, 346 transgenic green fluorescence protein posi-
European Institutes of Health guidelines for the care and use of tive (GFPþ) animals (C57BL/6-Tg ACTB-enhanced green fluo-
laboratory animals and approved by local authorities. For all rescence protein (EGFP), 1Osb/J; JAX Laboratory, Bar Harbor,
experiments, male C57BL/6 mice (11–13 weeks, 23–27 g; Maine, www.jax.org; male, 6–8 weeks old) were used. GFP
Charles River, Sulzfeld, Germany (www.criver.com) were used. expression was under control of the actin promoter, which allows
The total number of animals was 437, which were assigned to reliable and stable tracking of transplanted NPCs. The SVZ was
eight treatment paradigms (Table 1). Treatment was performed in microdissected under stereomicroscopic control (Zeiss, Jena, Ger-
a blinded manner. A schematic display for intracerebral transplan- many, www.zeiss.de), minced into small pieces, and then
Doeppner, Ewert, T€onges et al. 1299

mechanically triturated and dissociated into a single-cell suspen- Induction of Transient Focal Cerebral Ischemia
sion. Cells were cultured in serum-free basic Dulbecco’s modified
Cerebral ischemia was induced using middle cerebral artery
Eagle’s medium (DMEM)/F12 (PAA, Linz, Austria, www.paa.
(MCA) occlusion [19]. Animals were anesthetized (0.8%–1.5%
com) supplemented with epidermal growth factor (EGF, 2 lg/
isofluran, 30% O2, and remainder N2O), and rectal temperature
ml), basic fibroblast growth factor (bFGF, 2 lg/ml), and penicil-
was maintained at 36.5 C–37.0 C using a feedback-controlled
lin-streptomycin (Invitrogen, Frankfurt, Germany, www.
heating system under continuous control of blood flow changes
invitrogen.com). Cells were incubated with 5% CO2 at 37 C. The
by means of a laser Doppler flow (LDF) system (Perimed, Jar-
growth factors were supplemented every 2–3 days and cells were
falla, Sweden, www.perimed-instruments.com). Occlusion of the
passaged via accutase (Invitrogen) digestion for 30 minutes at
left MCA was achieved using a 7-0 silicon-coated nylon monofil-
37 C with a resuspension every 10 minutes. Thereafter, cells
ament (180 lm tip diameter; Doccol, Redlands, CA, www.
were centrifuged and resuspended in conditioned media. Neuro-
doccol.com), which was withdrawn after 45 minutes to induce
sphere passages were done every 7–10 days and cells used for
transient ischemia. LDF recordings continued for additional 15
transplantation were derived from passage 4.
minutes to monitor appropriate reperfusion.

Transplantation of NPCs
Quantitative Analyses of NPCs and Neurosphere
Mice received either intravenous (systemic) or intrastriatal (intra-
Differentiation Assay cerebral) injections of NPCs or PBS 6 hours after induction of
For estimation of neurosphere numbers, 10,000 primary NPCs stroke. Systemic injection of NPCs (106 cells in 100 ll PBS) or
(P0) and cells from passage 4 (P4) were plated in six-well plates PBS (100 ll) was performed via cannulation of the right femoral
and cultured for 7 days with/without TAT-HA (250 nM) or TAT- vein. For intracerebral stereotactic injections of PBS (5 ll) or
Hsp70 (250 nM). Thereafter, neurosphere numbers and neuro- NPCs (5  105 cells in 5 ll PBS), surgeries were performed in
sphere diameters were quantified. Cells were then treated with mice that were anesthetized with ketamine (10 mg/kg) and
accutase in order to achieve single-cell suspensions followed by xylazine (25 mg/kg). Thereafter, animals were placed in a
determination of total cell numbers. For differentiation analysis, stereotactic apparatus (Kopf Instruments, Tujunga, CA, www.
NPCs from P0 and P4 that had been cultured for 7 days (native kopfinstruments.com) and fixed accordingly. The skull was
or transduced with 250 nM TAT-HA or 250 nM TAT-Hsp70) exposed, and a hole was drilled at the appropriate position on the
were gently triturated and plated onto 24-well plates (40 cells per ischemic left hemisphere. Injections (0.4 mm anterior, 1.8 mm
microliter) containing coverslips coated with 1 mg/ml poly(D-ly- lateral and 3.5 mm ventral from bregma) were performed using
sine) (Sigma-Aldrich, Taufkirchen, Germany, www.sigmaaldrich. 10 ll Hamilton syringes (Bonaduz, Switzerland, www.hamilton-
com) according to modified protocols [38, 39]. Differentiation company.com) with a rate of 1 ll/minute. The syringe was kept
medium consisted of DMEM/F12 supplemented with 2% B27 in place for additional 5 minutes after injection before removal.
(Invitrogen), 1% fetal bovine serum, and penicillin-streptomycin.
For Western blot analysis, cells were lysed (50 mM Tris at pH Analysis of Poststroke Brain Injury and
8.0, 150 mM NaCl, and Triton 1%) on day 7 postplating. Lysates Immunohistochemistry
were centrifuged and supernatants were used for SDS-PAGE.
Equal amounts of protein (75 lg) were diluted in 6 sample Infarct volumes were analyzed on day 4 (n ¼ 5 per group), for
buffer, boiled and loaded onto 10% polyacrylamide gels. Proteins which brains were removed and cut into slices of 2 mm each. Sli-
were transferred onto polyvinylidene difluoride membranes, ces were stained with 2,3,5-triphenyltetrazolium chloride (2%),
which were immersed in blocking solution (5% dry milk powder and a computer-based analysis of infarct volumes was done using
in TBS-T [0.1% Tween 20 þ Tris-buffered saline]; 1 hour at the freely available software ImageJ (NIH; http://rsbweb.nih.gov/
room temperature [RT]). The following primary antibodies were ij) by subtracting the area of the nonlesioned ipsilateral hemi-
used: polyclonal rabbit antinestin (1:2,000), polyclonal rabbit sphere from that of the contralateral side. Infarct volume sizes
antiglial fibrillary acidic protein (GFAP; 1:10,000), polyclonal were calculated by integration of the lesioned areas. Postischemic
rabbit anti-20 ,30 -cyclic nucleotide 30 -phosphodiesterase (CNPase; brain edema was measured as the increase of ipsilateral hemi-
1 lg/ml), and antibeta tubulin III (1 lg/ml; all obtained from spheric volume in comparison to the contralateral hemisphere.
Abcam, Cambridge, UK, www.abcam.com). Membranes were For immunohistochemical analysis, animals were i.p. injected
incubated with a peroxidase-coupled, goat anti-rabbit secondary with chloralhydrate (420 mg/kg b.wt.) and transcardially perfused
antibody (1:2,000; Abcam), washed several times, immersed in with 4% paraformaldehyde at days 4 (n ¼ 5–6), 14 (n ¼ 7), 28
enhanced chemiluminescence solution, and exposed to enhanced (n ¼ 8–9), and 56 (n ¼ 7). The brains were removed, shock-fro-
chemiluminescence-Hyperfilm (Amersham Biosciences, Freiburg, zen in liquid nitrogen, and 16-lm thick coronal cryostat sections
Germany, www.amershambiosciences.com). were prepared. Quantitative analyses for immunohistochemical
stainings were performed defining regions of interest (ROIs)
within the ischemic basal ganglia. Stereotactic coordinates were
0.14 mm anterior, 2.5–3.25 mm ventral, and 1.5–2.25 mm lateral
Oxygen-Glucose-Deprivation of Cultured NPCs from bregma. Three sections per animal and ROI were used. For
Experimental procedures were performed as previously described quantitative analysis of proliferating BrdUþ cells, sections were
[19]. NPCs were passaged and 100,000 cells were first preincu- exposed to blocking solution and subsequently stained with a
bated for 4 hours with conditioned cell culture medium contain- monoclonal mouse anti-BrdU antibody (1:400; Roche, Mannheim,
ing TAT-Hsp70 (250 nM). Then, the medium was substituted by Germany, www.roche.de) or a monoclonal rat anti-BrdU antibody
a glucose-free crystalloid solution (‘‘Thomajodin’’ plus 1 mM (1:400; Abcam). Since quenching of fluorescence signal of GFPþ
mannitol; Deltapharm, Dortmund, Germany) containing TAT- NPCs occurs during section processing, a polyclonal rabbit anti-
Hsp70 (250 nM). Cells were incubated in a hypoxic chamber GFP antibody (1:2,500; Abcam) was used to enhance GFP signal
(1% O2; remainder 5% CO2 and 94% N2) for 45 minutes and intensity. For differentiation analysis of GFPþ or BrdUþ cells, dou-
reincubated in normal glucose and TAT-Hsp70 (250 nM) contain- ble staining was performed against BrdU/GFP and a polyclonal
ing cell culture medium for 24 hours. Cell viability was assessed goat antidoublecortin antibody (1:50; Santa Cruz Biotechnology,
using a LIVE/DEAD-Viability/Cytotoxicity-Assay kit (Lonza, Heidelberg, Germany, www.scbt.com), a polyclonal rat anti-GFAP
Basel, Switzerland, www.lonza.com). TAT-HA (250 nM; nega- antibody (1:500; Zymed, Germany), a monoclonal mouse anti-
tive control) and PBS served as controls for TAT-Hsp70. Con- CNPase antibody (1:400; Millipore, Abingdon, UK, www.
trols were kept under normal cell conditions without oxygen-glu- millipore.com), a monoclonal mouse anti-NeuN antibody (1:1,000;
cose-deprivation (OGD) for the duration of the experiment. Millipore), or a monoclonal mouse antinestin antibody (GFP
www.StemCells.com
1300 Transplantation of TAT-Hsp70-Transduced NPCs

counterstaining only; 1:500; Millipore). In order to exclude engulf- preferentially turn to the left, nonimpaired body side [41, 42].
ment of GFPþ-transplanted NPCs by microglia, double staining The laterality index was calculated according to the following
against GFP and IB4 was done on day 4 using a rat biotin-conju- formula: (number of left turns  number of right turns)/10.
gated anti-IB4 antibody (1:100; Vector, Peterborough, UK,
www.vectorlabs.com). All antibodies were incubated for 18 hours Zymography of Matrix Metalloproteases
at 4 C. Thereafter, the sections were incubated for 1 hour at RT. Left ischemic hemispheres (n ¼ 4 per condition; 24 hours post-
For double staining with BrdU, the following secondary antibodies stroke) were homogenized in cold lysis buffer (basic buffer) con-
were used: goat anti-mouse Cy-3 (1:400; Dianova, Hamburg, Ger- taining 50 mmol/l Tris-HCl (pH 7.6), 150 mmol/l NaCl, 5 mmol/l
many, www.dianova.com) or goat anti-rat Alexa 594 (1:400; Dia- CaCl2, 0.05% BRIJ-35, 0.02% NaN3, and 1% Triton X-100. Ho-
nova) for BrdU staining, goat anti-rat Alexa 488 (1:250; Invitro- mogenates were centrifuged at 4 C at 12,000g for 5 minutes and
gen, Germany) or donkey anti-goat Alexa 488 (1:250; Invitrogen) supernatants were incubated with a 1:10 volume of gelatine-
for GFAP or doublecortin (Dcx) staining, goat anti-mouse Alexa Sepharose 4B for 1 hour at 4 C. After centrifugation, pellets
488 (1:100; Jackson ImmunoResearch, Newmarket, UK, www. were resuspended in elution buffer (basic buffer containing 10%
jireurope.com) for CNPase staining, and goat anti-mouse Alexa dimethyl sulfoxide and 20% volume of lysis buffer); purified
488 (1:400; Invitrogen) for NeuN staining. For double staining samples were analyzed by zymography. Protein concentrations
with GFP, the secondary antibodies were as follows: goat anti- were determined by the bicinchoninic acid method (BCA kit,
mouse Cy-3 (1:100; Jackson ImmunoResearch) for NeuN, CNPase Thermo Scientific, Karlsruhe, Germany, www.thermoscientific.
and nestin staining as well as goat anti-rat Cy-3 antibody (1:200, com). Separation of matrix metalloprotease (MMP)-2 and MMP-9
Abcam) for GFAP staining. Double staining against Dcx was done as pro-form and active form was performed using Novex Zymo-
using a donkey anti-goat Cy-3 secondary antibody (1:500; Dia- gram Gels (Invitrogen) according to the manufacturer’s instruc-
nova). Photos for differentiation analysis with subsequent three- tions. Samples were incubated in nonreducing sample buffer (0.4
dimensional reconstruction were made using a Zeiss microscope mol/l Tris, pH 6.8, 5% SDS, 20% glycerol, and 0.05% bromphe-
equipped with an Apotome and the corresponding AxioVision nol blue) for 10 minutes at RT and then loaded onto 10% SDS
software. polyacrylamide electrophoresis gels containing 0.1% gelatin. Af-
For assessment of brain injury on day 4, terminal deoxynucleo- ter electrophoresis, samples were incubated with 2.5% Triton X-
tidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) 100 twice for 20 minutes, equilibrated with developing buffer
was done. The staining was performed incubating the sections with (Invitrogen), and incubated for 18 hours at 37 C. Gels were
proteinase K (7 minutes at 37 C) followed by exposure to the TdT stained with Coomassie Blue for 30 minutes and destained in
enzyme reaction according to the manufacturer’s manual (Roche). washing solution (30% methanol and 10% acetic acid). White
Sections were stained with a streptavidin-Alexa-488-conjugated bands on a dark background indicated zones of digestion corre-
secondary antibody (2 hours at RT; Invitrogen) and analyzed. In sponding to the presence of pro-MMPs and activated MMPs on
order to assess viability of BrdUþ cells, double staining using the the basis of their molecular weight. As standards, 0.1 ng of
rat anti-BrdU antibody and the TUNEL-protocol was performed. human pro-MMP-9 and human pro-MMP-2 (Merck Biosciences,
Further analysis of brain injury on days 4, 28, and 56 was per- Darmstadt, Germany, www.merckgroup.com) and 0.01 ng of acti-
formed by determination of neuronal density, that is, after quantita- vated MMP-9 and activated MMP-2 (Merck Biosciences) were
tive analysis of NeuNþ cells within defined ROIs as stated above. used. Gels were scanned and densitometrically analyzed.
Cell numbers for each specific staining were recalculated and are
given as total amount of cells per square millimeter. Analysis of Blood-Brain-Barrier Permeability
Assessment of glial scar surrounding the ischemic territory Mice (n ¼ 6 per condition) received intravenous bolus injections
was measured in four defined ROIs after GFAP-staining using a of 2% Evans blue dye (2 ml/kg b.wt.) via tail vein cannulation
modified protocol as previously described [8]. Images were 22 hours poststroke [43]. Two hours later, animals were sacrificed
obtained from the indicated ROIs, and the gliotic area was meas- by transcardiac perfusion with PBS. Left hemispheres from nonis-
ured for each ROI using Image J software. Data are given as chemic animals that had received no treatment served as refer-
mean area (mm2) out of four ROIs. ence for extravasal Evans blue dye contents. Brains were
removed and separated into hemispheres. Left (ischemic) hemi-
Assessment of Poststroke Functional Recovery spheres were weighed, homogenized in 2 ml of 50% trichloroace-
Motor coordination deficits were analyzed using the rota rod, tic acid, and centrifuged at 10,000 rpm for 20 minutes. The
tight rope, and the corner turn test. All behavioral tests were per- extracted Evans blue dye was further diluted with ethanol, and
formed on the same animals, which were also used for immuno- the fluorescence signal was measured with a luminescence spec-
histochemical analysis on day 56. Behavioral tests were per- trophotometer (exc. ¼ 620 nm, em. ¼ 680 nm). An external
formed in a blinded manner. One day before induction of stroke, standard (62.5–500 ng/ml) was used for calculation of Evans blue
animals were trained before the beginning of the actual tests on dye contents, which is given as (lg) Evans blue dye per (g)
day 7, 14, 28, and 56. Both rota rod and tight rope test were per- tissue.
formed as previously described [40]. Using the rota rod test, ani-
mals were put on an accelerating treadmill (TSE Systems, Bad
Determination of Thiobarbituric Acid-Reactive
Homburg, Germany, www.tse-systems.com; 3-cm diameter) with Substances
an accelerating speed of 4–40 rpm. The maximum speed was Oxidative stress was assessed 24 hours poststroke in brain ho-
achieved after 260 seconds, and maximum testing time was 300 mogenates from left ischemic hemispheres (n ¼ 4 per condition),
seconds. The time until animals dropped was registered and stat- which were obtained using lysis buffer as described above. For-
istically analyzed. For the tight rope test, animals were placed on mation of reactive oxygen species (ROS) leads to peroxidation of
a 60-cm long rope grasping the string with their forepaws. Maxi- fatty acids of phospholipids contained within the cell membrane.
mum test time was 60 seconds, and results were scored from 0 During peroxidation, thiobarbituric acid (TBA) reactive substan-
(minimum) to 20 (maximum) according to a validated score [40], ces (TBARS) are generated such as malondialdehyde (MDA).
depending on the time animals spent on the rope and whether or MDA reacts with TBA resulting in a chromogenic compound
not they reached the platform. Rota rod and tight rope tests were whose absorption is photometrically measured at k ¼ 532 nm
performed twice at each time point and means were calculated. [44]. Samples were incubated with cold 80% trichloroacetate so-
For the corner turn test, two vertical boards were attached at one lution (volume ratio of 5:1) and centrifuged at 7,500g for 5
side with an angle of 30 , and each mouse was tested for the side minutes. Supernatants were incubated with 1% TBA solution (pH
chosen over 10 trials per test day. Whereas healthy animals leave 7.0; volume ratio of 2:1) at 95 C for 10 minutes followed by
the corner without side preference, mice suffering from stroke additional centrifugation at 5,000g and subsequent photometric
Doeppner, Ewert, T€onges et al. 1301

analysis. The extent of TBARS formation is expressed as MDA TAT-Hsp70 Enhances Numbers of Transplanted
equivalents using 1,1,3,3-tetramethoxypropan as standard. NPCs After Intracerebral Injection Without Affect-
ing Cell Differentiation
ELISA for Measurement of Growth Factors Native or transduced GFPþ NPCs were transplanted systemi-
For ELISA experiments (n ¼ 4), samples were obtained on days cally or intracerebrally 6 hours after stroke (Fig. 2). Cells
4 and 56 as described for measurement of TBARS. Levels of were treated ex vivo before transplantation with either TAT-
growth factors such as vascular endothelial growth factor (VEGF; HA or TAT-Hsp70. Intracerebral transplantation within the is-
R&D Systems, Minneapolis, MN, www.rndsystems.com), nerve chemic striatum resulted in high numbers of GFPþ NPCs
growth factor (Promega, Mannheim, Germany, www.promega.- under each condition (Fig. 2A). Although the number of
com), brain-derived neurotrophic factor (BDNF; Promega), glial
grafted NPCs gradually declined over time in all experimental
cell line-derived neurotrophic factor (GDNF; Promega), bFGF
(R&D Systems), and EGF (R&D Systems, Minneapolis, MN,
groups, animals that had received TAT-Hsp70-transduced
www.rndsystems.com) were measured using commercial mouse NPCs always showed significantly more transplanted cells
ELISA kits according to the manufacturer’s instructions. (Fig. 2A). The latter typically formed agglomerations at the
site of transplantation without significant migration (Fig. 2C–
2I). On the contrary, systemic transplantation of NPCs
Statistics (native/transduced) yielded lower numbers of NPCs under
All data are given as mean 6 SD. For comparison between two each experimental condition as compared to intracerebral cell
groups, the Student’s t test was used, whereas for comparison delivery (Fig. 2B). Likewise, the number of transplanted cells
between multiple groups, a one-way analysis of variance followed gradually decreased over time. Transduction of NPCs with
by the Tukey’s post hoc test was performed. A p value of <.05 TAT-Hsp70, however, did not significantly increase the num-
was considered to be statistically significant. ber of transplanted cells after systemic injection within the is-
chemic hemisphere.
Differentiation states of transplanted NPCs (Supporting In-
formation Fig. S3) were not affected by TAT-Hsp70 on days 4
RESULTS and 56 (Fig. 2J, 2K). No colocalization between GFP and the
mature neuronal marker NeuN was observed. Noteworthy, coun-
TAT-Hsp70 Does Not Affect NPC Proliferation and terstaining between GFP and the microglial (and also monocyte)
Differentiation but Protects from Hypoxic-Hypogly- marker IB4 [46] revealed very low rates of colocalizations on
cemic Cell Injury day 4 after stroke (Fig. 2J), suggesting that transplanted NPCs
Protein delivery using TAT technology is highly efficient and are not significantly engulfed by activated microglia.
has been successfully tested on various cell types, with often
NPC-Mediated Poststroke Neuroprotection
100% transduction rates in cultivated cells [45]. Previously,
treatment of neuroblastoma cells with TAT-Hsp70 (250 nM) Infarct volume analysis revealed that both intracerebral and
achieved at least fourfold higher intracellular levels of TAT- systemic transplantation of NPCs yielded significant neuropro-
Hsp70 compared to endogenous Hsp70. In this study, we tection on day 4 poststroke, which was independent of NPC
again detected TAT-Hsp70 in cellular lysates after treatment transduction states (Fig. 3A–3C). Likewise, edema formation
with recombinant protein (Supporting Information Materials was significantly decreased in animals that had received either
and Methods; Supporting Information Fig. S2). Thereafter, intracerebral or systemic transplantation of native or trans-
effects of both the recombinant protein and cell passaging on duced NPCs (Fig. 3D). In line with this, analysis of neuronal
NPC proliferation, differentiation, and susceptibility to OGD density and TUNELþ cells on day 4 yielded increased neuro-
were analyzed in vitro before cell transplantation. nal density and reduced numbers of TUNELþ cells after ei-
Neurosphere formation rates were independent of both ther intracerebral or systemic injection of native/transduced
TAT-Hsp70 and cell passaging (Fig. 1A–1C). Likewise, neu- NPCs (Fig. 3E, 3F). Assessment of long-term neuroprotection
rosphere diameters did not significantly differ between treat- after intracerebral transplantation, however, revealed that only
ment groups. As for P0, mean neurosphere diameters were mice receiving TAT-Hsp70-transduced NPCs showed a signif-
89.9 6 27.2 (native NPCs), 107.3 6 34.1 (NPCs þ TAT- icantly increased neuronal density (Fig. 3G, 3H). On the other
HA), and 98.5 6 24.7 nm (NPCs þ TAT-Hsp70). In P4, hand, sustained neuroprotection after systemic transplantation
mean diameters were 93.6 6 19.4 (native NPCs), 96.9 6 of NPCs was observed in animals that had received either
27.8 (NPCs þ TAT-HA), and 101.3 6 29.1 nm (NPCs þ native or transduced NPCs. These data suggest that sustained
TAT-Hsp70). Furthermore, total cell numbers did not signifi- neuroprotection by intracerebral transplantation of NPCs is
cantly differ between nontreated and transduced NPCs from only achieved via in vitro transduction with TAT-Hsp70,
both passages (Fig. 1D). whereas systemic transplantation of NPCs induces long-term
Assessment of NPC differentiation (Fig. 1E, 1F) revealed neuroprotection independent of cell transduction.
high protein abundance of the neural stem/progenitor cell
marker nestin and the astroglial marker GFAP, whereas pro- Transplantation of NPCs Reduces Poststroke Neuro-
tein abundance of the neuronal marker b-tubulin III and the logical Impairment
oligodendroglial marker CNPase was low. However, no dif- Neurological deficits in mice were assessed using the rota rod
ference between the experimental groups was observed, that (Fig. 4A, 4B), the tight rope (Fig. 4C, 4D), and the corner
is, neither cell passaging nor TAT transduction affected cell turn test (Fig. 4E, 4F). Although intracerebral transplantation
differentiation. of either native or transduced NPCs reduced functional
TAT-Hsp70-mediated neuroprotection was analyzed in impairment in all behavioral tests as early as day 7, only ani-
NPCs from P0 and P4 that were exposed to a 45-minute mals that were treated with TAT-Hsp70-transduced NPCs
OGD with subsequent recultivation under standard cell culture showed sustained reduced functional deficits. However, sys-
conditions (Fig. 1G). Whereas TAT-Hsp70-transduced NPCs temic injection of either native or transduced NPCs resulted
from passage P0/P4 showed little cell injury, less than 30% of in sustained improved functional outcome in these animals as
native and TAT-HA-transduced NPCs survived OGD injury. compared to PBS controls.
www.StemCells.com
1302 Transplantation of TAT-Hsp70-Transduced NPCs

Figure 1. In vitro characterization of cultivated adult NPCs. (A–C): Neurosphere formation rates were analyzed in both primary passage (P0)
cells (i.e., 7 days after cell culturing) and in NPCs from passage 4 (P4). Typical examples of neurosphere formation in P0 and in P4 are depicted
in (A) and (B), respectively. (D): Neurospheres used for experiments described in (A–C) were dissociated by means of accutase followed by
determination of total NPC numbers within each six wells. (E, F): Cell differentiation was analyzed using Western blotting against nestin,
CNPase, GFAP, and b-tubulin III in P0 and P4 neurospheres. (F): Data from (E) are presented as arbitrary units after densitometric analysis. (G):
NPCs (P0 and P4) were exposed to 45 minutes of oxygen-glucose-deprivation (OGD) and recultivated for 24 hours at standard cell culture condi-
tions followed by subsequent analysis of cell injury. Controls were incubated under standard cell culture conditions without OGD. Scale bars ¼
100 lm. *Significantly different from controls, p < .05. Abbreviations: CNPase, 20 ,30 -cyclic nucleotide 30 -phosphodiesterase; GFAP, glial fibril-
lary acidic protein; NPC, neural precursor cell; TAT-HA, TAT-hemagglutinin.

Neurogenesis Is Enhanced in Mice Stereotactically each experimental condition (representative image in Fig.
Grafted with TAT-Hsp70-Transduced NPCs 5C), suggesting that BrdUþ cells do survive initially. BrdUþ
cells were typically located in the SVZ and scattered in the is-
Cerebral ischemia was associated with detection of enhanced chemic striatum (Fig. 5D–5N). Systemic transplantation of
numbers of proliferating BrdUþ cells for up to 8 weeks within native or transduced NPCs did not alter the amount of BrdUþ
the ischemic hemisphere of each experimental group (Fig. cells within the ischemic hemisphere (Fig. 5A, 5B). However,
5A, 5B). Noteworthy, colocalizations of proliferating BrdUþ intracerebral transplantation of TAT-Hsp70-transduced NPCs
cells with TUNELþ cells on day 4 were less than 3% for resulted in a sustained postischemic increase of BrdUþ cells
Doeppner, Ewert, T€onges et al. 1303

Figure 2. TAT-Hsp70 increases numbers of grafted NPCs after intracerebral transplantation without affecting cell differentiation. NPCs were
transplanted 6 hours after stroke via stereotactic injection of 500,000 GFPþ cells into the ischemic striatum (intracerebral injection; A) or via in-
travenous injection of 106 cells into the femoral vein (systemic injection; B). Photographs depicted represent typical orientations of TAT-Hsp70-
transduced NPCs after intracerebral injection on day 56 in low magnification (C–E; green ¼ GFP and blue ¼ DAPI; scale bar ¼ 50 lm) and
high magnification (F–H; scale bar ¼ 20 lm). (I): Three-dimensional reconstruction from merged photo (H). (J, K): Differentiation profile of
transplanted GFPþ NPCs on day 4 (J) and day 56 (K) after stroke. Note that GFP/IB4 staining was only performed on day 4. Colocalization of
GFPþ cells with NeuN was not observed at any time point analyzed. *Significantly different from animals treated with native or TAT-HA-trans-
duced NPCs, p < .05. Abbreviations: CNPase, 20 ,30 -cyclic nucleotide 30 -phosphodiesterase; GFAP, glial fibrillary acidic protein; GFP, green fluo-
rescence protein; NPC, neural precursor cell; TAT-HA, TAT-hemagglutinin; DAPI, 4’,6-diamidino-2-phenylindole.

(Fig. 5A, 5B). The latter might be a consequence of either duced NPCs did not significantly affect neuronal differentia-
enhanced stimulation of postischemic cell proliferation or tion of proliferating cells.
enhanced survival of proliferating cells due to intracerebrally
grafted TAT-Hsp70-transduced NPCs. In line with this, a dif- NPC-Induced Poststroke Mechanisms Vary with
ferentiation analysis of BrdUþ cells (Fig. 5O–5Z) revealed Transplantation Routes
that only intracerebral transplantation of TAT-Hsp70-trans- We next analyzed putative mechanisms underlying NPC-
duced NPCs yielded enhanced colocalizations with the imma- mediated neuroprotection after intracerebral and systemic
ture neuronal marker Dcx and the mature neuronal marker transplantation. Among different events, which contribute to
NeuN. On the other hand, systemic transplantation of trans- early ischemic injury, MMP-induced breakdown of the blood-
www.StemCells.com
1304 Transplantation of TAT-Hsp70-Transduced NPCs

Figure 3. Analysis of brain injury after cerebral ischemia. Infarct volume analysis after occlusion of the left middle cerebral artery was performed
on day 4 after intracerebral (A, C) or systemic (B, C) injection of NPCs or PBS, respectively. Likewise, edema formation (D) and number of
TUNELþ cells within the lesion site (E) were analyzed on day 4 after intracerebral and systemic injection protocols. Neuronal density, that is, the
number of NeuNþ cells within the ischemic lesion site, was determined on day 4 (F), day 28 (G), and on day 56 (H). *Significantly different from
PBS control mice, p < .05. #Significantly different from mice treated with intracerebral injections of native NPCs or TAT-HA-transduced NPCs,
p < .05. Abbreviations: NPC, neural precursor cell; PBS, phosphate buffered saline; TAT-HA, TAT-hemagglutinin; TUNEL, dUTP nick end labeling.

brain-barrier (BBB) is one decisive key factor [47]. As such, nor TAT-Hsp70-transduced NPCs affected MMP-9 activity,
analysis of MMP gel zymography at 24 hours poststroke (Fig. whereas systemic injection of NPCs (native/transduced) yielded
6A, 6B) revealed a detection of MMP-9 activity in control ani- decreased MMP-9 activity. MMP-2 activity was below detec-
mals. However, intracerebral transplantation of neither native tion threshold under all experimental paradigms (Fig. 6A).
Doeppner, Ewert, T€onges et al. 1305

Figure 4. NPC-mediated functional recovery after cerebral ischemia. Postischemic functional deficits were analyzed for up to 8 weeks after trans-
plantation of NPCs or injection of PBS, respectively. Data are given for animals that received either intracerebral (A, C, E) or systemic (B, D, F)
injections. Motor coordination was analyzed using the rota rod (A, B), the tight rope (C, D), and the corner turn test (E, F). Using the rota rod test,
maximal test time was 300 seconds, whereas for the tight rope test, a validated score between 0 (min) and 20 (max) was defined. For the corner turn
test, the laterality index (LI) was defined as (number of left turns  number of right turns)/10. Whereas healthy animals (not depicted here) show
random side preference (i.e., a LI of approximately 0.5), mice suffering from stroke leave the corner toward the nonimpaired left side of the body.
*Significantly different from PBS animals, p < .05. #Significantly different from mice treated with intracerebral injections of native NPCs or
TAT-HA-transduced NPCs, p < .05. Abbreviations: NPC, neural precursor cell; PBS, phosphate buffered saline; TAT-HA, TAT-hemagglutinin.

Further analysis revealed that BBB integrity was significantly transduced NPCs reduced glial scar formation (Fig. 6E). On the
enhanced after systemic application of native/transduced NPCs contrary, only TAT-Hsp70-transduced NPCs mediated reduced
at 24 hours poststroke (Fig. 6C). Moreover, measurement of glial scar formation after intracerebral transplantation.
TBARS formation at 24 hours showed fewer TBARS in mice Since transplanted NPCs might affect sustained changes
systemically treated with native/transduced NPCs (Fig. 6D). In- within the ischemic lesion site via by-stander effects, we
tracerebral injection of NPCs had, however, no effect on TBARS analyzed intracerebral contents of growth factors on day 4
formation. Analysis of long-term effects like astroglial scar for- (Fig. 6F) and on day 56 (Fig. 6G). Whereas systemic transplan-
mation revealed that scar formation was independent from trans- tation of native/transduced NPCs did not significantly affect
duction states of systemically transplanted cells; both native and growth factor contents, intracerebral transplantation of NPCs
www.StemCells.com
1306 Transplantation of TAT-Hsp70-Transduced NPCs

Figure 5. Analysis of postischemic cell proliferation and differentiation. Poststroke cell proliferation was analyzed using BrdU staining on day
28 (A) and on day 56 (B). (C): Double staining of BrdUþ cells (red) with TUNELþ cells (green) within the ischemic striatum on day 4 as shown
exemplarily from animals treated with intracerebral transplantation of TAT-Hsp70-transduced NPCs (scale bar ¼ 20 lm). (D–G): Typical local-
ization of BrdUþ cells (red) with DAPI signal (blue) on day 56 within the SVZ (arrows; scale bar ¼ 10 lm) including three-dimensional (3D)
reconstruction (G). Representative image after BrdU staining (red) plus DAPI signal (blue) taken from the aforementioned animal within the is-
chemic striatum both in low (H–J; scale bar ¼ 50 lm) and high magnification (K–M; scale bar ¼ 20 lm). (N): 3D reconstruction from merged
photo (M). Differentiation analysis of BrdUþ cells (O–Z) was performed using staining against Dcx (Q–U) and NeuN (V–Z) with subsequent
quantitative analysis within the SVZ and the ischemic striatum followed by a calculation of total cell numbers. Closed columns from (O–P) represent
intracerebral injections, whereas open columns reflect systemic injections. Stainings include BrdU signal (red), DAPI (blue), and Dcx or NeuN (green)
signal with subsequent 3D-reconstruction of merged photos (U and Z). Data are given as percentages of colocalizations of BrdU and the specific marker
referred to total numbers of BrdUþ cells. Scale bars ¼ 20 lm. *Significantly different from mice treated with PBS, native NPCs, or TAT-HA-trans-
duced NPCs, p < .05. Abbreviations: BrdU, 5-bromo-2-desoxyuridine; NPC, neural precursor cell; PBS, phosphate buffered saline; SVZ, subventricular
zone; TAT-HA, TAT-hemagglutinin.

(native/transduced) increased contents of BDNF, VEGF, and


GDNF on day 4. No significant difference between the various DISCUSSION
intracerebral treatment groups was observed at that time point.
On the contrary, growth factor levels on day 56 were signifi- The efficacy of stem and progenitor cell-based therapies is
cantly increased only in mice treated with intracerebral trans- well established and has already lead to clinical trials [13, 28,
plantation of TAT-Hsp70-transduced NPCs. These results sug- 29, 31, 48–50]. In this context, NPCs that can differentiate
gest that NPC-mediated beneficial effects against stroke depend into both glia and neurons might be advantageous, albeit neu-
on the transplantation route chosen. ral cell replacement seems to be not a prerequisite for cell-
Doeppner, Ewert, T€onges et al. 1307

Figure 6. NPC-induced neuroprotective mechanisms vary with transplantation routes. (A): Gelatin zymography of brain homogenates at 24 hours after
induction of cerebral ischemia. Human pro-MMPs (0.1 ng) and activated MMPs (0.01 ng) served as standards. (B): Densitometric analysis of activated
MMP-9 from gelatin zymography at 24 hours poststroke. (C): Assessment of blood-brain-barrier permeability was assessed 24 hours poststroke by means of
Evans blue dye extravasation. Nonischemic animals (‘‘No ischemia’’) that had received no treatment served as reference. (D): Oxidative stress was indirectly
measured determining the amount of TBARS in brain homogenates of left ischemic hemispheres at 24 hours after induction of stroke. (E): Glial scar forma-
tion was analyzed using GFAP-staining on day 56 after stroke. (F, G): Determination of selected growth factors on day 4 and day 56 after stroke. Brain
homogenates from left ischemic hemispheres were prepared for measurement of BNDF, EGF, FGF, GDNF, NGF, and VEGF by means of ELISA. Closed
columns represent data after intracerebral injections, whereas open columns show data for systemic injections. Data are presented as means 6 SD. #Signifi-
cantly different from mice treated with systemic injections of any kind as well as from mice treated with intracerebral injection of PBS, p < .05. *Significantly
different from mice treated with intracerebral injections of PBS, native NPCs, or TAT-HA-transduced NPCs as well as from mice receiving any kind of
systemic injection, p < .05. Abbreviations: BDNF, brain-derived neurotrophic factor; EGF, epidermal growth factor; FGF, fibroblast growth factor; GDNF,
glial cell line-derived neurotrophic factor; MMP, matrix metalloprotease; NGF, nerve growth factor; NPC, neural precursor cell; PBS, phosphate buffered
saline; SVZ, subventricular zone; TAT-HA, TAT-hemagglutinin; TBARS, thiobarbituric acid-reactive substances; VEGF, vascular endothelial growth factor.

www.StemCells.com
1308 Transplantation of TAT-Hsp70-Transduced NPCs

based stroke therapy [20]. Nevertheless, therapeutic ferent mechanisms by which systemic and intracerebral trans-
approaches are limited due to low survival of transplanted plantation of NPCs reduce postischemic brain injury.
cells. We therefore transduced adult NPCs in vitro with the Systematic studies on NPC-induced neuroprotection
chaperone TAT-Hsp70 in order to enhance resistance of against cerebral ischemia after both systemic and local trans-
grafted cells within the postischemic milieu. Bearing in mind plantation do not exist apart from the aforementioned works
that the best cell delivery route still remains unknown, NPCs [33, 34]. Since our study indicates that systemic transplanta-
were injected either systemically or intracerebrally. Our data tion of native and transduced NPCs resulted in sustained neu-
show that sustained neuroprotection after intracerebral trans- roprotection despite low intracerebral numbers of grafted
plantation of NPCs correlates with increased survival of cells, NPC-induced modulation of extracerebral injurious
grafted cells due to TAT-Hsp70 transduction. On the other mechanisms might be intriguing. Among mechanisms leading
hand, systemic transplantation of NPCs mediates acute neuro- to acute poststroke brain injury, breakdown of the BBB is one
protection via different mechanisms, which may include stabi- key factor. This study shows that systemic transplantation of
lization of the BBB and reduction of ROS despite low intra- NPCs induces a reduced formation of ROS during reperfusion
cerebral numbers of grafted cells. and a reduction of BBB leakage. Along with this, activation
Cell-penetrating peptides like TAT-Hsp70 are an elegant of MMP-9, which is critically involved in BBB breakdown
tool to deliver cargo across intact biological membranes [51]. [47], was significantly reduced in animals that had received
In this context, we have previously shown that in vitro trans- either native or TAT-transduced NPCs. Nevertheless, one has
duction of NPCs with TAT-Bcl-xL is efficient and results in to keep in mind that a 45-minute stroke induces relatively
sustained neuroprotection against stroke after intracerebral small infarct sizes that affect both the striatum and parts of
transplantation of TAT-Bcl-xL-transduced NPCs [19]. the dorsolateral cortex resulting in rather mild breakdown of
Although Hsp70-induced neuroprotection has been shown the BBB [59].
before [10, 52–54], this study shows for the first time that the Intracerebral transplantation of NPCs did not affect BBB
fusion protein TAT-Hsp70 enhances resistance of NPCs leakage or formation of ROS as compared to systemic trans-
against hypoxic-hypoglycemic injury. Since neuroprotection plantation of NPCs. Rather, transplantation of TAT-Hsp70-
by the TAT domain itself has been described in vitro recently transduced NPCs resulted in sustained enhancement of post-
[55, 56], we used TAT-HA as a negative control in order to stroke neurogenesis as suggested by expression of the neuro-
exclude effects of the TAT domain itself on cell viability, nal markers Dcx and NeuN in BrdUþ-proliferating cells.
neurosphere formation rates, NPC numbers, and differentia- However, TAT-Hsp70 itself does neither induce neuronal
tion patterns of NPCs. differentiation of SVZ-derived NPCs in vitro nor affect dif-
Although the number of transplanted NPCs gradually ferentiation of grafted cells in vivo. Thus, enhanced post-
declined under each experimental condition, intracerebral cell stroke neurogenesis as observed by differentiation analysis
delivery always yielded higher intracerebral NPC numbers of BrdUþ cells is most likely due to the antiapoptotic prop-
than systemic cell delivery. However, intracerebral transplan- erties of the fusion protein itself. In other words, TAT-
tation of only TAT-Hsp70-transduced NPCs resulted in signif- Hsp70 protects intracerebrally transplanted NPCs from sec-
icantly enhanced cell numbers within the ischemic striatum ondary cell death resulting in sustained neuroprotection.
for up to 8 weeks when compared with controls. Despite the Taken into account that general mature neuronal differentia-
fact that transplanted NPCs were not exposed to cerebral is- tion rates of BrdUþ cells after intracerebral transplantation
chemia itself, secondary cell death of transplanted NPCs in of TAT-Hsp70-transduced NPCs were low with no mature
the process of a proinflammatory and proapoptotic ischemic neuronal phenotype of exogenous NPCs observable, neuronal
milieu has already been described before [18, 22]. Therefore, cell replacement or integration of grafted cells within the
enhanced numbers of grafted NPCs after TAT-Hsp70 trans- neural network is not likely. Rather, indirect by-stander
duction are most likely due to the antiapoptotic properties of effects orchestrating neurorestorative responses of the ische-
the fusion protein itself. mic milieu might be responsible for NPC-induced neuropro-
Analysis of brain injury and functional impairment after tection after intracerebral transplantation. As such, intracere-
intracerebral transplantation of NPCs correlated with the bral transplantation of TAT-Hsp70-transduced NPCs was
amount of grafted cells within the peri-infarct area. As such, associated with increased levels of growth factors after 8
only animals that had been transplanted with TAT-Hsp70- weeks poststroke, possibly being critically involved in NPC-
transduced NPCs showed sustained neuroprotection that was mediated long-term neuroprotection and stimulation of en-
associated with improved motor coordination. On the other dogenous neurogenesis as observed by enhanced numbers of
hand, systemic delivery of native and transduced NPCs signif- new-born mature neuronal cells. Enhanced secretion of
icantly reduced brain injury and motor coordination deficits at growth factors is also likely to be involved in mediation of
any time point analyzed, albeit only a small number of scat- subacute neuroprotection after intracerebral transplantation
tered NPCs were found within the ischemic hemisphere. of NPCs, where growth factor levels were always signifi-
Although stem cell transplantation extends the therapeutic cantly increased independent of transduction states.
time window as compared to thrombolysis, the optimal time Increased growth factor levels on day 4 after intracerebral
point for transplantation is still elusive and depends on the transplantation correlated with intracerebral cell numbers,
focus of the therapeutic aim. If reduction of brain injury and which were at that time point still high under each experi-
infarct volume is to be achieved, acute cell delivery of cells mental condition. On the contrary, reduction of growth fac-
might be most critical [20]. Consequently, we have chosen a tor contents after intracerebral transplantation of native or
6-hour time window for this study. On the other hand, manip- TAT-HA-transduced NPCs 8 weeks poststroke is likely due
ulation of endogenous repair mechanisms such as neuroplas- to secondary cell loss of these nontransduced NPCs. There-
ticity might allow cell transplantation at subacute or even fore, it is well conceivable that intracerebrally grafted NPCs
later time points [8, 29, 57, 58]. In this context, different cell act via indirect by-stander mechanisms as discussed by us
delivery routes also affect the time window for transplanta- and others [7, 8, 17, 19, 20], for which sufficiently high
tion. Whereas systemic transplantation of NPCs could benefit numbers of residing grafted cells as induced by TAT-Hsp70
from cell homing via inflammation, the latter might induce are a prerequisite. Nevertheless, a causal relation between
cell death after local cell transplantation pointing toward dif- growth factor secretion and NPC-mediated neuroprotection
Doeppner, Ewert, T€onges et al. 1309

still has to be established. On the other hand, systemic trans- initiates sustained neuroprotection despite low intracerebral
plantation of NPCs mediates acute neuroprotection from the numbers of grafted cells via different mechanisms, like stabili-
luminal side of the vessels for which high intracerebral cell zation of the BBB and reduction of ROS during early reperfu-
numbers seem to be not necessary. This observation is in sion. Therefore, systemic NPC delivery might be more feasible
line with previous reports where systemic transplantation of for clinical stroke concepts because of its simplicity and due to
stem and precursor cells initiated beneficial effects despite lower intracerebral cell numbers needed, lacking a need of in
low cell numbers or even no detectable cells within the is- vitro manipulation of NPCs such as TAT-Hsp70 transduction.
chemic brain [60–63]. Our study points out the need of a more adequate recognition
of NPC actions related to cell delivery routes.

CONCLUSIONS
DISCLOSURE OF POTENTIAL
This work shows that TAT-Hsp70 successfully enhances sur- CONFLICTS OF INTEREST
vival of NPCs after intracerebral transplantation culminating in
long-term neuroprotection against cerebral ischemia. Although G.P.H.D. is currently affiliated with the Department 851,
cell dosages for intracerebral transplantation did not vary Neurodegeneration II, H. Lundbeck A/S, Valby, Denmark. No
within this study, it is intriguing that the effects seen after in- conflict of interest results from this employment. The remaining
tracerebral transplantation depend on high intracerebral num- authors have nothing to disclose and also have no conflict of
bers of grafted cells. On the other hand, systemic NPC delivery interest.

20 Bliss TM, Andres RH, Steinberg GK. Optimizing the success of


REFERENCES cell transplantation therapy for stroke. Neurobiol Dis 2010;37:
275–283.
21 Mergenthaler P, Dirnagl U, Meisel A. Pathophysiology of stroke: Les-
1 Alvarez-Buylla A, Lois C. Neuronal stem cells in the brain of adult sons from animal models. Metab Brain Dis 2004;19:151–167.
vertebrates. Stem Cells 1995;13:263–272. 22 Dirnagl U, Iadecola C, Moskowitz MA. Pathobiology of ischaemic
2 Gross CG. Neurogenesis in the adult brain: Death of a dogma. Nat stroke: An integrated view. Trends Neurosci 1999;22:391–397.
Rev Neurosci 2000;1:67–73. 23 Brown IR. Heat shock proteins and protection of the nervous system.
3 Taupin P. Neurogenesis in the adult central nervous system. C R Biol Ann N Y Acad Sci 2007;1113:147–158.
2006;329:465–475. 24 Giffard RG, Han RQ, Emery JF et al. Regulation of apoptotic and
4 Arvidsson A, Collin T, Kirik D et al. Neuronal replacement from en- inflammatory cell signaling in cerebral ischemia: The complex roles
dogenous precursors in the adult brain after stroke. Nat Med 2002;8: of heat shock protein 70. Anesthesiology 2008;109:339–348.
963–970. 25 Chu K, Kim M, Chae SH et al. Distribution and in situ proliferation
5 Liu J, Solway K, Messing RO et al. Increased neurogenesis in the patterns of intravenously injected immortalized human neural stem-
dentate gyrus after transient global ischemia in gerbils. J Neurosci like cells in rats with focal cerebral ischemia. Neurosci Res 2004;50:
1998;18:7768–7778. 459–465.
6 Parent JM. Injury-induced neurogenesis in the adult mammalian brain. 26 Chu K, Kim M, Park KI et al. Human neural stem cells improve sen-
Neuroscientist 2003;9:261–272. sorimotor deficits in the adult rat brain with experimental focal ische-
7 Bacigaluppi M, Pluchino S, Martino G et al. Neural stem/precursor mia. Brain Res 2004;1016:145–153.
cells for the treatment of ischemic stroke. J Neurol Sci 2008;265: 27 Locatelli F, Bersano A, Ballabio E et al. Stem cell therapy in stroke.
73–77. Cell Mol Life Sci 2009;66:757–772.
8 Bacigaluppi M, Pluchino S, Peruzzotti Jametti L et al. Delayed post- 28 Kelly S, Bliss TM, Shah AK et al. Transplanted human fetal neural
ischaemic neuroprotection following systemic neural stem cell trans- stem cells survive, migrate, and differentiate in ischemic rat cerebral
plantation involves multiple mechanisms. Brain 2009;132: cortex. Proc Natl Acad Sci USA 2004;101:11839–11844.
2239–2251. 29 Andres RH, Horie N, Slikker W et al. Human neural stem cells
9 Haas S, Weidner N, Winkler J. Adult stem cell therapy in stroke. Curr enhance structural plasticity and axonal transport in the ischaemic
Opin Neurol 2005;18:59–64. brain. Brain 2011;134:1777–1789.
10 Doeppner TR, Nagel F, Dietz GPH et al. TAT-Hsp70-mediated neuro- 30 Capone C, Frigerio S, Fumagalli S et al. Neurosphere-derived cells
protection and increased survival of neuronal precursor cells after exert a neuroprotective action by changing the ischemic microenviron-
focal cerebral ischemia in mice. J Cereb Blood Flow Metab 2009;29: ment. PLoS One 2007;2:e373.
1187–1196. 31 Darsalia V, Allison SJ, Cusulin C et al. Cell number and timing of
11 Lindvall O, Kokaia Z. Stem cell research in stroke: How far from the transplantation determine survival of human neural stem cell grafts in
clinic? Stroke 2011;42:2369–2375. stroke-damaged rat brain. J Cereb Blood Flow Metab 2011;31:
12 Ma DK, Bonaguidi MA, Ming GL et al. Adult neural stem cells in the 235–242.
mammalian central nervous system. Cell Res 2009;19:672–682. 32 Ishibashi S, Sakaguchi M, Kuroiwa T et al. Human neural stem/pro-
13 Hicks AU, Lappalainen RS, Narkilahti S et al. Transplantation of genitor cells, expanded in long-term neurosphere culture, promote
human embryonic stem cell-derived neural precursor cells and functional recovery after focal ischemia in Mongolian gerbils. J Neu-
enriched environment after cortical stroke in rats: Cell survival and rosci Res 2004;78:215–223.
functional recovery. Eur J Neurosci 2009;29:562–574. 33 Li L, Jiang Q, Ding G et al. Effects of administration route on migra-
14 Mochizuki N, Takagi N, Kurokawa K et al. Injection of neural pro- tion and distribution of neural progenitor cells transplanted into rats
genitor cells improved learning and memory dysfunction after cerebral with focal cerebral ischemia, an MRI study. J Cereb Blood Flow
ischemia. Exp Neurol 2008;211:194–202. Metab 2010;30:653–662.
15 Burns TC, Verfaillie CM, Low WC. Stem cells for ischemic brain 34 Jin K, Sun Y, Xie L et al. Comparison of ischemia-directed migration
injury: A critical review. J Comp Neurol 2009;515:125–144. of neural precursor cells after intrastriatal, intraventricular, or intrave-
16 Zhang ZG, Chopp M. Neurorestorative therapies for stroke: Underly- nous transplantation in the rat. Neurobiol Dis 2005;18:366–374.
ing mechanisms and translation to the clinic. Lancet Neurol 2009;8: 35 Nagel F, Falkenburger BH, Tonges L et al. Tat-Hsp70 protects dopa-
491–500. minergic neurons in midbrain cultures and in the substantia nigra in
17 Bliss T, Guzman R, Daadi M et al. Cell transplantation therapy for models of Parkinson’s disease. J Neurochem 2008;105:853–864.
stroke. Stroke 2007;38:817–826. 36 Nagel F, Dohm CP, Bahr M et al. Quantitative evaluation of chaper-
18 Hicks AU, MacLellan CL, Chernenko GA et al. Long-term assessment one activity and neuroprotection by different preparations of a cell-
of enriched housing and subventricular zone derived cell transplanta- penetrating hsp70. J Neurosci Methods 2008;171:226–232.
tion after focal ischemia in rats. Brain Res 2008;1231:103–112. 37 Dietz GPH, B€ahr M. Synthesis of cell-penetrating peptides and their
19 Doeppner TR, El Aanbouri M, Dietz GP et al. Transplantation of application in neurobiology. Methods Mol Biol 2007;399:181–198.
TAT-Bcl-xL-transduced neural precursor cells: Long-term neuropro- 38 Bouab M, Paliouras GN, Aumont A et al. Aging of the subventricular
tection after stroke. Neurobiol Dis 2010;40:265–276. zone neural stem cell niche: Evidence for quiescence-associated

www.StemCells.com
1310 Transplantation of TAT-Hsp70-Transduced NPCs

changes between early and mid-adulthood. Neuroscience 2011;173: 51 Dietz GPH. Protection by neuroglobin, cell-penetrating peptide-medi-
135–149. ated delivery in vivo: A decade of research Comment on Cai et al.
39 Chojnacki A, Weiss S. Production of neurons, astrocytes and oligo- TAT-mediated delivery of neuroglobin protects against focal cerebral
dendrocytes from mammalian CNS stem cells. Nat Protoc 2008;3: ischemia in mice. Exp Neurol 2011;231:1–10.
935–940. 52 Badin RA, Lythgoe MF, van der Weerd L et al. Neuroprotective
40 Doeppner TR, Bretschneider E, Doehring M et al. Enhancement of en- effects of virally delivered HSPs in experimental stroke. J Cereb
dogenous neurogenesis in ephrin-B3 deficient mice after transient Blood Flow Metab 2006;26:371–381.
focal cerebral ischemia. Acta Neuropathol 2011;122:429–442. 53 Zheng Z, Kim JY, Ma H et al. Anti-inflammatory effects of the 70
41 Zhang L, Schallert T, Zhang ZG et al. A test for detecting long-term kDa heat shock protein in experimental stroke. J Cereb Blood Flow
sensorimotor dysfunction in the mouse after focal cerebral ischemia. J Metab 2008;28:53–63.
Neurosci Methods 2002;117:207–214. 54 Dietz GPH. Cell-penetrating peptide technology to deliver chaperones
42 Li X, Blizzard KK, Zeng Z et al. Chronic behavioral testing after and associated factors in diseases and basic research. Curr Pharm Bio-
focal ischemia in the mouse: Functional recovery and the effects of technol 2010;11:167–174.
gender. Exp Neurol 2004;187:94–104. 55 Meade AJ, Meloni BP, Mastaglia FL et al. AP-1 inhibitory peptides
43 Chiba Y, Sasayama T, Miyake S et al. Anti-VEGF receptor antagonist attenuate in vitro cortical neuronal cell death induced by kainic acid.
(VGA1155) reduces infarction in rat permanent focal brain ischemia. Brain Res 2010;1360:8–16.
Kobe J Med Sci 2008;54:E136–146. 56 Wei X, Miou Z, Baudry M. Neuroprotection by cell permeable TAT-
44 Noll T, de Groot H, Sies H. Distinct temporal relation among oxygen mGluR1 peptide in ischemia: Synergy between carrier and cargo
uptake, malondialdehyde formation, and low-level chemiluminescence sequences. Neuroscientist 2008;14:409–414.
during microsomal lipid peroxidation. Arch Biochem Biophys 1987; 57 Carmichael ST. Cellular and molecular mechanisms of neural repair
252:284–291. after stroke: Making waves. Ann Neurol 2006;59:735–742.
45 Dietz GPH, B€ahr M. Delivery of bioactive molecules into the cell: 58 Jin K, Mao X, Xie L et al. Delayed transplantation of human neural
The trojan horse approach. Mol Cell Neurosci 2004;27:85–131. precursor cells improves outcome from focal cerebral ischemia in
46 Lee CH, Moon SM, Yoo KY et al. Long-term changes in neuronal aged rats. Aging Cell 2010;9:1076–1083.
degeneration and microglial activation in the hippocampal ca1 region 59 Chiang T, Messing RO, Chou WH. Mouse model of middle cerebral
after experimental transient cerebral ischemic damage. Brain Res artery occlusion. J Vis Exp 2011;48:pii:2761.
2010;1342:138–149. 60 Borlongan CV, Hadman M, Sanberg CD et al. Central nervous
47 Jin R, Yang G, Li G. Molecular insights and therapeutic targets for system entry of peripherally injected umbilical cord blood cells is
blood-brain barrier disruption in ischemic stroke: Critical role of ma- not required for neuroprotection in stroke. Stroke 2004;35:
trix metalloproteinases and tissue-type plasminogen activator. Neuro- 2385–2389.
biol Dis 2010;38:376–385. 61 Guzman R, Choi R, Gera A et al. Intravascular cell replacement ther-
48 Wechsler LR. Clinical trials of stroke therapy: Which cells, which apy for stroke. Neurosurg Focus 2008;24:E15.
patients? Stroke 2009;40:S149–151. 62 Hicks A, Jolkkonen J. Challenges and possibilities of intravascu-
49 Chen J, Li Y, Wang L et al. Therapeutic benefit of intravenous admin- lar cell therapy in stroke. Acta Neurobiol Exp (Wars) 2009;69:
istration of bone marrow stromal cells after cerebral ischemia in rats. 1–11.
Stroke 2001;32:1005–1011. 63 Vendrame M, Cassady J, Newcomb J et al. Infusion of human umbili-
50 Schwarting S, Litwak S, Hao W et al. Hematopoietic stem cells cal cord blood cells in a rat model of stroke dose-dependently rescues
reduce postischemic inflammation and ameliorate ischemic brain behavioral deficits and reduces infarct volume. Stroke 2004;35:
injury. Stroke 2008;39:2867–2875. 2390–2395.

See www.StemCells.com for supporting information available online.

You might also like