You are on page 1of 10

Experimental Neurology 226 (2010) 90–99

Contents lists available at ScienceDirect

Experimental Neurology
j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / y e x n r

Increased subventricular zone-derived cortical neurogenesis after ischemic lesion


Maria Kreuzberg a,1, Evgeny Kanov a,b,1, Oleg Timofeev b, Markus Schwaninger c,
Hannah Monyer a, Konstantin Khodosevich a,⁎
a
Department of Clinical Neurobiology, Interdisciplinary Center for Neurosciences, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
b
Trans-Technologies, Ltd, St. Petersburg, Pesochny settl., Leningradskaya ul., 70/4, build. 6, Russia
c
Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany

a r t i c l e i n f o a b s t r a c t

Article history: In adult rodents stroke enhances neurogenesis resulting in the addition of neurons to forebrain regions such
Received 31 March 2010 as striatum or cortex where postnatal neurogenesis under normal conditions plays a negligible role. In the
Revised 3 August 2010 cortex, new neurons are generated either from local cortical precursors that are activated by stroke or from
Accepted 7 August 2010
precursors residing in the subventricular zone (SVZ) of lateral ventricles that under normal conditions
Available online 14 August 2010
supply neuroblasts by and large only for the olfactory bulb. In this study we used 5HT3A-EGFP transgenic
Keywords:
mice in which all neuroblasts originating in the SVZ are EGFP-labeled. We induced stroke in these mice and
Stroke by combination of EGFP detection with BrdU injections we labeled all post-stroke-generated SVZ-derived
SVZ neuroblasts. We showed an increase in SVZ-derived neuroblasts 14 and 35 days after stroke in the ipsilateral
Neurogenesis hemisphere. Post-stroke-generated SVZ-derived neuroblasts migrated to the cortex and survived for at least
Cortex 35 days representing 2% of BrdU-positive cells in peri-infarct area where they differentiate into mature
Neuroblasts neurons. Thus, stroke enhances SVZ neurogenesis and attracts newborn neurons to the injury zone.
© 2010 Elsevier Inc. All rights reserved.

Introduction neurogenic regions and promote migration of neuroblasts to the


ischemic boundary in the cortex and striatum (Arvidsson et al., 2002;
Stroke, one of the most common causes of death and disability in Jin et al., 2001; Parent et al., 2002). In the striatum it has been
adulthood, is triggered by occlusion of a cerebral artery leading to described that these newborn neuroblasts differentiate into GABAer-
brain infarction. The resulting functional neurological deficits are gic medium spiny neurons or calretinin-positive GABAergic aspiny
mainly due to the subsequent massive loss of neurons, suggesting that inhibitory interneurons (Arvidsson et al., 2002; Hou et al., 2008; Liu et
cell replacement therapy might reduce the long-term consequences in al., 2009; Parent et al., 2002; Yang et al., 2008) and that the latter are
stroke patients. integrated into neuronal networks receiving synaptic input and firing
There is an increasing body of evidence that stroke, which action potentials (Hou et al., 2008; Yamashita et al., 2006). In contrast,
experimentally can be induced in rats and mice by transient or relatively little is known about the fate of SVZ-derived neuroblasts
permanent middle cerebral artery occlusion, can trigger neurogenesis that are directed to the cortex following cortical stroke.
in the subventricular zone (SVZ) of the lateral ventricle and the In the present study we analyzed the contribution of SVZ
subgranular zone (SGZ) of the dentate gyrus (Arvidsson et al., 2002; progenitor cells to cortical neurogenesis in a cortex-specific stroke
Dempsey et al., 2003; Jin et al., 2001; Komitova et al., 2005; Parent model caused by middle cerebral artery occlusion (MCAO) using
et al., 2002; Zhang et al., 2001), reviewed in (Wiltrout et al., 2007). 5HT3A-EGFP transgenic mice, where EGFP is expressed under
Under physiological conditions, neuroblasts generated in the SVZ promoter of serotonin receptor 5HT3A (Inta et al., 2008). In these
migrate tangentially along the rostral migratory stream (RMS) to the mice all SVZ-derived doublecortin-positive cells (=neuroblasts) are
olfactory bulb where they continue to migrate radially to reach their EGFP-labeled. Upon maturation EGFP expression decreases in the
final destination and differentiate into GABAergic periglomerular and majority of neurons, but persists in those neurons, that normally
granule neurons (Ghashghaei et al., 2007; Khodosevich et al., 2009; express the 5HT3A receptor (e.g., periglomerular cells in olfactory
Lledo et al., 2006). Neurogenesis in the SGZ gives rise to glutamatergic bulb or layer 1 cortical interneurons). In addition, EGFP never co-
granule cells in the dentate gyrus (Abrous et al., 2005; Kempermann localizes with glial markers. Thus, by combining EGFP detection and
et al., 2004). Ischemic brain injury can increase neurogenesis in both BrdU injections we were able to follow the migratory routes of post-
stroke-generated SVZ-derived neuroblasts. We found an increase in
⁎ Corresponding author. Fax: + 49 6221 561397.
post-stroke-generated neuroblasts in the SVZ of ischemic mice in both
E-mail address: khodosevich@urz.uni-hd.de (K. Khodosevich). the ipsilateral and contralateral hemisphere. However, while the
1
Equal contribution. increase in neuroblast generation in the ipsilateral SVZ could be

0014-4886/$ – see front matter © 2010 Elsevier Inc. All rights reserved.
doi:10.1016/j.expneurol.2010.08.006
M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99 91

detected already 14 days after stroke, in the contralateral SVZ it was incubated for 1 h with 5% normal goat serum (NGS) or 5% BSA in 0.1%
observed only 35 days after stroke. In the peri-ischemic zone, 1% and Triton-X-100 and subsequently overnight at 4 °C with primary
2% of all BrdU-positive cells were EGFP/BrdU double-positive antibodies. After washing steps with PBS, sections were incubated
neuroblasts/neurons 14 days and 35 days after stroke, respectively. for 2 h with the respective secondary antibodies and mounted.
In the cortex the majority of EGFP/BrdU double-positive neuroblasts For BrdU double or triple labeling sections were washed in PBS and
survived for at least 35 days after stroke. Many of them expressed incubated with 1 N HCl for 45 min followed by neutralization in
mature neuronal marker NeuN and were not labeled by apoptotic 10 mM Tris–HCl pH 8.5 for 15 min. Sections were washed in PBS,
marker activated caspase-3. In the frontal cortex, a portion of post- incubated for 1 h with 5% BSA in 0.2% Triton X-100 and subsequently
stroke generated SVZ-derived neurons co-expressed calretinin, a overnight at 4 °C with primary anti-BrdU antibodies. After washing
marker for a GABAergic interneuron subclass. steps with PBS, sections were incubated for 2 h with the respective
secondary antibodies.
Materials and methods Sections were mounted on microscopy glass slides using Mowiol
(Sigma-Aldrich, Germany) and were analyzed with Olympus BX51TF
Animals fluorescent (Olympus, Germany) and Zeiss LSM510 confocal micro-
scopes (Carl Zeiss, Germany).
For all our experiments, we used 5HT3A-EGFP transgenic mice
(Inta et al., 2008). All procedures with animals were performed Statistical analysis
according to the Heidelberg University Animal Care Committee.
The size of the areas containing EGFP-positive neuroblasts/
Materials and reagents neurons in the subventricular zone and rostral migratory stream
was measured using ImageJ software and statistically analyzed.
The following antibodies were used in the study: rabbit anti-EGFP Images were analyzed on every 3rd section through the whole SVZ
(1:10000; Molecular Probes, USA), chicken anti-EGFP (1:1500; (13 sections for each brain, 7–9 brains for each condition) at high
Abcam, UK), mouse anti-calretinin (1:1000; Swant, Switzerland), magnification. All green fluorescent cells (up to individual neuro-
rabbit anti-calretinin (1:1500; Swant, Switzerland), rabbit anti- blasts) were outlined in ImageJ and included into analysis. If data
activated caspase-3 (1:1000; R&D Biosystems, USA), rat anti-BrdU were not normally distributed according to d'Agostino and Shapiro–
(1:500; Accurate, USA), mouse anti-NeuN (1:500; BD Biosciences, Wilk tests, we used Kruskal–Wallis test for multiple comparisons and
USA), goat anti-doublecortin (1:500; Santa Cruz, Germany), mouse Mann–Whitney test with Bonferroni correction for comparison
anti-GFAP (1:4000; Sigma-Aldrich, Germany), goat anti-Olig2 (1:500; between single groups. For normally distributed data t-test was
R&D Biosystems, USA), mouse anti-Mash1 (1:500; BD Biosciences, used. Differences were considered significant at p b 0.05. All values in
USA), rabbit anti-nestin (1:500; Abcam, UK), mouse anti-PSA-NCAM graphs are mean ± standard deviation. For the expression analysis of
(1:1000, Millipore, USA), Cy3-conjugated goat anti-rabbit, donkey BrdU, BrdU/EGFP, BrdU/EGFP/activated caspase-3, BrdU/EGFP/Cr
anti-goat and goat anti-rat (1:1000; Jackson Immuno, Germany), Cy5- and BrdU/EGFP/NeuN, at least 5 brains for each condition were used
conjugated goat anti-rat, goat anti-mouse, donkey anti-goat and goat and cell count was performed on 5–12 coronal sections in peri-infarct
anti-rabbit (1:1000; Jackson Immuno, Germany), Alexa 488-conju- area. The peri-infarct area was defined as a 300-μm brain region
gated goat anti-chicken and goat anti-rabbit (1:1000; Molecular surrounding the ischemic scar.
Probes, USA).
Results
Middle cerebral artery occlusion (MCAO)
Generation of SVZ neuroblasts is enhanced after ischemic stroke
For MCAO, 16–18 weeks old male 5HT3A-EGFP transgenic animals
(Inta et al., 2008) were used. MCAO was performed as previously Although several studies demonstrated an increase in SVZ
described (Zhang et al., 2005). Briefly, mice were anesthetized by neurogenesis after ischemic stroke, they did not directly estimate
intraperitoneal injection of 150–200 μl 2.5% avertin solution per 10 g the increase in SVZ-derived neuroblasts in the cortex (Arvidsson et al.,
body weight. A skin incision was made between the left ear and the 2002; Jiang et al., 2001; Jin et al., 2003; Leker et al., 2007; Parent et al.,
orbit, and the temporal muscle was removed by electrical coagulation 2002; Zhang et al., 2006, 2001; Ziv et al., 2007). We investigated
(Erbe, Germany). The skull was opened using a driller and the middle cortical neurogenesis after stroke combining BrdU injections and
cerebral artery was occluded by electrical coagulation. The wound EGFP detection in SVZ-derived neurons. BrdU injections allow the
was closed with a skin suture. During surgery animals were kept at labeling of proliferating cells and their descendants. However, since
body temperature using a heating pad. Only those animals that had the vast majority of BrdU-labeled cells in the ischemic zone are
the similar extent of brain damage were used for the analysis. microglia and astrocytes (Wang et al., 2004), the study of neurogen-
esis requires a combinatorial approach using in addition to BrdU
BrdU application and dissection of brains appropriate neuroblast markers. Here we utilized 5HT3A-EGFP mice,
thus profiting from the EGFP expression in all neuroblasts originating
On days 1–6 after MCAO, mice obtained BrdU (Sigma-Aldrich in the SVZ (Inta et al., 2008) and investigated the extent of SVZ
GmbH, Germany) to label dividing cells. BrdU was administered at a response to stroke.
concentration of 50 μg per g of body weight twice a day by We injected 5HT3A-EGFP mice with BrdU on days 1–6 after stroke
intraperitoneal injections. One or 4 weeks after the last BrdU and analyzed post-stroke neurogenesis in the SVZ 1 or 4 weeks after
injection, animals were deeply anesthetized with 2.5% avertin the last BrdU injection (Fig. 1A). Fourteen days after stroke, the area of
solution and perfused with 4% PFA. Brains were dissected and EGFP-positive cells (neuroblasts) in the SVZ and RMS was signifi-
postfixated with 4% PFA overnight at 4 °C. cantly increased in the ischemic hemisphere compared to the non-
ischemic hemisphere or sham-operated controls (7–9 animals per
Immunohistochemistry group) (Fig. 1B–D, for confocal images of EGFP/BrdU cells see Fig. 4).
We observed an increase in SVZ neuroblast production in response to
Fifty-μm-thick sections were made using a Leica VT100S vibratome stroke in both RMS and SVZ (Fig. 1C, D). Neuroblast generation in the
(Leica Microsystems GmbH, Germany). Sections were washed in PBS, non-ischemic hemisphere was also slightly enhanced (Fig. 1B–D).
92 M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99

Fig. 1. SVZ neurogenesis after cerebral ischemia in 5HT3A-EGFP mice. (A) Scheme of experiment. BrdU injections started 1 day after MCAO and continued for the 6 following days.
The analysis was performed 7 and 28 days after the last BrdU injection. (B–D) 14 days post-ischemia there was an increase in neuroblasts in the ischemic hemisphere in comparison
to the non-ischemic hemisphere and sham-operated animals. There was also a slight increase in the number of neuroblasts in the non-ischemic hemisphere in comparison to sham-
operated animals (*p b 0.001). (E–G) 35 days after MCAO, the number of neuroblast in the ischemic hemisphere was comparable to that at 14 days after MCAO, while neuroblast
number in the non-ischemic hemisphere increased significantly and almost reached that of the ischemic hemisphere (*p b 0.001). Scale bars—100 μm.

Five weeks after stroke, the area of EGFP-positive neuroblasts in in the ischemic hemisphere was similar in size at 14 and 35 days after
both SVZ and RMS was still significantly larger in the ischemic stroke, in the non-ischemic hemisphere the area of SVZ-derived
hemisphere compared to that in sham-operated mice (Fig. 1E–G, for neuroblasts was larger after 35 days compared to the earlier time-
confocal images of EGFP/BrdU cells see Fig. 4). In addition, the area of point at 14 days post stroke (Fig. 1C, D and F, G, respectively). At the
EGFP-positive neuroblasts in the non-ischemic hemisphere was later time-point the area of SVZ-derived neuroblasts in the non-
also significantly larger compared to that in sham operated mice ischemic hemisphere was almost comparable in size with the
(Fig. 1E–G). Interestingly, while the area of SVZ-derived neuroblasts corresponding area in the ischemic hemisphere (Fig. 1F, G).
M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99 93

Neuroblasts produced in SVZ after stroke migrate and are retained in the BrdU/EGFP-double-positive cell count = 53± 29]. However, since the
injured hemisphere number of BrdU-positive cells was dramatically decreased 35 days after
MCAO relative to 14 days after MCAO (Fig. 4C and B, respectively), the
Two weeks after stroke we observed a stream of neuroblasts absolute number of EGFP/BrdU-double positive cells 35 days after
descending from the SVZ into the ischemic area (Fig. 2A, B). In the MCAO was only slightly lower than 14 days after MCAO.
“ischemic” stream, all EGFP-positive cells co-expressed PSA-NCAM
and DCX (doublecortin), neuroblast markers (Fig. 2B1 and B2, Many post-stroke-generated SVZ-derived cortical neurons do not die by
respectively). Concomitantly, in the SVZ and RMS there was 100% apoptosis
co-localization between EGFP and DCX cells (Fig. 3A). Also, all EGFP/
BrdU-double positive cells in peri-infarct area co-expressed DCX Previous studies described that many neurons generated in the
2 weeks, but not 5 weeks after stroke (data not shown). However, at cortex following stroke did not survive, and indicated that the
both time-points analyzed, EGFP-positive cells were never co-labeled neurons died, most probably by caspase-dependent apoptosis
with Mash1 and nestin (Mash1 is a marker of transit-amplifying (Arvidsson et al., 2002; Thored et al., 2007). Hence, we examined
precursors and nestin is a marker for neural stem cells and transit- the survival of post-stroke born SVZ-derived neurons that migrated to
amplifying precursors in SVZ (Abrous et al., 2005; Lledo et al., 2006), the peri-infarct area by immunolabeling with apoptotic markers. Of all
Fig. 3B and C, respectively), as well as with glial markers - GFAP EGFP/BrdU double-positive cells, only 5% and 3% were also positive
(astrocytic marker, Fig. 3D) and Olig2 (oligodendrocytic marker, for activated caspase-3, at 14 and 35 days after MCAO, respectively
Fig. 3E). In Fig. 3F we have summarized the expression of EGFP in (Fig. 5A–D) [for 14 days n = 5, 10 sections for each brain (every 3rd
5HT3A-EGFP transgenic mouse brain using data from this study and section of infarcted cortex), BrdU/EGFP cells = 112 ± 26, BrdU/EGFP/
from (Inta et al., 2008; Vucurovic et al., 2010). Note that EGFP is caspase-3 cells = 6 ± 4; for 35 days n = 5, 10 sections for each brain
expressed in practically all neuroblasts born in caudal ganglionic (every 3rd section of infarcted cortex), BrdU/EGFP cells = 102 ± 59,
eminence and postnatal SVZ (Inta et al., 2008), while after maturation BrdU/EGFP/caspase-3 cells = 4 ± 2]. Thus, many of SVZ-derived
EGFP expression ceases in the majority of neurons. neurons do not die via apoptosis and survive in the peri-infarct area
We next analyzed the extent of post-stroke-generated neuroblast at least 5 weeks after stroke (note that in experiments shown in Fig. 5
migration to the injured cortex and whether they survive after several we used twice more brain sections in comparison to Fig. 3 experi-
weeks. BrdU incorporation in EGFP-positive cells of 5HT3A-EGFP mice ments, which resulted in more BrdU/EGFP cell counts).
provides direct evidence that they were generated after stroke. Two
weeks after MCAO there were a number of post-stroke-generated SVZ- Post-stroke-born SVZ-derived cortical neurons express markers of
derived neuroblasts in the peri-infarct area (Fig. 4A, B, B'). The majority mature cells
of EGFP/BrdU-double positive cells had only one process (Fig. 4B') and
in total these cells accounted for around 1% of BrdU-positive cells in the Finally, we analyzed whether neuroblasts generated in the SVZ
peri-infarct area (Fig. 4D) [n = 5, 5 sections for each brain (every 6th after stroke mature to cortical interneurons. Thirty-five days after
section of infarcted cortex), BrdU-positive cell count = 3811 ± 752, MCAO only 50% of EGFP/BrdU cells in the cortex were NeuN-positive
BrdU/EGFP-double-positive cell count = 58 ± 14]. (Fig. 6A, B). However, this very likely is an underestimation of cortical
Thirty-five days after MCAO there were still a significant number of post-stroke-generated SVZ-derived neurons because of two factors:
EGFP/BrdU-double-positive cells in peri-infarct area—around 2% of first, in 5HT3A-EGFP mice a large fraction of postnatally generated
BrdU-positive cells (Fig. 4C, C', D) [n = 6, 5 sections for each brain (every neurons (granule cells of the olfactory bulb) loses EGFP expression
6th section of infarcted cortex), BrdU-positive cell count = 2572 ± 774, upon maturation (data not shown) and second, the majority of SVZ-

Fig. 2. SVZ neuroblasts migrate to ischemic zone. (A–B) A stream of EGFP-positive SVZ neuroblasts migrating to the ischemic zone 2 weeks after MCAO. Cells in the stream express
neuroblast markers—PSA-NCAM (B1) and DCX (B2). In (A) white line borders the ischemic scar. Scale bars: in (A) 100 μm, in (B) 50 μm.
94 M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99

Fig. 3. Characterization of EGFP-positive cells in the ischemic brain. All EGFP-positive cells in SVZ co-express neuroblast marker DCX (A), but never express transit-amplifying
precursor marker Mash1 (B) or neural stem cell marker nestin (C). (D) EGFP-positive cells in the peri-infarct area do not express astrocytic marker GFAP. (E) EGFP-positive cells in
the peri-infarct area do not express oligodendrocytic marker Olig2. Note that overview images in (D) and (E) are projections of 5 stacks of 2.5-μm width while single cell confocal 3D
images are one 2.5-μm stack. In (E) white line borders the ischemic scar. (F) A scheme of EGFP expression in the brain of 5HT3A-EGFP mice relative to different stages of neuronal
differentiation produced using data obtained in this work as well as in (Inta et al., 2008; Vucurovic et al., 2010). Note that a small proportion of 5HT3A-EGFP neuroblasts continue to
express EGFP when mature. Scale bars—50 μm.

derived cortical neurons generated postnatally under normal condi- population in lower cortical layers which comprises the majority of
tions do not express NeuN, but express the interneuronal marker postnatally born cortical neurons—these EGFP/BrdU/Cr + neurons do
calretinin (Cr) [in (Inta et al., 2008) see EGFP/BrdU/Cr + neuronal not express NeuN (Khodosevich, Monyer, unpublished observation)].
M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99 95

Fig. 4. Post-stroke-generated SVZ-derived neuroblasts/neurons in the cortex. (A) EGFP/BrdU double-positive neuroblasts in the ipsilateral hemisphere migrate beyond the common
migratory pathways to the infarcted cortex. Arrows indicate double-positive cells. (B), (B') and (C), (C') post-stroke-generated SVZ-derived neuroblasts/neurons in the cortex
14 days and 35 days after MCAO, respectively. Arrowheads indicate double-positive cells. (D) Percentage of EGFP/BrdU double-positive cells relative to all BrdU-positive cells in the
peri-infarct area 14 and 35 days after MCAO. Scale bars: in (A) 50 μm, in (B) and (C) 100 μm, in (B') and (C') 20 μm.

The lack of NeuN expression has been reported previously for neurons the nearby RMS was much higher in the ischemic hemisphere in
in several brain regions, e.g. cerebellum, olfactory bulb, substantia comparison to the non-ischemic hemisphere 35 days after MCAO
nigra (Cannon and Greenamyre, 2009; Mullen et al., 1992; Weyer and (Fig. 7A–E) [n = 8, 12 sections for each brain (every 3rd section of
Schilling, 2003). frontal cortex), BrdU/EGFP/Cr cells in ischemic hemisphere= 28 ± 14,
To assess the maturation of post-stroke-generated SVZ-derived BrdU/EGFP/Cr cells in non-ischemic hemisphere = 8 ± 4]. Thus, a
neurons, we analyzed the population of neurons in 5HT3A-EGFP portion of SVZ-derived neuroblasts, generated after stroke, migrates
transgenic mice that still express EGFP when mature. Using 5HT3A- to the frontal cortex and differentiates into Cr-positive interneurons.
EGFP mice, we have previously shown that upon maturation
postnatally SVZ-generated neurons in deep cortical layers of the Discussion
frontal cortex (Inta et al., 2008) express the interneuronal marker Cr.
Therefore we investigated the fate of EGFP-positive neuroblasts after In the present study we investigated post-stroke neurogenesis and
cerebral ischemia and focused on the population of Cr-positive its contribution to the generation of neurons in the cortex using the
interneurons in deep frontal cortex layers. We found that the number MCAO model in 5HT3A-EGFP transgenic mice with EGFP expression in
of triple-positive EGFP/BrdU/Cr interneurons in the frontal cortex and SVZ-derived neurons. These transgenic mice in combination with
96 M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99

Fig. 5. Apoptosis in cortical post-stroke-generated SVZ-derived neuroblasts/neurons. (A), (C) and (B), (D) Only few EGFP/BrdU double-positive cells are also activated caspase-3
(casp-3)-positive 14 and 35 days after stroke, respectively. Arrowheads—EGFP/BrdU double-positive, but activated caspase-3-negative cells. Arrow—EGFP/BrdU/activated caspase-3
triple-positive cell. Scale bars—20 μm.

BrdU injections provided us a novel tool to track SVZ-derived cortical SVZ-derived neuroblasts, generated in response to the stroke,
neuroblasts produced during the first days after stroke. We found differentiated into mature neurons expressing NeuN or calretinin.
that following stroke the neuroblast production in the SVZ is Although many studies described cortical neurogenesis after
enhanced in both the ipsilateral and contralateral hemisphere. We stroke (Arvidsson et al., 2002; Jiang et al., 2001; Jin et al., 2003;
showed that neuroblasts migrated to the cortex and could be detected Leker et al., 2007; Parent et al., 2002; Zhang et al., 2006, 2001; Ziv
35 days after the infarct. The majority of SVZ-derived cortical et al., 2007), they did not address the extent of SVZ-derived
neuroblasts/neurons did not express apoptotic markers and many neuroblast recruitment to the cortex in response to stroke. To control

Fig. 6. Maturation of post-stroke-generated SVZ-derived neurons in ischemic brain. (A) and (B) EGFP/BrdU/NeuN triple-positive cells in the cortex 35 days after MCAO. Scale
bars—10 μm.
M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99 97

Fig. 7. Maturation of post-stroke-generated SVZ-derived neurons in frontal cortex of ischemic hemisphere. EGFP/BrdU and interneuronal marker calretinin (Cr) triple-positive
neurons in the contralateral (A) and the ipsilateral (B) to the stroke hemisphere (in the area adjacent to RMS). Note higher number of triple-positive cells in the ipsilateral
hemisphere. (C and D) Examples of EGFP/BrdU/Cr triple-positive cells in frontal cortex. (E) The number of EGFP/BrdU/Cr triple-positive cells in frontal cortex is dramatically
increased in the ipsilateral hemisphere compared to the contralateral side (*p b 0.005). Scale bars: in (A) and (B), 20 μm; in (C) and (D), 10 μm.
98 M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99

both birth-dating and the origin of neurons, combined BrdU injections increased number of newborn Cr-positive interneurons in the
with transgenic or viral labeling of SVZ-derived neuroblasts is ipsilateral hemisphere. However, we cannot clarify whether this
required. Such an approach was used by Burns et al. (2007) to trace increase in Cr-positive interneurons is a result of the overall increased
SVZ-derived neuroblasts in the striatum after stroke. The authors proliferation in the ipsilateral SVZ only or whether in addition certain
found only few BrdU-positive cells that were produced in the SVZ, regional cues might be present in the cortex of the ipsilateral
which indicates only a small contribution of the SVZ to striatal post- hemisphere, facilitating the integration of newborn neuroblasts into
stroke neurogenesis. However, this can be an underestimation, cortical circuits.
because tamoxifen-activated Cre recombination, which was used for According to our data there are 50–100 Cr-positive cells per
cell tracking, might not label all precursor cells since in some adult Cre hemisphere that are generated in SVZ during the first days following
expressing strains the transgenic promoter is methylated and Cre stroke (28 ± 14 in every third section per ischemic hemisphere).
recombination is inhibited (see e.g. (Long and Rossi, 2009)). In our Although the number of cells is not that high, activation of neuronal
study, we used transgenic mice expressing EGFP under the control of generation after injury can reflect the plasticity of adult/mature brain
the 5HT3A promoter that allowed the tracing of all neuroblasts and its adaptation to external stimuli. These Cr-positive cells migrate
generated in the SVZ. We combined MCAO experiments with BrdU to the cortical region that is far away from the injury zone and might
injections, thus, labeling all EGFP-positive neuroblasts that were represent an intrinsic brain attempt to recover from the injury.
produced in the SVZ in response to stroke. The contribution of the SVZ However, it remains to be investigated whether these post-stroke
to cell replacement in injured cortex was significant given that 1% of generated cells are integrated in the local circuits.
BrdU-positive cells in the peri-infarct area were also EGFP-positive, i.e. It was shown in several studies that cell proliferation in the
neuroblasts/neurons, 14 days after MCAO and 2% of BrdU-positive ipsilateral SVZ was increased already 1 to 2 weeks after stroke and
cells were EGFP-positive 35 days after MCAO (Fig. 4D). The increase returned to baseline 6 weeks after stroke (Arvidsson et al., 2002;
that at a first glance might be puzzling can be accounted for by the fact Zhang R.L. et al., 2004; Zhang R. et al., 2004). In the current study, the
that BrdU labeling also includes a number of astrocytes and microglia, number of neuroblasts in the ipsilateral SVZ was twice that of the
many of which do not survive (Wang et al., 2004). The absolute contralateral SVZ 2 weeks after stroke. However, 5 weeks after the
number of EGFP/BrdU double-positive cells in post-ischemic cortex stroke, the number of neuroblasts in the contralateral SVZ was almost
was slightly higher at the earlier time point. comparable to that of the ipsilateral SVZ. Although Leker et al. (2007)
We showed in a previous study (Inta et al., 2008) and confirmed it showed an increase in newborn cells in the contralateral hemisphere
here that all EGFP-positive cells generated in the SVZ co-express 30 days after stroke, the effect was milder than in the ipsilateral
neuroblast marker DCX. Thus, all cells that are migrating to the hemisphere. In our study, the area of neuroblasts in both ipsilateral
ischemic zone are neuroblasts. The reactivation of 5HT3A promoter, and contralateral SVZ was approximately the same and 2 times larger
and subsequent labeling of non-SVZ-derived cells, in some local compared to sham-operated mice. Thus, we showed that in the
precursors is very unlikely due to several reasons. First, following hemisphere contralateral to stroke, the SVZ also reacts to brain injury,
stroke we always observed a stream of migrating EGFP-positive but later than ipsilaterally. This might be due to the longer time
neuroblasts starting in the SVZ and ending in the peri-infarct area (see required for some factors to diffuse to the contralateral SVZ to trigger
Fig. 2A, B). Second, local cortical precursors generate many glial cells an increase in neuroblast production. Alternatively, some regional
in response to stroke, while EGFP-positive cells never co-express glial factors first influence the ipsilateral hemisphere and stimulate
markers. And finally, SVZ precursors and adult cortical precursors are neurogenesis and later systemic factors modulate the activity in the
quite different cells, and in 5HT3A-EGFP mice only those cells that whole brain.
were born in postnatal SVZ and prenatal caudal ganglionic eminence In spite of major efforts to develop treatment following stroke, it
are labeled (Inta et al., 2008). has remained one of the leading causes of death and disability in
In the striatum around 80% of post-stroke-generated neurons die adults. There is an increasing number of studies showing that
between 2 and 6 weeks after stroke (Arvidsson et al., 2002). This progenitor cells residing in the SVZ are potentially able to replace
massive cell death is most likely caspase-dependent since inhibition the loss of nervous tissue after stroke. Thus, modulation of intrinsic
of caspase function remarkably increased neuronal survival in the neurogenesis in SVZ following stroke is a promising approach aiming
striatum (Thored et al., 2007). We showed in this study that many of at functional restoration of damaged brain circuits and increased
cortical post-stroke-generated neurons did not express activated recovery after stroke.
caspase-3 and survived at least 5 weeks after stroke.
We demonstrated that at least half of the BrdU/EGFP cells 35 days Acknowledgments
after MCAO were mature neurons as they exhibited expression of the
neuronal marker NeuN. However, it is possible that in these transgenic This work was supported in part by the DFG (SFB488 grant), the
mice we underestimated the number of SVZ-derived mature neurons BMBF (RUS 09/B38), and European Union's Seventh Framework
since the majority of cortical neurons generated postnatally in SVZ Programme (FP7/2007-2013) under grant agreements 201024 and
under non-pathological conditions are Cr-positive, but NeuN-negative 202213 (European Stroke Network).
[see above and (Inta et al., 2008)]. Also, in other brain regions such as the
olfactory bulb we could observe that 5HT3A-EGFP-positive neuroblasts
References
upon differentiation lose EGFP expression (data not shown). Interest-
ingly, Leker et al (Leker et al., 2007) showed in the distal MCAO rat Abrous, D.N., Koehl, M., Le Moal, M., 2005. Adult neurogenesis: from precursors to
model that 3 months after MCAO 12% of BrdU-positive cells were also network and physiology. Physiol. Rev. 85, 523–569.
Arvidsson, A., Collin, T., Kirik, D., Kokaia, Z., Lindvall, O., 2002. Neuronal replacement
NeuN-positive. This discrepancy might result from species differences
from endogenous precursors in the adult brain after stroke. Nat. Med. 8, 963–970.
(rat versus mouse) or different time points of analysis (90 versus Burns, K.A., Ayoub, A.E., Breunig, J.J., Adhami, F., Weng, W.L., Colbert, M.C., Rakic, P.,
35 days after MCAO). Also, it is possible that the majority of post-stroke- Kuan, C.Y., 2007. Nestin-CreER mice reveal DNA synthesis by nonapoptotic neurons
following cerebral ischemia hypoxia. Cereb. Cortex 17, 2585–2592.
generated neurons are derived from local cortical precursors rather than
Cannon, J.R., Greenamyre, J.T., 2009. NeuN is not a reliable marker of dopamine neurons
SVZ cells, and, thus, omitted from our analysis. in rat substantia nigra. Neurosci. Lett. 464, 14–17.
Previously we have shown that many Cr-positive interneurons in Dempsey, R.J., Sailor, K.A., Bowen, K.K., Tureyen, K., Vemuganti, R., 2003. Stroke-induced
the frontal cortex are generated postnatally from SVZ precursors (Inta progenitor cell proliferation in adult spontaneously hypertensive rat brain: effect of
exogenous IGF-1 and GDNF. J. Neurochem. 87, 586–597.
et al., 2008). This study reveals, that the generation of these Cr- Ghashghaei, H.T., Lai, C., Anton, E.S., 2007. Neuronal migration in the adult brain: are we
positive interneurons is stimulated after stroke resulting in an there yet? Nat. Rev. Neurosci. 8, 141–151.
M. Kreuzberg et al. / Experimental Neurology 226 (2010) 90–99 99

Hou, S.W., Wang, Y.Q., Xu, M., Shen, D.H., Wang, J.J., Huang, F., Yu, Z., Sun, F.Y., 2008. Vucurovic, K., Gallopin, T., Ferezou, I., Rancillac, A., Chameau, P., van Hooft, J.A., Geoffroy,
Functional integration of newly generated neurons into striatum after cerebral H., Monyer, H., Rossier, J., Vitalis, T., 2010. Serotonin 3A receptor subtype as an early
ischemia in the adult rat brain. Stroke 39, 2837–2844. and protracted marker of cortical interneuron subpopulations. Cereb. Cortex.
Inta, D., Alfonso, J., von Engelhardt, J., Kreuzberg, M.M., Meyer, A.H., van Hooft, J.A., Wang, K., Bekar, L.K., Furber, K., Walz, W., 2004. Vimentin-expressing proximal reactive
Monyer, H., 2008. Neurogenesis and widespread forebrain migration of distinct astrocytes correlate with migration rather than proliferation following focal brain
GABAergic neurons from the postnatal subventricular zone. Proc. Natl Acad. Sci. injury. Brain Res. 1024, 193–202.
USA 105, 20994–20999. Weyer, A., Schilling, K., 2003. Developmental and cell type-specific expression of the
Jiang, W., Gu, W., Brannstrom, T., Rosqvist, R., Wester, P., 2001. Cortical neurogenesis in neuronal marker NeuN in the murine cerebellum. J. Neurosci. Res. 73, 400–409.
adult rats after transient middle cerebral artery occlusion. Stroke 32, 1201–1207. Wiltrout, C., Lang, B., Yan, Y., Dempsey, R.J., Vemuganti, R., 2007. Repairing brain after
Jin, K., Minami, M., Lan, J.Q., Mao, X.O., Batteur, S., Simon, R.P., Greenberg, D.A., 2001. stroke: a review on post-ischemic neurogenesis. Neurochem. Int. 50, 1028–1041.
Neurogenesis in dentate subgranular zone and rostral subventricular zone after Yamashita, T., Ninomiya, M., Hernandez Acosta, P., Garcia-Verdugo, J.M., Sunabori, T.,
focal cerebral ischemia in the rat. Proc. Natl Acad. Sci. USA 98, 4710–4715. Sakaguchi, M., Adachi, K., Kojima, T., Hirota, Y., Kawase, T., Araki, N., Abe, K., Okano,
Jin, K., Sun, Y., Xie, L., Peel, A., Mao, X.O., Batteur, S., Greenberg, D.A., 2003. Directed H., Sawamoto, K., 2006. Subventricular zone-derived neuroblasts migrate and
migration of neuronal precursors into the ischemic cerebral cortex and striatum. differentiate into mature neurons in the post-stroke adult striatum. J. Neurosci. 26,
Mol. Cell. Neurosci. 24, 171–189. 6627–6636.
Kempermann, G., Jessberger, S., Steiner, B., Kronenberg, G., 2004. Milestones of Yang, Z., You, Y., Levison, S.W., 2008. Neonatal hypoxic/ischemic brain injury induces
neuronal development in the adult hippocampus. Trends Neurosci. 27, 447–452. production of calretinin-expressing interneurons in the striatum. J. Comp. Neurol.
Khodosevich, K., Seeburg, P.H., Monyer, H., 2009. Major signaling pathways in 511, 19–33.
migrating neuroblasts. Front. Mol. Neurosci. 2, 7. Zhang, R.L., Zhang, Z.G., Zhang, L., Chopp, M., 2001. Proliferation and differentiation of
Komitova, M., Mattsson, B., Johansson, B.B., Eriksson, P.S., 2005. Enriched environment progenitor cells in the cortex and the subventricular zone in the adult rat after focal
increases neural stem/progenitor cell proliferation and neurogenesis in the cerebral ischemia. Neuroscience 105, 33–41.
subventricular zone of stroke-lesioned adult rats. Stroke 36, 1278–1282. Zhang, R., Zhang, Z., Wang, L., Wang, Y., Gousev, A., Zhang, L., Ho, K.L., Morshead, C.,
Leker, R.R., Soldner, F., Velasco, I., Gavin, D.K., Androutsellis-Theotokis, A., McKay, R.D., Chopp, M., 2004a. Activated neural stem cells contribute to stroke-induced
2007. Long-lasting regeneration after ischemia in the cerebral cortex. Stroke 38, neurogenesis and neuroblast migration toward the infarct boundary in adult rats.
153–161. J. Cereb. Blood Flow Metab. 24, 441–448.
Liu, F., You, Y., Li, X., Ma, T., Nie, Y., Wei, B., Li, T., Lin, H., Yang, Z., 2009. Brain injury does Zhang, R., Zhang, Z., Zhang, C., Zhang, L., Robin, A., Wang, Y., Lu, M., Chopp, M., 2004b.
not alter the intrinsic differentiation potential of adult neuroblasts. J. Neurosci. 29, Stroke transiently increases subventricular zone cell division from asymmetric to
5075–5087. symmetric and increases neuronal differentiation in the adult rat. J. Neurosci. 24,
Lledo, P.M., Alonso, M., Grubb, M.S., 2006. Adult neurogenesis and functional plasticity 5810–5815.
in neuronal circuits. Nat. Rev. Neurosci. 7, 179–193. Zhang, W., Potrovita, I., Tarabin, V., Herrmann, O., Beer, V., Weih, F., Schneider, A.,
Long, M.A., Rossi, F.M., 2009. Silencing inhibits Cre-mediated recombination of the Z/AP Schwaninger, M., 2005. Neuronal activation of NF-kappaB contributes to cell death
and Z/EG reporters in adult cells. PLoS ONE 4, e5435. in cerebral ischemia. J. Cereb. Blood Flow Metab. 25, 30–40.
Mullen, R.J., Buck, C.R., Smith, A.M., 1992. NeuN, a neuronal specific nuclear protein in Zhang, R., Xue, Y.Y., Lu, S.D., Wang, Y., Zhang, L.M., Huang, Y.L., Signore, A.P., Chen, J.,
vertebrates. Development 116, 201–211. Sun, F.Y., 2006. Bcl-2 enhances neurogenesis and inhibits apoptosis of newborn
Parent, J.M., Vexler, Z.S., Gong, C., Derugin, N., Ferriero, D.M., 2002. Rat forebrain neurons in adult rat brain following a transient middle cerebral artery occlusion.
neurogenesis and striatal neuron replacement after focal stroke. Ann. Neurol. 52, Neurobiol. Dis. 24, 345–356.
802–813. Ziv, Y., Finkelstein, A., Geffen, Y., Kipnis, J., Smirnov, I., Shpilman, S., Vertkin, I., Kimron,
Thored, P., Wood, J., Arvidsson, A., Cammenga, J., Kokaia, Z., Lindvall, O., 2007. Long- M., Lange, A., Hecht, T., Reyman, K.G., Marder, J.B., Schwartz, M., Yoles, E., 2007. A
term neuroblast migration along blood vessels in an area with transient novel immune-based therapy for stroke induces neuroprotection and supports
angiogenesis and increased vascularization after stroke. Stroke 38, 3032–3039. neurogenesis. Stroke 38, 774–782.

You might also like