Ahlenius 2012 JN
Ahlenius 2012 JN
Ahlenius 2012 JN
Development/Plasticity/Repair
Stem Cell Laboratory, 5and Division of Neuropathology, Lund Stem Cell Center, Lund University Hospital, SE-221 84 Lund, Sweden
Ischemic stroke causes transient increase of neural stem and progenitor cell (NSPC) proliferation in the subventricular zone (SVZ), and
migration of newly formed neuroblasts toward the damaged area where they mature to striatal neurons. The molecular mechanisms
regulating this plastic response, probably involved in structural reorganization and functional recovery, are poorly understood. The
adaptor protein LNK suppresses hematopoietic stem cell self-renewal, but its presence and role in the brain are poorly understood. Here
we demonstrate that LNK is expressed in NSPCs in the adult mouse and human SVZ. Lnk ⫺/⫺ mice exhibited increased NSPC proliferation
after stroke, but not in intact brain or following status epilepticus. Deletion of Lnk caused increased NSPC proliferation while overex-
pression decreased mitotic activity of these cells in vitro. We found that Lnk expression after stroke increased in SVZ through the
transcription factors STAT1/3. LNK attenuated insulin-like growth factor 1 signaling by inhibition of AKT phosphorylation, resulting in
reduced NSPC proliferation. Our findings identify LNK as a stroke-specific, endogenous negative regulator of NSPC proliferation, and
suggest that LNK signaling is a novel mechanism influencing plastic responses in postischemic brain.
Figure 1. LNK is expressed in SVZ NSPCs in vivo and in vitro. A, RT-PCR and Q-PCR analysis of Lnk expression in tissue and neurospheres derived from mouse SVZ, and in FAC-sorted neuroblasts
(Dcx-GFP), NSPCs (Sox2-GFP), and microglia (Iba1-GFP) from SVZ and hippocampus (HPC) of reporter mice. B, Confocal photomicrographs showing LNK immunoreactivity (Figure legend continues.)
Ahlenius et al. • LNK, Stroke and Neurogenesis J. Neurosci., April 11, 2012 • 32(15):5151–5164 • 5153
with migration and maturation of newly formed neurons and parallel to the injection site, after applying conductive gel (Parker Labo-
ultimately their integration in established networks. We have ratories). A square-wave electroporator CUY21 EDIT (NepaGene) was
previously identified TNF-␣ signaling through TNF-R1 as a neg- used to deliver three 50 ms pulses of 200 V with an interval of 950 ms
ative regulator of insult-induced NSPC proliferation (Iosif et al., (Barnabé-Heider et al., 2008). After 72 h, mice were injected with bro-
modeoxyuridine (BrdU) four times with 2 h intervals, and 2 h after last
2006, 2008), establishing TNF-␣ as an important factor for the
injection mice were perfused.
neurogenic response to injury. Given the potent action of Lnk in
Fluorescence-activated cell sorting. Neurospheres were passaged and
inhibiting cytokine-induced proliferation in the hematopoietic SVZ tissue dissociated as described above. Cells were washed and resus-
system, and that several known LNK targets including EPO and pended in PBS containing 30 mM glucose and 5% FBS. Suspensions were
C-KIT have been shown to regulate NSPCs, we hypothesized that passed through a 70 m filter and cells were sorted using a fluorescence-
LNK might be a negative regulator also of NSPCs. activated cell sorting (FACS) Aria (BD Biosciences). The sorting gate was
Our findings here for the first time identify LNK as a novel set around the main GFP⫹ population, at least 1 log higher than GFP⫺
endogenous negative regulator of NSPC proliferation, which controls. For BrdU incorporation and cell cycle analysis WT and Lnk ⫺/⫺
could have an important role for brain plasticity during recovery neurospheres were pulsed with BrdU and cells harvested at 3, 6, and 10 h
after stroke. and stained for BrdU and labeled with propidium iodide. Data were
acquired on an LSR II flow cytometer, with Diva software version 6.0
Materials and Methods (Becton Dickinson). Data analysis was done on FlowJo software ver-
Animals. Experimental procedures were approved by the Malmö-Lund sion 9.4.9 (TreeStar).
Ethical Committee. Generation of Lnk ⫺/⫺ mice has been described Immunocytochemistry in vitro. Neurospheres or cells attached to slides
(Takaki et al., 2000). Sox2-GFP mice were provided by Dr. L. Pevny, were fixed with 4% paraformaldehyde (PFA), preincubated in potassium
Dcx-GFP mice from Drs L. Aigner and S. Coulliard-Despres, and Iba1- PBS (KPBS) with 0.025% Triton X-100 and 5% serum for 1 h, and
GFP mice from Dr. Kohsaka. Mice were backcrossed into C57BL6 incubated with primary antibodies overnight. Primary antibodies in-
(Charles River), which were used as wild-type (WT) controls. A total of cluded rabbit anti-p-H3 (1:400; Millipore), rabbit anti-SOX2 (1:500;
10 Sox2-GFP, 12 Iba1-GFP, 10 Dcx-GFP mice of either sex, and 55 WT Millipore), and goat anti-LNK (1:100; Santa Cruz Biotechnology). After
C57BL6 and 35 Lnk ⫺/⫺ male mice were used for this study. washing, slides were incubated with goat anti-rabbit Alexa Fluor 488 or
Human tissue. Ganglionic eminence (GE) was subdissected from donkey anti rabbit Cy3 for p-H3 staining. For LNK/SOX2 double-
brains of dead aborted 6- to 9-week-old human fetuses. Tissue was ob- staining, biotinylated horse anti-goat together with donkey anti-rabbit
tained from Lund University hospital after informed consent in accor- Cy3 secondary antibodies followed by streptavidin-Alexa Fluor 488 was
dance with guidelines set by the Lund/Malmö Ethical Committee. used.
Anonymized SVZ brain samples were obtained, according to national Proliferation and survival assay. Neurospheres or sorted cells were
regulations, from autopsies of adult humans in which neuropathological plated on PDL/laminin-coated coverslips in proliferative medium. For
investigations was performed as part of clinical diagnostic routine. assessing mitosis, neurospheres or cells were stained for p-H3. The per-
Dissection and cell culture. Neurospheres were generated from SVZ of centage of p-H3⫹ cells of the total number of Hoechst⫹ cells was quan-
intact 8- to 12-week-old mice as described previously (Iosif et al., 2008). tified. Apoptosis was quantified with TUNEL staining (Roche) with
For neurosphere formation, cells were grown at clonal density (10 cells/ Hoechst counterstaining. The percentage of TUNEL⫹ cells with apopto-
l). Differentiation of neurospheres was performed by plating on poly- tic Hoechst morphology of the total Hoechst⫹ cells was estimated.
D-lysine (PDL)/laminin coated coverslips and culturing in medium
Induction of stroke. The MCAO filament model was used (Iosif et al.,
without growth factors but with N2 and 1% fetal bovine serum (FBS). 2008). Briefly, the left external carotid was ligated and temporary sutures
Human fetal GE was mechanically dissociated and cultured in DMEM/ were placed around the common and internal carotid arteries. An 8 – 0
F12 with N2, 2 g/ml heparin, 10 ng/ml leukemia inhibitory factor monofilament (Alcon) coated with silicone was advanced through the
(Sigma), and 20 ng/ml epidermal growth factor (EGF) and basic fibro- internal carotid artery until it blocked the blood flow in the MCA. For
blast growth factor (bFGF). reperfusion, mice were re-anesthetized after 40 min of occlusion and the
Retroviral infection, transient transfection, and in vivo electroporation. filament was removed. We observed no differences between Lnk ⫺/⫺ and
Expanded neurospheres from WT mice were infected with retroviruses WT mice in initial neurological deficits, or in body weight and motor
expressing either MSCV-IRES-GFP or Lnk (MIG-Lnk) (Tong and Lo- behavior at 1 week after MCAO (Iosif et al., 2008).
dish, 2004) (provided by Drs. J. Lodish and W. Tong,) using a multiplic- Induction of SE. Mice were anesthetized and implanted with a stimu-
ity of infection of 2. Cells were allowed to grow for an additional 5 days in lating/recording electrode (Plastics One) unilaterally into ventral hip-
vitro before cell sorting. Constitutively active (CA)- or dominant- pocampal CA1–CA3 region. Ten days later, mice were subjected to
negative (DN)-STAT1 plasmids (provided by Drs. T. Ouchi and J. Dar- electrically induced SE (Iosif et al., 2006). Animals received 1 h of su-
nell, through Addgene, respectively) were transiently expressed in prathreshold stimulation consisting of 10 s trains of 1 ms biphasic square
neurospheres using lipofectamine (Invitrogen) and 48 h later expression wave pulses at a frequency of 50 Hz. Stimulation was interrupted for 1
of Lnk was assessed by Q-PCR. For in vivo transfection of SVZ cells, 6 g min every 10 min to allow for electroencephalographic (EEG) recording
of CA- or DN-AKT (provided by Dr. M. Hung, through Addgene) plas- and measurement of afterdischarges (MacLab). After ending the stimu-
mids were injected into the lateral ventricle of 8- to 12-week-old WT lation, all mice exhibited self-sustained, continuous ictal EEG activity,
mice. Immediately after injection, mice were electroporated using 5 mm and associated motor behavioral convulsions (Iosif et al., 2006). Epileptic
disk electrodes, which were placed laterally on the skin covering the skull activity was arrested with pentobarbital at 2 h after stimulation offset.
BrdU administration. BrdU (50 mg/kg, i.p.), dissolved in PBS, was
4 given four times with a 2 h interval to intact mice and at 7 d after SE, and
animals were killed 2 h thereafter. Mice subjected to stroke received BrdU
(Figure legend continued.) in mouse SVZ neurospheres (NS) costained for SOX2. Double- once daily for 7 d after the insult and were killed the following day. For
positive cell depicted by arrowhead on low-magnification images is shown also at higher mag- BrdU label retention studies, mice received two daily BrdU injections
nification. C, RT-PCR analysis of LNK expression in GE tissue and NS derived from human fetal GE during day 9 and 10 after stroke and were killed 8 weeks later.
(hGE). Photomicrographs showing LNK immunoreactivity in human NS (hNS) costained for Immunohistochemistry in vivo. Animals were transcardially perfused
SOX2. Arrowhead indicates example of double-labeled cell. D, RT-PCR analysis of LNK in three with ice-cold 4% PFA. Free-floating sections (30 m) were preincubated
different specimens of adult human SVZ tissue. E, Overview of adult human SVZ stained for LNK in 0.25% Triton X-100 in KPBS containing 5% donkey or goat serum for
(green) and GFAP (red). Boxed areas shown in higher magnification, top box (F–I) and lower 1 h. Sections were incubated with rat anti-BrdU (1:100; Sigma), rabbit
box (J–M) depict LNK⫹ (F, J), GFAP⫹ (G, K), LNK⫹/GFAP⫹ (H, I), and Hoechst⫹ (I, M) cells. anti-p-H3, rabbit anti-IBA1 (1:1000; Wako), goat anti-Dcx (1:400; Santa
Arrowheads indicate double-labeled cells. Scale bars: B, 50 m; (in C) C–D, 20 m; E, 15 m, Cruz Technology), rabbit anti-GFAP (1:400; Zymed), mouse anti-Nestin
and (in M) F–M, 5 m. (1:200; Millipore Bioscience Research Reagents), mouse anti-FoxJ1 (1:
5154 • J. Neurosci., April 11, 2012 • 32(15):5151–5164 Ahlenius et al. • LNK, Stroke and Neurogenesis
4
and Lnk ⫺/⫺ mice. Photomicrographs (D) and percentage of
p-H3⫹ (E) and apoptotic TUNEL⫹ (F) neurosphere cells out
of a total number of Hoechst⫹ cells. (G, H) FACS analysis of
BrdU incorporation and cell cycle in WT and Lnk ⫺/ ⫺ NSPCs.
Neurospheres were pulsed with BrdU and analyzed 3, 6, and
10 h thereafter. Histograms showing increased percentage of
BrdU⫹ cells in Lnk ⫺/⫺ (bottom) as compared with WT (top)
neurospheres (G). Bivariate (x-axis, propidium iodide; y-axis,
BrdU) cell cycle distribution analysis of WT (top) and Lnk ⫺/⫺
neurosphere cells. Density plots showing increased per-
centage of cells in S- and G2/M-phase in Lnk ⫺/⫺ (bottom)
as compared with WT (top) (H). Means ⫾S EM, n ⫽ 5 (A,
Figure 2. Deletion of Lnk leads to increased in vitro proliferation of SVZ NSPCs by shortening cell cycle duration. Number (A) and B) and 4 (E, F). *p ⬍ 0.05, Student’s unpaired t test. Scale
size (B) of primary and secondary neurospheres formed from WT and Lnk ⫺/⫺ SVZ. C, Photomicrographs of neurospheres from WT bars: C, 100 m; D, 10 m.
Ahlenius et al. • LNK, Stroke and Neurogenesis J. Neurosci., April 11, 2012 • 32(15):5151–5164 • 5155
Figure 4. Proliferation of NSPCs in SVZ is enhanced after stroke in Lnk ⫺/⫺ mice. Number and distribution of cells expressing p-H3 (A, C) and BrdU (B, D) in SVZ of intact WT and Lnk ⫺/⫺ mice
(A, B) or 7 d after stroke (C, D). Representative confocal images of BrdU⫹/Dcx⫹ (E) and BrdU⫹/Sox2⫹ (F) cells in the SVZ after stroke. Number of proliferating Dcx⫹ and Sox2⫹ cells after stroke
(G). Size and distribution of lesioned area after stroke (H). BrdU was injected 4 times 2 h apart in intact animals (A, B) or once daily following MCAO for 7 d (C–F). Ipsilateral denotes side of stroke.
Means ⫾ SEM, n ⫽ 4 and 5 for WT and Lnk ⫺/⫺ mice, respectively. *p ⬍ 0.05, t test (A, B, G, H) or two-way ANOVA with Scheffé’s post hoc test (C–D). Scale bar, 100 m.
Ahlenius et al. • LNK, Stroke and Neurogenesis J. Neurosci., April 11, 2012 • 32(15):5151–5164 • 5157
mice was also similar (Fig. 2 B, C). However, with prolonged time
in culture from second passage, cells from Lnk ⫺/⫺ mice exhibited
an increased percentage of cells in mitosis as detected by p-H3
staining. The fraction of p-H3⫹ cells in neurospheres from
Lnk ⫺/⫺ mice was 181% higher as compared with that from WT
animals (Fig. 2 D, E). The increased percentage of p-H3⫹ cells
was not due to improved survival since we did not find any dif-
ference in the percentage of apoptotic TUNEL⫹ cells between
WT and Lnk ⫺/⫺ neurospheres (Fig. 2 F). To exclude the possibil-
ity that the increased number of p-H3⫹ cells was due to cell cycle
arrest in G2/M phase, we pulsed WT and Lnk ⫺/⫺ NSPCs with
BrdU and analyzed BrdU incorporation and cell cycle status by
FACS at 3, 6, and 10 h. Both WT and Lnk ⫺/⫺ NSPCs increased the
percentage of BrdU⫹ cells over time. Strikingly, in Lnk ⫺/⫺
NSPCs the percentage of BrdU⫹ cells was higher at all time
points increasing to 21.6, 33.4, and 40.6%, respectively (Fig. 2G)
as compared with 14.4, 15.2, and 18.1% in WT. The percentage of
cells in S-phase increased in WT from 16.6% to 19.2 and 21.2%
and in Lnk ⫺/⫺ from 19.9, 30.1, and 35.3% at 3, 6, and 10 h,
respectively. The number of WT cells in G0/G1 was similar (74.9,
71.0, and 72.3%) at all time points, but cells in G2/M decreased
from 1.5 and 1.4% at 3 and 6 h, respectively, to 0.5% at 10 h. In
contrast the number of Lnk ⫺/⫺ cells in G0/G1 decreased from Figure 7. Deletion of Lnk does not alter number of microglia in SVZ. A, Number of Iba1⫹
76.1% to 65.1 and 58.5% at 3, 6, and 10 h, respectively, while the microglia in intact SVZ, and ipsilateral and contralateral to the ischemic lesion or stimulating
percentage of cells in G2/M remained unchanged (1.5, 1.3, and electrode for stroke and SE, respectively, in WT and Lnk ⫺/⫺ mice. Means ⫾ SEM, n ⫽ 8 and 10
(Intact), 9 and 7 (Stroke), and 7 and 9 (SE) for WT and Lnk ⫺/⫺, respectively. No differences
1.2%) (Fig. 2 H).Together, our data indicate that Lnk ⫺/⫺ cells
between WT and Lnk ⫺/⫺ mice, p ⬎ 0.05 Student’s unpaired t test or two-way ANOVA with
proliferate faster by shortening the G0/G1 phase. This conclusion Scheffé’s post hoc test. B, Photomicrograph showing the distribution of IBA⫹ cells (IBA1 in red,
is supported by previous reports that decreasing G1 length ex- Hoechst in blue) in SVZ of intact WT and Lnk ⫺/⫺ mice. Scale bar, 30 m. Str, striatum;. LV,
pands NSPC populations in the developing and adult brain lateral ventricle.
(Lange et al., 2009; Artegiani et al., 2011).
To provide further evidence for a suppressant role of LNK in
NSPC proliferation, we infected SVZ neurospheres from WT the peak of the increased proliferation, 7 d after stroke. BrdU was
mice with retroviruses to express either GFP alone or GFP to- injected once daily following MCAO. In contrast to the lack of
gether with Lnk. Five days thereafter, GFP⫹ cells were sorted and change in the intact brain, stroke increased the number of p-H3⫹
Lnk gene expression was assessed by Q-PCR, as well as neuro- cells by 60 and 63% (Fig. 4C) and BrdU⫹ cells by 32 and 37%
sphere formation, proliferation, and survival were analyzed. In (Fig. 4 D) in SVZ of Lnk ⫺/⫺ compared with WT mice, ipsilateral
the Lnk-infected cells, we detected a threefold increase in Lnk and contralateral to MCAO, respectively.
gene expression (Fig. 3A) and a dramatic, 96 and 61% decrease in We then analyzed whether NSPCs and neuroblasts, which
the number and size of neurospheres, respectively (Fig. 3B–D). In were both expressing LNK, contributed to stroke-enhanced cell
agreement, the number of cells in mitosis was 63% lower (Fig. proliferation in the SVZ of Lnk ⫺/⫺ mice. Sections were double-
3 E, F ). Lnk overexpression also influenced survival, the percent- stained for BrdU and the NSPC marker Sox2 or the neuroblast
age of TUNEL⫹ apoptotic cells being 107% higher as compared marker Dcx (Fig. 4 E, F ). Loss of Lnk increased poststroke prolif-
with that in control cells (Fig. 3G). Our findings after deletion eration of both NSPCs and neuroblasts in SVZ by 25 and 35%,
and overexpression of Lnk clearly show a role of this adaptor respectively, although in contrast to the Sox2⫹ NSPCs, the
protein as a negative regulator of in vitro proliferation of NPSCs. change in number of Dcx⫹ cells did not reach statistical signifi-
cance (Fig. 4G).
Stroke-induced proliferation of NSPCs is negatively regulated To explore the possibility that the higher cell proliferation in
by LNK SVZ caused by Lnk deletion was not confined to NSPCs but in-
Next we explored whether LNK also regulates NSPC proliferation cluded also other cell types, we analyzed the proliferation of
in the SVZ in vivo. The proliferative marker BrdU was injected FoxJ1⫹ ependymal, CD31⫹ endothelial, and Iba1⫹ microglial
four times 2 h apart in intact Lnk ⫺/⫺ mice and WT controls, and cells using BrdU injections at 1 week after MCAO. However, we
animals were killed 2 h thereafter. To reveal changes in cell cycle did not detect any colabeled cells in the SVZ (data not shown),
duration or cell division of NSPC in SVZ we analyzed BrdU in- providing evidence that at this time point after stroke, the vast
corporation in replicated DNA and p-H3 expression. No differ- majority of proliferating cells are NSPCs, and that Lnk deletion
ences in number of p-H3⫹ or BrdU⫹ cells were detected enhances the proliferation of this cell type.
between WT and Lnk ⫺/⫺ mice (Fig. 4 A, B), indicating that LNK Given the importance of the vascular niche in regulating
does not regulate proliferation in the healthy, intact SVZ in vivo. NSPCs, and the previously reported effect of LNK on endothelial
We have previously shown that stroke caused by MCAO cells and vasculature (Kwon et al., 2009; Kamei et al., 2010; Gold-
induces a transient increase of NSPC proliferation in SVZ man and Chen, 2011), we also assessed the overall staining pat-
(Arvidsson et al., 2002; Thored et al., 2006). To determine tern of CD31 in the SVZ and striatum. We measured the area
whether LNK influences NSPC proliferation in SVZ under path- covered by CD31 immunoreactivity in randomly selected areas of
ological conditions, we compared NSPC proliferation and early SVZ and striatum of WT and Lnk ⫺/ ⫺ mice at 1 week after stroke.
survival of newly formed cells in SVZ of WT and Lnk ⫺/⫺ mice at Arguing against an involvement of an altered vascular environ-
Ahlenius et al. • LNK, Stroke and Neurogenesis J. Neurosci., April 11, 2012 • 32(15):5151–5164 • 5159
alteration of IGF1-mediated Akt phosphorylation and gene ex- stroke in rats (Thored et al., 2009) and has been reported to be a
pression of stem cell regulators. However, this is most likely not mediator of the increased NSPC proliferation after stroke (Yan et
the only way that LNK may influence stroke-induced neurogen- al., 2006). The low levels of both Lnk and growth factors in SVZ of
esis. LNK is also known to affect endothelial cell proliferation and intact and SE animals probably explain the lack of change in
angiogenesis, to inhibit several cytokine signaling pathways in the NSPC proliferation under these conditions in the Lnk ⫺/⫺ mice.
hematopoietic system, and to exacerbate myeloproliferative dis- In contrast, we detected increased proliferation in the neuro-
ease. Furthermore, LNK is expressed in many cell types other sphere cultures from Lnk ⫺/⫺ mice. This in vitro environment is
than NSPCs. Therefore, it is conceivable that LNK acts not only more likely to mimic the SVZ niche after stroke with high levels of
on NSPC proliferation but also can influence stroke-induced growth factor signaling, which could explain why LNK has an
neurogenesis through other mechanisms. It is possible that LNK effect in vitro but not in vivo under basal conditions. LNK is
acts on a global level by fine-tuning the response to growth factor
known to inhibit RTK signaling by interacting either directly with
or cytokine stimulation.
the growth factor receptors or with downstream kinases, result-
We obtained no evidence for differences between WT and
ing in decreased phosphorylation (Takaki et al., 2000; Tong et al.,
Lnk ⫺/ ⫺ mice in endothelial cell proliferation or vascularization
in SVZ after stroke. However, due to the importance of the vas- 2005; Bersenev et al., 2008). We found that Lnk deletion in NSPCs
cular niche for NSPCs and the previously reported influence of gave rise to an increase in the phosphorylation of ERK1/2 in
LNK on endothelial cells, one cannot completely exclude that response to EGF receptor and FGF receptor stimulation and a
discrete changes in the vascular niche in Lnk ⫺/ ⫺ mice might more pronounced increase in the phosphorylation of AKT fol-
contribute to the NSPC proliferative effect observed here. lowing IGF1 receptor stimulation. We also showed that changes
The action of LNK as a negative regulator of NSPC prolifera- in AKT activation induced by transfection of CA-AKT or DN-
tion in the SVZ after stroke is most likely exerted in the NSPCs AKT plasmids into SVZ gave rise to increased and decreased
themselves and not indirectly through other cell types. First, we NSPC proliferation, respectively. Consistent with our data, LNK
found that the NSPCs express LNK. Second, although our data inhibits AKT and ERK1/2 signaling in hematopoietic stem/pro-
indicated that also microglia express Lnk, we obtained no in vivo genitor and endothelial cells, respectively (Tong et al., 2005; Wan
evidence that deletion of Lnk influenced the activation of SVZ et al., 2006; Seita et al., 2007). Thus, LNK can suppress NSPC
microglia. Third, our in vitro data clearly showed that altering the proliferation following stroke by inhibiting phosphorylation of
levels of Lnk in NSPCs, in the absence of immune cells and mi- kinases downstream of growth factor receptors, in particular
croglia, gave rise to the expected changes of cell proliferation. IGF1R. Hypothetically, the reduction of ERK1/2 and AKT signal-
We observed that the proliferation of NSPCs in SVZ of intact ing could lead to the decreased gene expression of Hes and Gli1,
and SE-subjected animals was not influenced by the deletion of which are involved in NSPC proliferation (Cayuso et al., 2006;
Lnk in the transgenic mice. In contrast, we detected increased Stecca and Ruiz i Altaba, 2009; Gregory et al., 2010). In support of
proliferation in neurosphere cultures from Lnk ⫺/ ⫺ mice as well this hypothesis, ERK and AKT activation positively regulates Gli
as in the NSPCs in SVZ of animals subjected to stroke. This expression in chicken neural explants and 3T3 cells (Riobó et al.,
discrepancy in the effect of LNK deficiency is most probably due 2006). The most prominent pathway discovered here is IGF1-
to the low levels of growth factors in SVZ of intact WT and
AKT signaling. However, given the broad spectrum of growth
SE-subjected animals and the high levels of growth factors in
factors and cytokine pathways known to be influenced by LNK, it
neurosphere culture medium and in the SVZ after stroke. Thus,
is possible that LNK also inhibits other pathways in NSPCs fol-
LNK is known to inhibit RTK signaling by interacting either di-
rectly with the growth factor receptors or with downstream ki- lowing stroke.
nases, resulting in decreased phosphorylation (Takaki et al., 2000; Here we have for the first time demonstrated the presence of
Tong et al., 2005; Bersenev et al., 2008). LNK in the adult brain and identified LNK signaling as a negative
The suppressant effect of LNK on NSPC proliferation was regulator of NSPC proliferation. This mechanism has so far been
specific for stroke and not observed in intact or SE animals and, shown to operate only after stroke, i.e., in response to a severe
accordingly, Lnk was upregulated after stroke but downregulated brain injury. Since stroke gives rise to NSPC proliferation in the
after SE. This finding is the first evidence that the expression of human brain (Martí-Fàbregas et al., 2010), and we found LNK to
Lnk is regulated in the adult brain. Our findings indicate that be expressed in human NSPCs, it is conceivable that LNK acts to
STAT1 and STAT3 induce the elevated Lnk expression in SVZ suppress the enhanced NSPC proliferation not only in rodents
NSPCs after stroke. We found that Stat1 was upregulated follow- but also in patients after stroke. Our findings raise the possibility
ing stroke but not SE in NSPCs, consistent with its previously that LNK action is a key player for cellular plasticity and a poten-
reported activation in neurons following ischemia (Takagi et al., tial novel therapeutic target in the postischemic brain.
2002). Furthermore, STAT3 has been shown to be activated in the
peri-infarct areas following ischemia, a process that was inhibited
by blocking IL-6 signaling (Yamashita et al., 2005). Stroke in- References
duces several cytokines including IL-6 and IFN ␣ and ␥ (Ya- Aerts S, Van Loo P, Thijs G, Mayer H, de Martin R, Moreau Y, De Moor B
mashita et al., 2005; Liesz et al., 2011), which are known to (2005) TOUCAN 2: the all-inclusive open source workbench for regula-
activate STAT family members (Takeda and Akira, 2000). Thus, tory sequence analysis. Nucleic Acids Res 33:W393–W396.
it is conceivable that the upregulation of LNK seen after stroke is Artegiani B, Lindemann D, Calegari F (2011) Overexpression of cdk4 and
cyclinD1 triggers greater expansion of neural stem cells in the adult mouse
mediated by STAT1/3 activation by cytokines released after the
brain. J Exp Med 208:937–948.
insult. Arvidsson A, Collin T, Kirik D, Kokaia Z, Lindvall O (2002) Neuronal re-
Stroke but not SE gave rise to increased expression of several placement from endogenous precursors in the adult brain after stroke.
growth factor ligands and/or receptors in SVZ which could trig- Nat Med 8:963–970.
ger NSPC proliferation, e.g., EPO, FGF, EGF, and IGF1. For ex- Barnabé-Heider F, Meletis K, Eriksson M, Bergmann O, Sabelström H, Har-
ample, Igf1 is upregulated in SVZ both short- and long-term after vey MA, Mikkers H, Frisen J (2008) Genetic manipulation of adult
Ahlenius et al. • LNK, Stroke and Neurogenesis J. Neurosci., April 11, 2012 • 32(15):5151–5164 • 5163
mouse neurogenic niches by in vivo electroporation. Nat Methods Martí-Fàbregas J, Romaguera-Ros M, Gómez-Pinedo U, Martínez-Ramírez
5:189 –196. S, Jiménez-Xarrié E, Marín R, Martí-Vilalta JL, García-Verdugo JM
Bengzon J, Kokaia Z, Elmér E, Nanobashvili A, Kokaia M, Lindvall O (1997) (2010) Proliferation in the human ipsilateral subventricular zone after
Apoptosis and proliferation of dentate gyrus neurons after single and ischemic stroke. Neurology 74:357–365.
intermittent limbic seizures. Proc Natl Acad Sci U S A 94:10432–10437. Mayor C, Brudno M, Schwartz JR, Poliakov A, Rubin EM, Frazer KA, Pachter
Bersenev A, Wu C, Balcerek J, Tong W (2008) Lnk controls mouse hemato- LS, Dubchak I (2000) VISTA: visualizing global DNA sequence align-
poietic stem cell self-renewal and quiescence through direct interactions ments of arbitrary length. Bioinformatics 16:1046 –1047.
with JAK2. J Clin Invest 118:2832–2844. Moskowitz MA, Lo EH, Iadecola C (2010) The science of stroke: mecha-
Bersenev A, Wu C, Balcerek J, Jing J, Kundu M, Blobel GA, Chikwava KR, nisms in search of treatments. Neuron 67:181–198.
Tong W (2010) Lnk constrains myeloproliferative diseases in mice. Nomura T, Göritz C, Catchpole T, Henkemeyer M, Frisén J (2010) EphB
J Clin Invest 120:2058 –2069. signaling controls lineage plasticity of adult neural stem cell niche cells.
Buza-Vidas N, Antonchuk J, Qian H, Månsson R, Luc S, Zandi S, Anderson K, Cell Stem Cell 7:730 –743.
Takaki S, Nygren JM, Jensen CT, Jacobsen SE (2006) Cytokines regulate Oh ST, Simonds EF, Jones C, Hale MB, Goltsev Y, Gibbs KD Jr, Merker JD,
postnatal hematopoietic stem cell expansion: opposing roles of thrombo- Zehnder JL, Nolan GP, Gotlib J (2010) Novel mutations in the inhibi-
poietin and LNK. Genes Dev 20:2018 –2023. tory adaptor protein LNK drive JAK-STAT signaling in patients with
Carlén M, Meletis K, Göritz C, Darsalia V, Evergren E, Tanigaki K, Amendola myeloproliferative neoplasms. Blood 116:988 –292.
M, Barnabé-Heider F, Yeung MS, Naldini L, Honjo T, Kokaia Z, Shuplia- Oishi K, Watatani K, Itoh Y, Okano H, Guillemot F, Nakajima K, Gotoh Y
kov O, Cassidy RM, Lindvall O, Frisén J (2009) Forebrain ependymal (2009) Selective induction of neocortical GABAergic neurons by the
cells are Notch-dependent and generate neuroblasts and astrocytes after PDK1-Akt pathway through activation of Mash1. Proc Natl Acad Sci
stroke. Nat Neurosci 12:259 –267. U S A 106:13064 –13069.
Cayuso J, Ulloa F, Cox B, Briscoe J, Martí E (2006) The Sonic hedgehog Parent JM, Yu TW, Leibowitz RT, Geschwind DH, Sloviter RS, Lowenstein
pathway independently controls the patterning, proliferation and sur- DH (1997) Dentate granule cell neurogenesis is increased by seizures
vival of neuroepithelial cells by regulating Gli activity. Development and contributes to aberrant network reorganization in the adult rat hip-
133:517–528. pocampus. J Neurosci 17:3727–3738.
Doetsch F, Caillé I, Lim DA, García-Verdugo JM, Alvarez-Buylla A (1999) Parent JM, Valentin VV, Lowenstein DH (2002) Prolonged seizures in-
Subventricular zone astrocytes are neural stem cells in the adult mamma- crease proliferating neuroblasts in the adult rat subventricular zone-
lian brain. Cell 97:703–716. olfactory bulb pathway. J Neurosci 22:3174 –3188.
Ekdahl CT, Kokaia Z, Lindvall O (2009) Brain inflammation and adult neu- Riobó NA, Lu K, Ai X, Haines GM, Emerson CP Jr (2006) Phosphoinositide
rogenesis: the dual role of microglia. Neuroscience 158:1021–1029. 3-kinase and Akt are essential for Sonic Hedgehog signaling. Proc Natl
Ferron SR, Andreu-Agullo C, Mira H, Sanchez P, Marques-Torrejon MA,
Acad Sci U S A 103:4505– 4510.
Farinas I (2007) A combined ex/in vivo assay to detect effects of exoge-
Rudd CE (2001) Lnk adaptor: novel negative regulator of B cell lymphopoi-
nously added factors in neural stem cells. Nat Protoc 2:849 – 859.
esis. Sci STKE 2001:pe1.
Goldman SA, Chen Z (2011) Perivascular instruction of cell genesis and fate
Seita J, Ema H, Ooehara J, Yamazaki S, Tadokoro Y, Yamasaki A, Eto K,
in the adult brain. Nat Neurosci 14:1382–1389.
Takaki S, Takatsu K, Nakauchi H (2007) Lnk negatively regulates self-
Gregory GD, Miccio A, Bersenev A, Wang Y, Hong W, Zhang Z, Poncz M,
renewal of hematopoietic stem cells by modifying thrombopoietin-
Tong W, Blobel GA (2010) FOG1 requires NuRD to promote hemato-
mediated signal transduction. Proc Natl Acad Sci U S A 104:2349 –2354.
poiesis and maintain lineage fidelity within the megakaryocytic-erythroid
Stecca B, Ruiz i Altaba A (2009) A GLI1–p53 inhibitory loop controls neural
compartment. Blood 115:2156 –2166.
stem cell and tumour cell numbers. EMBO J 28:663– 676.
Iosif RE, Ekdahl CT, Ahlenius H, Pronk CJ, Bonde S, Kokaia Z, Jacobsen SE,
Subramaniam S, Unsicker K (2006) Extracellular signal-regulated kinase as
Lindvall O (2006) Tumor necrosis factor receptor 1 is a negative regula-
an inducer of non-apoptotic neuronal death. Neuroscience
tor of progenitor proliferation in adult hippocampal neurogenesis. J Neu-
138:1055–1065.
rosci 26:9703–9712.
Iosif RE, Ahlenius H, Ekdahl CT, Darsalia V, Thored P, Jovinge S, Kokaia Z, Sun L, Lee J, Fine HA (2004) Neuronally expressed stem cell factor induces
Lindvall O (2008) Suppression of stroke-induced progenitor prolifera- neural stem cell migration to areas of brain injury. J Clin Invest
tion in adult subventricular zone by tumor necrosis factor receptor 1. 113:1364 –1374.
J Cereb Blood Flow Metab 28:1574 –1587. Takagi Y, Harada J, Chiarugi A, Moskowitz MA (2002) STAT1 is activated
Iseki M, Takaki S, Takatsu K (2000) Molecular cloning of the mouse APS as in neurons after ischemia and contributes to ischemic brain injury.
a member of the Lnk family adaptor proteins. Biochem Biophys Res Com- J Cereb Blood Flow Metab 22:1311–1318.
mun 272:45–54. Takaki S, Sauer K, Iritani BM, Chien S, Ebihara Y, Tsuji K, Takatsu K, Perl-
Jin K, Wang X, Xie L, Mao XO, Zhu W, Wang Y, Shen J, Mao Y, Banwait S, mutter RM (2000) Control of B cell production by the adaptor protein
Greenberg DA (2006) Evidence for stroke-induced neurogenesis in the lnk. Definition of a conserved family of signal-modulating proteins. Im-
human brain. Proc Natl Acad Sci U S A 103:13198 –13202. munity 13:599 – 609.
Kamei N, Kwon SM, Alev C, Ishikawa M, Yokoyama A, Nakanishi K, Yamada Takaki S, Tezuka Y, Sauer K, Kubo C, Kwon SM, Armstead E, Nakao K,
K, Horii M, Nishimura H, Takaki S, Kawamoto A, Ii M, Akimaru H, Katsuki M, Perlmutter RM, Takatsu K (2003) Impaired lymphopoiesis
Tanaka N, Nishikawa S, Ochi M, Asahara T (2010) Lnk deletion rein- and altered B cell subpopulations in mice overexpressing Lnk adaptor
forces the function of bone marrow progenitors in promoting neovascu- protein. J Immunol 170:703–710.
larization and astrogliosis following spinal cord injury. Stem Cells Takeda K, Akira S (2000) STAT family of transcription factors in
28:365–375. cytokine-mediated biological responses. Cytokine Growth Factor Rev
Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM, 11:199 –207.
Haussler D (2002) The human genome browser at UCSC. Genome Res Thored P, Arvidsson A, Cacci E, Ahlenius H, Kallur T, Darsalia V, Ekdahl CT,
12:996 –1006. Kokaia Z, Lindvall O (2006) Persistent production of neurons from
Kwon SM, Suzuki T, Kawamoto A, Ii M, Eguchi M, Akimaru H, Wada M, adult brain stem cells during recovery after stroke. Stem Cells 24:739 –747.
Matsumoto T, Masuda H, Nakagawa Y, Nishimura H, Kawai K, Takaki S, Thored P, Heldmann U, Gomes-Leal W, Gisler R, Darsalia V, Taneera J,
Asahara T (2009) Pivotal role of lnk adaptor protein in endothelial pro- Nygren JM, Jacobsen SE, Ekdahl CT, Kokaia Z, Lindvall O (2009) Long-
genitor cell biology for vascular regeneration. Circ Res 104:969 –977. term accumulation of microglia with proneurogenic phenotype concom-
Lange C, Huttner WB, Calegari F (2009) Cdk4/cyclinD1 overexpression in itant with persistent neurogenesis in adult subventricular zone after
neural stem cells shortens G1, delays neurogenesis, and promotes the stroke. Glia 57:835– 849.
generation and expansion of basal progenitors. Cell Stem Cell 5:320 –331. Tong W, Lodish HF (2004) Lnk inhibits Tpo-mpl signaling and Tpo-
Liesz A, Zhou W, Mracskó É, Karcher S, Bauer H, Schwarting S, Sun L, Bruder mediated megakaryocytopoiesis. J Exp Med 200:569 –580.
D, Stegemann S, Cerwenka A, Sommer C, Dalpke AH, Veltkamp R Tong W, Zhang J, Lodish HF (2005) Lnk inhibits erythropoiesis and Epo-
(2011) Inhibition of lymphocyte trafficking shields the brain against del- dependent JAK2 activation and downstream signaling pathways. Blood
eterious neuroinflammation after stroke. Brain 134:704 –720. 105:4604 – 4612.
5164 • J. Neurosci., April 11, 2012 • 32(15):5151–5164 Ahlenius et al. • LNK, Stroke and Neurogenesis
Tsai PT, Ohab JJ, Kertesz N, Groszer M, Matter C, Gao J, Liu X, Wu H, 6 signaling aggravates ischemic cerebral damage in mice: possible in-
Carmichael ST (2006) A critical role of erythropoietin receptor in neu- volvement of Stat3 activation in the protection of neurons.
rogenesis and post-stroke recovery. J Neurosci 26:1269 –1274. J Neurochem 94:459 – 468.
Velazquez L, Cheng AM, Fleming HE, Furlonger C, Vesely S, Bernstein A, Yan YP, Sailor KA, Vemuganti R, Dempsey RJ (2006) Insulin-like growth
Paige CJ, Pawson T (2002) Cytokine signaling and hematopoietic ho- factor-1 is an endogenous mediator of focal ischemia-induced neural
meostasis are disrupted in Lnk-deficient mice. J Exp Med 195:1599 –1611. progenitor proliferation. Eur J Neurosci 24:45–54.
Wan M, Li Y, Xue H, Li Q, Li J (2006) TNF-alpha induces Lnk expression Ye P, D’Ercole AJ (2006) Insulin-like growth factor actions during develop-
through PI3K-dependent signaling pathway in human umbilical vein en- ment of neural stem cells and progenitors in the central nervous system.
dothelial cells. J Surg Res 136:53–57. J Neurosci Res 83:1– 6.
Wang TC, Chiu H, Chang YJ, Hsu TY, Chiu IM, Chen L (2011) The adaptor Young KM, Fogarty M, Kessaris N, Richardson WD (2007) Subventricu-
protein SH2B3 (Lnk) negatively regulates neurite outgrowth of PC12 cells lar zone stem cells are heterogeneous with respect to their embryonic
and cortical neurons. PLoS One 6:e26433. origins and neurogenic fates in the adult olfactory bulb. J Neurosci
Welsh M, Songyang Z, Frantz JD, Trüb T, Reedquist KA, Karlsson T, Mi- 27:8286 – 8296.
yazaki M, Cantley LC, Band H, Shoelson SE (1998) Stimulation through Yousaf N, Deng Y, Kang Y, Riedel H (2001) Four PSM/SH2-B alternative
the T cell receptor leads to interactions between SHB and several signaling splice variants and their differential roles in mitogenesis. J Biol Chem
proteins. Oncogene 16:891–901. 276:40940 – 40948.
Yamashita T, Sawamoto K, Suzuki S, Suzuki N, Adachi K, Kawase T, Zhao C, Deng W, Gage FH (2008) Mechanisms and functional implications
Mihara M, Ohsugi Y, Abe K, Okano H (2005) Blockade of interleukin- of adult neurogenesis. Cell 132:645– 660.