You are on page 1of 9

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/273152215

Coupling of Neurogenesis and Angiogenesis


After Ischemic Stroke

Article in Brain Research February 2015


DOI: 10.1016/j.brainres.2015.02.042 Source: PubMed

CITATIONS READS

16 84

4 authors, including:

Linhui Ruan Brian Wang


University of North Texas HSC at Fort Worth University of North Texas HSC at Fort Worth
10 PUBLICATIONS 104 CITATIONS 15 PUBLICATIONS 148 CITATIONS

SEE PROFILE SEE PROFILE

Qichuan Zhuge
38 PUBLICATIONS 334 CITATIONS

SEE PROFILE

All content following this page was uploaded by Brian Wang on 04 December 2015.

The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document
and are linked to publications on ResearchGate, letting you access and read them immediately.
brain research 1623 (2015) 166173

Available online at www.sciencedirect.com

www.elsevier.com/locate/brainres

Research Report

Coupling of neurogenesis and angiogenesis


after ischemic stroke

Linhui Ruana,1, Brian Wangb,c,1, Qichuan ZhuGea, Kunlin Jina,b,c,n


a
Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Afliated Hospital, Wenzhou
Medical University, Wenzhou, China
b
Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie
Boulevard, Fort Worth, TX 76107, USA
c
Institute of Aging and Alzheimer's Disease Research, University of North Texas Health Science Center at Fort Worth, TX
76107, USA

ar t ic l e in f o abs tra ct

Article history: Stroke is a leading cause of mortality and severe long-term disability worldwide. Development
Accepted 20 February 2015 of effective treatment or new therapeutic strategies for ischemic stroke patients is therefore
Available online 28 February 2015 crucial. Ischemic stroke promotes neurogenesis by several growth factors including FGF-2,

Keywords: IGF-1, BDNF, VEGF and chemokines including SDF-1, MCP-1. Stroke-induced angiogenesis is

Stroke similarly regulated by many factors most notably, eNOS and CSE, VEGF/VEGFR2, and Ang-1/

Neurogenesis Tie2. Important ndings in the last decade have revealed that neurogenesis is not the stand-

Angiogenesis alone consideration in the ght for full functional recovery from stroke. Angiogenesis has been

Ischemia also shown to be critical in improving post-stroke neurological functional recovery. More than

Cell interaction that, recent evidence has shown a highly possible interplay or dependence between stroke-
induced neurogenesis and angiogenesis. Moving forward, elucidating the underlying mechan-
isms of this coupling between stroke-induced neurogenesis and angiogenesis will be of great
importance, which will provide the basis for neurorestorative therapy.
This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
& 2015 Elsevier B.V. All rights reserved.

1. Introduction it must be administered within 4.5 h after the onset of stroke for
rtPA to be effective. As a result, the short therapeutic time
Stroke is a leading cause of mortality and severe disability window and the potential complication from intracranial
worldwide. Currently, the number of patients suffering from hemorrhage benet only a minority of stroke patients (Zhang
stroke is steadily increasing. The only proven therapy for acute and Chopp, 2009). Even with successful rtPA thrombolysis, most
ischemic stroke approved by the FDA is systemic thrombolysis stroke survivors still suffer from permanent neurological func-
with recombinant tissue plasminogen activator (rtPA). However, tional decits. Therefore, extending the therapeutic time

n
Corresponding author at: Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500
Camp Bowie Boulevard, Fort Worth, TX 76107, USA. fax: 1 817 735 0480.
E-mail address: kunlin.jin@unthsc.edu (K. Jin).
1
These authors contributed equally to this work.

http://dx.doi.org/10.1016/j.brainres.2015.02.042
0006-8993/& 2015 Elsevier B.V. All rights reserved.
brain research 1623 (2015) 166173 167

window of rtPA or uncovering new therapeutic strategies for cells (Zhang et al., 2006). As reported by Zhang et al., the pro-
the treatment of ischemic stroke is highly sought after. portion of actively dividing SVZ NSCs is about 1521% in the
In the past decade, several independent research groups have adult rat brain. Stroke increases the proportion of proliferating
reported that neurogenesis continues into adulthood (Wang and SVZ cells to 24% just two days after stroke, and this proportion
Jin, 2014). Neurogenesis is the process of producing new func- reaches a maximum level of 31% 7 days after stroke (Zhang et al.,
tional neurons from neural stem/progenitor cells (NSCs), includ- 2006). The length of the SVZ NSC cell cycle is 1821 h in the
ing proliferation of endogenous NSCs, migration, and normal rat brain throughout its lifetime. However, stroke reduces
differentiation into mature functional neurons. It is well accepted the length of the cell cycle to 11 h at 2 days after stroke onset.
now that neurogenesis occurs at two distinct regions in the Switching from asymmetric to symmetric NSC division may
intact brain throughout life: the subventricular zone (SVZ) of the be another underlying mechanism, which contributes to the
lateral ventricles and subgranular zone (SGZ) in the dentate expansion of the NSC pool. Symmetric divisions generate two
gyrus of the hippocampus (Wang and Jin, 2014). In pathological identical daughter cells that go into maintaining the NSC pool
conditions such as ischemic stroke, enhanced neurogenesis has while asymmetric or self-renewing divisions generate one
been reported in animal models of stroke and even in stroke daughter cell and a differentiated cell such as a neuron or non-
patients (Jin et al., 2001; Jin et al., 2006; Thored et al., 2006), stem-cell progenitor (Chenn and McConnell, 1995; Gotz and
suggesting a potential avenue for the treatment of ischemic Huttner, 2005; Smart, 1973). It was shown that in the adult rat,
stroke. However, it is now apparent that neurogenesis is not the stroke briey increases the number of dividing SVZ NSCs with
stand-alone consideration in the ght for full functional recovery vertical cleavage orientation, and decreases the number of SVZ
from stroke. One should now also factor in the role of angiogen- NSCs with horizontal cleavage orientation (Zhang et al., 2004).
esis as it has been shown to be critical in improving post-stroke This suggests that the NSCs switch from asymmetric to sym-
neurological functional recovery. metric division to expand the NSC pool, whose numbers were
Angiogenesis, dened as new microvessel formation via shown to be signicantly increased after stroke. Interestingly, 4
branching off from pre-existing vessels (Carmeliet and Jain, days after stroke, the frequency of neuronal phenotype in
2011), is a multi-step biological process, including proliferation symmetrically divided cells increased to 47% from 33% in
and sprouting of endothelial cells, formation of tube-like vascular asymmetrically divided cells. Thus, it is possible to conclude that
structures, branching and anastomosis (Risau, 1997). Angiogen- stroke increases the neuronal phenotype though no evidence
esis is found in the penumbra of the brain infarct region in was shown to what degree this phenotype is preferred versus
animal models of stroke and even in the brains of stroke patients other glial cell fates.
(Hayashi et al., 2003; Krupinski et al., 1994; Zhang et al., 2002). It Several extracellular signals such as growth factors that
has been reported that neurogenesis and angiogenesis occur in regulate stroke-induced proliferation of NSCs have been
the brains of stroke patients and a positive correlation was seen identied and extensively examined (Fig. 1).
between patient survival and density of microvessels (Krupinski
et al., 1994). Several ndings in addition to this prove that 2.1.1. FGF-2
neurogenesis and angiogenesis are coupled processes after an The mRNA expression of broblast growth factor-2 (FGF-2) is
insult such as ischemic stroke, and should be acknowledged and upregulated signicantly after ischemic stroke injury in the adult
pursued as concurrent and non-mutually exclusive events to rat brain (Naylor et al., 2005) as well as in patients who died from
further develop neurorestorative therapy. acute ischemic stroke (Navaratna et al., 2009). It was also shown
This mini-review aims to establish the underlying mechan-
isms of how ischemic stroke induces endogenous neurogenesis
and angiogenesis, and then address the interplay between Penumbra
neurogenesis and angiogenesis after ischemic stroke. Angiogenesis

2. Mechanisms underlying stroke-induced OB SVZ eNOS/CSE


VEGF/VEGFR2
neurogenesis DG Ang-1/Tie2

RMS
In the following sections, we review cellular and molecular
mechanisms underlying stroke-induced neurogenesis and
how factors released by endothelial cells may participate in
the process. Neurogenesis

2.1. Proliferation Proliferation Migration

Endogenous NSC proliferation is the rst step in stroke-induced


neurogenesis. It was demonstrated that ischemic stroke is
FGF-2 SDF-1
sufcient to increase the endogenous NSC proliferation to result IGF-1 MCP-1
in the expansion of the NSC pool (Tang et al., 2009). Ischemic BDNF MMPs
VEGF
stroke injury promotes the proliferation of NSCs and expands the
NSC pool by regulating NSC cytokinetics such as shortening the Fig. 1 Key pathways potentially involved in the coupling of
length of the cell cycle to increase the percentage of proliferating neurogenesis and angiogenesis after ischemic stroke.
168 brain research 1623 (2015) 166173

that FGF-2 was localized to endothelial cells in these stroke (Li et al., 2014; Shin et al., 2004). The mechanism wherein
patients (Issa et al., 2005). To illustrate the impact of the loss of eNOS regulates BDNF secretion is still largely unclear.
endogenous FGF-2, Yoshimura et al. used mice genetically
decient in FGF-2 and showed that knocking out of FGF-2 2.1.4. VEGF
resulted in reduction of ischemia-induced progenitor prolifera- Vascular endothelial growth factor (VEGF) is an angiogenic
tion when compared with wild type mice (Yoshimura et al., protein that binds to its receptor on endothelial cells to
2001). However, after overexpression of FGF-2 by intraventricular induce angiogenesis. Evidence from recent studies suggests
(ICV) injection with a herpes simplex virus-1 amplicon vector that VEGF also act directly on neuronal progenitor cells to
carrying the FGF-2 gene, the number of proliferating NSCs in produce neurogenic effect. VEGF receptors, such as Flk-1 and
post-stroke mice increased signicantly when compared to the Flt-1 are expressed on neural progenitors in the adult SVZ
wild type mice (Yoshimura et al., 2001). Furthermore, transplan- and hippocampus (Maurer et al., 2003; Yang et al., 2003). A
tation of FGF-2 gene-modied MSCs to ischemic rats resulted in study by Jin et al. revealed that ICV infusion of VEGF into the
enhanced neurogenesis and increased functional recovery (Ikeda adult rat brain promotes proliferation of NSCs in the SGZ and
et al., 2005). Based on these ndings, endogenous production of the SVZ area (Jin et al., 2002). Further, Sun and colleagues
FGF-2 by endothelial cells plays an important role in regulating revealed that ICV administration of VEGF after stroke injury
post-stroke induced NSC proliferation. led to improved neurological performance and a signicant
reduction in infarct volume, suggesting that VEGF has an
important role in post-stroke neurogenesis as well as angio-
2.1.2. IGF-1
genesis (Sun et al., 2003).
Insulin-like growth factor-1 (IGF-1), primarily produced in the
liver, plays a major role in brain development. It was reported
2.2. Migration
that IGF-1 had a direct effect on proliferation of adult hippo-
campal NSCs in vitro, and the proliferative effect is relied upon
In order for proliferating NSCs to contribute to functional
the activation of the MAPK signaling pathway (Kalluri et al.,
recovery, it is necessary for these NSCs to migrate from their
2007). Like FGF-2, the expression of IGF-1 protein and its
birthplace to the ischemic region. In the normal adult brain, the
receptor signicantly increased after ischemic stroke insult
SVZ neuroblasts are destined to migrate to the olfactory bulb
(Yan et al., 2006). Inhibiting IGF-1 expression by ICV adminis-
through the rostral migratory stream. In the stroke brain, many
tration of the IGF-1 antibody led to the blockage of the stroke-
of these SVZ neuroblasts migrate from the neurogenic region of
induced proliferation of NSCs (Yan et al., 2006). In a more
the SVZ through the brain parenchyma into the boundary of the
recent study Zhu et al. (2009) demonstrated neurotrophic and
infarct region (Arvidsson et al., 2002; Jin et al., 2003; Thored et al.,
angiogenic properties of IGF-1 in vivo using a mouse model
2006). This redirected migration is associated with cellular
of permanent focal ischemia and overexpression of IGF-1 by
interactions between immature migrating neuroblasts, astrocy-
stereotaxic injection with an adeno-associated viral (AAV)
tic processes and blood vessels (Yamashita et al., 2006). How-
vector containing the IGF-1 gene. The results were two-fold.
ever, ischemic stroke also upregulates inhibitory molecules such
They reported signicant increases in vascular density at 8
as chondroitin sulfate proteoglycans (CSPGs), which block the
weeks post-stroke in the peri-infarct region, which corre-
migration of neuroblasts (Carmichael, 2005). On the other hand,
sponded with improved vascular perfusion locally, and in
stroke not only upregulates chemotactic factors for neuroblast
neurogenesis at 7 days post-stroke. The question to ask here
migration, but also produce peri-infarct scar and barrier mole-
is: could it be possible that improved vascular density, and
cules to impede neuroblast migration. The net neuroblast
hence, perfusion, leads to increased neurogenesis?
migration is thus dependent upon the balance between inhibi-
tory molecules and chemotactic factors.
2.1.3. BDNF The mechanism underlying this injury-induced redirected
Among the many neurotrophic factors, the brain-derived migration is unclear. Several receptor-ligand signaling path-
neurotrophic factor (BDNF) has been most extensively stu- ways and molecular factors involved in the stroke-induced
died for its role in adult neurogenesis. Intrahippocampal endogenous NSCs migration have been identied (Fig. 1).
administration of BDNF in adult rats results in increased These include stromal cell-derived factor-1 (SDF-1) and CXC
neurogenesis in the dentate gyrus and ICV infusion of BDNF chemokine receptor 4 (CXCR4), monocyte chemoattractant
promotes the production of new neurons in the adult olfac- protein-1 (MCP-1), and matrix metalloproteinases (MMPs).
tory bulb in the intact brain (Scharfman et al., 2005; Zigova
et al., 1998). As demonstrated by Kokaia's group, ischemic 2.2.1. SDF-1 and CXCR4
stroke insult induced the upregulation of BDNF and its Under physiological conditions, high levels of SDF-1 (CXCL12)
receptor expression (Kokaia et al., 1998). Continuous intras- secreted from ependymal cells perpetuates NSC quiescence
triatal delivery of BDNF via recombinant AAV gene transfer (Sawada et al., 2014). After ischemic stroke, SDF-1 that is released
before ischemia in adult rats resulted in enhanced neurogen- from reactive astrocytes and vascular cells increases the state of
esis and better neurological functional recovery (Andsberg activation of Type C and activated Type B NSCs (Kokovay et al.,
et al., 2002). A well-known study by Chen et al. found that 2010). SDF-1 is a member of the alpha chemokine family, which
endothelial nitric oxide synthase (eNOS) knock-out mice have has a crucial role in the mobilization and homing of hemato-
decreased BDNF expression after stroke, suggesting the poietic stem cells to the bone marrow with its receptor CXCR4
involvement of eNOS in the regulation of BDNF expression (Hattori et al., 2003). A role for SDF-1 and its receptor CXCR4 in
(Chen et al., 2005) mediated by endothelial cells and neurons the directional migration of neuroblasts has been reported as
brain research 1623 (2015) 166173 169

well. Robin et al. showed that CXCR4 is expressed on NSCs and new vessels is to increase collateral circulation as rst line
migrating neuroblasts after stroke. Inhibiting SDF-1 expression defense against ischemia. Mounting evidence indicate that
using an antibody against CXCR4 signicantly reduced stroke- angiogenesis play a crucial role in ischemic stroke brain repair
induced NSC migration (Robin et al., 2006). Ohab et al. also found and long-term functional recovery. In animal models of ischemic
that ICV administration of SDF-1 in ischemic mice promoted stroke, proliferation of endothelial cells at 1224 h after injury
neuroblast migration after stroke and contributed to behavioral was seen (Hayashi et al., 2003; Marti et al., 2000). Capillary
recovery (Ohab et al., 2006). Through a series of thorough sprouting and new vessels were reported to continue to grow
investigations, Kokovay et al. further reported that neuroblasts for at least 3 weeks in the ischemic boundary zones (Hayashi
homed to the vasculature, and were attracted by factors released et al., 2003; Zhang et al., 2002). In ischemic stroke patients,
by endothelial cells (Kokovay et al., 2010). Taken together, these Krupinski and group analyzed post-mortem brain tissues and
studies demonstrated that neuroblast migration toward the found that angiogenesis also occur in the boundary of the
infarct boundary is dependent upon an SDF-1/CXCR4 mechan- ischemic site. They also noted that angiogenic activity occurred
ism through binding with the neurovasculature. at 34 days after stroke (Krupinski et al., 1994). Thus, the
activated angiogenesis is benecial for the stroke-injured brain,
2.2.2. MCP-1 as high levels of new vessel formation following stroke is
Monocyte chemoattractant protein-1 (MCP-1), a chemokine of the correlated with better functional recovery and prolonged survival
CC family, was previously shown to interact with its receptor (Krupinski et al., 1994). The presence of angiogenic activity in the
CCR2, which is widely expressed on NSCs to increase NSC long run however, is still not examined. Since we know that
migration in vitro (Wang and Jin, 2014). It was also reported to angiogenesis is coupled to neurogenesis (Teng et al., 2008; Zhang
have a crucial role in guiding neuroblast migration after ischemic et al., 2014), and that neurogenesis is dependent on new vessels
stroke. Yan and colleagues demonstrated that ischemic stroke in for long-term survival (Font et al., 2010), it is possible to posit,
rats caused an increase of MCP-1 mRNA expression in activated albeit nave, that angiogenic activity decreases in the long run,
astrocytes and microglia, which lasted for at least 3 days after which makes it a possible causal factor for the high death rate of
reperfusion (Yan et al., 2007). This study also conrmed that neuroblasts (at least 80%) in the ischemic boundary zone.
migrating neuroblasts expressed its respective receptor, CCR2, in Several excellent reviews have been written regarding angio-
the adult rodent brain. Furthermore, after focal ischemia, knock- genesis under physiological conditions and after stroke; we urge
out mice lacking either MCP-1 or CCR2 showed signicantly the reader to refer to them accordingly (Beck and Plate, 2009;
decreased numbers of migrating neuroblasts from the ipsilateral Carmeliet and Jain, 2011; Ergul et al., 2012; Font et al., 2010; Xiong
SVZ to the injured striatum. So far, the mechanism for MCP-1/ et al., 2010). Here, we highlight a few well-studied factors that
CCR2-dependent migration of neuroblasts has yet to be eluci- regulate post-stroke angiogenesis, including eNOS and CSE, VEGF
dated though activation of the PI3 kinase pathway seems to be a and its receptor VEGFR2, and angiopoietin-1 (Ang-1) and its
likely candidate (Katakowski et al., 2003; Turner et al., 1998). receptor Tie2 (Fig. 1).

2.2.3. MMPs 3.1. eNOS and CSE


Matrix metalloproteinases (MMPs), a family of proteinases, are
known to have a role in extracellular matrix (ECM) degradation Upon proliferation and migration of endothelial cells, angiogen-
and cell migration. Recently, the role of MMP in guiding SVZ esis and vasodilation occurs, which are regulated by nitric oxide
neuroblasts migration has also been implicated. The expression (NO) through eNOS generation and more recently, by hydrogen
of MMPs-3 and -9 in neuroblasts has been observed. Inhibition sulde (H2S) through cystathionine--lyase (CSE), an enzyme
of function of these MMPs resulted in the reduction of post- required for H2S synthesis (Coletta et al., 2012). Coletta et al.
stroke neuroblast migration (Barkho et al., 2008). Using a showed that the cooperative action of these two gases are
coculture system consisting SVZ NSCs and brain endothelial required for angiogenesis and vasodilation to occur. By inhibiting
cells from the adult mouse to mimic the microenvironment eNOS or protein kinase G-I (PKG-I), the H2S-stimulated angiogenic
within the peri-infarct region, Wang et al. showed that the response and vasorelexation were inhibited. Contrariwise, CSE
secretion of MMPs-2 and -9 on the vasculature via the activa- silencing in bEnd3 immortalized mouse brain microvascular
tion of PI3K/Akt and ERK1/2 signaling pathways also contri- endothelial cells with shRNA abolished NO-stimulated accumu-
butes to the migration of neuroblasts (Wang et al., 2006). These lation of cGMP, angiogenesis, and acetylcholine-induced vasor-
ndings indicate that neuroblasts may digest their way elexation, suggesting the critical role of H2S in NO vascular
through the ECM as they migrate by secreting MMPs. More activity. In aiming to improve angiogenesis after stroke, one
work is needed to determine how cellcell interactions could might nd it helpful to consider nding ways to reinstate the
utilize MMPs to aid in the migration of neuroblasts to the homeostatic balance of H2S and NO in the vasculature.
ischemic lesion.
3.2. VEGF/VEGFR2 and Ang-1/Tie2

3. Mechanisms underlying stroke-induced VEGF/VEGFR2 and the Ang-1/Tie2 have been shown to reg-
angiogenesis ulate angiogenesis in the ischemic boundary zone.
VEGF and the mRNA for its receptor VEGFR2 were shown to
New vessels are generated in the brain through several ways, be upregulated after stroke, which exerted remarkable pro-
including angiogenesis, vasculogenesis, and collateral vessel angiogenic effects (Zhang et al., 2000). Zhang et al. found that
growth. The overarching purpose for the generation of these VEGF administration 48 h after stroke promoted the formation
170 brain research 1623 (2015) 166173

of newly formed vessels, increased vascular permeability, and with regards to nding a suitable angiogenic-targeted treatment
hence, increased cerebral microvascular plasma perfusion in for stroke. Yet, we are hopeful moving forward that researchers
the penumbra of the ischemic rat cortex (Zhang et al., 2000). will continue to uncover more subtleties in order to understand
Congruent with their ndings, they reported an improvement the mechanisms and effectively treat stroke.
in neurological function in ischemic rats after late VEGF
administration. Sun et al. also administered VEGF via ICV
and found that von Willebrand factor-immunoreactive endo- 4. Coupling between neurogenesis and
thelial cell numbers increased, suggesting the enhancement of angiogenesis after ischemic stroke
postischemic angiogenesis (Sun et al., 2003).
On the other hand, Lin et al. noted that after the induction Recent evidence has shed more light on the role of the brain
of stroke, the temporal proles of Ang-1/Tie2 were vastly vasculature in neurogenesis (Lacar et al., 2012; Shen et al., 2008;
different compared to other angiogenic genes like VEGF/ Tavazoie et al., 2008). Capillaries in the SVZ are shown to be
VEGFR (Lin et al., 2000), in that Ang-1/Tie2 exert their actions permeable to diffusible molecules released from endothelial
during the late stages of vascular development vascular cells such as VEGF (Jin et al., 2002), FGF-2 (Biro et al., 1994),
remodeling and maturation. Tie2 is found predominantly in amongst many others. This is possible because pericytes and
endothelial cells, and is critical for vascular formation and astrocytic end feet do not tightly envelope the capillaries in the
maintenance. Beck and colleagues showed that after ische- region of the SVZ, thereby creating a special incomplete BBB
mia, the increase of Ang-1/Tie2 expression resulted in the (Tavazoie et al., 2008). Adult NSCs were also shown to express
maturation of newly formed vessels to stabilize functional the laminin receptor 61 integrin (VLA6), which regulates NSC
brain vessels (Beck et al., 2000). In a follow up study, Lin et al. binding to endothelial cells, and its expression shown to
noted an upregulation on both the mRNA and protein levels decrease as the NSCs differentiate (Shen et al., 2008). Taken
of Tie2 receptors in the new vessels in the ischemic cortex together, this suggests that regulation of neurogenesis is
just a few hours after stroke. They saw another peak at 3 dependent upon the brain vasculature. To date, there is a fair
days, which persisted for a week post-stroke (Lin et al., 2001). amount of literature regarding vasculature-dependent adult
Further, Lin and group showed that the Ang-1 mRNA was neurogenesis, but we expect even more studies to be conducted
transiently expressed just after stroke, but reported a large since this is a relatively new concept introduced just a little
increase 12 weeks post-stroke (Lin et al., 2000). over a decade ago by Louissant and colleagues' pivotal work in
Interestingly, some groups have also reported that Ang-1/ adult canaries (Louissaint et al., 2002).
Tie2 and endogenous stimulation or exogenous administra- The processes of neurogenesis and angiogenesis after stroke
tion of VEGF can act in combination to enhance angiogenesis are linked together and coordinated. For instance, neuroblasts
and vascular integrity (Lin et al., 2001; Zacharek et al., 2007; migrate from the SVZ region to the infarct boundary where post-
Zhang and Chopp, 2002). Lin et al. found that VEGF shared a stroke angiogenesis occurs, and these neuroblasts migrate clo-
similar expression pattern with Ang-2, another protein of the sely with cerebral vessels (Ohab et al., 2006; Thored et al., 2007).
angiopoietin family that can interact with Tie2; it was co- Many migrating neuroblasts are found to localize specically to
localized with Tie2 in the ischemic cortex where active vessel blood vessels in areas of active vascular sprouting and remodel-
remodeling occurred (Lin et al., 2001). Zhu and colleagues ing in the infarct boundary. Blocking normal and post-ischemic
further showed that Ang-2 and VEGF worked in concert to angiogenesis with 1 week of endostatin treatment showed
enhance angiogenesis after stroke, but this came at a cost: signicant decreases in the number of newborn endothelial cells
increased MMP-9 activity and inhibition of zonular occludens- and overall vascular density in the peri-infarct cortex compared
1 (ZO-1) expression resulting in BBB disruption (Zhu et al., to control (Ohab et al., 2006). This resulted in a 10-fold reduction
2005). In a follow up study, Zhu and colleagues found that in neuroblasts in the peri-infarct cortex 7 days after stroke
overexpressing VEGF and Ang-1 in adult male CD-1 mice compared to control, suggesting that angiogenesis and neuro-
increased vascular density, but maintained ZO-1 protein genesis are causally linked within the post-stroke neuro-
expression after stroke, suggesting differential effects of Ang- vascular niche (Ohab et al., 2006). A further study by Thored
1 and Ang-2 with VEGF, in that Ang-1 maintains BBB integrity and colleagues revealed that even after 16 weeks from the stroke
unlike Ang-2 (Zhu et al., 2006). In fact, enhanced angiogenesis insult, there were occurrences of low-grade angiogenesis and
were reported by combining VEGF and Ang-2 compared to increased vessel density in the adjacent injured striatum,
VEGF alone (Zhu et al., 2005) or administering VEGF and Ang-1 whereby neuroblasts preferentially migrated toward (Thored
at submaximal doses compared to maximal doses of either et al., 2007), indicating that angiogenesis as well as the vascu-
alone (Chae et al., 2000). Another study by Zacharek et al. lature is important for migration during long-term neurogenesis.
investigated VEGF and Ang-1/Tie2 effects in bone marrow Using a co-culture system, Teng et al. endeavored to show that
stromal cell (MSC) treatment of stroke (Zacharek et al., 2007). angiogenesis and neurogenesis are coupled processes (Teng
They found that MSC treatment increased VEGF and Ang-1/ et al., 2008). To do this, they rst co-cultured normal SVZ cells
Tie2 expression in the ischemic boundary zone. In addition, and rat brain endothelial cells (RBECs) taken from stroke rats to
knockdown of Tie2 expression in brain endothelial cells determine the effect of stroke-induced RBECs on SVZ cell
reduced MSC-induced capillary tube formation signicantly, proliferation and differentiation. Next, they determined the effect
suggesting Tie2's critical role in MSC-induced angiogenesis. of soluble proteins released by stroke-induced NSCs on RBECs via
Although stroke-induced angiogenesis has been extensively incubation with the supernatant collected from a culture of
studied, the array of proteins involved and the supposed inter- normal and stroke-induced SVZ cells. The results demonstrated
actions with one another signicantly increases the complexity that stroke-induced RBECs increased NSC proliferation by 28%.
brain research 1623 (2015) 166173 171

Compared with normal RBECs, stroke-induced RBECs also Stroke-induced angiogenesis is regulated by many factors
expanded the neuronal population by 46% via reducing NSC most notably eNOS and CSE, VEGF/VEGFR2, and Ang-1/Tie2.
differentiation into astrocytes through the upregulation and Furthermore, stroke-induced angiogenesis has been shown to
downregulation of Hes6 and Sox2, respectively. Furthermore, couple with and enhance post-stroke neurogenesis. Moving
stroke-induced NSCs were reported to secrete high levels of forward, elucidating the underlying mechanisms of the cou-
VEGF, which enhanced capillary tube formation, but with the pling between stroke-induced neurogenesis and angiogenesis
introduction of a VEGFR2 antagonist, angiogenesis was comple- will be of great importance, which will provide the basis for
tely abolished. Taken together, these results suggest the inter- neurorestorative therapy.
dependence of neurogenesis and angiogenesis after stroke, and
that VEGF mediates this coupling, though the presence of other
angiogenic factors that can be released by stroke-induced SVZ
cells cannot be ruled out. Fairly recently, a study by Zhang et al. Conict of interest
demonstrated using a whole-mount preparation of the lateral
wall of the lateral ventricle that endothelial cell proliferation in The authors have no conict of interests to declare.
the SVZ increased from 2% (in non-stroke mice) to 12% at 7 days
and 14% at 14 days after stroke, and that newly-produced
neuroblasts seen along the lateral wall of the lateral ventricle
were intimately associated with the newly formed vessels Acknowledgments
(Zhang et al., 2014). Vascular volume also steadily increased
from 2.6% of the total volume before the insult to 4.2, 4.9, and This work is supported by the National Natural Science Founda-
5.7% at 14, 30, and 90 days after insult, respectively, of which the tion of China (81371396) to QZ, National Natural Science Founda-
majority of newly formed vessels were capillaries. All in all, this tion of China (81400954) to LR, and by the National Institute of
study revealed long-term alterations in the NSC and vascular Health (NIH) Grants AG21980 and NS057186 to KJ.
architecture of the adult SVZ.
The underlying mechanisms of how stroke-induced neuro- r e f e r e nc e s
genesis and angiogenesis are coupled together are yet to be
elucidated (Fig. 1). The endothelial cells activated by ischemic
stroke in the ischemic boundary zone secrete several factors to Andsberg, G., et al., 2002. Neuropathological and behavioral
regulate the biological activities of NSCs, especially the migra- consequences of adeno-associated viral vector-mediated
continuous intrastriatal neurotrophin delivery in a focal
tion of neuroblasts. These endothelial cells produce SDF1 and
ischemia model in rats. Neurobiol. Dis. 9, 187204.
MMPs. SDF1 is a CXC chemokine, which guide neuroblast Arvidsson, A., et al., 2002. Neuronal replacement from
migration towards the peri-infarct region by binding to its endogenous precursors in the adult brain after stroke. Nat.
receptor CXCR4 found on neuroblasts (Robin et al., 2006). Med. 8, 963970.
MMP, which digest the extracellular matrix and enable cells to Barkho, B.Z., et al., 2008. Endogenous matrix metalloproteinase
penetrate, has been implicated in guiding neuroblast migration (MMP)-3 and MMP-9 promote the differentiation and
migration of adult neural progenitor cells in response to
from the neurogenic region to the ischemic boundary (Lee et al.,
chemokines. Stem Cells. 26, 31393149.
2006). In addition to secreting chemokines to guide neuroblast
Beck, H., et al., 2000. Expression of angiopoietin-1, angiopoietin-2,
migration, activated endothelial cells in the ischemic boundary and tie receptors after middle cerebral artery occlusion in the
secrete VEGF to promote neurogenesis. VEGF is not only an rat. Am. J. Pathol. 157, 14731483.
angiogenic growth factor, but also a strong neurogenic growth Beck, H., Plate, K.H., 2009. Angiogenesis after cerebral ischemia.
factor. VEGF has been shown to promote the proliferation of Acta Neuropathol. 117, 481496.
NSCs both in vitro and in the adult rat brain (Jin et al., 2002). Biro, S., et al., 1994. Expression and subcellular distribution of
basic fibroblast growth factor are regulated during migration
NSCs in the SVZ can also promote angiogenesis by secreting
of endothelial cells. Circ. Res. 74, 485494.
several angiogenic factors such as VEGFR2, Ang-2, and FGF (Liu
Carmeliet, P., Jain, R.K., 2011. Molecular mechanisms and clinical
et al., 2007). Taken together, these data suggest that neurogen- applications of angiogenesis. Nature 473, 298307.
esis and angiogenesis are highly coordinated and coupled to Carmichael, S.T., 2005. Rodent models of focal stroke: size,
enhance brain repair after ischemic stroke injury. mechanism, and purpose. NeuroRx: J. Am. Soc. Exp.
NeuroTher. 2, 396409.
Chae, J.K., et al., 2000. Coadministration of angiopoietin-1 and
vascular endothelial growth factor enhances collateral
5. Conclusion
vascularization. Arterioscler. Thromb. Vasc. Biol. 20,
25732578.
Ischemic stroke induces coordinated endogenous neurogen- Chen, J., et al., 2005. Endothelial nitric oxide synthase regulates
esis and angiogenesis, which contributes to brain repair. The brain-derived neurotrophic factor expression and
potential mechanisms that trigger augmented endogenous neurogenesis after stroke in mice. J. Neurosci. 25, 23662375.
neurogenesis and angiogenesis after stroke are becoming Chenn, A., McConnell, S.K., 1995. Cleavage orientation and the
asymmetric inheritance of Notch1 immunoreactivity in
increasingly known. Ischemic stroke promotes the prolifera-
mammalian neurogenesis. Cell 82, 631641.
tion of NSCs by a variety of growth factors including FGF-2,
Coletta, C., et al., 2012. Hydrogen sulfide and nitric oxide are
IGF-1, BDNF, and VEGF. After NSC proliferation, neuroblasts mutually dependent in the regulation of angiogenesis and
migrate from the SVZ neurogenic niche to the injured brain endothelium-dependent vasorelaxation. Proc. Natl. Acad. Sci.
region through the production of SDF-1, MCP-1, and MMPs. USA 109, 91619166.
172 brain research 1623 (2015) 166173

Ergul, A., Alhusban, A., Fagan, S.C., 2012. Angiogenesis: a Louissaint Jr., A., et al., 2002. Coordinated interaction of
harmonized target for recovery after stroke. Stroke 43, neurogenesis and angiogenesis in the adult songbird brain.
22702274. Neuron 34, 945960.
Font, M.A., Arboix, A., Krupinski, J., 2010. Angiogenesis, Marti, H.J., et al., 2000. Hypoxia-induced vascular endothelial
neurogenesis and neuroplasticity in ischemic stroke. Curr. growth factor expression precedes neovascularization after
Cardiol. Rev. 6, 238244. cerebral ischemia. Am. J. Pathol. 156, 965976.
Gotz, M., Huttner, W.B., 2005. The cell biology of neurogenesis. Maurer, M.H., et al., 2003. Expression of vascular endothelial
Nat. Rev. Mol. Cell Biol. 6, 777788. growth factor and its receptors in rat neural stem cells.
Hattori, K., Heissig, B., Rafii, S., 2003. The regulation of Neurosci. Lett. 344, 165168.
hematopoietic stem cell and progenitor mobilization by Navaratna, D., et al., 2009. Mechanisms and targets for angiogenic
chemokine SDF-1. Leuk. lymphoma 44, 575582. therapy after stroke. Cell Adhes. Migr. 3, 216223.
Hayashi, T., et al., 2003. Temporal profile of angiogenesis and Naylor, M., et al., 2005. Preconditioning-induced ischemic
expression of related genes in the brain after ischemia. tolerance stimulates growth factor expression and
J. Cereb. Blood Flow Metab. 23, 166180. neurogenesis in adult rat hippocampus. Neurochem. Int. 47,
Ikeda, N., et al., 2005. Bone marrow stromal cells that enhanced 565572.
fibroblast growth factor-2 secretion by herpes simplex virus Ohab, J.J., et al., 2006. A neurovascular niche for neurogenesis
vector improve neurological outcome after transient focal after stroke. J. Neurosci.: Off. J. Soc. Neurosci. 26, 1300713016.
cerebral ischemia in rats. Stroke; J. Cereb. Circ. 36, 27252730. Risau, W., 1997. Mechanisms of angiogenesis. Nature 386,
Issa, R., et al., 2005. Expression of basic fibroblast growth factor 671674.
mRNA and protein in the human brain following ischaemic Robin, A.M., et al., 2006. Stromal cell-derived factor 1alpha
stroke. Angiogenesis 8, 5362. mediates neural progenitor cell motility after focal cerebral
Jin, K., et al., 2001. Neurogenesis in dentate subgranular zone and ischemia. J. Cereb. Blood Flow Metab.: Off. J. Int. Soc. Cereb.
rostral subventricular zone after focal cerebral ischemia in the Blood Flow Metab. 26, 125134.
rat. Proc. Natl. Acad. Sci. USA 98, 47104715. Sawada, M., Matsumoto, M., Sawamoto, K., 2014. Vascular
Jin, K., et al., 2002. Vascular endothelial growth factor (VEGF) regulation of adult neurogenesis under physiological and
stimulates neurogenesis in vitro and in vivo. Proc. Natl. Acad.
pathological conditions. Front. Neurosci. 8, 53.
Sci. USA 99, 1194611950. Scharfman, H., et al., 2005. Increased neurogenesis and the
Jin, K., et al., 2003. Directed migration of neuronal precursors into
ectopic granule cells after intrahippocampal BDNF infusion in
the ischemic cerebral cortex and striatum. Mol. Cell. Neurosci.
adult rats. Exp. Neurol. 192, 348356.
24, 171189.
Shen, Q., et al., 2008. Adult SVZ stem cells lie in a vascular niche:
Jin, K., et al., 2006. Evidence for stroke-induced neurogenesis in
a quantitative analysis of niche cellcell interactions. Cell
the human brain. Proc. Natl. Acad. Sci. USA 103, 1319813202.
Stem Cell 3, 289300.
Kalluri, H.S., Vemuganti, R., Dempsey, R.J., 2007. Mechanism of
Shin, T., et al., 2004. Immunohistochemical localization of
insulin-like growth factor I-mediated proliferation of adult
endothelial and inducible nitric oxide synthase within
neural progenitor cells: role of Akt. Eur. J. Neurosci. 25,
neurons of cattle with rabies. J. Vet. Med. Sci. 66, 539541.
10411048.
Smart, I.H., 1973. Proliferative characteristics of the ependymal
Katakowski, M., et al., 2003. Phosphoinositide 3-kinase promotes
layer during the early development of the mouse neocortex: a
adult subventricular neuroblast migration after stroke. J.
pilot study based on recording the number, location and plane
Neurosci. Res. 74, 494501.
of cleavage of mitotic figures. J. Anat. 116, 6791.
Kokaia, Z., et al., 1998. Rapid alterations of BDNF protein levels in
Sun, Y., et al., 2003. VEGF-induced neuroprotection, neurogenesis,
the rat brain after focal ischemia: evidence for increased
and angiogenesis after focal cerebral ischemia. J. Clin.
synthesis and anterograde axonal transport. Exp. Neurol. 154,
Investig. 111, 18431851.
289301.
Tang, H., et al., 2009. Effect of neural precursor proliferation level
Kokovay, E., et al., 2010. Adult SVZ lineage cells home to and leave
the vascular niche via differential responses to SDF1/CXCR4 on neurogenesis in rat brain during aging and after focal
signaling. Cell Stem Cell 7, 163173. ischemia. Neurobiol. Aging 30, 299308.
Krupinski, J., et al., 1994. Role of angiogenesis in patients with Tavazoie, M., et al., 2008. A specialized vascular niche for adult
cerebral ischemic stroke. Stroke; J. Cereb. Circ. 25, 17941798. neural stem cells. Cell Stem Cell 3, 279288.
Lacar, B., et al., 2012. Neural progenitor cells regulate capillary Teng, H., et al., 2008. Coupling of angiogenesis and neurogenesis
blood flow in the postnatal subventricular zone. J. Neurosci. in cultured endothelial cells and neural progenitor cells after
32, 1643516448. stroke. J. Cereb. Blood Flow Metab. 28, 764771.
Lee, S.R., et al., 2006. Involvement of matrix metalloproteinase in Thored, P., et al., 2006. Persistent production of neurons from
neuroblast cell migration from the subventricular zone after adult brain stem cells during recovery after stroke. Stem Cells
stroke. J. Neurosci.: Off. J. Soc. Neurosci. 26, 34913495. 24, 739747.
Li, S.T., et al., 2014. Endothelial nitric oxide synthase protects Thored, P., et al., 2007. Long-term neuroblast migration along
neurons against ischemic injury through regulation of brain- blood vessels in an area with transient angiogenesis and
derived neurotrophic factor expression. CNS Neurosci. Ther. increased vascularization after stroke. Stroke 38, 30323039.
20, 154164. Turner, S.J., et al., 1998. The CC chemokine monocyte chemotactic
Lin, T.N., et al., 2000. Induction of angiopoietin and Tie receptor peptide-1 activates both the class I p85/p110
mRNA expression after cerebral ischemia-reperfusion. phosphatidylinositol 3-kinase and the class II PI3K-C2alpha.
J. Cereb. Blood Flow Metab. 20, 387395. J. Biol. Chem. 273, 2598725995.
Lin, T.N., et al., 2001. Induction of Tie-1 and Tie-2 receptor protein Wang, B., Jin, K., 2014. Current perspectives on the link between
expression after cerebral ischemia-reperfusion. J. Cereb. Blood neuroinflammation and neurogenesis. Metab. Brain Dis. (Epub
Flow Metab. 21, 690701. ahead of print).
Liu, X.S., et al., 2007. Stroke induces gene profile changes Wang, L., et al., 2006. Matrix metalloproteinase 2 (MMP2) and MMP9
associated with neurogenesis and angiogenesis in adult secreted by erythropoietin-activated endothelial cells promote
subventricular zone progenitor cells. J. Cereb. blood flow neural progenitor cell migration. J. Neurosci.: Off. J. Soc. Neurosci.
Metab.: Off. J. Int. Soc. Cereb. Blood Flow Metab. 27, 564574. 26, 59966003.
brain research 1623 (2015) 166173 173

Xiong, Y., Mahmood, A., Chopp, M., 2010. Angiogenesis, stroke. J. Cereb. Blood Flow Metab.: Off. J. Int. Soc. Cereb. Blood
neurogenesis and brain recovery of function following injury. Flow Metab. 26, 857863.
Curr. Opin. Investig. Drugs. 11, 298308. Zhang, R.L., et al., 2014. Stroke increases neural stem cells and
Yamashita, T., et al., 2006. Subventricular zone-derived angiogenesis in the neurogenic niche of the adult mouse.
neuroblasts migrate and differentiate into mature neurons in PLoS One 9, e113972.
the post-stroke adult striatum. J. Neurosci.: Off. J. Soc. Zhang, Z., Chopp, M., 2002. Vascular endothelial growth factor
Neurosci. 26, 66276636. and angiopoietins in focal cerebral ischemia. Trends
Yan, Y.P., et al., 2006. Insulin-like growth factor-1 is an Cardiovasc. Med. 12, 6266.
endogenous mediator of focal ischemia-induced neural Zhang, Z.G., et al., 2000. VEGF enhances angiogenesis and
progenitor proliferation. Eur. J. Neurosci. 24, 4554. promotes blood-brain barrier leakage in the ischemic brain.
Yan, Y.P., et al., 2007. Monocyte chemoattractant protein-1 plays a J. Clin. Investig. 106, 829838.
critical role in neuroblast migration after focal cerebral Zhang, Z.G., et al., 2002. Correlation of VEGF and angiopoietin
ischemia. J. Cereb. Blood Flow Metab.: Off. J. Int. Soc. Cereb. expression with disruption of blood-brain barrier and
Blood Flow Metab. 27, 12131224. angiogenesis after focal cerebral ischemia. J. Cereb. Blood
Yang, S.Z., et al., 2003. Distribution of Flk-1 and Flt-1 receptors in Flow Metab.: Off. J. Int. Soc. Cereb. Blood Flow Metab. 22,
neonatal and adult rat brains. Anat. Rec. Part A: Discov. Mol. 379392.
Cell. Evol. Biol. 274, 851856. Zhang, Z.G., Chopp, M., 2009. Neurorestorative therapies for
Yoshimura, S., et al., 2001. FGF-2 regulation of neurogenesis in stroke: underlying mechanisms and translation to the clinic.
adult hippocampus after brain injury. Proc. Natl. Acad. Sci. Lancet Neurol. 8, 491500.
USA 98, 58745879. Zhu, W., et al., 2009. Postischemic IGF-1 gene transfer promotes
Zacharek, A., et al., 2007. Angiopoietin1/Tie2 and VEGF/Flk1 neurovascular regeneration after experimental stroke.
induced by MSC treatment amplifies angiogenesis and J. Cereb. Blood Flow Metab. 29, 15281537.
vascular stabilization after stroke. J. Cereb. Blood Flow Metab. Zhu, Y., et al., 2005. Angiopoietin-2 facilitates vascular
27, 16841691. endothelial growth factor-induced angiogenesis in the mature
Zhang, R., et al., 2004. Stroke transiently increases subventricular mouse brain. Stroke 36, 15331537.
zone cell division from asymmetric to symmetric and Zhu, Y., et al., 2006. Effects of angiopoietin-1 on vascular
increases neuronal differentiation in the adult rat. J. Neurosci. endothelial growth factor-induced angiogenesis in the mouse
24, 58105815. brain. Acta Neurochir. Suppl. 96, 438443.
Zhang, R.L., et al., 2006. Reduction of the cell cycle length by Zigova, T., et al., 1998. Intraventricular administration of BDNF
decreasing G1 phase and cell cycle reentry expand neuronal increases the number of newly generated neurons in the adult
progenitor cells in the subventricular zone of adult rat after olfactory bulb. Mol. Cell. Neurosci. 11, 234245.

View publication stats

You might also like