You are on page 1of 7

Journal of Investigative Surgery, 23, 149–155, 2010

Copyright  C Informa Healthcare USA, Inc.


ISSN: 0894-1939 print / 1521-0553 online
DOI: 10.3109/08941930903469482

ORIGINAL RESEARCH

VEGF Promotes Angiogenesis and Functional


Recovery in Stroke Rats
Ji-Ping Yang Huai-Jun Liu Xin-Feng Liu
Department of Medical Imaging, The Department of Medical Imaging, The Depatment of Neurology, Jinling
Second Hospital of Hebei Medical Second Hospital of Hebei Medical Hospital, Nanjing University School of
University, Shijiazhuang, Hebei University, Shijiazhuang, Hebei Medicine, Nanjing, Jiangsu Province,
Province, China and Depatment of Province, China China
J Invest Surg Downloaded from informahealthcare.com by Mcgill University on 11/17/14

Neurology, Jinling Hospital, Nanjing


University School of Medicine, Nanjing,
Jiangsu Province, China

ABSTRACT
We evaluated the effects of intranasal vascular endothelial growth factor VEGF on neurological
For personal use only.

function and angiogenesis in ischemic boundary following cerebral ischemia. Sprague–Dawley rats
were randomized into sham operation group (n = 9), VEGF group (n = 18), and control group
(n = 18). The VEGF and control rats were intranasally administered (IN) with VEGF or saline,
starting three days after middle cerebral artery occlusion (MCAO) and daily. Neurological scores
were obtained at 1, 7, and 14 days after MCAO. Rats were sacrificed at 14 days, the von Willebrand
factor (vWF) immunoreactive, BrdU+ /vWF+ cells, and microvessels were evaluated respectively.
Compared to the control rats, intranasal administration of VEGF improved behavioral recovery,
and increased the number of vWF+ , BrdU+ /vWF+ cells, and FITC-dextran perfused microvessels in
ischemic boundary (p < .01). Our data suggest that intranasal administration of VEGF may induce
angiogenesis in ischemic boundary and improve behavioral recovery following cerebral ischemia
in rats, which may provide a powerful strategy for stroke.

Keywords; intranasal administration, middle cerebral artery occlusion, vascular endothelial growth factor,
functional recovery, angiogenesis, cerebral ischemia

INTRODUCTION der zone start to proliferate as early as 12–24 hr follow-


ing vessel occlusion [1–3]. This in turn already leads to
Studies using either mice or rats with transient or an increase of vessels in the ischemic border zone three
permanent occlusion of middle cerebral artery (MCA) days following cerebral ischemia. Vascular endothelial
demonstrated that endothelial cells in the ischemic bor- growth factor (VEGF) has been extensively character-
ized as an angiogenic polypeptide growth factor and
Received June 10, 2009; accepted August 3, 2009. is known to modulate numerous endothelial functions
This study was supported by China Postdortor Science Foundation that are mediated by its receptor, KDR (kinase insert
(grant no. 20070411051, Dr. Yang). Jiangsu Postdoctor Science Foun-
domain receptor) [4]. VEGF also plays an important
dation (grant no. 0701003A, Dr.Yang).
Address correspondence to Xin-Feng Liu, MD, Department of Neu- role in the nervous system [5–7].
rology, Jinling Hospital, Nanjing University School of Medicine, 305
East Zhongshan Road, Nanjing, Jiangsu Province, 210002 China.
Intranasal (IN) administration provides a method to
E-mail: xinfliu@yahoo.com.cn bypass the blood-brain barrier (BBB), and directly

149
J.-P. Yang et al.

deliver therapeutic drugs to the central nervous system previously [11]. Three days after MCAO rats were
(CNS) [8–10]. Results from our previous studies have anesthetized with 10% chloral hydrate (350 mg/kg, in-
documented that intranasally delivered VEGF can by- traperitoneally) and laid on their backs, with their neck
pass the BBB via olfactory- and trigeminal-associated elevated by rolled-up 4 cm × 4 cm gauze, and body tem-
extracellular pathways to directly entry into the CNS perature was maintained with a heating pad and rectal
[11]. thermal probe set at 37◦ C. Under the best aseptic con-
ditions, a total of 100 µl VEGF (200 µg/ml) per rat was
The present study was designed to evaluate the effects given in olfactory pathway [13], 10 µl at a time, alternat-
of IN VEGF on functional recovery and angiogenesis in ing the nostrils, with an interval of 2 min between each
ischemic boundary following cerebral ischemia in rats. administration, over a total of 18.5 min. During these
procedures, the mouth and the opposite nostril were
shut. This was repeated once daily until one day before
MATERIALS AND METHODS
sacrifice. The same volume of saline served as the treat-
Animals ment for saline-treated group. Animals subjected to
the same surgery without vascular occlusion served as
Adult male Sprague-Dawley rats (220–250 g) were used
J Invest Surg Downloaded from informahealthcare.com by Mcgill University on 11/17/14

sham-operated group. All rats were over-anesthetized


in all experiments. Rats were housed in cages and the with 10% chloral hydrate (400 mg/kg, IP) and sacri-
room was kept at 24 ± 1◦ C temperature and 50–60% ficed by decapitation on the 14th day after MCAO.
humidity, under a 12:12-hr light-dark cycle and with
access to food and water ad libitum. All procedures
were performed under the guideline approved by the BrdU Administration
National Institutes of Health Guide for the Care and
For identification of cell proliferation, BrdU (Sigma,
Use of Laboratory Animals (NIH Publications No. 80-
USA) was injected intraperitoneally at a dosage of
23, revised 1996). Every effort was made to minimize
50 mg/kg body weight starting at 24 hr after MCAO
pain and discomfort.
For personal use only.

once daily until the day of sacrifice.

Middle Cerebral Artery Occlusion (MCAO) Neurological Function


Model
The modified Neurological Severity Scores (mNSS) test
Rats were anesthetized by 10% chloral hydrate (normal score is 0; maximal deficit score 18) was per-
(350 mg/kg, intraperitoneally). The right MCA was formed 1, 7, and 14 days after MCAO by an investigator
occluded by means of a monofilament suture as de- who was blinded to the experimental groups as previ-
scribed previously [12]. Briefly, under aseptic condi- ously described [14].
tions, the wound was anesthetized with Lidocaine 2%
(0.1 ml) and a 4–0 surgical monofilament nylon suture
with rounded tip was introduced into the right inter- Infarct Volume Measurement
nal carotid artery through the external carotid artery For quantitative infarct volume, rats (Sham group,
stump, advanced 17.5–18.5 mm past the carotid artery n = 3, VEGF group, n = 6, and Control group, n =
bifurcation to occlude the origin of the right MCA. Af- 6) were perfused transcardially with saline followed
ter 90 min of occlusion, the suture was withdrawn to by 4% paraformaldehyde under deep anesthesia at
restore blood flow. Rectal temperature was monitored 14 days after MCAO, and brains were removed. Brains
and maintained at 37◦ C with heating pads throughout were postfixed in 4% paraformaldehyde for six hr and
the surgical procedures. Since the procedure was so cryoprotected in 30% sucrose solution overnight. In all
short, no analgesics or antibiotics were used. 20 µm coronal sections were obtained on a cryostat
and stored at −80◦ C. Hematoxylin and eosin-stained
specimens were used to measure infarct volume from
Experimental Groups and Intranasal
seven coronal sections spaced 2 mm apart per rat, as de-
Administration scribed previously [15]. Infarct volume was presented
All rats were randomized into three groups: sham op- as a volume percentage of the infarct compared with
erated group (n = 9), VEGF-treated group (n = 18), the contralateral hemisphere [15].
and saline-treated (Control) group (n = 18). VEGF and
control rats were intranasally administered recombi- Immunohistochemical Assessment
nant human VEGF 165 (rhVEGF165, molecular weight
38.2 kDa, PeproTech Corporation, USA) or saline. In- To determine von Willebrand factor (vWF) expression,
tranasal administration was performed as described rats were fixed in 4% paraformaldehyde then washed
150
VEGF Promotes Functional Recovery

in phosphate buffered saline (PBS). The coronal paraf-


fin sections were incubated in 3% H2 O2 in PBS for
10 min, in blocking solution (10% goat serum in PBS)
for 30 min at room temperature, and with rabbit
polyclonal anti-vWF (1:75, Santa Cruz Biotechnology
Inc. Santa Cruz, California, USA) at 4◦ C overnight. Sec-
tions were washed and incubated. The reaction was
stopped with diaminobenzidine (DAB). The positive
cells of vWF in nonreplicated fields were measured
with the Meta Morph micro image analysis system un-
der 400-fold light microscope.
For double-label immunocytochemistry, sections were
fixed with 4% paraformadehyde in PBS, and incubated
in 2 mol/l HCl at 37◦ C for 30 min. Sections were incu-
bated in blocking solution with primary antibodies at
J Invest Surg Downloaded from informahealthcare.com by Mcgill University on 11/17/14

4◦ C overnight and with secondary antibodies at room Figure 1. Quantitative effect of IN VEGF on neurological
temperature for two hr. Primary antibodies were as function after MCAO (mean ±SD). ∗ vs. Control group,
follows: mouse monoclonal anti-BrdU antibody (1:800, p < .01.
Sigma) and rabbit polyclonal anti-vWF antibody (1:200,
Santa Cruz Biotechnology Inc. Santa Cruz, Califor-
nia, USA). The secondary antibodies were fluorescein scores were analyzed by two-way repeated ANOVA.
isothiocyanate (FITC)-conjugated goat antimouse IgG Results were considered significant at p < .05.
(1:100, Jackson Immunoresearch) and tetramethylrho-
damine isothiocyanate (TRITC)-conjugated goat an-
For personal use only.

tirabbit IgG (1:100, Jackson Immunoresearch). BrdU RESULTS


and vWF double labeled cells were detected using a Neurologic Outcome
confocal laser scanning microscope (Leica TCS SP2,
Germany). Results were presented as number of dou- No neurological impairments were observed in the
ble labeled cells. sham-operated group. There was no significant differ-
ence in behavioral recovery between VEGF-treated and
saline-treated group at day 1 after MCAO (p > .05).
Vascular Density Detection Compared to IN saline, VEGF significantly improved
behavioral recovery at days 7 and 14 after MCAO (p <
At 14 days after MCAO, rats (Sham group, n = 3, VEGF .01, Figure 1).
group, n = 6, and Control group, n = 6) were subjected
to FITC-dextran (2 × 106 molecular weight, Sigma;
1 ml of 50 mg/ml preparation) administration via the Infarct Volume
tail vein five min before sacrifice under deep anesthe-
sia with chloral hydrate (350 mg/kg, IP). The brains No infarct volume was observed in the sham-operated
were rapidly removed and placed in 4% paraformalde- group. Compared to IN saline, VEGF significantly re-
hyde at 4◦ C for 48 hr. The green fluorochromes (FITC- duced infarct volume at day 14 after MCAO (p < .01,
dextran) on the sections were excited by a laser beam at Figure 2).
488 nm. Five 50-µm coronal sections at 2 mm intervals
from each rat and eight views in each section adjacent
Angiogenesis in the Ischemic Boundary
to the ischemic lesion were selected to observe by laser-
scanning microscope. The number of microvessels was Cerebral blood vessels were immunostained with the
measured by Image-Pro Plus Version 6.0 software [16, antibody against vWF, an endothelial cell marker.
17]. Number of cerebral vessels increased in the ischemic
boundary regions in rats treated with VEGF, compared
to the sham-operated and saline-treated rats (p < .01,
Statistical Analysis Figure 3).
Data were presented as mean ± standard deviation Three-dimensional measurements of FITC-dextran
(SD). Histological outcome measures were compared perfused cerebral microvessels revealed that VEGF sig-
using one-way analysis of variance (ANOVA) followed nificantly increased the number of microvessels in the
by Tukey’s test for multiple comparisons. Neurological boundary regions of ischemia compared to stroke rats
151
J.-P. Yang et al.

DISCUSSION
Angiogenesis, the growth of new blood vessels, could
be interpreted as a natural defense mechanism help-
ing to restore oxygen and nutrient supply to the is-
chemic brain tissue [18]. Besides, angiogenic vessels
provide neurotrophic support to newly generated neu-
rons [19], and neuroblasts have been found to be con-
centrated around blood vessels following stroke [20].
In the meantime, greater microvessel density in the is-
chemic border correlates with longer survival in stroke
patients [21].
VEGF is the most important mitogen in the process of
angiogenesis. The binding of VEGF to its specific re-
ceptors VEGFR-1(flt-1) and VEGFR-2(flk-1) on the sur-
J Invest Surg Downloaded from informahealthcare.com by Mcgill University on 11/17/14

face of endothelial cells activates intracellular tyrosine


kinases, triggering multiple downstream signals that
Figure 2. Quantitative effect of IN VEGF on infarct volume promote angiogenesis [18]. Previous reports showed
after MCAO (mean ±SD). ∗ vs. Control group, p < .01. an upregulation of VEGF in the penumbra following
experimental cerebral ischemia [3, 22, 23]. An increase
with saline treatment and sham-operated rats (p< .01, in VEGF expression in the infarcted hemisphere was
Figure 4). confirmed as early as three hr after an ischemic injury
and continued up to three [3] or seven days [22, 23].
Only few BrdU colocalized with vWF in the sham- The expression level of VEGFR-1 increased in the in-
For personal use only.

operated rats. As expected, compared to the sham- farct core from the 3rd day up to the 14th day [24] and
operated and saline-treated rats, a greater number of in the ischemic border zone at 48 hr following cere-
BrdU+ /vWF+ immunoreactive cells were detected in bral ischemia [3, 23]. VEGFR-2 mediates microvascular
the VEGF-treated rats at day 14 after MCAO (p < .01, permeability, endothelial cell proliferation, invasion,
Figure 5). migration, and survival [25]. Several reports described

Figure 3. VEGF enhanced the number of microvascular. Cerebral blood vessels immunostained with the antibody against vWF.
Number of cerebral vessels increased in the ischemic boundary regions in rats treated with VEGF (c), compared to those of the
sham-operated group (a), and rats in the saline-treated (Control) group (b). Bar = 40 µm. D, Quantitative data of vWF+ cells
(mean ±SD). ∗ vs. Control group, p < .01. # vs. Sham-operated group, p < .01.
152
VEGF Promotes Functional Recovery
J Invest Surg Downloaded from informahealthcare.com by Mcgill University on 11/17/14
For personal use only.

Figure 4. FITC-dextran perfused vessels in the ischemic boundary regions of VEGF-treated (c), saline-treated (Control) group
(b), and sham-operated rats (a). VEGF significantly increased the number of microvessels in the boundary regions of ischemia
compared to saline-treated and sham-operated rats. Bar = 20 µm. D, Quantitative data of FITC-dextran perfused microvessels
(mean ±SD). ∗ vs. Control group, p < .01. #vs. Sham-operated group, p < .01.

Figure 5. IN VEGF enhanced the angiogenesis in the boundary regions of ischemia. Part a through c shows colocalization of
BrdU and vWF in the ischemic boundary of rats treated with VEGF. Bar = 40 µm. D, Quantitative data of BrdU+ /vWF+
immunoreactive cells (mean ±SD). ∗ vs. Control group, p < .01. #vs. Sham-operated group, p < .01.
153
J.-P. Yang et al.

an upregulation of VEGFR-2 following experimental mised the beneficial neuroprotective actions of VEGF
cerebral ischemia [3, 22, 26]. [38]. However, late administration of VEGF, i.e., on
days 1–3 of reperfusion improved neurological out-
In hypoxic and ischemic brain, the temporal-spatial ex- come [31, 32]. In our study, VEGF was intranasally de-
pression of VEGF has been shown to correlate with vas- livered on days 3–13 of ischemia which could enhance
culogenesis [27, 28]. VEGF expression in the ischemic angiogenesis and improve functional recovery.
border zone increased progressively between 2 and
14 days after MCAO, and evidence of new vessel forma-
tion was present during days 7 to 28 [28]. Intracerebral
infusion of VEGF stimulates angiogenesis detectable by
CONCLUSION
laminin immunostaining in rat cerebral cortex 3–7 days In summary, IN VEGF can improve angiogenesis in the
later [29, 30]. In the ischemic rat brain, intravenously ischemic border zone and functional recovery follow-
administered VEGF also triggers angiogenesis, as man- ing cerebral ischemia in rats. IN VEGF may provide a
ifested by an increase in the number and volume of powerful strategy for stroke.
FITC-dextran-perfused cerebral cortical microvessels
7 days later [31]. Sun et al. [32] also found an increase
J Invest Surg Downloaded from informahealthcare.com by Mcgill University on 11/17/14

in the density of vWF-immunoreactive blood vessels DECLARATION OF INTEREST


7–28 days after focal ischemia plus VEGF treatment in
the striatal ischemic penumbra. The authors report no conflict of interest. The authors
alone are responsible for the content and writing of this
Increases in expression of VEGF/VEGF receptors at the paper.
penumbra region after ischemia plus VEGF treatment
are coincident with the time and distribution of neovas-
cularization in ischemic brain, suggesting that VEGF is
temporally and spatially correlated with angiogenesis
REFERENCES
For personal use only.

[28]. Therefore, exogenous VEGF may promote thera- 1. Beck H, Acker T, Wiessner C, et al. Expression of angiopoietin-
peutic angiogenesis in the ischemic rats, and restoration 1, angiopoietin-2, and tie receptors after middle cerebral artery
occlusion in the rat. Am J Pathol. 2000;157:1473–1483.
of cerebral microvascular circulation is important in the
2. Hayashi T, Noshita N, Sugawara T, et al. Temporal profile of
ischemic brain for functional recovery after stroke. angiogenesis and expression of related genes in the brain after
ischemia. J Cereb Blood Flow Metab. 2003;23:166–180.
In our study, an increase of microvessels in the 3. Marti HJ, Bernaudin M, Bellail A, et al. Hypoxia-induced vas-
ischemic border zone was confirmed by detecting cular endothelial growth factor expression precedes neovascu-
more FITC-dextran-perfused microvessels, vWF+ cells, larization after cerebral ischemia. Am J Pathol. 2000;156:965–976.
and BrdU+ / vWF+ cells in the rats treated with IN 4. Petrova TV, Makinen T, Alitalo K. Signaling via vascular en-
rhVEGF165 when compared to IN saline rats. Thus, dothelial growth factor receptors. Exp Cell Res. 1999;253:117–130.
5. Raab S, Plate KH. Different networks, common growth factors:
these data indicated that IN rhVEGF165 significantly shared growth factors and receptors of the vascular and the
enhanced angiogenesis in ischemic brain. nervous system. Acta Neuropathol. 2007;113:607–626.
6. Yang JP, Liu XF, Liu HJ, et al. Extracellular signal-regulated
In the literature on stroke, much attention has been kinase involved in NGF/VEGF-induced neuroprotective effect.
focused on the penumbra [33]. Excitotoxic damage, in- Neurosci Lett. 2008;434:212–217.
flammation, and ultimately cell death in the penum- 7. Yang JP, Liu HJ, Li Y. Effect of endoplasmic reticulum stress in
bra are possible targets for therapy [34]. Reports from VEGF-induced neuroprotection. J Invest Surg. 2009;22:29–34.
8. Thorne RG, Pronk GJ, Padmanabhan V, et al. Delivery of insulin-
stroke models and patients document the proliferation like growth factor-I to the rat brain and spinal cord along olfac-
of vascular cells and expression of markers for angio- tory and trigeminal pathways following intranasal administra-
genesis surrounding infarcts [21, 35–37]. However, the tion. Neuroscience. 2004;127:481–496.
relationship between angiogenesis and functional re- 9. Liu XF, Fawcett JR, Thorne RG, et al. Intranasal administration
covery in the brain after focal ischemia has not been of insulin-like growth factor-I bypasses the blood-brain barrier
and protects against focal cerebral ischemic damage. J Neurol
characterized. Our data demonstrated that late admin- Sci. 2001;187:91–97.
istration of IN rhVEGF165 to ischemic rats significantly 10. Thorne RG, Frey WH. 2nd. Delivery of neurotrophic factors
improved neurological function and reduced infarct to the central nervous system: pharmacokinetic considerations.
size. Therefore, treatment with VEGF to potentiate new Clin Pharmacokinet. 2001;40:907–946.
vessel formation may be an important therapy for be- 11. Yang JP, Liu HJ, Cheng SM, et al. Direct transport of VEGF from
the nasal cavity to brain. Neurosci Lett. 2009;449:108–111
havioral recovery during the repair process after stroke. 12. Longa EZ, Weinstein PR, Carlson S, et al. Reversible middle
cerebral artery occlusion without craniectomy in rats. Stroke.
The effect of VEGF depends on the dose, route, and time
1989;20:84–91.
of administration in relation to brain ischemia. When 13. Yang JP, Liu HJ, Wang ZL, et al. The dose-effectiveness of in-
VEGF was delivered in the acute stage after stroke, the tranasal VEGF in treatment of experimental stroke. Neurosci Lett.
BBB was damaged and this damage may have compro- 2009;461:212–216.
154
VEGF Promotes Functional Recovery

14. Chen J, Sanberg PR, Li Y, et al. Intravenous administration of 27. Ogunshola OO, Stewart WB, Mihalcik V, et al. Neuronal
human umbilical cord blood reduces behavioral deficits after VEGF expression correlates with angiogenesis in postnatal
stroke in rats. Stroke. 2001;32:2682–2688. developing rat brain. Brain Res Dev Brain Res. 2000;119:139–
15. Swanson RA, Morton MT, Tsao-Wu G, et al. A semiautomated 153.
method for measuring brain infarct volume. J Cereb Blood Flow 28. Zhang ZG, Zhang L, Tsang W, et al. Correlation of VEGF and
Metab. 1990;10:290–293. angiopoietin expression with disruption of blood-brain barrier
16. Barros LF, Belfort R Jr. The effects of the subconjunctival injec- and angiogenesis after focal cerebral ischemia. J Cereb Blood Flow
tion of bevacizumab (Avastin) on angiogenesis in the rat cornea. Metab. 2002;22:379–392.
An Acad Bras Cienc. 2007;79:389–394. 29. Rosenstein JM, Mani N, Silverman WF, et al. Patterns of
17. Blatt RJ, Clark AN, Courtney J, et al. Automated quantitative brain angiogenesis after vascular endothelial growth factor ad-
analysis of angiogenesis in the rat aorta model using Image-Pro ministration in vitro and in vivo. Proc Nat Acad Sci U S A.
Plus 4.1. Comput Methods Programs Biomed. 2004;75:75–79. 1998;95:7086–7091.
18. Beck H, Plate KH. Angiogenesis after cerebral ischemia. Acta 30. Krum JM, Mani N, Rosenstein JM. Angiogenic and astroglial
Neuropathol. 2009;117:481–496. responses to vascular endothelial growth factor administration
19. Leventhal C, Rafii S, Rafii D, et al. Endothelial trophic support in adult rat brain. Neuroscience. 2002;110:589–604.
of neuronal production and recruitment from the adult mam- 31. Zhang ZG, Zhang L, Jiang Q, et al. VEGF enhances angiogenesis
malian subependyma. Mol Cell Neurosci. 1999;13:450–464. and promotes blood–brain barrier leakage in the ischemic brain.
20. Yamashita T, Ninomiya M, Hernández Acosta P, et al. Subven- J Clin Invest. 2000;106:829–838.
J Invest Surg Downloaded from informahealthcare.com by Mcgill University on 11/17/14

tricular zone-derived neuroblasts migrate and differentiate into 32. Sun Y, Jin K, Xie L, et al. VEGF-induced neuroprotection, neu-
mature neurons in the post-stroke adult striatum. J Neurosci. rogenesis, and angiogenesis after focal cerebral ischemia. J Clin
2006;26:6627–6636. Invest. 2003;111:1843–1851.
21. Krupinski J, Kaluza J, Kumar P, et al. Role of angiogenesis in pa- 33. Astrup J, Siesjo BK, Symon L. Thresholds in cerebral ischemia:
tients with cerebral ischemic stroke. Stroke. 1994;25:1794–1798. the ischemic penumbra. Stroke. 1981;12:723–725.
22. Hayashi T, Abe K, Suzuki H, et al. Rapid induction of vascular 34. Dirnagl U, Iadecola C, Moskowitz MA. Pathobiology of
endothelial growth factor gene expression after transient middle ischaemic stroke: an integrated view. Trends Neurosci.
cerebral artery occlusion in rats. Stroke. 1997;28:2039–2044. 1999;22:391–397.
23. Plate KH, Beck H, Danner S, et al. Cell type specific up- 35. Chen HH, Chien CH, Liu M. Correlation between angiogenesis
regulation of vascular endothelial growth factor in an MCA- and basic fibroblast growth factor expression in experimental
occlusion model of cerebral infarct. J Neuropathol Exp Neurol. brain infarct. Stroke. 1994;25:1651–1657.
For personal use only.

1999;58:654–666. 36. Krupinski J, Kumar P, Kumar S, et al. Increased expression


24. Kovács Z, Ikezaki K, Samoto K, et al. VEGF and flt. Expression of TGF-beta 1 in brain tissue after ischemic stroke in humans.
time kinetics in rat brain infarct. Stroke. 1996;27:1865–1872. Stroke. 1996;27:852–857.
25. Hicklin DJ, Ellis LM. Role of the vascular endothelial growth 37. Okada Y, Copeland BR, Hamann GF, et al. Integrin alphav-
factor pathway in tumor growth and angiogenesis. J Clin Oncol. beta3 is expressed in selected microvessels after focal cerebral
2005;23:1011–1027. ischemia. Am J Pathol. 1996;149:37–44.
26. Lennmyr F, Ata KA, Funa K, et al. Expression of vascular en- 38. van Bruggen N, Thibodeaux H, Palmer JT, et al. VEGF an-
dothelial growth factor (VEGF) and its receptors (Flt-1 and Flk-1) tagonism reduces edema formation and tissue damage after
following permanent and transient occlusion of the middle cere- ischemia/reperfusion injury in the mouse brain. J Clin Invest.
bral artery in the rat. J Neuropathol Exp Neurol. 1998;57:874–882. 1999;104:1613–1620.

155

You might also like