You are on page 1of 7

From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

Plenary paper

Bifunctional role for VEGF-induced heme oxygenase-1 in vivo: induction


of angiogenesis and inhibition of leukocytic infiltration
Benedetta Bussolati, Asif Ahmed, Helen Pemberton, R. Clive Landis, Francesco Di Carlo, Dorian O. Haskard, and Justin C. Mason

Heme-oxygenases (HOs) catalyze the con- bition abrogated VEGF-driven angiogen- LPS-induced model of inflammatory an-
version of heme into carbon monoxide esis. Two murine models of angiogenesis giogenesis, induction of HO-1 with cobalt
and biliverdin. HO-1 is induced during were used: (1) angiogenesis initiated by protoporphyrin significantly inhibited leu-
hypoxia, ischemia/reperfusion, and in- addition of VEGF to Matrigel and (2) a kocyte invasion into LPS-conditioned Ma-
flammation, providing cytoprotection and lipopolysaccharide (LPS)–induced model trigel and thus prevented the subsequent
inhibiting leukocyte migration to inflam- of inflammatory angiogenesis in which angiogenesis. We therefore propose that
matory sites. Although in vitro studies angiogenesis is secondary to leukocyte during chronic inflammation HO-1 has 2
have suggested an additional role for invasion. Pharmacologic inhibition of roles: first, an anti-inflammatory action
HO-1 in angiogenesis, the relevance of HO-1 induced marked leukocytic infiltra- inhibiting leukocyte infiltration; and sec-
this in vivo remains unknown. We investi- tion that enhanced VEGF-induced angio- ond, promotion of VEGF-driven nonin-
gated the involvement of HO-1 in angio- genesis. However, in the presence of an flammatory angiogenesis that facilitates
genesis in vitro and in vivo. Vascular anti-CD18 monoclonal antibody (mAb) to tissue repair. (Blood. 2004;103:761-766)
endothelial growth factor (VEGF) induced block leukocyte migration, VEGF-induced
prolonged HO-1 expression and activity angiogenesis was significantly inhibited
in human endothelial cells and HO-1 inhi- by HO-1 antagonists. Furthermore, in the © 2004 by The American Society of Hematology

Introduction
An increasing body of evidence indicates that cells from the innate concentration in the brain, testis, and vascular EC, while HO-1 is more
immune system such as monocytes/macrophages are important widely distributed.16 HO-1 is rapidly induced during hypoxia, ischemia/
regulators of angiogenesis.1,2 Granulocytes, whose role is fre- reperfusion, hyperthermia, and endotoxic shock, providing cytoprotec-
quently underestimated, also appear to play a primary role in the tion during the resolution of stress-induced inflammatory injury.17 The
initial phases of the angiogenic process.3,4 Angiogenesis is closely importance of this is demonstrated by the severe and persistent
associated with inflammation in several diseases including rheuma- endothelial damage observed in human HO-1 deficiency18 and in
toid arthritis, atherosclerosis, carcinoma, and hematologic malignan- gene-targeted mice deficient in HO-1.19 Furthermore, induction of
cies.5,6 Moreover, several angiogenic growth factors are not HO-1 may directly regulate EC activation, preventing adhesion
endothelial cell (EC) specific. In particular, vascular endothelial molecule expression and chronic graft rejection.20-22 In vitro, HO-1
growth factor (VEGF), recognized to be fundamental to the process of protects ECs from hydrogen peroxide–mediated cell death23 and
angiogenesis,7 may also activate and recruit monocytes8 and has been from tumor necrosis factor ␣ (TNF␣) cytotoxicity.24 In addition to
directly implicated in the inflammatory angiogenesis associated with its role in vascular cytoprotection, it has been suggested that HO-1
diseases such as atherosclerosis and rheumatoid arthritis.9-12 may play a role in angiogenesis as induction of HO-1 increases EC
Recent studies have shown that heme oxygenase-1 (HO-1) may VEGF synthesis25 and overexpression of HO-1 induces prolifera-
exert anti-inflammatory effects including prolongation of cardiac xeno- tion and formation of capillary-like structures.26 However, the
graft graft survival13 and inhibition of leukocyte transendothelial migra- importance of HO-1 in VEGF-driven angiogenesis in vivo is
tion during complement-dependent inflammation14 and in response to currently unknown.
low-density lipoprotein (LDL) oxidation.15 Heme oxygenases are rate- In the present study, we have investigated both in vitro and in
limiting enzymes that catalyze the conversion of heme into carbon vivo the involvement of HO-1 in VEGF-dependent angiogenesis
monoxide (CO) and biliverdin. Human HO exists in 2 main isoforms, and specifically the effect of HO-1 activation on inflammatory
HO-1 (inducible) and HO-2 (constitutive). HO-2 is present in high angiogenesis in vivo.

From the Department of Biology and Clinical Science, University of Torino, and “Oncology,” Compagnia San Paolo, Torino, Italy. J.C.M. is an Arthritis Research
Research Center for Experimental Medicine (CeRMS), Ospedale S. Giovanni Campaign Senior Fellow.
Battista, Torino, Italy; Department of Reproductive and Vascular Biology, The
Reprints: Asif Ahmed, Department of Reproductive and Vascular Biology, The
Medical School, University of Birmingham, Edgbaston, Birmingham, United
Medical School, University of Birmingham, Edgbaston, Birmingham, B12 2TG,
Kingdom; and British Heart Foundation Cardiovascular Medicine Unit, The Eric
United Kingdom; e-mail: a.s.ahmed@bham.ac.uk; and Benedetta Bussolati,
Bywaters Center, Imperial College London, Hammersmith Hospital, London,
Department of Biology and Clinical Science, University of Torino, Az
United Kingdom.
Ospedaliera San Luigi, Regione Gonzole 10-10043 Orbassano, Torino, Italy;
Submitted June 18, 2003; accepted September 22, 2003. Prepublished online as e-mail: benedetta.bussolati@unito.it.
Blood First Edition Paper, October 2, 2003; DOI 10.1182/blood-2003-06-1974. The publication costs of this article were defrayed in part by page charge
payment. Therefore, and solely to indicate this fact, this article is hereby
Supported by grants from Arthritis Research Campaign grant M0620, British
marked ‘‘advertisement’’ in accordance with 18 U.S.C. section 1734.
Heart Foundation Programme grant RG/98/0003, and the Italian Ministry of
University and Research (MIUR) ex 60%, and by the Special Project © 2004 by The American Society of Hematology

BLOOD, 1 FEBRUARY 2004 䡠 VOLUME 103, NUMBER 3 761


From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

762 BUSSOLATI et al BLOOD, 1 FEBRUARY 2004 䡠 VOLUME 103, NUMBER 3

ng/mL) was added in the absence or presence of SnPP at a range of


concentrations and ECs were cultured for 72 hours with 3H-labeled
Materials and methods thymidine added 16 hours prior to the end of the assay. ECs were harvested
Reagents and proliferation quantified with an automated Betaplate 96-well harvester
(Wallac Oy, Turku, Finland). EC viability was assessed by trypan blue
The HO-1 inhibitors tin (IV) protoporphyrin IX dichloride (SnPP) and zinc exclusion and found to be more than 90%. EC tube formation was assessed
(II) protoporphyrin IX (ZnPP) and the HO-1 activator cobalt (III) protopor- by adaptation of a previously described assay.32 Segments (5 mm ⫻ 5 mm)
phyrin IX chloride (CoPP) were purchased from Porphyrin Products of C57BL/6 murine abdominal wall muscle were placed in Matrigel in
(Logan, UT). Metalloporphyrins were dissolved in 0.2 M NaOH and the pH 24-well plates. Dulbecco modified Eagle medium (DMEM) supplemented
adjusted to 7.4. Acidic fibroblast growth factor (aFGF) and VEGF were with 10% FBS, heparin (10 U/mL), and VEGF in the absence and presence
from PeproTech EC (London, United Kingdom). The rat antimouse ␤2 of SnPP (20 ␮M) was added and changed every 2 days. After 9 days EC
integrin (CD18) monoclonal antibody (mAb) M18/2 (immunoglobulin G 2a tube length was measured from the edge of the embedded tissue using
[IgG2a]) was obtained from the American Type Culture Collection (Manas- Optimas 6.1 Software (Optimas UK, West Malling, United Kingdom). In
sas, VA). Lipopolysaccharide (LPS) from Escherichia coli (0111:B4) was individual experiments each treatment was assessed in triplicate and the
purchased from Sigma-Aldrich (St Louis, MO) and stock solutions were length of 10 EC tubes for each tissue segment was measured. The
prepared by suspending 10 mg in 2 mL 20 mM ethylenediaminetetraacetic endothelial nature of the tubes was confirmed by staining with fluorescein
acid (EDTA), sonicating until clarified, and freezing at ⫺20°C. LPS isothiocyanate (FITC) anti–von Willebrand factor (anti-VWF; Serotec,
working dilutions were prepared in 10 mM HEPES (N-2-hydroxyethylpipera- Oxford, United Kingdom) and the lectin Griffonia simplicifolia isolectin B4
zine-N⬘-2-ethanesulfonic acid; Gibco Laboratories, Grand Island, NY). (Vector Laboratories, Peterborough, United Kingdom). In all experiments
Matrigel basement membrane matrix was obtained from Becton Dickinson appropriate carrier controls were included.
Labware (Bedford, MA).

Endothelial cells In vivo murine angiogenesis assay

Human umbilical vein ECs (HUVECs) and the human microvascular Angiogenesis was quantified by the Matrigel plug assay as previously
endothelial cell 1 (HMEC-1) dermal microvascular line27 (a kind gift from described.33 Matrigel (8.13 mg/mL), in liquid form at 4°C, was mixed with
Dr E Ades, Centers for Disease Control, Atlanta, GA) were cultured as heparin (64 U/mL, Sigma) and the experimental substances and injected
previously described in detail.28,29 (0.25 mL) into the abdominal subcutaneous tissue of mice, along the
peritoneal midline. To evaluate the effect of HO-1 blockade or activation,
Western blotting analysis SnPP (20 ␮M), ZnPP (20 ␮M), or CoPP (25 ␮M) was added to Matrigel.
CoPP was also injected intraperitoneally (5 mg/kg) at day 0. To inhibit ␤2
HMECs or HUVECs, treated for up to 48 hours with VEGF or aFGF, were integrins, mAb M18/2 or isotype-matched control mAbs were added to
lysed (4 mM EDTA, 50 mM Tris (tris[hydroxymethyl]aminomethane)– Matrigel at a final concentration of 20 ␮g/mL and in addition 100 ␮g of
HCL, pH7.4 in 150 mM NaCl with 25 mM sodium deoxycholic acid, 200 mAb M18/2 or isotype-matched control mAb was injected intraperitoneally
␮M sodium orthovanadate, 10 mM sodium pyrophosphate, 100 mM on days 0, 3, and 5. At day 6, mice were killed and gels were processed for
sodium fluoride, 1% Triton X-100, 1 mM phenylmethysulfonyl fluoride, light microscopy and immunohistochemistry, as described.34 Rabbit anti-
and 5% protease inhibitor cocktail) and the protein content determined VWF Ab, as well as control rabbit and mice IgG (Sigma), were used as
using the Bio-Rad Dc protein assay (Bio-Rad, Hercules, CA). Cell lysates primary antibodies for indirect immunofluorescence. FITC-conjugated
were subjected to sodium dodecyl sulfate–polyacrylamide gel electrophore- antirabbit IgG and antirat IgG were used as secondary antibodies (all from
sis (SDS-PAGE) on 12.5% gels and separated proteins transferred to Sigma). Direct immunofluorescence staining was performed with antimy-
Immobilon-P transfer membranes (Millipore Corporation, Bedford, MA). eloperoxidase (anti-MPO) phycoerythrin (PE)–conjugated monoclonal anti-
HO-1 was detected with polyclonal anti–HO-1 (Stressgen Biotechnologies, bodies (Cedarlane, Hornby, ON, Canada). The total Matrigel area and the
Victoria, BC, Canada) and actin was detected as a loading control with mAb area occupied by vessels were planimetrically assessed from cross-sections
C-2 (Santa Cruz Biotechnology, Santa Cruz, CA). The blots were developed of Matrigel plugs observed by light microscopy as previously described.33
with an enhanced chemiluminescence substrate (Amersham Pharmacia Results were expressed as percentage ⫾ SE of the vessel area with respect
Biotech UK, Little Chalfont, United Kingdom). to the total Matrigel area.

Heme oxygenase activity assay


Statistics
HMECs were harvested by scraping, centrifuged at 2000g for 10 minutes,
and stored at ⫺80°C until required. Following 3 freeze/thaw cycles the All data are expressed as mean ⫾ SD. Nonparametric statistical analysis
protein concentration was determined using the Bio-Rad Dc protein assay. A was performed by the Kruskal-Wallis test for analysis of variance (ANOVA).
chromatographic assay was then performed30 in which the samples were
added to a reagent mix containing 0.2 mM MgCl2, 100 mM phosphate-
buffered saline (PBS) pH7.4, 0.5 mg/mL of liver cytosol, 1.1 mM
glucose-6-phosphate, 0.84 U/mL glucose-6-phosphate dehydrogenase, 28 Results
␮M hemin, and 40 mM nicotinamide adenine dinucleotide phosphate
(NADPH). Following vortexing, the reagents were left to react at 37°C for 1 VEGF, but not aFGF, induces HO-1 expression
hour. The reaction was terminated with chloroform and the samples
vortexed and centrifuged at 900g for 5 minutes. After vortexing and further The expression of HO-1 by ECs following treatment with VEGF
centrifugation at 1100g the extracted bilirubin was quantified by calculating and aFGF was assessed by Western blotting. An increase in HO-1
the difference in absorbance at 464 nm and 530 nm (extinction coefficient expression following exposure to VEGF was seen at 24 hours and
for bilirubin 40 mM⫺1cm⫺1). HO-1 activity was expressed as pmol of was maximal at 48 hours in HMECs (Figure 1A). Similar results
bilirubin formed per mg of protein per hour. were obtained with HUVECs (Figure 1B) and bovine aortic ECs
(not shown). In contrast aFGF, at a concentration sufficient to
Endothelial cell proliferation and tube formation assays induce EC proliferation, was incapable of inducing a significant
Cell proliferation was measured as previously described.31 HUVECs change in HO-1 expression after 48 hours (Figure 1C). Endothelial
(5 ⫻ 103 cells/well) were cultured in M199/10% fetal bovine serum (FBS) expression of HO-1 after treatment with VEGF was coupled with
in the absence of growth factor for 24 hours in 96-well plates. VEGF (25 an increase in HO-1 activity (Figure 1D).
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

BLOOD, 1 FEBRUARY 2004 䡠 VOLUME 103, NUMBER 3 DUAL ROLE OF VEGF-INDUCED HO-1 IN ANGIOGENESIS 763

Figure 1. Expression and activity of HO-1 induced by VEGF in human ECs. Lysates were prepared from HMECs and HUVECs treated with VEGF (25 ng/mL) or aFGF (100
ng/mL) for up to 48 hours and proteins were separated by SDS-PAGE and transblotted on to nitrocellulose membranes. Immunoblots were probed with a polyclonal Ab against
HO-1 and mAb C-2 against actin as a loading control. (A) HMECs and (B) HUVECs. Lane 1, unstimulated ECs; 2, VEGF 24 hours; 3, VEGF 48 hours. (C) HMECs. Lane 1,
unstimulated ECs; 2, aFGF 24 hours; 3, aFGF 48 hours. (D) Heme oxygenase activity was measured in lysates of HMECs cultured in the presence or absence of VEGF (25
ng/mL) for 24 hours. Data presented as mean ⫾ SEM.

Blockade of HO-1 inhibits VEGF-induced in vitro angiogenesis dependent manner. To investigate this further, murine abdominal muscle
was implanted in Matrigel and capillary sprouting and neovessel
The role of HO-1 induction in VEGF-driven angiogenesis was first
development in response to VEGF was analyzed. As seen in Figure
assessed in vitro. As seen in Figure 2A, the induction of HO-1 with
2B-C, inhibition of HO-1 with both SnPP and ZnPP (not shown)
CoPP alone resulted in EC proliferation comparable to that seen with
completely inhibited the development of VEGF-induced neovessels.
VEGF. The presence of the HO-1 inhibitors SnPP or ZnPP (not shown)
significantly inhibited VEGF-induced EC proliferation in a dose- HO-1 blockade promotes VEGF-induced inflammation
and angiogenesis

The murine Matrigel model was used to investigate the role of


HO-1 in VEGF-induced angiogenesis in vivo. Surprisingly, HO-1
inactivation by SnPP (20 ␮M) increased the angiogenic effect
induced by VEGF (Figure 3A). In contrast, no such effect was
observed when SnPP was added to Matrigel plugs containing aFGF
(Figure 3A). Further experiments with ZnPP (20 ␮M) confirmed
these results (Figure 3B). Histologic examination of the implants
containing VEGF plus SnPP or ZnPP showed the formation of
enlarged hemorrhagic vessels when compared with the neovessels
seen in plugs containing VEGF alone (Figure 4A-B). In addition,
the presence of a dense inflammatory infiltrate was noted within
implants treated with VEGF ⫹ SnPP or ZnPP (Figure 4C) and was
confirmed by immunofluorescence staining for myeloperoxidase
(Figure 4D). Quantification demonstrated a significant leukocytic
infiltrate in plugs treated with a combination of VEGF and SnPP or
ZnPP when compared with VEGF alone (P ⬍ .05) and a smaller
but significant infiltrate was also seen with SnPP or ZnPP alone
(P ⬍ .05; Figure 3C). Interestingly, in a recent study of wound
healing, heme-induced influx of leukocytes was also significantly
elevated following pharmacologic inhibition of HO-1.35
In order to investigate further the influence of the migrating
leukocytes on angiogenesis the inhibitory anti-␤2 mAb M18/2 was
used. Treatment of mice with M18/2 did not affect VEGF-induced
angiogenesis per se but abrogated the previously observed enhance-
ment of VEGF-induced angiogenesis in the presence of SnPP
(P ⬍ .05; Figure 5A). This was related to a specific suppression of
leukocytic infiltration into Matrigel plugs by M18/2, as an isotype-
matched control mAb did not affect the proinflammatory effects of
Figure 2. Role of HO-1 in VEGF-induced angiogenesis in vitro. (A) HUVECs were SnPP (Figure 5B) and no effect of mAb M18/2 was seen on
cultured in M199 with 10% FBS for 24 hours prior to the addition of VEGF (25 ng/mL) VEGF-induced angiogenesis (Figure 5A). Furthermore, in the
and CoPP (12.5 and 25 ␮M) both for 72 hours. In test wells, ECs were preincubated absence of leukocyte infiltration, SnPP completely inhibited VEGF-
with SnPP at the concentrations shown for 60 minutes prior to the addition of VEGF.
3H-thymidine (0.037 MBq/mL [1 ␮Ci/mL]) was added 16 hours before the end of the induced angiogenesis (Figure 5A) supporting a role for HO-1 in
assay and the plates were read on an automated plate reader. Bars show the mean ⫹ angiogenesis directly driven by VEGF.
SD uptake of 3H-thymidine (n ⫽ 4). (B-F) Murine abdominal muscle was implanted in
Matrigel and cultured in DMEM supplemented with 10% FBS and heparin (10 U/mL). HO-1 activation inhibits leukocyte recruitment and
VEGF (25 ng/mL) was added to the medium in the absence and presence of SnPP inflammation-related angiogenesis
(20 ␮M). (B) After 9 days EC tube length was measured by image analysis. Data
presented as mean ⫹ SD tube length (n ⫽ 3). Panels C-F show representative To explore these findings further, lipopolysaccharide was added to
micrographs of the effect of SnPP on neovessels formed in the presence of VEGF.
Panels C and E show VEGF alone and panels D and F show VEGF ⫹ SnPP. Original Matrigel plugs as a representative model of inflammatory angiogen-
magnification: low power ⫻ 20 (C-D) and high power ⫻ 40 (E-F). esis in vivo. This model is characterized by an intense leukocytic
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

764 BUSSOLATI et al BLOOD, 1 FEBRUARY 2004 䡠 VOLUME 103, NUMBER 3

Figure 3. Role of HO-1 during in vivo angiogenesis. (A) Quantitative evaluation of neovessels infiltrating Matrigel after injection of aFGF (50 ng/mL), VEGF (40 ng/mL), or
vehicle alone in the absence or presence of SnPP (20 ␮M). The results are expressed as percentage ⫾ SE of the vessel area to the total Matrigel area. (B) Effect of SnPP (20
␮M) and ZnPP (20 ␮M) on angiogenesis induced by VEGF (40 ng/mL). (C) Quantitative evaluation of inflammatory cells infiltrating Matrigel that were counted in sections
stained with hematoxylin and eosin. The results are expressed as the mean ⫾ SE of cells/field (⫻ 400). Six mice were used per condition in each experiment.

infiltration and an associated angiogenic response.36 As expected, One of the products of HO-1 activation, CO, is an endogenously
the inclusion of LPS alone induced leukocytic infiltration into the produced vasorelaxant molecule that activates soluble guanylate cyclase
Matrigel plug with significant angiogenesis (Figure 4E and 6). To leading to an increase in intracellular cyclic guanosine monophosphate
investigate the effect of HO-1 activation in this model of angiogen- (cGMP).40 Recent work has demonstrated that treatment with CO
esis mice were treated with the CoPP. As seen in Figure 4F and prevents arteriosclerosis in a model of aortic transplantation in which
Figure 6, addition of CoPP significantly inhibited leukocyte- CO exerts a marked inhibitory effect on graft leukocyte infiltration and
induced angiogenesis, suggesting that HO-1 may act to suppress activation and reduces vascular smooth muscle cell proliferation via
inflammatory angiogenesis in vivo. generation of cGMP and activation of p38 mitogen-activated protein
kinase (MAPK).41 Activation of guanylate cyclase is also induced by
nitric oxide (NO), the major secondary mediator of VEGF effects.42 It is
Discussion therefore possible that HO-1 and NO synthase exert a synergistic effect

Previous in vitro studies have reported a role for HO-1 in EC survival,


proliferation, and tube formation, suggesting that HO-1 induction may
promote angiogenesis and that this is mediated, at least in part, by
CO.25,26 A recent study has also reported HO-1 induction in the chicken
chorioallantoic membrane following treatment with VEGF.37 We found
that VEGF, a potent angiogenic factor, induced HO-1 expression and
increased HO-1 activity in human ECs. The potential involvement of
endothelial HO-1 in VEGF-driven angiogenesis was demonstrated by
the ability of the HO-1 antagonists SnPP and ZnPP to significantly
inhibit EC proliferation and tube formation in vitro. Furthermore, the
results presented herein demonstrate for the first time that HO-1 plays an
important role in VEGF-driven angiogenesis in vivo and acts as a key
regulator in the control of the anti-inflammatory versus proinflammatory
actions of VEGF.
VEGF is known to act on both leukocytes and ECs. However, in
vivo models have suggested that VEGF is principally an inducer of
noninflammatory angiogenesis.38 This may reflect a predominant
influence of VEGF on ECs rather than leukocytes. In our in vivo
experiments inhibition of HO-1 with SnPP shifted VEGF-driven
angiogenesis from noninflammatory to proinflammatory, as evi-
denced by the presence of a dense leukocytic infiltrate. This
suggests that induction of HO-1 by VEGF may act to both promote
endothelial proliferation and to limit proinflammatory leukocytic
infiltration. This hypothesis was confirmed in vivo by the effect of
the inhibitory anti-␤2 integrin mAb M18/2. M18/2 did not affect
VEGF-induced angiogenesis per se but completely abrogated the
VEGF-induced leukocytic infiltration observed in the presence of
HO-1 inhibitors. Moreover, when leukocyte infiltration was inhib-
ited by M18/2, the HO-1 antagonists ZnPP and SnPP inhibited Figure 4. Histologic and immunohistochemical analysis of Matrigel implants.
VEGF-induced angiogenesis confirming the role of HO-1 in this Representative hematoxylin and eosin–stained sections of Matrigel containing VEGF
(A) or VEGF plus SnPP (B) implanted in mice and excised 6 days after injection. A
process. Furthermore, experiments performed with aFGF, which dense inflammatory infiltrate around vessels was observed in plugs containing VEGF
did not induce HO-1, showed that blockade of HO-1 did not plus SnPP when compared with VEGF alone (original magnification ⫻ 160). (C-D)
influence aFGF-induced angiogenesis, thus demonstrating that the Double immunofluorescence staining for VWF (C) and MPO (D) showing the
presence of leukocytes around vessels in plugs with VEGF plus SnPP (original
HO-1 inhibitors themselves do not affect angiogenesis. These data
magnification ⫻ 250). (E-F) Representative hematoxylin and eosin–stained sections
support previous studies showing that VEGF and FGF induce of Matrigel containing LPS (E) or LPS in the presence of CoPP, which prevented
angiogenesis through distinct pathways.39 leukocyte-dependent angiogenesis (F) (original magnification ⫻ 160).
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

BLOOD, 1 FEBRUARY 2004 䡠 VOLUME 103, NUMBER 3 DUAL ROLE OF VEGF-INDUCED HO-1 IN ANGIOGENESIS 765

Figure 5. Effect of anti–␤2-integrin Ab M18/2 on angiogen-


esis and leukocyte infiltration. M18/2 or a isotype-
matched control mAb was added to Matrigel and injected
intraperitoneally every other day, as described in “Materials
and methods.” (A) Quantitative evaluation of neovessels
infiltrating Matrigel after treatment with SnPP, VEGF, or
VEGF plus SnPP in presence of M18/2 or a control mAb. The
results are expressed as percentage ⫾ SE of the vessel area
with respect to the total Matrigel area. (B) Quantitative
evaluation of inflammatory cells infiltrating Matrigel counted
in hematoxylin and eosin–stained sections. The results are
expressed as the mean ⫾ SE of cells/field (⫻ 400). Four
mice were used per condition in each experiment. ANOVA
with Dunnett multicomparison test was performed between
control Ab and M18/2 (*P ⬍ .05).

during VEGF-induced angiogenesis. Moreover, HO-1 may play a role recruitment represents an important mechanism for the control of
in different angiogenic settings, such as during hypoxia, where NO angiogenesis associated with inflammation.
production and endothelial NO synthase expression are suppressed.43 The data presented herein, and in other recent studies, suggests
Indeed, a role for HO-1 in angiogenesis during hypoxia has been that many anti-inflammatory and cytoprotective molecules act
suggested by the observation that HO-1 activation enhances VEGF through a mechanism dependent upon the expression of HO-1 and
production and that VEGF induced by hypoxia was down-regulated by that the products of HO-1 facilitate the protective effects.46 This is
HO-1 inhibitors but not by inhibitors of NO synthase.44 supported by the demonstration that HO-1 expression, or treatment
HO-1 breaks down heme to equimolar amounts of CO, biliver- with CO, reproduces these beneficial actions. Thus, HO-1 and CO
din, and free iron. Biliverdin is rapidly reduced to bilirubin by the have been shown to mediate the protective effects of interleukin-10
action of biliverdin reductase. Although less is known about the in LPS-induced septic shock in mice.49 Further examples of the
anti-inflammatory and cytoprotective effects of bilirubin, these pivotal role of HO-1 include its importance in the antiproliferative
have been clearly demonstrated.45,46 Hence, the relative contribu- effects of rapamycin,50 the anti-inflammatory actions of 15-Deoxy-
tion of bilirubin and CO in mediating the effects of VEGF-induced 12,14-prostaglandin J2,51 and the cytoprotective effects of heat-
HO-1 in angiogenesis merits further investigation. shock preconditioning.52
An anti-inflammatory effect of HO-1 has been demonstrated in vitro VEGF may act both as a potent proangiogenic factor and as a
and in vivo. The induction of HO-1 decreases EC expression of the leukocyte chemoattractant. We propose that HO-1 induced by
cellular adhesion molecule ICAM-122 and inhibits monocyte chemo- VEGF acts to promote angiogenesis while inhibiting the local
taxis.15 Pretreatment with HO-1 agonists attenuates tissue damage in a recruitment of leukocytes (Figure 7). Thus, in the setting of
variety of models including carrageenan-induced pleuritis, endotoxic inflammatory diseases associated with VEGF-driven angiogenesis,
shock, and oxidant-induced lung injury.14,47,48 Moreover, increased such as rheumatoid arthritis, the action of VEGF-induced HO-1
leukocyte adhesion to the vessel wall and spontaneous perivascular may be to maximize angiogenesis associated with the resolution of
infiltration of leukocytes into the liver, lungs, and kidneys was observed tissue injury while inhibiting leukocyte adhesion and transmigra-
in HO-1–deficient mice.19 To investigate the role of HO-1 in inflamma- tion. However, HO-1 induction may be impaired during inflamma-
tory angiogenesis we used LPS-conditioned Matrigel. In this model,36 tion, as recently reported in human chronic graft rejection.53 Thus,
neutrophils invade and begin to degrade the gel creating clefts and this is therapeutic induction of HO-1, or delivery of CO or bilirubin, may
followed by migration of macrophages. Growth factors, such as VEGF be beneficial in the treatment of chronic inflammatory diseases, not
and basic FGF (bFGF), released by neutrophils and macrophages only through their anti-inflammatory actions but also via a proan-
subsequently induce EC migration and tube formation. Thus, this model giogenic effect enhancing tissue repair.
of angiogenesis is directly dependent upon leukocyte migration. Treat-
ment of mice with the HO-1 inducer CoPP significantly inhibited
LPS-induced leukocyte infiltration and the subsequent angiogenic
response. This suggests that HO-1–mediated inhibition of leukocyte

Figure 6. Effect of HO-1 induction on inflammatory angiogenesis in vivo.


Matrigel containing LPS (10 ng/mL) plus CoPP (25 ␮M) was injected subcutaneously
into C57BL/6 mice. After 6 days plugs were explanted and fixed in formalin and
paraffin embedded. Quantification of neovascularization was performed on hematoxy-
lin and eosin–stained sections as described in “Materials and methods.” The results Figure 7. Proposed mechanism for the actions of HO-1 in vivo. HO-1 activation
are expressed as percentage ⫾ SE of the vessel area with respect to the total by VEGF favors endothelial cell proliferation while inhibiting leukocyte migration and
Matrigel area. Nine mice were used per condition in each experiment. ANOVA with activity, thus resulting in a predominantly noninflammatory angiogenesis. However,
Dunnett multicomparison test was performed between LPS and LPS plus CoPP when HO-1 is inhibited, there is increased leukocyte migration with subsequent local
(*P ⬍ .05). release of growth factors and induction of inflammatory angiogenesis.
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

766 BUSSOLATI et al BLOOD, 1 FEBRUARY 2004 䡠 VOLUME 103, NUMBER 3

References
1. Sunderkotter C, Steinbrink K, Goebeler M, Bhard- prevented by graft expression of anti-oxidant and 37. Fernandez M, Bonkovsky HL. Vascular endothe-
waj R, Sorg C. Macrophages and angiogenesis. anti-apoptotic genes. Nat Med. 1998;4:1392- lial growth factor increases heme oxygenase-1
J Leukoc Biol. 1994;55:410-422. 1396. protein expression in the chick embryo cho-
2. Polverini PJ. Role of the macrophage in angio- 21. Soares MP, Brouard S, Smith RN, Bach FH. rioallantoic membrane. Br J Pharmacol. 2003;
genesis-dependent diseases. In: Goldberg IP, Heme oxygenase-1, a protective gene that pre- 139:634-640.
Rosen ER, eds. Regulation of angiogenesis. Vol vents the rejection of transplanted organs. Immu- 38. Leung DW, Cachianes G, Kuang WJ, Goeddel
79. Basel, Switzerland: Birkhaüser Verlag; 1997: nol Rev. 2001;184:275-285. DV, Ferrara N. Vascular endothelial growth factor
11-28. 22. Wagener FA, da Silva JL, Farley T, et al. Differen- is a secreted angiogenic mitogen. Science. 1989;
3. Cassatella MA. Neutrophil-derived proteins: sell- tial effects of heme oxygenase isoforms on heme 246:1306-1309.
ing cytokines by the pound. Adv Immunol. 1999; mediation of endothelial intracellular adhesion 39. Giavazzi R, Sennino B, Coltrini D, et al. Distinct
73:369-509. molecule 1 expression. J Pharmacol Exp Ther. role of fibroblast growth factor-2 and vascular en-
4. Kasama T, Kobayashi K, Yajima N, et al. Expres- 1999;291:416-423. dothelial growth factor on tumor growth and an-
sion of vascular endothelial growth factor by sy- 23. Motterlini R, Foresti R, Intaglietta M, Winslow RM. giogenesis. Am J Pathol. 2003;162:1913-1926.
novial fluid neutrophils in rheumatoid arthritis NO-mediated activation of heme oxygenase: en- 40. Morita T, Perrella MA, Lee ME, Kourembanas S.
(RA). Clin Exp Immunol. 2000;121:533-538. dogenous cytoprotection against oxidative stress Smooth muscle cell-derived carbon monoxide is
5. Folkman J. Angiogenesis in cancer, vascular, to endothelium. Am J Physiol. 1996;270:H107- a regulator of vascular cGMP. Proc Natl Acad Sci
rheumatoid and other disease. Nat Med. 1995;1: H114. U S A. 1995;92:1475-1479.
27-31. 24. Polte T, Oberle S, Schroder H. The nitric oxide 41. Otterbein LE, Zuckerbraun BS, Haga M, et al.
6. Carmeliet P, Jain RK. Angiogenesis in cancer and donor SIN-1 protects endothelial cells from tumor Carbon monoxide suppresses arteriosclerotic
other diseases. Nature. 2001;407:249-257. necrosis factor-alpha-mediated cytotoxicity: pos- lesions associated with chronic graft rejection and
7. Ferrara N, Carver-Moore K, Chen H, et al. Het- sible role for cyclic GMP and heme oxygenase. J with balloon injury. Nat Med. 2003;9:183-190.
erozygous embryonic lethality induced by tar- Mol Cell Cardiol. 1997;29:3305-3310. 42. Ziche M, Morbidelli L, Choudhuri R, et al. Nitric
geted inactivation of the VEGF gene. Nature. 25. Józkowicz A, Huk I, Nigisch A, et al. Heme oxy- oxide synthase lies downstream from vascular
1996;380:439-442. genase and angiogenic activity of endothelial endothelial growth factor-induced but not basic
8. Barleon B, Sozzani S, Zhou D, et al. Migration of cells: stimulation by carbon monoxide and inhibi- fibroblast growth factor-induced angiogenesis.
human monocytes in response to vascular endo- tion by tin protoporphyrin-IX. Antioxid Redox Sig- J Clin Invest. 1997;99:2625-2634.
thelial growth factor (VEGF) is mediated via the nal. 2003;5:155-162. 43. McQuillan LP, Leung GK, Marsden PA, Kostyk
VEGF receptor flt-1. Blood. 1996;87:3336-3343. 26. Deramaudt BM, Braunstein S, Remy P, Abraham SK, Kourembanas S. Hypoxia inhibits expression
9. Fava RA, Olsen NJ, Spencer-Green G, et al. Vas- NG. Gene transfer of human heme oxygenase of eNOS via transcriptional and post-transcrip-
cular permeability factor/endothelial growth factor into coronary endothelial cells potentially pro- tional mechanisms. Am J Physiol. 1994;267:
(VPF/VEGF): accumulation and expression in motes angiogenesis. J Cell Biochem. 1998;68: H1921-H1927.
human synovial fluids and rheumatoid synovial 121-127. 44. Dulak J, Jozkowicz A, Foresti R, et al. Heme oxy-
tissue. J Exp Med. 1994;180:341-346. 27. Ades EW, Candal FJ, Swerlick RA, et al. genase activity modulates vascular endothelial
10. Koch AE, Harlow LA, Haines GK, et al. Vascular HMEC-1: establishment of an immortalized hu- growth factor synthesis in vascular smooth
endothelial growth factor: a cytokine modulating man microvascular endothelial cell line. J Invest muscle cells. Antioxid Redox Signal. 2002;4:229-
endothelial function in rheumatoid arthritis. J Im- Dermatol. 1992;99:683-690. 240.
munol. 1994;152:4149-4156. 28. Wellicome SM, Thornhill MH, Pitzalis C, et al. A 45. Nakagami T, Toyomura K, Kinoshita T, Morisawa
11. Luttun A, Tjwa M, Moons L, et al. Revasculariza- monoclonal antibody that detects a novel antigen S. A beneficial role of bile pigments as an endog-
tion of ischemic tissues by PlGF treatment, and on endothelial cells that is induced by tumor ne- enous tissue protector: anti-complement effects
inhibition of tumor angiogenesis, arthritis and ath- crosis factor, IL-1, or lipopolysaccharide. J Immu- of biliverdin and conjugated bilirubin. Biochim
erosclerosis by anti-Flt1. Nat Med. 2002;8:831- nol. 1990;144:2558-2565. Biophys Acta. 1993;1158:189-193.
840. 29. Mason JC, Yarwood H, Sugars K, et al. Induction 46. Otterbein LE, Soares MP, Yamashita K, Bach FH.
12. Zhao Q, Egashira K, Inoue S, et al. Vascular en- of decay-accelerating factor by cytokines or the Heme oxygenase-1: unleashing the protective
dothelial growth factor is necessary in the devel- membrane-attack complex protects vascular en- properties of heme. Trends Immunol. 2003;24:
opment of arteriosclerosis by recruiting/activating dothelial cells against complement deposition. 449-455.
monocytes in a rat model of long-term inhibition Blood. 1999;94:1673-1682. 47. Otterbein L, Sylvester SL, Choi AM. Hemoglobin
of nitric oxide synthesis. Circulation. 2002;105: 30. Foresti R, Sarathchandra P, Clark JE, Green CJ, provides protection against lethal endotoxemia in
1110-1115. Motterlini R. Peroxynitrite induces haem oxygen- rats: the role of heme oxygenase-1. Am J Respir
13. Soares MP, Lin Y, Anrather J, et al. Expression of ase-1 in vascular endothelial cells: a link to apo- Cell Mol Biol. 1995;13:595-601.
heme oxygenase-1 can determine cardiac xeno- ptosis. Biochem J. 1999;339:729-736. 48. Otterbein LE, Kolls JK, Mantell LL, et al. Exog-
graft survival. Nat Med. 1998;4:1073-1077. 31. Mason JC, Lidington EA, Yarwood H, Lublin DM, enous administration of heme oxygenase-1 by
14. Willis D, Moore AR, Frederick R, Willoughby DA. Haskard DO. Induction of endothelial cell decay- gene transfer provides protection against hyper-
Heme oxygenase: a novel target for the modula- accelerating factor by vascular endothelial growth oxia-induced lung injury. J Clin Invest. 1999;103:
tion of the inflammatory response. Nat Med. factor: a mechanism for cytoprotection against 1047-1054.
1996;2:87-90. complement-mediated injury during inflammatory 49. Lee TS, Chau LY. Heme oxygenase-1 mediates
15. Ishikawa K, Navab M, Leitinger N, Fogelman AM, angiogenesis. Arthritis Rheum. 2001;44:138-150. the anti-inflammatory effect of interleukin-10 in
Lusis AJ. Induction of heme oxygenase-1 inhibits 32. Hiraoka N, Allen E, Apel IJ, Gyetko MR, Weiss mice. Nat Med. 2002;8:240-246.
the monocyte transmigration induced by mildly SJ. Matrix metalloproteinases regulate neovascu- 50. Visner GA, Lu F, Zhou H, et al. Rapamycin in-
oxidized LDL. J Clin Invest. 1997;100:1209-1216. larization by acting as pericellular fibrinolysins. duces heme oxygenase-1 in human pulmonary
16. Maines MD. The heme oxygenase system: a Cell. 1998;95:365-377. vascular cells: implications in the antiproliferative
regulator of second messenger gases. Annu Rev 33. Bussolati B, Biancone L, Cassoni P, et al. PAF response to rapamycin. Circulation. 2003;107:
Pharmacol Toxicol. 1997;37:517-554. produced by human breast cancer cells promotes 911-916.
17. Balla J, Jacob HS, Balla G, Nath K, Vercellotti migration and proliferation of tumor cells and neo- 51. Lee TS, Tsai HL, Chau LY. Induction of heme oxy-
GM. Endothelial cell heme oxygenase and ferritin angiogenesis. Am J Pathol. 2000;157:1713-1725. genase-1 expression in murine macrophages is
induction by heme proteins: a possible mecha- 34. Biancone L, De Martino A, Orlandi V, et al. Devel- essential for the anti-inflammatory effect of low
nism limiting shock damage. Trans Assoc Am opment of inflammatory angiogenesis by local dose 15-deoxy-Delta 12,14-prostaglandin J2.
Physicians. 1992;105:1-6. stimulation of Fas in vivo. J Exp Med. 1997;186: J Biol Chem. 2003;278:19325-19330.
18. Yachie A, Niida Y, Wada T, et al. Oxidative stress 147-152. 52. Redaelli CA, Tian YH, Schaffner T, et al. Ex-
causes enhanced endothelial cell injury in human 35. Wagener FA, Van Beurden HE, Von Den Hoff JW, tended preservation of rat liver graft by induction
heme oxygenase-1 deficiency. J Clin Invest. Adema GJ, Figdor CG. The heme-heme oxygen- of heme oxygenase-1. Hepatology. 2002;35:
1999;103:129-135. ase system: a molecular switch in wound healing. 1082-1092.
19. Poss KD, Tonegawa S. Heme oxygenase 1 is re- Blood. 2003;102:521-528. 53. Avihingsanon Y, Ma N, Csizmadia E, et al. Ex-
quired for mammalian iron reutilization. Proc Natl 36. Benelli R, Morini M, Carrozzino F, et al. Neutro- pression of protective genes in human renal allo-
Acad Sci U S A. 1997;94:10919-10924. phils as a key cellular target for angiostatin: impli- grafts: a regulatory response to injury associated
20. Hancock WW, Buelow R, Sayegh MH, Turka LA. cations for regulation of angiogenesis and inflam- with graft rejection. Transplantation. 2002;73:
Antibody-induced transplant arteriosclerosis is mation. FASEB J. 2002;16:267-269. 1079-1085.
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

2004 103: 761-766


doi:10.1182/blood-2003-06-1974 originally published online
October 2, 2003

Bifunctional role for VEGF-induced heme oxygenase-1 in vivo: induction


of angiogenesis and inhibition of leukocytic infiltration
Benedetta Bussolati, Asif Ahmed, Helen Pemberton, R. Clive Landis, Francesco Di Carlo, Dorian O.
Haskard and Justin C. Mason

Updated information and services can be found at:


http://www.bloodjournal.org/content/103/3/761.full.html
Articles on similar topics can be found in the following Blood collections
Hemostasis, Thrombosis, and Vascular Biology (2485 articles)
Phagocytes (969 articles)
Plenary Papers (483 articles)

Information about reproducing this article in parts or in its entirety may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests

Information about ordering reprints may be found online at:


http://www.bloodjournal.org/site/misc/rights.xhtml#reprints

Information about subscriptions and ASH membership may be found online at:
http://www.bloodjournal.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society
of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
Copyright 2011 by The American Society of Hematology; all rights reserved.

You might also like