ArticlePDF Available

Complete structure of an increasing capillary permeability protein (ICPP) purified from Vipera lebetina venom - ICPP is angiogenic via vascular endothelial growth factor receptor signaling

Authors:

Abstract and Figures

The partial sequence of the increasing capillary permeability protein (ICPP) purified from Vipera lebetina venom revealed a strong homology to vascular endothelial growth factor (VEGF)-A. We now report its complete amino acid sequence determined by Edman degradation and its biological effects on mouse and human vascular endothelial cells. ICPP is a homodimeric protein linked by cysteine disulfide bonds of 25115 Da revealed by mass spectrometry. Each monomer is composed of 110 amino acids including eight cysteine residues and a pyroglutamic acid at the N-terminal extremity. ICPP shares 52% sequence identity with human VEGF but lacks the heparin binding domain and Asn glycosylation site. Besides its strong capillary permeability activity, ICPP was found to be a potent in vitro angiogenic factor when added to mouse embryonic stem cells or human umbilical vein endothelial cells. ICPP was found to be as potent as human VEGF165 in activating p42/p44 MAPK, in reinitiation of DNA synthesis in human umbilical vein endothelial cells, and in promoting in vitro angiogenesis of mouse embryonic stem cells. All these biological actions, including capillary permeability in mice, were fully inhibited by 1 microm of a new specific VEGF receptor tyrosine kinase inhibitor (ZM317450) from AstraZeneca that belongs to the anilinocinnoline family of compounds. Indeed, up to a 30 times higher concentration of inhibitor did not affect platelet-derived growth factor, epidermal growth factor, FGF-2, insulin, alpha-thrombin, or fetal calf serum-induced p42/p44 MAPK and reinitiation of DNA synthesis. Therefore, we conclude that this venom-derived ICPP exerts its biological action (permeability and angiogenesis) through activation of VEGF receptor signaling (VEGF-R2 and possibly VEGF-R1).
Content may be subject to copyright.
Complete Structure of an Increasing Capillary Permeability Protein
(ICPP) Purified from Vipera lebetina Venom
ICPP IS ANGIOGENIC VIA VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR SIGNALING*
Received for publication, March 6, 2002, and in revised form, May 13, 2002
Published, JBC Papers in Press, May 20, 2002, DOI 10.1074/jbc.M202202200
Ammar Gasmi‡
, Christine Bourcier§, Zohra Aloui‡, Najet Srairi‡, Sandrine Marchetti§,
Clotilde Gimond§, Stephen R. Wedge
, Laurent Hennequin
, and Jacques Pouysse´ gur§**
From the Laboratoire des Venins et Toxines, Institut Pasteur de Tunis, B. P. 74, 1002 Tunis-Belvede`re, Tunisia,
Cancer and Infection Research, AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, United Kingdom,
and the §Institute of Signaling, Developmental Biology and Cancer Research, CNRS UMR 6543, Centre A. Lacassagne,
33 Avenue Valombrose, 06189 Nice, France
The partial sequence of the increasing capillary per-
meability protein (ICPP) purified from Vipera lebetina
venom revealed a strong homology to vascular endothe-
lial growth factor (VEGF)-A. We now report its complete
amino acid sequence determined by Edman degradation
and its biological effects on mouse and human vascular
endothelial cells. ICPP is a homodimeric protein linked
by cysteine disulfide bonds of 25115 Da revealed by mass
spectrometry. Each monomer is composed of 110 amino
acids including eight cysteine residues and a pyroglu-
tamic acid at the N-terminal extremity. ICPP shares 52%
sequence identity with human VEGF but lacks the hep-
arin binding domain and Asn glycosylation site. Besides
its strong capillary permeability activity, ICPP was
found to be a potent in vitro angiogenic factor when
added to mouse embryonic stem cells or human umbili-
cal vein endothelial cells. ICPP was found to be as po-
tent as human VEGF165 in activating p42/p44 MAPK, in
reinitiation of DNA synthesis in human umbilical vein
endothelial cells, and in promoting in vitro angiogenesis
of mouse embryonic stem cells. All these biological ac-
tions, including capillary permeability in mice, were
fully inhibited by 1
M of a new specific VEGF receptor
tyrosine kinase inhibitor (ZM317450) from AstraZeneca
that belongs to the anilinocinnoline family of com-
pounds. Indeed, up to a 30 times higher concentration of
inhibitor did not affect platelet-derived growth factor,
epidermal growth factor, FGF-2, insulin,
-thrombin, or
fetal calf serum-induced p42/p44 MAPK and reinitiation
of DNA synthesis. Therefore, we conclude that this
venom-derived ICPP exerts its biological action (perme-
ability and angiogenesis) through activation of VEGF
receptor signaling (VEGF-R2 and possibly VEGF-R1).
Angiogenesis is a tightly regulated process occurring physi-
ologically during embryonic development, during the men-
strual cycle, and in wound healing. It is also associated with a
number of pathological situations including diabetic retino-
pathy, inflammation, brain edema following ischemic stroke,
solid tumor growth, and metastasis. A number of polypeptide
growth factors have been demonstrated to induce and regulate
angiogenesis in vivo, among them fibroblast growth factor
(FGF),
1
platelet-derived growth factor (PDGF), epidermal
growth factor (EGF), and vascular endothelial growth factor
(VEGF)-A (1, 2). Although the mode of activation at the recep-
tor level differs, all these mitogens activate the ubiquitously
expressed isoforms of mitogen-activated protein kinases re-
ferred to as p42/p44 MAPK or Erk, two essential transducers of
growth, survival, and differentiation signals.
VEGF was the first mitogenic growth factor proven to have
endothelial cell specificity and to be critical for blood vessel
formation. The vascular endothelium-specific growth factors
are now known to include five members of the VEGF family,
four members of the angiopoietin family, and at least one
member of the large ephrin family (3). VEGF is a multifunc-
tional cytokine that is produced by virtually every tissue and
overexpressed upon hypoxic stress and oncogenic transforma-
tion (4). It is a homodimeric glycoprotein, expressed as several
spliced variants; the major forms contain 121, 165, 189, and
206 amino acids. VEGF121 differs from the larger VEGF iso-
forms in that it is the only VEGF type that does not possess
heparin binding ability (5). These isoforms act in a coordinate
fashion to recruit and expand the tumor vasculature (6). The
main receptors that seem to be involved in initiating signal
transduction cascades in response to VEGFs comprise a family
of closely related receptor tyrosine kinases that are expressed
almost exclusively on neovasculature and on the tumor endo-
thelium. They consist of three members now termed VEGFR-1
(Fms-like tyrosine kinase, Flt-1), VEGFR-2 (kinase insert
domain-containing receptor (KDR)), and the VEGF-C and -D
receptor, VEGFR-3 (known previously as Flt-3). Some other
accessory receptors (neuropilins) that seem to be involved pri-
marily in modulating binding to the main receptors have also
been reported (7). Their roles in signaling have not yet been
fully elucidated (3). VEGFR-2, however, via activation of in-
trinsic tyrosine kinase activity, appears to mediate all the
* This work was supported by grants from the Centre National de la
Recherche Scientifique, Le Ministe`re de l’Education, de la Recherche et
de la Technologie, La Ligue Nationale Contre le Cancer (e´quipe labe-
lise´e J. P.). The costs of publication of this article were defrayed in part
by the payment of page charges. This article must therefore be hereby
marked advertisement in accordance with 18 U.S.C. Section 1734
solely to indicate this fact.
To whom correspondence may be addressed. Fax: 216-71-791833;
E-mail: ammar.gasmi@pasteur.rns.tn.
** To whom correspondence may be addressed. Fax: 33-492-03-1225;
pouysseg@unice.fr.
1
The abbreviations used are: FGF, fibroblast growth factor; VEGF,
vascular endothelial growth factor; svVEGF, snake venom VEGF;
VEGFR, VEGF receptor; VTKI, VEGF receptor tyrosine kinase inhibi-
tor; PDGF, platelet-derived growth factor; ICPP, increasing capillary
permeability protein; KDR, kinase insert domain-containing receptor;
MAPK, mitogen-activated protein kinase; ERK, extracellular signal-
regulated kinase; MEK, MAPK/ERK kinase; ES, embryonic stem; HF,
hypotensive factor; SFM, serum-free media; PBS, phosphate-buffered
saline; HPLC, high pressure liquid chromatography.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 277, No. 33, Issue of August 16, pp. 29992–29998, 2002
© 2002 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
This paper is available on line at http://www.jbc.org29992
by guest on July 17, 2015http://www.jbc.org/Downloaded from
major actions of VEGF: capillary permeability, chemotaxis, cell
survival, and cell division (8).
The possibility that vascular growth factors may help pre-
vent or repair damaged and leaky vessels offers therapeutic
hope for ischemic diseases, diabetic retinopathy, or inflamma-
tory setting (9, 10). In opposition, new antitumoral approaches
targeting the tumor vasculature via inhibition of VEGF signal-
ing are actively being developed; they include neutralizing
anti-VEGF antibodies, anti-VEGF receptor antibodies, soluble
VEGF receptors, antisense VEGF techniques, and VEGF re-
ceptor tyrosine kinase inhibitors (11).
We have previously isolated a protein from Vipera lebetina
venom based on its potent ability to increase capillary perme-
ability. The partial sequence of this protein, referred to as
increasing capillary permeability protein (ICPP), revealed a
VEGF-like structure (12). In this report, we present the com-
plete amino acid sequence of ICPP and demonstrate that this
venom related-VEGF is capable of inducing p42/p44 MAPK
activity and DNA synthesis in human umbilical vein endothe-
lial cells (HUVEC) and of promoting in vitro angiogenesis.
Interestingly, all these ICPP-induced biological actions are
fully inhibited by a new VEGF receptor tyrosine kinase inhib-
itor, as presented here.
EXPERIMENTAL PROCEDURES
Materials—ICPP was purified from V. lebetina venom (12). Reverse
phase analytical columns C8 (5
m, 4.6 250 mm) were purchased
from Beckman Instruments. Endoproteinases Asp-N, Arg-C, and Lys-C
were of sequencing grade and were obtained from Roche Molecular
Biochemicals. Recombinant human FGF-2 and VEGF165 were pro-
duced in our laboratory from Escherichia coli and Pichia pastoris,
respectively, after purification on heparin binding affinity columns,
whereas human recombinant PDGF
and EGF were from Sigma. All
other reagents used were of analytical grade from commercial sources.
The AstraZeneca compound 4-fluro-5-{[6-methoxy-7-(2-methoxye-
thoxy) cinnolin-4-yl]amino}-2-methylphenol (ZM317450), referred to
here as VEGF receptor tyrosine kinase inhibitor (VTKI), belongs to the
anilinocinnoline family of compounds. It was prepared according to the
protocol presented in the patent WO 9734876 by A. P. Thomas and
L. F. Hennequin.
Reduction and Alkylation of ICPP—Reduction was performed by
incubating ICPP for1hat37°Cin6
M guanidine-HCl, 0.5 M Tris-HCl,
2m
M EDTA, 1.4
M DTT (dithiothreitol), pH 7.5. Then, alkylation
occurred following addition of 4-vinylpyridine (9
mol final concentra-
tion) and terminated after 5 min by addition of DTT to a final concen-
tration of 14
M. The mixture was desalted on a reverse phase HPLC on
a C8 column. Solvents A and B were 0.1% trifluoroacetic acid (v/v) and
0.1% trifluoroacetic acid (v/v) in 100% acetonitrile, respectively. Protein
was eluted from the column by a linear gradient of 10 80% of solvent
B in 60 min at a flow rate of 1 ml/min and monitored at 214 nm.
Enzymatic Digestions of ICPP—Digestions of reduced and alkylated
ICPP were carried out by the endoproteinases Arg-C, Lys-C, or Asp-N.
The denatured protein was incubated in appropriate buffer medium.
The suitable time, temperature, enzyme/substrate ratio, and termina-
tion of the reactions were performed according to the manufacturers
instructions. Urea (2
M) was added to the reaction mixture to ensure
solubility. The resulting peptides were subjected to a reverse phase
chromatography on C8 column and eluted by an increasing gradient of
10 60% of solvent B in 60 min. Effluent was continuously monitored at
214 nm, and peaks were collected manually and subjected to sequence
analysis.
Amino Acid Analysis and Sequence Comparison—The sequences of
N-terminal subunits were determined after chemically unblocking with
HCl in anhydrous methanol (13) by Edman degradation with an Ap-
plied Biosystems 470A liquid-phase sequencer equipped with on-line
phenylthiohydantoin reverse HPLC using an RP18 column. The se-
quences of peptides obtained from enzymatic digestions of reduced and
alkylated ICPP were performed as described previously. A search for
similar proteins was performed following computer analysis with the
BLAST data base search program.
Mass Spectral Analysis—Determination of the molecular mass of
native ICPP was carried out on a Voyager DE-RP matrix-assisted laser
desorption ionization time-of-flight mass spectrometer (PerSeptive Bio-
systems, Inc., Framingham, MA). A sinapinic acid matrix at 10 mg/ml
in 50% acetonitrile/50% H
2
O/0.1% trifluoroacetic acid was used.
Culture, MAPK Activity, and DNA Replication of HUVECs—
HUVECs were isolated from umbilical cord veins by collagenase perfu-
sion as described previously (14) and cultivated in SFM (Invitrogen)
supplemented with 20% fetal calf serum, 20 ng of FGF-2, and 10 ng of
EGF/ml (EGF provided by Sigma). For reinitiation of DNA synthesis,
HUVEC were serum-starved for 24 h, trypsinized, and replated in
24-well plates coated with gelatin at 50,000 cells/well in 0.5 ml of SFM.
Six h later, cells were incubated in 0.5 ml of SFM containing 1
Ci of
tritiated thymidine and stimulated with growth factors or ICPP in the
presence or absence of various concentrations of VTKI. After 24 h,
TCA-insoluble material was collected, and radioactivity was counted.
For the measurement of p42/p44 MAPK activation, HUVEC were
serum-starved for 24 h, trypsinized, and replated in 12-well plates at
10
5
cells in 1 ml of SFM. After 6 h, cells were stimulated with growth
factors or ICPP with or without VTKI; VTKI was preincubated 15 min
prior to stimulation. Stimulation was arrested after 10 min of stimula-
tion with cold PBS washing, and cells were harvested in 100
lof
Laemmli sample buffer followed by protein separation on SDS-10%
polyacrylamide gel electrophoresis (15). Western blotting was per-
formed as described previously (16). The blots were incubated with a
1/5000 dilution of the anti-phospho p42/p44 MAPK monoclonal anti-
body (Sigma).
Receptor Tyrosine Kinase Assays—The ability of VTKI to inhibit the
kinase activity associated with the VEGF receptors R1 (Flt-1) and R2
(KDR), the FGF receptor FGFR1, and the EGF receptor was determined
using a previously described enzyme-linked immunosorbent assay (17).
Receptor tyrosine kinases used in isolated enzyme assays were gener-
ated as insect cell lysates following cell infection with recombinant
baculoviruses containing kinase domains.
Briefly, compounds were incubated with enzyme, 10 m
M MnCl
2
, and
2m
M ATP in 96-well plates coated with a poly(Glu,Ala,Tyr) 6:3:1
random copolymer substrate (Sigma). The ATP concentration used was
at, or just below, the respective K
m
value. Phosphorylated tyrosine was
detected by sequential incubation with mouse IgG anti-phosphotyrosine
antibody (Upstate Biotechnology Inc., Lake Placid, NY), a horse radish
peroxidase-linked sheep anti-mouse Ig antibody (Amersham Bio-
sciences), and 2,2-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid
(Roche Molecular Biochemicals). Microcal Origin software (Version
3.78, Microcal Software Inc., Northhampton, MA) was used to interpo-
late IC
50
values by non-linear regression (four-parameter logistic
equation).
Protein Determination—Protein concentration was determined by
the procedure of Lowry et al. (18) with the folin phenol reagent and with
bovine serum albumin as a standard.
In Vitro Angiogenesis—Culture and differentiation of embryonic
stem (ES) cells was used as an angiogenesis test in vitro. Mouse 129/
OLA ES cells (E14Tg2A.IV clone, initially provided by Dr. M. Hooper,
Edinburgh, UK and subcloned by Dr. A. Smith, Edinburgh, UK) were
grown in Dulbeccos modified Eagles medium with Glutamax-1 and
sodium pyruvate (Invitrogen) containing 10% fetal calf serum
(Dutscher, Brumath, France), 50 units/ml penicillin, 50
g/ml strepto-
mycin, 0.1 m
M
-mercaptoethanol, and non-essential amino acids (all
reagents from Invitrogen). They were kept undifferentiated by the
addition of either 10
3
units/ml recombinant leukemia inhibitory factor
purchased from Sigma or 100 units/ml leukemia inhibitory factor pro-
duced in COS cells as described previously (19).
For differentiation, ES cells were cultured in hanging drops as de-
scribed previously (20) with some modifications. Briefly, ES cells were
detached in trypsin/EDTA and aggregated into embryoid bodies in the
above described Dulbeccos modified Eagles medium lacking supple-
mental leukemia inhibitory factor. Aggregation was performed in 20-
l
drops hanging from the lids of bacteriological Petri dishes and contain-
ing 800 cells. The lids were then placed over PBS-filled dishes and
incubated at 37 °C. This was designated as day 0. At day 3, the result-
ing embryoid bodies were then transferred to gelatin-coated 24-well
tissue culture plates. When indicated, the medium was supplemented
with human 10 ng/ml rVEGF165 (Sigma) or various concentrations of
purified ICPP. After 12 days of differentiation, embryoid bodies were
fixed in 4% paraformaldehyde (in PBS) for 20 min at room temperature,
permeabilized with 0.2% Triton X-100 for 5 min, and blocked in PBS
containing 10% fetal calf serum for 2 h. Cells were then incubated with
a rat anti-CD31 antibody (clone MEC13.3; BD PharMingen) for1hat
room temperature. Staining was revealed by incubating first with a
biotin-conjugated donkey anti-rat antibody (Jackson ImmunoResearch
Laboratories, West Grove, PA), and second with Alexa Fluor-conjugated
Structure and Biology of Vipera lebetina Venom VEGF 29993
by guest on July 17, 2015http://www.jbc.org/Downloaded from
streptavidin (Molecular Probes, Eugene, OR). Preparations were
mounted in PBS:glycerol (1:9) and viewed under a Leica microscope.
Vascular Permeability AssaysCapillary permeability activities
were tested using slight modifications of the Miles permeability assay
(21) in female MF1 mice (Harlan France; 34 weeks of age). Prior to the
assay, mice were injected intravenously (tail vein) with 50
lofa1%
Evans Blue solution. Immediately afterward, 50
l of PBS, or an
equivalent volume of various concentrations of VEGF165 or ICPP in
PBS, were injected intradermally on the back of mice. To evaluate the
inhibitory action of VTKI, the compound (50
lat10
M in PBS) was
preinjected (intradermally), 60 min prior to administration of a cyto-
kine, at the same site. Animals were sacrificed 30 min after cytokine
injection, and the dorsal epithelium was dissected and photographed.
To quantify the leakage of the dye, the skin patches were eluted at 56 °C
with formalin and measured with a spectrophotometer (A
600
).
RESULTS
Amino Acid Sequence and Molecular Mass Determination of
ICPPReduced and alkylated ICPP was subjected to separate
enzymatic digestions by Asp-N, Lys-C, and Arg-C. These diges-
tions yielded many peptides designated N, L, and R, respec-
tively. The resulting fragments were purified by reverse phase
chromatography on a C8 column (data not shown), numbered
according to their order of elution from the column, and sub-
jected to sequencing by Edman degradation. Unblocking of the
N-terminal amino acid sequence was performed as described
under Experimental Procedures, and 52 residues were clearly
identified. ICPP was found to be a homodimeric protein, each
monomer consists of 110 residues with 8 half-cystines/mol, and
the entire sequence and the sequence of the peptide fragments
determined are presented in Fig. 1. The amino acid data were
corroborated with mass spectrometric data. The molecular
mass of ICPP calculated from the sequence is 25099 Da and is
in good agreement with that determined by mass spectrometry,
which is 25115 Da.
Sequence Comparison of ICPP with Hypotensive Factor (HF),
Snake Venom VEGFs (svVEGFs), and Human VEGFThe
search for similar proteins by computer analysis of the se-
quence data of ICPP revealed that this protein had a high
similarity with the VEGF of many animal species and some
similarity with PDGF. Fig. 2 shows the alignment of amino
acid sequences of ICPP with those of human VEGF (22), an HF
purified from Vipera aspis aspis (23), and svVEGF derived from
Bothrops insularis (Fig. 2, Bins svVEGF) and Bothrops jara-
raca (Fig. 2, Bjar svVEGF) venoms (24). Based on a BLAST
search, ICPP shares sequence identities with these proteins
with rates of 52, 95, 65, and 67%, respectively. Sequence data
of ICPP exhibit many residues identified by site-directed mu-
tagenesis of human VEGF as being important for receptor
binding (25). It is interesting to note that most of the residues
implicated as being important for VEGF binding to VEGFR-1/
R-2 were also conserved (26). The heparin binding site in hu-
man VEGF has been localized to the C-terminal 55 residues of
the VEGF165 spliced form (27). Interestingly, ICPP lacks the
heparin binding domain, and unlike all VEGF isoforms, it does
not contain any Asn-linked glycosylation site defined by the
consensus sequence: NX(S/T). Perhaps this subtle difference
with VEGF may result in distinct biological activities.
ICPP Stimulates p42/p44 MAP Kinase Activity in HUVEC,
Implication of VEGF ReceptorsSince ICPP and human VEGF
have a high sequence similarity, it was appropriate to compare
the effect of these proteins on signaling and angiogenic poten-
tial using HUVEC. The Raf-1MEKp42/p44 MAPK has been
demonstrated to rapidly convey growth and survival signals
from a variety of receptor tyrosine kinases. We therefore tested
the biological action of ICPP on HUVEC that are responsive to
VEGF and FGF for growth and differentiation. Fig. 4A shows
that like human VEGF165 and FGF-2, V. lebetina ICPP stim-
ulates p42/p44 MAP kinases in HUVEC. This action is rapid,
detected within 2 min, peaks around 1015 min, and decreases
to basal level after4hofstimulation (data not shown). This
temporal action as well as the maximal intensity of MAPK
activation parallel that observed with human VEGF165. In the
same cells, however, p42/p44 MAPK activation could reach 35
times higher levels in response to FGF-2. We then compared
the potency of ICPP and human VEGF165 using MAP kinase
activation as a reporter system. On a molar basis, ICPP puri-
fied to homogeneity is at least 1.5 times more potent than
human recombinant VEGF165 obtained from Sigma or freshly
prepared from P. pastoris and purified on heparin affinity
columns (data not shown).
We then sought to determine whether ICPP can signal
through endogenous VEGF receptors. To answer this point, we
exploited the specificity of tyrosine kinase inhibitors developed
by AstraZeneca. These compounds attain selectivity by compet-
ing with the non-conserved hydrophobic pocket of the ATP
binding site in kinases. The inhibitor used in this study
(ZM317450) is a new molecule of the anilinocinnoline family of
compounds (Fig. 3), referred to here as VTKI. In isolated en-
zyme assays, VTKI is a potent inhibitor of VEGFR-2 tyrosine
kinase (IC
50
50 nM) with submicromolar activity versus the
kinase activity of VEGFR-1 (Table I). In comparison with its
inhibitory activity versus VEGFR-2 tyrosine kinase, VTKI
demonstrated 2000-fold selectivity versus that associated
with FGFR1 and EGFR (Table I). Selectivity is also conserved
in endothelial cells (HUVEC); 10
M VTKI ablates fully ICPP-
and VEGF-induced MAPK activation but does not affect that
stimulated by FGF-2 (Fig. 4A). Inhibition of ICPP-stimulated
MAPK activation was even found to be complete at 1
M. The
dose response of VTKI was explored on HUVEC stimulated
with equally potent growth factors: VEGF (30 ng/ml) or ICPP
(10 ng/ml). The VTKI concentration inhibiting half of the re-
sponse (EC
50
) is 100 nM for both VEGF (Fig. 4B) and ICPP
(data not shown). Next we explored the specificity of VTKI by
evaluating its action on the ER22, a derivative of the fibroblas-
tic cell line CCL39 that express several receptor tyrosine
kinases, PDGF-R, FGF-R, and EGF-R (28). Fig. 5 shows that
MAPK stimulation by all agonists remains unaffected by VTKI
at a concentration 300 times greater than the EC
50
for inhibi
-
tion MAPK stimulated by VEGF. In addition, we showed that
VTKI did not affect MAP kinase activation in response to the G
protein-coupled receptor agonist,
-thrombin, or fetal calf
serum (Fig. 5). These results demonstrate that VTKI is not a
FIG.1. Complete amino acid se-
quence of ICPP. The N-terminal amino
acid sequence was determined after
chemical unblocking of the intact protein.
Peptides obtained from enzymatic diges-
tions with Asp-N, Lys-C and Arg-C were
designated A, L, and R, respectively.
Structure and Biology of Vipera lebetina Venom VEGF29994
by guest on July 17, 2015http://www.jbc.org/Downloaded from
general blocker of MAPK signaling and highlight the specificity
toward VEGF receptor kinase. Collectively, these data suggest
that ICPP mediates its effect on MAPK signaling via VEGF
receptor signaling.
ICPP Is a Potent Angiogenic Factor through VEGF Receptor
SignalingSince we demonstrated in the previous section that
ICPP can signal through VEGF receptors, the next question
was to investigate whether, besides MAPK signaling, ICPP is
capable of mimicking all the biological actions of VEGF. We
first tested its capacity to induce angiogenesis in two in vitro
systems, DNA proliferation in HUVEC and growth and differ-
entiation of mouse vascular endothelial cells. Fig. 6A shows
that serum-starved HUVEC are able to reinitiate DNA synthe-
sis in response to VEGF165 or ICPP. FGF-2, a more potent
agonist for stimulation of MAPK signaling, is also a much more
potent mitogen for HUVEC (Fig. 6A). In our assays, DNA
synthesis was stimulated 3-fold above basal with ICPP and
510-fold above basal with FGF-2. Interestingly the mitogenic
action of ICPP or VEGF is fully suppressed by VTKI, whereas the
activity of FGF is practically unaffected. We regularly observed a
10 15% inhibition of FGF-induced mitogenic action that we at-
tributed to the autocrine production of VEGF in response to FGF,
a proposal consistent with a previous observation (29). This bio-
logical assay was then used to compare the sensitivity of ICPP
and VEGF toward the VTKI. As shown in Fig. 6B, reinitiation of
DNA synthesis was fully suppressed at 100 n
M of VTKI, indicat-
ing that reinitiation of DNA synthesis appears slightly more
sensitive than the MAPK inhibition. The VTKI dose-response
curves for ICPP and VEGF are identical, suggesting that ICPP
and VEGF are using the same receptor system for in vitro angio-
genesis. In addition, we concluded that ICPP triggers MAPK
activation and reinitiation of DNA synthesis exclusively via
VEGF-R2 signaling. Indeed, expression of VEGF-R2 alone in the
fibroblastic cell line CCL39, devoid of VEGF-R, was sufficient to
elicit MAPK activation and DNA synthesis in response to ICPP.
This action was fully abolished by VTKI (data not shown).
The second in vitro system exploited to assess ICPP-induced
angiogenesis is more demanding since it records the capacity of
a cytokine to promote the proliferation of embryonic vascular
endothelial cell precursors and their capacity to differentiate
into a vascular network. In comparison with FGF-2, VEGF has
been reported to be an extremely potent cytokine in this bio-
logical assay (30). Interestingly, as shown in Fig. 6C, ICPP is a
potent molecule that, like VEGF, can induce a vascular-like
network positive for the vascular specific marker CD31. Al-
though this assay does not allow an accurate quantitation,
ICPP has always been more potent than VEGF165 in estab-
lishing a vascular differentiated network. This ICPP-induced
growth and differentiation is again fully prevented by the
addition of VTKI (Fig. 6C).
ICPP-stimulated Increased Capillary Permeability Is Medi-
ated through VEGF Receptor SignalingICPP was isolated by
its capacity to stimulate capillary permeability on mice (12),
and indeed, when compared with VEGF, the best permeability
factor known to date, ICPP was found to be an extremely potent
permeability factor (Fig. 7A). This biological activity was ex-
amined using the Miles assay (21), which involved intradermal
injection of purified ICPP and VEGF165 in mice and measure-
ment of the leakage of Evans Blue dye into the extravascular
space (Fig. 7B). Intradermal injection of PBS was used as a
control. For this biological action, ICPP was found to be as
potent if not more potent than VEGF165. The ICPP- and
VEGF-induced permeability was inhibited by VTKI when the
inhibitor was preinjected 60 min before the peptide growth
factor (Fig. 7). This inhibition reflects VEGF receptor targeting
since VTKI is unable to prevent increased permeability trig-
FIG.2.Sequence alignment of ICPP (Vleb ICPP) with the hypotensive factor (Vasp HF) purified from V. aspis aspis venom (23),
the deduced svVEGF amino acid sequences characterized from B. insularis (Bins svVEGF) and B. jararaca (Bjar svVEGF) venoms
(24), respectively, and human VEGF (22). Human VEGF residues identified by site-directed mutagenesis as being important for receptor
binding (25) are in bold. Residues implicated in VEGF R2 (Flk-1 (fetal liver kinase-1)/KDR) binding along with the VEGF crystal structure are
underlined (26). The heparin binding domain (55 residues) is indicated by italics. The box indicates the N-glycosylation site, and the asterisks
indicate identical residues.
FIG.3.Structure of VTKI. See Experimental Procedures for the
preparation of the AstraZeneca compound (ZM317450).
TABLE I
Inhibition of isolated receptor tyrosine kinase activity
(IC
50
,
M) by VTKI
The ability of VTKI to inhibit recombinant kinase activity was exam-
ined using a 96-well ELISA assay with 2
M ATP. Data represent the
mean S.E. of at least three separate determinations.
VEGF-R1 VEGF-R2 EGFR FGFR1
0.50 0.04 0.05 0.01 100 100
Structure and Biology of Vipera lebetina Venom VEGF 29995
by guest on July 17, 2015http://www.jbc.org/Downloaded from
gered via the
-thrombin receptor peptide (31) activating
PAR-1.
2
DISCUSSION
Snake venoms represent an extraordinary source of biologi-
cal molecules that have been invaluable to our knowledge and
understanding of basic biological processes. Besides the toxins
acting directly in the nervous system, many venom proteins
target blood capillaries, preventing blood coagulation via dis-
integrins and enzymes degrading fibrinogens, whereas others
increase the permeability of blood capillaries. This is the case
for bradykinin potentiating factors purified from B. jararaca
that convert angiotensin I into angiotensin II and phospho-
lipase A2 purified from Trimeresurus mucrosquamatus, which
induces release of histamine from mast cell degranulation (32).
In this context, it is remarkable to see the emergence of a new
set of selected molecules with permeability capacity in snake
venoms. ICPP, an increasing capillary permeability protein of
V. lebetina venom, is indeed a VEGF-related molecule highly
homologous to the HF with vascular permeability activity (23).
During the preparation of this manuscript, another report
demonstrated the expression of related VEGF molecules in the
venom of Bothrops jararaca (pit viper) (24), a result extending
the notion that many snakes have evolved to specifically ex-
press in their venoms VEGF-like molecules, the most potent
permeability factors described so far in vertebrates. During
evolution, the co-selection in snake venoms of potent toxins
together with the most effective permeability factors was cer-
tainly crucial for rapid dissemination of the toxins in the gen-
eral circulation of the prey.
In the present study, we determined the complete amino acid
sequence of ICPP, confirming that it is structurally related to
the PDGF family. This protein consists of two homodimers of
110 amino acids having a molecular mass of 25115 Da. ICPP
shares the highest sequence identity with HF, the hypotensive
factor purified from V. aspis aspis (23), and the recently iden-
tified svVEGF from B. insularis pit viper venom (24). ICPP,
together with the other snake svVEGFs, shares about 50%
identity with human VEGF-A (22). ICPP, like HF and svVEGF,
differs in length from VEGF165, lacking a heparin binding
domain and a potential N-linked glycosylation site. The recom-
binant protein svVEGF from the B. insularis pit viper was
biologically characterized only by its ability to increase vascu-
lar permeability (24). However, in addition to increasing per-
meability, HF has been shown to also exert a strong hypoten-
sive effect and have a mitogenic effect on endothelial cells. The
mitogenic activity of HF was 510 times lower than that of
VEGF and was inhibited by cycloheximide. The authors spec-
ulated that this protein may induce a signaling response and
increase protein synthesis in endothelial cells, but studies ex-
amining the mechanism of action have not been reported (23).
VEGF functions by interacting with two well characterized
high affinity tyrosine kinase receptors, VEGF-R1 and VEGF-
R2, that are selectively expressed on endothelium. VEGF-R2
2
V. Vouret-Cravieri, unpublished results.
FIG.4. ICPP, like VEGF, activates
p42/p44 MAP kinases in HUVEC; the
dose response of the VTKI on VEGF-in-
duced MAP kinase activation is shown.
Serum-starved HUVEC were stimulated
for 15 min with 10 ng/ml VEGF165, ICPP
or FGF-2 in the absence (0) or presence
(10
M) of VTKI (A). The kinase inhibitor
(dissolved in Me
2
SO) was added 15 min
prior to stimulation, and Me
2
SO alone
was added to the media of cells that did
not receive VTKI (1% final concentration).
Total extracts (50
g of proteins) were
separated on SDS-polyacrylamide gel
electrophoresis and immunoblotted with
a specific anti-phospho p42/p44 MAP ki-
nase monoclonal antibody as described
under Experimental Procedures. The
doublet represents both ERK isoforms. C,
control. HUVEC were stimulated for 15
min with 10 ng/ml VEGF165 () and pre-
incubated with various concentrations of
VTKI (B). Me
2
SO was kept constant at
1%. Activation of p42/p44 MAPK was
monitored as indicated in Experimental
Procedures.
FIG.5. VTKI is a specific VEGF re-
ceptor antagonist. ER22 fibroblastic
cells, a derivative of CCL39 expressing
800,000 EGF receptors (28), were serum-
starved for 24 h and stimulated with five
different agonists: human PDGFb (10 and
30 ng/ml), human FGF-2 (10 and 30 ng/ml),
human EGF (10 and 30 ng/ml), 5% fetal
calf serum (FCS), or human
-thrombin
(THR) at 1 unit/ml. Where indicated, cells
were preincubated with either 1 or 10
M of
VTKI. Activation of p42/p44 MAPK was
monitored as described under Experimen-
tal Procedures.
Structure and Biology of Vipera lebetina Venom VEGF29996
by guest on July 17, 2015http://www.jbc.org/Downloaded from
appears to be the dominant signaling receptor in VEGF-
induced mitogenesis, and permeability increases (33, 34),
whereas the role of VEGF-R1 in endothelial cell function is
much less clear. Recent findings suggest that VEGF-R1 may
have a negative role, either by acting as a decoy receptor or by
suppressing signaling through VEGF-R2 (35).
Plasmin cleavage of VEGF165 generates a 110-residue long
N-terminal fragment (VEGF110) that lacks the heparin bind-
ing domains. VEGF110 is thought to maintain the ability to
bind to the VEGF receptors, but its endothelial cell mitogenic
potency is decreased substantially (100-fold) relative to
VEGF165, indicating that the heparin binding domains are
critical for stimulating endothelial cell proliferation (36). This
finding also concurs with experiments using VEGF121, which
does not bind to either heparan sulfates or to the extracellular
matrix. VEGF121 has been described as being 10100-fold less
potent than VEGF165 at inducing biological responses in en-
dothelial cells (37). The effect of glycosylation of VEGF165 on
receptor binding has also been studied by Keyt et al. (25) using
a constructed unglycosylated form of VEGF. They showed that
N-linked carbohydrate at Asn-75 does not appear to have an
effect in mediating VEGF receptor binding and the precise role
of glycosylation remains to be elucidated.
ICPP and VEGF165 were tested in parallel in various bioas-
says including MAP kinase activation, in vitro angiogenesis,
and capillary permeability. In this regard, our studies have
shown that ICPP, like VEGF165 and FGF-2, stimulates p42/
p44 MAP kinases in HUVEC. The activity of ICPP was found to
FIG.6. ICPP is angiogenic; it stimulates reinitiation of DNA
synthesis in HUVEC and promotes cell proliferation and differ-
entiation of mouse embryonic stem cells. In A and B, for reinitia-
tion of DNA synthesis, confluent and serum-starved HUVEC were
stimulated for 24 h (see Experimental Procedures) in SFM in the
presence of an agonist (VEGF165, ICPP, or FGF-2) and tritiated thy-
midine (1
Ci/ml). Where indicated (gray bars), cells were preincubated
with 10
M VTKI. Tritiated thymidine incorporation into acid-insoluble
material was measured in triplicate wells, and the average (less than
15% variation for each condition) was plotted. In B, the stimulation
conditions were identical to those in panel A, except that cells received
various concentrations of VTKI during the course of the stimulation. In
C, mouse ES cells were cultivated and differentiated as described under
Experimental Procedures.At day 3, the resulting embryo bodies were
then transferred to gelatin-coated 24-well tissue culture plates. Where
indicated, the medium was supplemented with 10 ng/ml VEGF165, 10
ng/ml ICPP, or no supplement (Control). After 12 days of differentia-
tion, embryoid bodies were fixed and revealed with a rat anti-CD31
antibody as indicated under Experimental Procedures.
FIG.7. Capillary permeability stimulated by ICPP is antago-
nized by VTKI. ICPP, VEGF165, or PBS was injected intradermally
into mice within a dorsal region (n 5 per group). To evaluate the
inhibitory action of VTKI, the compound (50
lat10
M in PBS) was
preinjected (60 min prior to administration) at the intended site of
cytokine administration. Animals were sacrificed 30 min after cytokine
injection, and the dorsal epithelium was dissected and photographed
(A). The leakage of the Evans Blue dye into the extravascular space was
quantified as indicated under Experimental Procedures (B). The val-
ues represent the mean of three independent experiments S.D. In the
picture shown, 50
l of VEGF165 at 30 ng/ml and ICPP at 10 ng/ml
were injected intradermally (see Experimental Procedures).
Structure and Biology of Vipera lebetina Venom VEGF 29997
by guest on July 17, 2015http://www.jbc.org/Downloaded from
be at least 1.5 times more potent than human recombinant
VEGF165 and mediated via signaling through endogenous
VEGF receptors. The requirement for VEGF receptor signaling
in transduction of a biological response to ICPP was demon-
strated using VTKI, an antagonist that can selectively inhibit
the kinase activity associated with VEGF-R2 and VEGF-R1.
This inhibitor ablates fully ICPP- and VEGF-induced MAPK
activation (IC
50
of 100 nM versus both) but does not affect that
induced by FGF-2. The specificity of VTKI has been further
demonstrated in ER22 cells, where it was found to not affect
PDGF-, EGF-, or FGF-2-mediated MAPK activation at a con-
centration of 30
M. Furthermore, VTKI did not inhibit the
MAP kinase activation in response to the G protein-coupled
receptor agonist,
-thrombin, or fetal calf serum.
When the mitogenic action of ICPP was studied, it was found
to be as potent if not slightly more potent than VEGF165 at
reinitiating DNA synthesis in HUVEC. This ICPP-induced re-
sponse was fully suppressed by VTKI. In addition, we have
observed that ICPP promotes DNA proliferation in HUVEC
and induces growth and differentiation of mouse embryonic
endothelial cells. VTKI can also inhibit these ICPP-induced
angiogenic and permeability responses, indicating an involve-
ment of VEGF receptor signaling. In each biological response
examined, ICPP was found to possess remarkably similar and
at least three times more potent bioactivity than VEGF165
despite the absence of a heparin binding domain. The struc-
tural differences between ICPP and VEGF, in particular the
absence in ICPP of heparin binding domains and consensus
N-glycosylation sites, suggest that some of their biological
activities might be different.
In conclusion, we have shown that the amino acid sequence
of ICPP displays a high similarity to that of VEGF and that
capillary permeability, angiogenesis, endothelial cell mitoge-
nicity, and MAP kinase activation induced by ICPP were
mediated through VEGF receptor signaling. These findings
provide the first evidence that ICPP is a novel member of the
family of VEGF-like growth factors. Moreover, considering the
therapeutic impact of VEGF-A in the treatment of coronary
heart disease or critical limb ischemia (38, 39), ICPP, which
appears more potent than VEGF, may represent a novel can-
didate for therapeutic angiogenic approaches aimed at growing
new vasculature.
AcknowledgmentsWe thank Dr. Z. Ben Lasfar (Veterinary Labora-
tory, Pasteur Institute of Tunis) for providing snake venom, P. Y.
Haumont from PerkinElmer Life Sciences for the mass determination
of ICPP by mass spectrometry, J. Kendrew (AstraZeneca, Alderley
Park, UK) for generating tyrosine kinase data, D. Roux and M. Hattab
(CNRS, Nice, France) for the CCL39 cell line expressing VEGF-R2 and
for the purification of recombinant FGF and VEGF.
REFERENCES
1. Folkman, J., and Klagsbrun, M. (1987) Science 235, 442 447
2. Carmeliet, P. (2000) Nat. Med. 6, 389 395
3. Yancopoulos, G. D., Davis, S., Gale, N. W., Rudge, J. S., Wiegand, S. J., and
Holash, J. (2000) Nature 407, 242248
4. Mazure, N. M., Chen, E. Y., Yeh, P., Laderoute, K. R., and Giaccia, A. J. (1996)
Cancer Res. 56, 34363440
5. Houck, K. A., Leung, D. W., Rowland, A. M., Winer, J., and Ferrara, N. (1992)
J. Biol. Chem. 267, 2603126037
6. Grunstein, J., Masbad, J. J., Hickey, R., Giordano, F., and Johnson, R. S.
(2000) Mol. Cell Biol. (2000) 20, 72827291
7. Soker, S., Takashima, S., Miao, H., Neufeld, G., and Klagsbrun, M. (1998) Cell
92, 735745
8. Matsumoto, T., and Claesson-Welsh, L. (2001) Sci. STKE. 2001 112, RE21
(www.stke.sciencemag.org)
9. Ferrara, N., and Alitalo, K. (1999) Nat. Med. 5, 1359 1364
10. Thurston, G., Rudge, J. S., Ioffe, E., Zhou, H., Ross, L., Croll, S. D., Glazer, N.,
Holash, J., McDonald, D. M., and Yancopoulos, G. D. (2000) Nat. Med. 6,
460 463
11. Gasparini, G., and Harris, A. L. (1995) J. Clin. Oncol. 13, 765782
12. Gasmi, A., Abidi, F., Srairi, N., Oijatayer A., Karoui, H., and Elayeb, M. (2000)
Biochem. Biophys. Res. Commun. 268, 6972
13. Kawasaki, I., and Itano, H. A. (1972) Anal. Biochem. 48, 546 565
14. Barbieri, B., Balconi, B., Dejana, E., and Donati, M. (1981) Proc. Soc. Exp. Biol.
Med. 168, 204207
15. Laemmli, U. K. (1970) Nature 277, 680 685
16. Vinals, F., Chambard, J-C., and Pouyssegur, J. (1999) J. Biol. Chem. 274,
26988 26995
17. Hennequin, L. F., Thomas, A. P., Johnstone, C., Stokes, E. S. E., Ple´ ,P.A.,
Lohmann, J. M., Ogilvie, D. J., Dukes, M., Wedge, S. R., Curwen, J. O.,
Kendrew, J., and Lambert-van der Brempt, C. (1999) J. Med. Chem. 42,
5369 5389
18. Lowry, O. H., Rosebrough, N. H., Farr, A. L., and Randall, R. J. (1951) J. Biol.
Chem. 193, 265275
19. Smith, A. (1991) J. Tissue Cult. Methods 13, 89 94
20. Fa¨ssler, R., Pfaff, M., Murphy, J., Noegel, A., Johansson, S., Timpl, R., and
Albrecht, R. (1995) J. Cell Biol. 128, 979 988
21. Miles, A. A., and Wilhelm, D. L. (1955) Br. J. Exp. Path. 36, 71 81
22. Leung, D. W., Cachiane, G., Kuang, W. J., Goddel, D. V., and Ferrara, N.
(1989) Science 246, 13061309
23. Komori, Y., Nikai, T., Taniguchi, T., Masuda, K., and Sugihara, H. (1999)
Biochemistry 38, 1176911803
24. Junqueira de Azevedo, I. L., Farsky, S. H., Oliveira, M. L., and Ho, P. L. (2001)
J. Biol. Chem. 276, 3983639842
25. Keyt, B. A., Nguyen, H. V., Berleau, L. T., Ouarta, C. M., Park, J., Chen H.,
and Ferrara, N. (1996) J. Biol. Chem. 271, 5638 5646
26. Muller, Y. A., Li, B., Christinger, H. W., Wells, J. A., Cunningham, B. C., and
de Vos, A. M. (1997) Proc. Natl. Acad. Sci. U. S. A. 94, 71927197
27. Fairbrother, W. J., Champe, M. A., Christinger, H. W., Keyt, B. A., and
Starovasnik, M. A. (1998) Structure 6, 637648
28. LAllemain, G., Seuwen, K., Velu, T., and Pouyssegur, J. (1989) Growth
Factors 1, 311321
29. Saadeh, P. B., Mehrara, B. J., Steinbrech, D. S., Spector, J. A., Greenwald,
J. A., Chin, G. S., Ueno, H., Gittes, G. K., and Longaker, M. T. (2000)
Endocrinology 141, 20752083
30. Vittet, D., Prandini, M. H., Berthier, R., Schweitzer, A., Martin-Sisteron, H.,
Uzan, G., and Dejana, E. (1996) Blood 88, 3424 3431
31. Vouret-Craviari, V., Van Obberghen-Schilling, E., Rasmussen, U. B., Pavirani,
A., Lecocq, J. P., and Pouyssegur, J. (1992) Mol. Biol. Cell 3, 95102
32. Markland, F. S. (1998) Toxicon 36, 1749 1800
33. Waltenberger, J., Claesson-Welsh, L., Siegbahn, A., Shibuya, M., and Heldin,
C. H. (1994) J. Biol. Chem. 269, 26988 26995
34. Korpelainen, E. I., and Alitalo, K. (1998) Curr. Opin. Cell Biol., 10, 159 164
35. Rahimi, N., Dayanir, V., and Lashkari, K. (2000) J. Biol. Chem. 275,
16986 16992
36. Keyt, B. A., Berleau L. T., Nguyen, H. V., Chen, H., Heinsohn, H., Vendleu, R.,
and Ferrara, N. (1996) J. Biol. Chem. 271, 7788 7795
37. Ogawa, S., Oku, A., Sawano, A., Yamaguchi, S., Yazaki, Y., and Shibuya, M.
(1998) J. Biol. Chem. 273, 3127331282
38. Waltenberger, J. (1997) Circulation 96, 4083 4094
39. Folkman, J. (1998) Circulation 97, 1108 1110
Structure and Biology of Vipera lebetina Venom VEGF29998
by guest on July 17, 2015http://www.jbc.org/Downloaded from
Hennequin and Jacques Pouysségur
Clotilde Gimond, Stephen R. Wedge, Laurent
Aloui, Najet Srairi, Sandrine Marchetti,
Ammar Gasmi, Christine Bourcier, Zohra
RECEPTOR SIGNALING
ENDOTHELIAL GROWTH FACTOR
IS ANGIOGENIC VIA VASCULAR
Venom: ICPPVipera lebetinaPurified from
Capillary Permeability Protein (ICPP)
Complete Structure of an Increasing
TRANSDUCTION:
MECHANISMS OF SIGNAL
doi: 10.1074/jbc.M202202200 originally published online May 20, 2002
2002, 277:29992-29998.J. Biol. Chem.
10.1074/jbc.M202202200Access the most updated version of this article at doi:
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted
When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/277/33/29992.full.html#ref-list-1
This article cites 39 references, 20 of which can be accessed free at
by guest on July 17, 2015http://www.jbc.org/Downloaded from
... VEGFs constitute a pivotal family of proteins with significant relevance in stimulating neovascularization in endothelial cells, thereby fostering the formation of novel blood vessels [25]. For svVEGFs, this function is poorly explored since there are, so far, few studies reporting their ability to form new vessels [28,44,57]. One of those is ICPP, a svVEGF from Macrovipera lebetina snake venom, which could induce neovascularization on HUVEC cells in a 10 ng/mL concentration, while human VEGF promoted the formation of new vessels with a 30 ng/mL concentration [57]. ...
... For svVEGFs, this function is poorly explored since there are, so far, few studies reporting their ability to form new vessels [28,44,57]. One of those is ICPP, a svVEGF from Macrovipera lebetina snake venom, which could induce neovascularization on HUVEC cells in a 10 ng/mL concentration, while human VEGF promoted the formation of new vessels with a 30 ng/mL concentration [57]. Also, CdcVEGFs, from C. d. collilineatus snake venom, were shown to induce formation of new vessels in a tube formation assay when tested at 20 ng/mL concentration, surpassing the potency of human FGF used at the same concentration [28]. ...
... The infiltration of leukocytes into the peritoneal cavity accompanied by a concurrent increase in total protein levels triggered by CdtVEGF offer compelling evidence of its capacity to incite local inflammation-a characteristic that remains unexplored within this particular protein class. The induction of vascular permeability from CdtVEGF corroborates reports of other svVEGFs that also led to an increase when tested in animal models at low concentrations (10 ng/mL, 0.5 nM) [18,26,31,57,64]. In another study, svVEGF IC2 from M. lebetina snake venom, when tested on C57Bl/6 mice, induced higher vascular permeability than when tested on Swiss lineage [42]. ...
Article
Full-text available
A pioneering study regarding the isolation, biochemical evaluation, functional assays and first PEGylation report of a novel vascular endothelial growth factor from Crotalus durissus terrificus venom (CdtVEGF and PEG-CdtVEGF). CdtVEGF was isolated from crude venom using two different chromatographic steps, representing 2% of soluble venom proteins. Its primary sequence was determined using mass spectrometry analysis, and the molecule demonstrated no affinity to heparin. The Brazilian crotalid antivenom recognized CdtVEGF. Both native and PEGylated CdtVEGF were able to induce new vessel formation and migration, and to increase the metabolic activity of human umbilical endothelial vascular cells (HUVEC), resulting in better wound closure (~50% within 12 h) using the native form. CdtVEGF induced leukocyte recruitment to the peritoneal cavity in mice, with a predominance of neutrophil influx followed by lymphocytes, demonstrating the ability to activate the immune system. The molecule also induced a dose-dependent increase in vascular permeability, and PEG-CdtVEGF showed less in vivo inflammatory activity than CdtVEGF. By unraveling the intricate properties of minor components of snake venom like svVEGF, this study illuminates the indispensable significance of exploring these molecular tools to unveil physiological and pathological processes, elucidates the mechanisms of snakebite envenomings, and could possibly be used to design a therapeutic drug
... It is described that svVEGFs have the ability to interact with receptors VEGFR-1 and 2 [16,17], being capable to induce angiogenesis, in which some snake-derived VEGFs proved being more potent than VEGF-A [11,17,18]. Here, we isolated and analyzed the biochemical and functional properties of novel VEGFs (CdcVEGF) from Crotalus durissus collilineatus venom, with potent angiogenic activity in vitro. ...
... Following their structural characterization, since most svVEGFs present the amino terminal residue blocked by pyroglutamic acid [11,18,42], we choose mass spectrometry analysis using digestion with trypsin in the attempt to determine its primary structure. After spectra analysis at PeaksStudio XPro software using databases containing broad amino acid sequences of proteins from snakes and amino acid sequences of proteins from Crotalus genus, we could obtain peptides corresponding only to svVEGFs class of proteins. ...
... Known as vascular endothelial growth factors, VEGFs are able to induce neovascularization on endothelial cells, promoting the formation of new blood vessels [49] and although it is one of the main functions of these molecules, for svVEGFs there are few studies describing their ability in inducing angiogenesis [18,42]. One method to analyze angiogenesis is the chick chorioallantoic membrane (CAM) assay, consisting in the treatment of CAM with VEGFs. ...
Article
Vascular endothelial growth factors (VEGFs) are crucial molecules involved in the modulation of angiogenesis. Snake venom-derived VEGFs (svVEGFs) are known to contribute significantly to the envenoming due to their capacity of increasing vascular permeability. In our work, we isolated and analyzed the biochemical and functional properties of the VEGF from Crotalus durissus collilineatus venom (CdcVEGF). The venom was fractionated by reversed phase chromatography on FPLC system (Fast Protein Liquid Chromatography) and the eluted fractions were submitted to an ELISA assay using an anti-VEGF-F antibody, for identification of svVEGF. Positive fractions for svVEGF were submitted to SDS-PAGE and to an anion exchange chromatography to isolate the molecule. The subfractions were analyzed by ELISA and SDS-PAGE and six of them presented svVEGFs, named CdcVEGF1 (Q23-3), CdcVEGF2 (Q24-3), CdcVEGF3 (Q24-4), CdcVEGF4 (Q25-3), CdcVEGF5 (Q25-4), and CdcVEGF6 (Q25-5). Their structural characterization was accomplished by mass spectrometry analysis using MALDI-TOF to determine their molecular masses and UPLC-ESI-QTOF to determine their amino acid sequence. Interestingly, all isolated CdcVEGFs induced angiogenesis on HUVEC cells through tube formation on Matrigel when compared to culture medium (negative control). Moreover, CdcVEGF2 and CdcVEGF3 also induced a significant increase in tube formation when compared to the positive control (basic fibroblast growth factor - bFGF). Additionally, crotalid antivenom produced by the Instituto Butantan was able to recognize CdcVEGFs, demonstrating to be immunogenic. This study demonstrates that snake venom cocktail can reveal novel and important molecules, which are potential molecular tools to study diverse biological processes, such as angiogenesis.
... VEGF-like snake venom molecules, such as vammin and VR-1, have demonstrated more potent pro-angiogenic activity than mammalian VEGFs [44]. The recently discovered Macrovipera lebetina viper venom protein, called Increasing Capillary Permeability Protein (ICPP, 27 kDa) [45,46], has structural homology to VEGF, displaying an N-terminal amino acid sequence with considerable similarity to that of VEGF and plateletderived growth factor (PDGF). ICPP has been shown to exert a potent pro-angiogenic VEGF-like action through VEGFR2 receptor stimulation in mouse embryonic stem cells or human umbilical vein endothelial cells [45,46]. ...
... The recently discovered Macrovipera lebetina viper venom protein, called Increasing Capillary Permeability Protein (ICPP, 27 kDa) [45,46], has structural homology to VEGF, displaying an N-terminal amino acid sequence with considerable similarity to that of VEGF and plateletderived growth factor (PDGF). ICPP has been shown to exert a potent pro-angiogenic VEGF-like action through VEGFR2 receptor stimulation in mouse embryonic stem cells or human umbilical vein endothelial cells [45,46]. Thereafter, we showed that ICPP, initially described as a vascular permeability factor, exerted a powerful protective effect against acute cardiac ischemia by improving ischemiainduced mitochondrial dysfunction, after a single intravenous injection [47]. ...
Article
Full-text available
The venoms of Tunisian wildlife snakes are complex mixtures containing proteins/peptides and non-protein molecules. Proteins and peptides are the most abundant compounds responsible for the biological effects of venoms. Snake venoms proteins have enzymatic or non-enzymatic activities, which are grouped into different families including C-type lectin proteins, disintegrins (long, medium and short disintegrins), Kunitz-type serine protease inhibitors, natriuretic-like peptides, vascular endothelial growth factor-related proteins, L-amino acid oxidases, phospholipases A2 and serine proteinases. With technological advancements, the toxic effects of venoms were turned into potential benefits for clinical diagnosis, basic research and development of new research tools and drugs of potential clinical use. Our research team has shown that Macrovipera lebetina and Cerastes cerastes venom components of Tunisian wildlife snakes had great potential for the development of new drugs for the treatment of cancer, angiogenesis disorders or cardiovascular diseases. This review is an overview on snake venom proteins from Macrovipera lebetina and Cerastes cerastes and their biochemical, pharmacological and molecular characterization and their importance as protein resources with therapeutic potential.
... In vitro permeability was studied as described [40]. Briefly, TIME cells were grown on membranes of 6.5 mm Transwell inserts (0.4 lm pore size, Corning Ò ). ...
Article
Full-text available
The efficacy of anti‐angiogenic treatment by targeting VEGF/VEGF receptors in metastatic clear cell renal cell carcinoma (ccRCC) varies from patient to patient. Discovering the reasons behind this variability could lead to the identification of relevant therapeutic targets. Thus, we investigated novel splice variants of VEGF that are less efficiently inhibited by anti‐VEGF/VEGFR targeting than the conventional isoforms. By in silico analysis we identified a novel splice acceptor in the last intron of the VEGF gene resulting in an insertion of 23 bp in VEGF mRNA. Such an insertion can shift the open reading frame in previously described splice variants of VEGF (VEGFXXX), leading to a change in the C‐terminal part of the VEGF protein. Next, we analyzed the expression of these alternatively spliced VEGF new isoforms (VEGFXXX/NF.) in normal tissues and in RCC cell lines by qPCR and ELISA, and investigated the role of VEGF222/NF (equivalent to VEGF165) in physiological and pathological angiogenesis. Our in‐vitro data demonstrated that recombinant VEGF222/NF stimulated endothelial cell proliferation and vascular permeability by activating VEGFR2. In addition, VEGF222/NF overexpression enhanced proliferation and metastatic properties of RCC cells, whereas downregulation of VEGF222/NF resulted in cell death. We also generated an in‐vivo model of RCC by implanting RCC cells overexpressing VEGF222/NF in mice, which we treated with polyclonal anti‐VEGFXXX/NF antibodies. VEGF222/NF overexpression enhanced tumor formation with aggressive properties and a fully functional vasculature , while treatment with anti‐VEGFXXX/NF antibodies slowed tumor growth by inhibiting tumor cell proliferation and angiogenesis. In a patient cohort from the NCT00943839 clinical trial we investigated the relationship between plasmatic VEGFXXX/NF levels, resistance to anti‐VEGFR therapy and survival. High plasmatic VEGFXXX/NF levels correlated with shorter survival and lower efficacy of anti‐angiogenic drugs. Our data confirmed the existence of new VEGF isoforms that could serve as novel therapeutic targets in patients with RCC that are resistant to anti‐VEGFR therapy.
... Images were analyzed using the Java-based ImageJ34 program and the distance was measured after 0, 3, 6, 9 and 12 hours. For permeability testing, con uent TIME cells were treated with VEGF 222/NF (100 ng/mL) for 20 minutes and the experiment was performed as described [52]. ...
Preprint
Full-text available
Background The efficacy of anti-VEGF/VEGF receptors in the treatment of metastatic clear cell renal cell carcinoma (ccRCC) varies from patient to patient. Discovering the reasons for this variability could lead to the identification of relevant therapeutic targets. We have investigated the possibility of splicing events leading to new forms of VEGF that are less efficiently inhibited by anti-VEGF/VEGFR targeting the conventional forms. Methods In silico analysis identified the presence of an unknown splice acceptor in the last intron of the VEGF gene resulting in an insertion of 23 bases in VEGF mRNA. Such an insertion can occur in previously described splice variants of VEGF (VEGFXXX) and shift the open reading frame, leading to a change in the c-terminal part of VEGF. We investigated the role of the resulting new major form of VEGF, VEGF222NF, in physiological and pathological angiogenesis. We analyzed the expression of these new alternatively spliced forms in normal tissue and in a series of RCC cells by qPCR and ELISA. We generated experimental RCC in mice by implanting ccRCC cells overexpressing VEGF222NF. The experimental RCC were also treated with polyclonal anti-VEGF/NF antibodies. The relationship between plasmatic VEGF/NF levels and resistance to anti-VEGFR and survival was also investigated in a cohort of patients from the NCT00943839 clinical trial. Results VEGF222/NF stimulated endothelial cell proliferation and vascular permeability through activation of VEGFR2. Overexpression of VEGF222/NF stimulated proliferation and metastatic properties of RCC cells, whereas its downregulation resulted in cell death. RCC cells overexpressing VEGF222/NF generated aggressive experimental tumors that developed functional blood and lymphatic vessels. Anti-VEGFXXX/NF antibodies slowed the growth of experimental RCC by inhibiting tumor cell proliferation and the development of blood and lymphatic vessels. High plasmatic VEGFXXX/NF levels correlated with shorter survival and lower efficacy of anti-angiogenic drugs. Conclusions The existence of new VEGF isoforms has shed new light on the VEGF field.
... Images were analyzed using the Java-based ImageJ34 program and the distance was measured after 0, 3, 6, 9 and 12 hours. For permeability testing, con uent TIME cells were treated with VEGF 222/NF (100 ng/mL) for 20 minutes and the experiment was performed as described [52]. ...
Preprint
Full-text available
Background The efficacy of anti-VEGF/VEGF receptors in the treatment of metastatic clear cell renal cell carcinoma (ccRCC) varies from patient to patient. Discovering the reasons for this variability could lead to the identification of relevant therapeutic targets. We have investigated the possibility of splicing events leading to new forms of VEGF that are less efficiently inhibited by anti-VEGF/VEGFR targeting the conventional forms. Methods In silico analysis identified the presence of an unknown splice acceptor in the last intron of the VEGF gene resulting in an insertion of 23 bases in VEGF mRNA. Such an insertion can occur in previously described splice variants of VEGF (VEGFXXX) and shift the open reading frame, leading to a change in the c-terminal part of VEGF. We investigated the role of the resulting new major form of VEGF, VEGF222NF, in physiological and pathological angiogenesis. We analyzed the expression of these new alternatively spliced forms in normal tissue and in a series of RCC cells by qPCR and ELISA. We generated experimental RCC in mice by implanting ccRCC cells overexpressing VEGF222NF. The experimental RCC were also treated with polyclonal anti-VEGF/NF antibodies. The relationship between plasmatic VEGF/NF levels and resistance to anti-VEGFR and survival was also investigated in a cohort of patients from the NCT00943839 clinical trial. Results VEGF222/NF stimulated endothelial cell proliferation and vascular permeability through activation of VEGFR2. Overexpression of VEGF222/NF stimulated proliferation and metastatic properties of RCC cells, whereas its downregulation resulted in cell death. RCC cells overexpressing VEGF222/NF generated aggressive experimental tumors that developed functional blood and lymphatic vessels. Anti-VEGFXXX/NF antibodies slowed the growth of experimental RCC by inhibiting tumor cell proliferation and the development of blood and lymphatic vessels. High plasmatic VEGFXXX/NF levels correlated with shorter survival and lower efficacy of anti-angiogenic drugs. Conclusions The existence of new VEGF isoforms has shed new light on the VEGF field.
... A homodimeric increasing capillary permeability protein (ICPP) has been found in some viper species (Gasmi et al., 2000). ICPP exerts its biological action (permeability and angiogenesis) through activation of vascular endothelial growth factor receptor signaling (VEGF-R2 and possibly VEGF-R1) (Gasmi et al., 2002). Vascular endothelial growth factor (VEGF) activates endothelial cells and promotes cell proliferation, migration, and an increase in vascular permeability. ...
Article
In this study, the early retinal and optic nerve blood flows of patients exposed to Viper bite were evaluated with non-invasive optical coherence tomography angiography (OCTA) and compared with healthy controls. The retinal and optic disc OCTA data of 31 victims of viper bite (group S) without systemic envenomation clinical symptoms and 31 healthy controls (group C) were compared. Only patients with early signs of envenomation were included in the study. Optical coherence tomography angiographies were performed with RTVue XR Avanti with AngioVue software. Vascular densities in the whole image, foveal, parafoveal regions at the superficial and the deep capillary plexus segments were acquired and statistically analyzed. The flow area parameters were measured in the superficial retinal capillary plexus, deep retinal capillary plexus, outer retinal capillary plexus, and choriocapillaris layers of the macula in 1-mm and 3-mm diameter areas. The peripapillary flow areas were measured for the optic nerve head, vitreous, radial peripapillary capillary (RPC), and choroid in a 4.50-mm diameter area. Foveal and parafoveal thicknesses were also measured and compared. The average hospital admission time of the patients in group S was 1.24 ± 0.75 (0.50–3.00) hours. Age (p = 0.103) and gender (p = 0.714) were similar in both groups. Superficial (p = 0.010), deep flow areas (p = 0.034), and superficial parafoveal vascular density (p = 0.001) were significantly reduced in group S compared to group C. The outer retinal flow area (p < 0.001) increased significantly in group S. Nerve head flow area (p = 0.035), one of the optic disc flow areas, was found to be decreased in group S. Notably, foveal (p < 0.001) and parafoveal (p = 0.003) thicknesses and superficial (p = 0.001) and deep (p < 0.001) foveal vascular densities were greater in group S. Compared to group C, the superficial (p = 0.009) and deep (p = 0.009) foveal flow areas in the central foveal area with a diameter of 1 mm increased significantly in group S. Viper venom may cause blood flow changes in the retina and optic disc and an increase in retinal thickness in the early period although there are no signs of systemic envenomation.
... ICPP was found to be a potent in vitro angiogenic factor when added to mouse embryonic stem cells or human umbilical vein endothelial cells. It was concluded that ICPP exerts its biological action (permeability and angiogenesis) through activation of VEGF receptor signaling (VEGF-R2 and possibly VEGF-R1) (Gasmi et al., 2002). Several distinct isoforms [IC1 (Increasing Capillary1) and IC2] of svVEGF have been isolated from M. lebetina venom. ...
Article
The isolation and characterization of individual snake venom components is important for a deeper understanding of the pathophysiology of envenomations, for improving the therapeutic procedures of patients, and it also opens possibilities for the discovery of novel toxins that might be useful as tools for understanding cellular and molecular processes. This review provides a summary of the different toxins that have been isolated and characterized from the venoms of Vipera lebetina (Macrovipera lebetina) subspecies Macrovipera lebetina cernovi, Macrovipera lebetina lebetina, Macrovipera lebetina obtusa, Macrovipera lebetina transmediterranea, Macrovipera lebetina turanica, the snake species causing the majority of human envenomings in Central Asia (Middle East) and North Africa. The venoms of these snakes contain proteins belonging to different families: Zn²⁺- metalloproteinases, serine proteinases, L-amino acid oxidase, 5′-nucleotidase, phosphodiesterase, phosphomonoesterase, nucleases, hyaluronidase, phospholipase A2, C-type lectin-like protein, disintegrin, DC-fragment, cystein-rich secretory protein, proteinase inhibitors, nerve growth factor (NGF), vascular endothelial growth factor (VEGF), low molecular weight peptides. Their main biochemical properties, toxic and pharmacological actions have been described. In this review we will provide an overview of the proteins and peptides from the venoms of M. lebetina subspecies, their biochemical, pharmacological and structural features and their role in snake venom toxinology. A lot of contributions have been made for better understandings of these venomous snakes, their venom, and their pharmacological effects. Many of these components are also fascinating models for drug design.
... Several VEGFs isolated from the venom of Macrovipera lebetina have been identified as preferentially binding the VEGF receptor [90,91]. Isolated VEGFs have also been shown to bind to VEGF co-receptors, NRP1 and NRP2. ...
Article
Full-text available
Snake venom serves as a tool of defense against threat and helps in prey digestion. It consists of a mixture of enzymes, such as phospholipase A2, metalloproteases, and l-amino acid oxidase, and toxins, including neurotoxins and cytotoxins. Beside their toxicity, venom components possess many pharmacological effects and have been used to design drugs and as biomarkers of diseases. Viperidae is one family of venomous snakes that is found nearly worldwide. However, three main vipers exist in the Middle Eastern region: Montivipera bornmuelleri, Macrovipera lebetina, and Vipera (Daboia) palaestinae. The venoms of these vipers have been the subject of many studies and are considered as a promising source of bioactive molecules. In this review, we present an overview of these three vipers, with a special focus on their venom composition as well as their biological activities, and we discuss further frameworks for the exploration of each venom.
Article
Vascular endothelial growth factor (VEGF) is a key regulator of angiogenesis, a physiological process characterized by the formation of new vessels from a preexisting endothelium. VEGF has also been implicated in pathologic states, such as neoplasias, intraocular neovascular disorders, among other conditions. VEGFs are distributed in seven different families: VEGF-A, B, C, D, and PIGF (placental growth factor), which are identified in mammals; VEGF-E, which are encountered in viruses; and VEGF-F or svVEGF (snake venom VEGF) described in snake venoms. This is the pioneer review of svVEGF family, exploring its distribution among the snake venoms, molecular structure, main functions, and potential applications.
Article
Full-text available
Vascular endothelial growth factor (VEGF) is a homodimeric peptide growth factor which binds to two structurally related tyrosine kinase receptors denoted Flt1 and KDR. In order to compare the signal transduction via these two receptors, the human Flt1 and KDR proteins were stably expressed in porcine aortic endothelial cells. Binding analyses using 125I-VEGF revealed Kd values of 16 pM for Flt1 and 760 pM for KDR. Cultured human umbilical vein endothelial (HUVE) cells were found to express two distinct populations of binding sites with affinities similar to those for Flt1 and KDR, respectively. The KDR expressing cells showed striking changes in cell morphology, actin reorganization and membrane ruffling, chemotaxis and mitogenicity upon VEGF stimulation, whereas Flt1 expressing cells lacked such responses. KDR was found to undergo ligand-induced autophosphorylation in intact cells, and both Flt1 and KDR were phosphorylated in vitro in response to VEGF, however, KDR much more efficiently than Flt1. Neither the receptor-associated activity of phosphatidylinositol 3'-kinase nor tyrosine phosphorylation of phospholipase C-gamma were affected by stimulation of Flt1 or KDR expressing cells, and phosphorylation of GTPase activating protein was only slightly increased. Members of the Src family such as Fyn and Yes showed an increased level of phosphorylation upon VEGF stimulation of cells expressing Flt1 but not in cells expressing KDR. The maximal responses in KDR expressing porcine aortic endothelial cells were obtained at higher VEGF concentrations as compared to HUVE cells, i.e. in the presence of Flt1. This difference could possibly be explained by the formation of heterodimeric complexes between KDR and Flt1, or other molecules, in HUVE cells.
Article
Full-text available
The vascular endothelial growth factor (VEGF) family encompasses four polypeptides that result from alternative splicing of mRNA. We have previously demonstrated differences in the secretion pattern of these polypeptides. Stable cell lines expressing VEGFs were established in human embryonic kidney CEN4 cells. VEGF121, the shortest form, was secreted and freely soluble in tissue culture medium. VEG189 was secreted, but was almost entirely bound to the cell surface or extracellular matrix. VEGF165 displayed an intermediary behavior. Suramin induced the release of VEGF189, permitting its characterization as a more basic protein with higher affinity for heparin than VEGF165 or VEGF121, but with similar endothelial cell mitogenic activity. Heparin, heparan sulfate, and heparinase all induced the release of VEGF165 and VEGF165 suggesting heparin-containing proteoglycans as candidate VEGF-binding sites. Finally, VEGF165 and VEGF189 were released from their bound states by treatment with plasmin. The released 34-kDa dimeric species are active as endothelial cell mitogens and as vascular permeability agents. We conclude that the bioavailability of VEGF may be regulated at the genetic level by alternative splicing that determines whether VEGF will be soluble or incorporated into a biological reservoir and also through proteolysis following plasminogen activation.
Article
Full-text available
A recent explosion in newly discovered vascular growth factors has coincided with exploitation of powerful new genetic approaches for studying vascular development. An emerging rule is that all of these factors must be used in perfect harmony to form functional vessels. These new findings also demand re-evaluation of therapeutic efforts aimed at regulating blood vessel growth in ischaemia, cancer and other pathological settings.
Article
A gene trap-type targeting vector was designed to inactivate the beta 1 integrin gene in embryonic stem (ES) cells. Using this vector more than 50% of the ES cell clones acquired a disruption in the beta 1 integrin gene and a single clone was mutated in both alleles. The homozygous mutant did not produce beta 1 integrin mRNA or protein, while alpha 3, alpha 5, and alpha 6 integrin subunits were transcribed but not detectable on the cell surface. Heterozygous mutants showed reduced beta 1 expression and surface localization of alpha/beta 1 heterodimers. The alpha V subunit expression was not impaired on any of the mutants. Homozygous ES cell mutants lacked adhesiveness for laminin and fibronectin but not for vitronectin and showed a reduced association with a fibroblast feeder layer. Furthermore, they did not migrate towards chemoattractants in fibroblast medium. None of these functions were impaired in heterozygous mutants. Scanning electron microscopy revealed that homozygous cells showed fewer cell-cell junctions and had many microvilli not usually found on wild type and heterozygous cells. This profound change in cell shape is not associated with gross alterations in the expression and distribution of cytoskeletal components. Unexpectedly, microinjection into blastocysts demonstrated full integration of homozygous and heterozygous mutants into the inner cell mass. This will allow studies of the consequences of beta 1 integrin deficiency in several in vivo situations.
Article
Normal bone growth and repair is dependent on angiogenesis. Fibroblast growth factor-2 (FGF-2), vascular endothelial growth fac- tor (VEGF), and transforming growth factor-b (TGFb) have all been implicated in the related processes of angiogenesis, growth, develop- ment, and repair. The purpose of this study was to investigate the relationships between FGF-2 and both VEGF and TGFb in nonim- mortalized and clonal osteoblastic cells. Northern blot analysis re- vealed 6-fold peak increases in VEGF mRNA a t6hi nfetal rat calvarial cells and MC3T3-E1 osteoblastic cells after stimulation with FGF-2. Actinomycin D inhibited these increases in VEGF mRNA, whereas cycloheximide did not. The stability of VEGF mRNA was not increased after FGF-2 treatment. Furthermore, FGF-2 induced dose- dependent increases in VEGF protein levels (P , 0.01). Although in MC3T3-E1 cells, TGFb1 stimulates a 6-fold peak increase in VEGF mRNA afte r3ho fstimulation, we found that both TGFb2 and TGFb3 yielded 2- to 3-fold peak increases in VEGF mRNA levels noted after 6 h of stimulation. Similarly, both TGFb2 and TGFb3 dose depen- dently increased VEGF protein production. To determine whether FGF-2-induced increases in VEGF mRNA may have occurred inde- pendently of TGFb, we disrupted TGFb signal transduction (using adenovirus encoding a truncated form of TGFb receptor II), which attenuated TGFb1 induction of VEGF mRNA, but did not impede FGF-2 induction of VEGF mRNA. In summary, FGF-2-induced VEGF expression by osteoblastic cells is a dose-dependent event that may be independent of concomitant FGF-2-induced modulation of TGFb ac- tivity. (Endocrinology 141: 2075-2083, 2000)
Article
A series of substituted 4-anilinoquinazolines and related compounds were synthesized as potential inhibitors of vascular endothelial growth factor (VEGF) receptor (Flt and KDR) tyrosine kinase activity. Enzyme screening indicated that a narrow structure−activity relationship (SAR) existed for the bicyclic ring system, with quinazolines, quinolines, and cinnolines having activity and with quinazolines and quinolines generally being preferred. Substitution of the aniline was investigated and clearly indicated that small lipophilic substituents such as halogens or methyl were preferred at the C-4‘ position. Small substituents such as hydrogen and fluorine are preferred at the C-2‘ position. Introduction of a hydroxyl group at the meta position of the aniline produced the most potent inhibitors of Flt and KDR tyrosine kinases activity with IC50 values in the nanomolar range (e.g. 10, 12, 13, 16, and 18). Investigation of the quinazoline C-6 and C-7 positions indicates that a large range of substituents are tolerated at C-7, whereas variation at the C-6 is more restricted. At C-7, neutral, basic, and heteroaromatic side chains led to very potent compounds, as illustrated by the methoxyethoxy derivative 13 (IC50 < 2 nM). Our inhibitors proved to be very selective inhibitors of Flt and KDR tyrosine kinase activity when compared to that associated with the FGF receptor (50- to 3800-fold). Observed enzyme profiles translated well with respect to potency and selectivity for inhibition of growth factor stimulated proliferation of human umbilical vein endothelial cells (HUVECs). Oral administration of selected compounds to mice produced total plasma levels 6 h after dosing of between 3 and 49 μM. In vivo efficacy was demonstrated in a rat uterine oedema assay where significant activity was achieved at 60 mg/kg with the meta hydroxy anilinoquinazoline 10. Inhibition of growth of human tumors in athymic mice has also been demonstrated: compound 34 inhibited the growth of established Calu-6 lung carcinoma xenograft by 75% (P < 0.001, one tailed t-test) following daily oral administration of 100 mg/kg for 21 days.
Article
Techniques are described for the culture of murine embryonic stem cells in the absence of heterologous feeder cells and for the induction of differentiation programs. The regulatory factor differentiation inhibiting activity/ leukaemia inhibitory factor (DIA/LIF) is produced at high concentration by transient expression in Cos cells and is used to suppress stem cell differentiation by addition to the culture medium. Differentiation is then induced in a controlled manner either by withdrawal of DIA/LIF or by exposure to the chemical inducers retinoic acid or 3-methoxybenzamide.
Article
In this issue of Circulation , Baumgartner et al1 ). The plasmid DNA was injected directly into the muscle of ischemic limbs. Anatomic and functional efficacy was demonstrated by increased serum levels of VEGF, improved hemodynamic measurements and angiographic evaluation, reduced pain, increased healing of ischemic ulcers, limb salvage, and immunohistochemical evidence of proliferating endothelial cells in tissue specimens. The authors emphasize that this is the first medical therapy to achieve an increase in limb perfusion that is equivalent to or greater than successful surgical or percutaneous intervention. Direct intramuscular gene transfer of plasmid DNA appears to effectively stimulate collateral vessel growth, despite the lower transfection efficiency that is usually associated with gene therapy in the absence of a viral vector. This result has implications for other clinical trials of gene therapy that use intramuscular naked DNA. The fact that Isner’s group could prepare the plasmid for human use in a university medical center laboratory dedicated to this purpose reveals why gene therapy has moved so rapidly from the laboratory to the clinic, in contrast to protein therapy, which requires expensive manufacturing facilities and years of scale-up effort. Although the plasmid was injected into muscle of the ischemic limb, VEGF levels were apparently elevated in the whole circulation, as evidenced by transient peaks of VEGF in the serum and by edema of the ischemic limb and in some patients, in the opposite limb. The increased collateral vessels, however, are localized to the ischemic limb and do not develop in other areas of the body. This may reflect in part the short half-life of VEGF in the circulation (minutes) as well as the upregulation of receptors for VEGF in ischemic tissue. …
Article
Vascular endothelial growth factor (VEGF), a major regulator of angiogenesis, binds to two receptor tyrosine kinases, KDR/Flk-1 and Flt-1. We now describe the purification and the expression cloning from tumor cells of a third VEGF receptor, one that binds VEGF165 but not VEGF121. This isoform-specific VEGF receptor (VEGF165R) is identical to human neuropilin-1, a receptor for the collapsin/semaphorin family that mediates neuronal cell guidance. When coexpressed in cells with KDR, neuropilin-1 enhances the binding of VEGF165 to KDR and VEGF165-mediated chemotaxis. Conversely, inhibition of VEGF165 binding to neuropilin-1 inhibits its binding to KDR and its mitogenic activity for endothelial cells. We propose that neuropilin-1 is a novel VEGF receptor that modulates VEGF binding to KDR and subsequent bioactivity and therefore may regulate VEGF-induced angiogenesis.