Sie sind auf Seite 1von 14

Cardiovascular Research 65 (2005) 550 – 563

www.elsevier.com/locate/cardiores

Review

The biology of vascular endothelial growth factors


Tuomas Tammela, Berndt Enholm, Kari Alitalo, Karri Paavonen*
Molecular/Cancer Biology Laboratory and Ludwig Institute for Cancer Research, Haartman Institute and Biomedicum Helsinki,
P.O. Box 63 (Haartmaninkatu 8) University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland

Received 30 September 2004; received in revised form 24 November 2004; accepted 2 December 2004
Time for primary review 18 days

Abstract

The discovery of the vascular endothelial growth factor (VEGF) family members VEGF, VEGF-B, placental growth factor (PlGF),
VEGF-C and VEGF-D and their receptors VEGFR-1, -2 and -3 has provided tools for studying the vascular system in development as
well as in diseases ranging from ischemic heart disease to cancer. VEGF has been established as the prime angiogenic molecule during
development, adult physiology and pathology. PlGF may primarily mediate arteriogenesis, the formation of collateral arteries from

Downloaded from by guest on December 22, 2014


preexisting arterioles, with potential future therapeutic use in for example occlusive atherosclerotic disease. VEGF-C and VEGF-D are
primarily lymphangiogenic factors, but they can also induce angiogenesis in some conditions. While many studies have addressed the role
of angiogenesis and the blood vasculature in human physiology, the lymphatic vascular system has until recently attracted very little
attention. In this review, we will discuss recent advances in angiogenesis research and provide an overview of the molecular players
involved in lymphangiogenesis.
D 2004 European Society of Cardiology. Published by Elsevier B.V. All rights reserved.

Keywords: Angiogenesis; Lymphangiogenesis; Growth factors; Endothelial receptors; VEGF

1. Introduction recently attracted very little attention. The vascular endo-


thelial growth factor (VEGF) family members VEGF,
Many human diseases are characterized by disorders of VEGF-B, placental growth factor (PlGF), VEGF-C and
the vasculature. In ischemic heart disease or peripheral VEGF-D bind their cognate receptors VEGFR-1, VEGFR-2
artery disease, insufficient blood vasculature leads to tissue and VEGFR-3 found on the vascular endothelium. VEGF is
ischemia. In cancer, tumor angiogenesis contributes to the prime hypoxia inducible angiogenic factor. VEGF
tumor growth and metastasis. Out of the many players in inhibition has been validated as clinical cancer therapy.
the angiogenesis field, the vascular endothelial growth Furthermore, several studies have reported beneficial effects
factors are by far the best characterized and several VEGFs of VEGF when used as a proangiogenic therapy in the
have already entered clinical use in a variety of human setting of tissue ischemia. PlGF may primarily mediate
conditions. While many studies have addressed the role of arteriogenesis, the formation of collateral arteries from
angiogenesis and the blood vasculature in human physiol- preexisting arterioles, and thus it has potential as a
ogy and pathology, the lymphatic vascular system has until therapeutic in for example occlusive atherosclerotic disease.
VEGF-C and VEGF-D are primarily lymphangiogenic
factors, inducing the growth of lymphatic vessels during
development as well as in the adult. Inhibition of VEGF-C
and VEGF-D signal transduction may prove crucial in the
* Corresponding author. Molecular/Cancer Biology Laboratory, Bio-
medicum Helsinki, P.O. Box 63 (Haartmaninkatu 8), 00014 University of
inhibition of lymphatic metastasis in cancer and stimulation
Helsinki, Finland. Tel.: +358 9 1912 5537; fax: +358 9 1912 5510. of this pathway may be effective in the treatment of
E-mail address: karri.paavonen@helsinki.fi (K. Paavonen). lymphedema.
0008-6363/$ - see front matter D 2004 European Society of Cardiology. Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.cardiores.2004.12.002
T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563 551

2. VEGF VEGF is first expressed mainly in the anterior portion of


mouse embryos and it directs the migration of VEGFR-1
VEGF binds VEGFR-1 and VEGFR-2 as well as and VEGFR-2 positive cells in embryonic tissues [15]. In
neuropilin-1 (Nrp-1) and Nrp-2; the latter are receptors general, VEGF expression is stronger at sites of active
for semaphorins, molecules involved in axonal guidance vasculogenesis and angiogenesis in embryos [16]. Homo-
during neuronal development [1,2]. VEGF induces pro- zygous VEGF knockout mice die at E8-E9 from defects in
liferation, sprouting, migration and tube formation of blood island formation, EC development and vascular
endothelial cells (ECs) [3]. VEGF is also a potent survival formation [13]. The levels of VEGF protein during develop-
factor for ECs during physiological and tumor angio- ment appear critical as mice lacking even a single VEGF
genesis and it has been shown to induce the expression of allele die at E11-E12, displaying defects in early vascular
antiapoptotic proteins in the ECs [4,5]. VEGF was development [13]. The different biological functions of
originally described as a permeability factor, as it increases VEGF isoforms were illustrated by studies on isoform-
permeability of the endothelium through the formation of specific VEGF knockout mice. Mice expressing only
intercellular gaps, vesico-vascular organelles, vacuoles and VEGF120 (homologue of human VEGF121) die soon after
fenestrations [6]. VEGF also causes vasodilatation through birth and those that survive succumb to ischemic cardiomy-
the induction of the endothelial nitric oxide synthase opathy and multiorgan failure [17]. Mice expressing only
(eNOS) and the subsequent increase in nitric oxide VEGF188 (human VEGF189) display impaired arteriolar
production [7,8]. development and approximately half die at birth [18]. Mice
Although VEGF acts mostly on ECs, it has been shown expressing only VEGF164 (human VEGF165) are viable and
to also bind VEGF receptors on hematopoietic stem cells healthy [18]. These studies underline the importance of
(HSCs), monocytes, osteoblasts and neurons [3]. Besides VEGF165 as the principal effector of VEGF action, with
angiogenesis, VEGF induces HSC mobilization from the intermediate diffusion and matrix-binding properties.
bone marrow, monocyte chemoattraction, osteoblast-medi- VEGF is strongly induced in hypoxic conditions via

Downloaded from by guest on December 22, 2014


ated bone formation and neuronal protection [3,9]. hypoxia inducible factor (HIF) regulated elements of the
Furthermore, VEGF stimulates inflammatory cell recruit- VEGF gene [19]. Constitutive degradation of hypoxia
ment and promotes the expression of proteases implicated inducible factor (HIF)-1a is blocked in hypoxia because
in pericellular matrix degradation in angiogenesis [10–12]. of the oxygen requirement of HIF prolyl hydroxylases,
Many cytokines including platelet-derived growth factor, followed by stabilization of HIF-1a and its heterodime-
epidermal growth factor, basic fibroblast growth factor and rization with HIF-1h, also called the aryl hydrocarbon
transforming growth factors induce VEGF expression in nuclear translocator (ARNT). These complexes then bind
cells [13]. hypoxia-responsive elements (HREs) in the promoters of
At least six VEGF isoforms of variable amino acid hypoxia inducible genes and initiate transcription of a set
number are produced through alternative splicing: of more than a hundred genes, including genes involved
VEGF121, VEGF145, VEGF165, VEGF183, VEGF189 and in glucose transport, glycolysis, and angiogenesis [19,20].
VEGF206 (Table 1) [3]. VEGF121, VEGF165 and VEGF189 Interestingly, Bartonella henselae, the causative agent of
are the major forms secreted by most cell types [14]. cat-scratch fever, can induce hypoxia via an intracellular
After secretion, VEGF121 may diffuse relatively freely in oxygen consumption mechanism, leading to VEGF
tissues, while approximately half of the secreted VEGF165 induction and an angiomatous tumor [21]. Examples of
binds to cell surface heparan sulfate proteoglycans other hypoxia-regulated genes include cyclooxygenase-2
(HSPGs). VEGF189 remains almost completely seques- (COX-2), MMP-2, VEGF and VEGFR-1 [19]. Deletion of
tered by HSPGs in the extracellular matrix making a HRE from the mouse VEGF gene promoter results in
HSPGs a reservoir of VEGF that can be mobilized via progressive motoneuron degeneration, presumably due to
proteolysis [3]. insufficient vascular perfusion of nervous tissue and

Table 1
A table showing the chromosomal localization, major mRNA transcript and major protein sizes of the VEGF receptors
Gene Sequence homology Chromosomal Splice Major mRNA Major protein
localization variants transcript size (kb) size (kDa)
VEGF 6p23.1 121,145, 165*, 3.7, 4.5 21
183*, 189*, 206*
VEGF-B 45% homology with VEGF-A 11q13 167*, 186 1.4 21, 30
VEGF-C 30% homology with VEGF-A165 4q34 – 2.4 20–21
VEGF-D 61% homology with VEGF-C; Xp22.31 – 2.2 20–21
31% with VEGF-A165
PlGF 42% homology with VEGF-A 14q24 131, 152*, 219 1.7, 1.2 38, 30
* Denotes splice variants that bind to heparan sulfate proteoglycans (HSPGs).
552 T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563

impaired motoneuron survival via loss of VEGF induction responses [44,45], while local administration of PlGF with
[22]. recombinant adenoviruses or as a recombinant protein
The skin has been widely used as a model for studying induces the formation of mature, leakage-resistant vessels
VEGF action in vivo; for example, transgenic mice over- in a macrophage-dependent manner [46–48].
expressing VEGF in the skin have abundant cutaneous
angiogenesis and an inflammatory skin condition resembling
psoriasis [23]. Overexpression of VEGF in mouse skin also 4. VEGF-B
accelerates experimental tumor growth [24]. In contrast, mice
with a targeted deletion of VEGF in the epidermis exhibit VEGF-B (also called VEGF-related factor/VRF) is a
delayed wound healing, while chemically induced skin ligand for VEGFR-1 and Nrp-1, and it can form heterodimers
papillomas developed less frequently in these animals [25]. with VEGF (Fig. 1) [49]. In humans, VEGF-B is expressed as
VEGF blocking monoclonal antibodies or VEGF receptor two different isoforms, VEGF-B167 and VEGF-B186 [50].
inhibition reduce the growth of experimental tumors in mice VEGF-B167 is nonglycosylated, binds HSPGs and is mostly
and humans [13,26,27]. In humans, VEGF is expressed in sequestered in the extracellular matrix while VEGF-B186 is
practically all solid tumors studied as well as in some O-glycosylated, and freely diffusible. In vivo, VEGF-B167 is
hematological malignancies [3]. In fact, correlations have the predominant form and is abundantly expressed in brown
been found between the level of VEGF expression, disease fat, in the myocardium and skeletal muscle (Fig. 2) [49].
progression and survival [28]. The precise role of VEGF-B in vivo is not known. As
The effects of VEGF on the lymphatic vasculature VEGF-B is highly expressed in striated muscle, myocardium
have also been recently studied. Adenoviral overexpres- and brown fat [51,52], its function may be linked to high
sion of the murine VEGF164 in the skin induced formation cellular energy metabolism. Interestingly, at least in one
of giant lymphatic vessels [29], while another study genetic background mice deficient in VEGF-B had smaller
employing the human VEGF165 isoform reported only hearts and impaired recovery after experimental myocardial

Downloaded from by guest on December 22, 2014


dilatation of cutaneous lymphatics (Fig. 3A) [30]. How- infarctions suggesting that the regeneration of coronary
ever, VEGF did not induce lymphangiogenesis in a collaterals through arteriogenesis could at least in part be
number of other tissue types [31–34]. The lymphangio- dependent on VEGF-B [53]. In another genetic background, a
genic effects of VEGF may be linked to the recruitment prolonged PQ interval was observed in the electrocardio-
of inflammatory cells, such as macrophages, which grams of VEGF-B gene targeted mice [54]. Furthermore, one
express VEGFR-1 and secrete lymphangiogenic factors report has claimed that VEGF-B knockout mice fail to
[35–37]. At least in midgestation mouse embryos, VEGF- develop pulmonary hypertension in response to hypoxia [55].
C but not VEGF had the capacity to induce migration of Conversely, overexpression of VEGF-B in the lung protected
endothelial cells committed to the lymphatic endothelial rats from hypoxic pulmonary hypertension in a manner
lineage [38]. comparable to VEGF [56]. VEGF-B knockout mice also
display reduced angiogenic responses in collagen-induced
arthritis, suggesting a role for VEGF-B in inflammatory
3. Placental growth factor angiogenesis [57]. Correspondingly, VEGF-B may have
subtle angiogenic effects, as overexpression of VEGF-B in
PlGF is predominantly expressed in the placenta, heart vivo by injection of recombinant plasmids in a mouse model
and lungs [39]. PlGF homodimers bind VEGFR-1 and of hind limb ischemia induced angiogenesis [58].
Nrp-1, while PlGF heterodimerization with VEGF may
also occur. Activation of VEGFR-1 by either PlGF or
VEGF induced different gene expression profiles and 5. VEGF-C
phosphorylation of distinct tyrosine residues in the tyrosine
kinase (TK) domain of VEGFR-1 [40], while combined Where VEGF-A and VEGF-B isoforms are formed
administration of these factors enhanced VEGF driven through alternative splicing, different forms of VEGF-C
angiogenesis [41]. This is probably due to the fact that and VEGF-D are the result of proteolytic processing.
PlGF/VEGF heterodimers bind VEGFR-2 and also VEGF-C is produced as a precursor protein, which is
VEGFR-1/VEGFR-2 heterodimers in vitro [42]. Of the activated by intracellular secretory proprotein convertases
three reported human isoforms of PlGF (PlGF-1, -2 and furin, PC5 and PC7 [59,60]. The secreted, VEGFR-3
-3), only PlGF-2 binds HSPGs [42]. PlGF knockout mice binding 31/29 kD subunits of VEGF-C are bound together
do not have an apparent phenotype. However, these mice by disulphide bonds, but the factor is further proteolyzed
recover poorly from experimental myocardial infarction in the extracellular environment by plasmin and other
and exhibit impaired collateral formation in response to proteases to generate a 21 kD non-disulfide-linked
hind limb ischemia [43]. Overexpression of PlGF in the homodimeric protein with high affinity for both VEGFR-
skin of transgenic mice results in a hypervascular 2 and VEGFR-3. The mature form of VEGF-C induces
phenotype with increased inflammatory and permeability mitogenesis, migration and survival of ECs [61]. During
T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563 553

Fig. 1. The VEGF family ligands and their receptors. VEGF, VEGF-B and PlGF bind VEGFR-1 and VEGFR-2 on the blood vascular endothelium. VEGF-C

Downloaded from by guest on December 22, 2014


and VEGF-D primarily bind VEGFR-3 on lymphatic endothelium.

development, VEGF-C is expressed along with its receptor E17.5 [38]. Loss of one VEGF-C allele results in
VEGFR-3 predominantly in regions where lymphatic lymphedema characterized by hypoplasia of the cutaneous
vessels develop [38,62]. The expression then decreases in lymphatic vessels indicating that the VEGF-C protein
most tissues, remaining high in the lymph nodes [63]. concentration is critical for the development of the
VEGF-C induces selective lymphangiogenesis without lymphatic vasculature [38]. However, VEGF-C is not
accompanying angiogenesis as demonstrated by early essential for the development of blood vessels unlike its
experiments in the chick chorioallantoic membrane assay receptors VEGFR-2 and VEGFR-3. Sprouting of endothe-
and in transgenic mice overexpressing VEGF-C in the skin lial cells committed to the lymphatic endothelial lineage in
[64,65]. Adenoviral VEGF-C gene transduction has been VEGF-C gene targeted mice could be rescued by applica-
shown to induce growth of functional lymphatic vessels in tion of recombinant VEGF-C and to a lesser degree by
several different animal models (Fig. 3C) [30,66]. In vivo, VEGF-D, but not with VEGF. This indicates that develop-
VEGFR-3 activation is sufficient to induce lymphangio- ment of the lymphatic vessels is dependent on VEGFR-3
genesis whereas the increase in blood vascular perme- and VEGF-C in a nonredundant fashion [38].
ability induced by VEGF-C and VEGF-D is mediated by VEGF-C mRNA transcription was induced in ECs in
VEGFR-2 [30,67]. response to proinflammatory cytokines [68]. The regulation
Mice lacking both VEGF-C alleles fail to develop of VEGF-C mRNA transcription by these cytokines
lymphatic vessels and succumb to tissue edema at E15.5– indicates that VEGF-C could regulate lymphatic vessel
function during inflammation, reflecting the role of the
lymphatic vasculature in the control of immune function and
VEGF165 leukocyte trafficking. The promoter of the VEGF-C gene
PIGF has been shown to contain putative NF-nB binding sites that
VEGF-B could mediate the activation of VEGF-C mRNA tran-
VEGF-C scription by proinflammatory cytokines [69]. A recent
report demonstrated that prostaglandin E2 mediated activa-
VEGF-D
tion of COX-2 increased VEGF-C mRNA transcription
40 30 20 10 0 suggesting that prostanoids could induce VEGF-C driven
lymphangiogenesis [70]. Recent reports have shown
Percentage of residue substitution
that inflammatory reactions in human kidney transplants
Fig. 2. A dendrogram showing the relationships VEGF family members undergoing rejection are accompanied by abundant lym-
estimated by the amino acid residue substitution analysis. phangiogenesis and that VEGFR-3 signaling contributes to
554 T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563

Downloaded from by guest on December 22, 2014


Fig. 3. Blood and lymphatic vascular effects of various adenovirally expressed human VEGFs in the mouse skin two weeks after treatment. (A) VEGF165
induces a prominent angiogenic response (blood vessels stained with antibodies for PECAM-1/CD31, green), accompanied by enlargement of lymphatic
vessels (stained with antibodies for the lymphatic marker LYVE-1, red). (B) VEGF-B167 does not induce either angiogenesis or lymphangiogenesis. (C) VEGF-
C is a powerful inducer of lymphangiogenesis, while its angiogenic activity is weak. (D) The AdLacZ control corresponds to nonstimulated, normal skin.

reactive lymphadenitis [71]. In a rabbit cornea model of stromal lymphangiogenesis and tumor cell invasion into
inflammatory angiogenesis and lymphangiogenesis, selec- lymphatic vessels [61,73,74]. Furthermore, overexpression
tive depletion of macrophages blocked lymphangiogenesis of VEGF-C in pancreatic islet cell tumors of transgenic mice
demonstrating that inflammatory cells can mediate the induced lymphangiogenesis and promoted lymph node
formation of lymphatic vessels [37]. The angiogenic and metastasis [75]. Interestingly, neutralization of VEGF-C
lymphangiogenic responses were blocked by a VEGF and VEGF-D in an experimental tumor model by systemic
inhibitor. The recruited macrophages expressed abundant overexpression of a soluble VEGFR-3 fused to the Fc domain
amounts of VEGF-C and VEGF-D, suggesting that the of immunoglobulin g chain (VEGFR-3-Ig) inhibited lym-
lymphangiogenic effect of VEGF is indirect and most likely phangiogenesis and lymph node metastasis [76,77]. Peritu-
mediated by inflammatory cytokines that induce the moral lymphatics were recently found to originate from the
expression of lymphangiogenic factors in the inflammatory preexisting lymphatic vasculature while circulating endothe-
infiltrate. In this regard, it is interesting to note that COX-2 lial progenitor cells did not contribute to the process [78].
up-regulated VEGF-C and promoted lymphangiogenesis in Although enlarged lymphatic vessels at the tumor edge have
human lung adenocarcinoma [70]. VEGF-C is also highly been reported in human cancers, functional intratumoral
expressed in arthritic joint synovium in patients with lymphatic vessels have not been reported [79]. Interestingly,
rheumatoid arthritis in the absence of lymphangiogenesis several clinical studies of cancer patients have shown a
per se, suggesting that the lymphangiogenic response is positive correlation between VEGF-C expression and lym-
insufficient in this disorder or that the induced VEGF-C phatic invasion, lymphatic metastasis and patient survival
participates in other functions [72]. [80]. However, although breast cancers frequently metasta-
In experimental models, tumor cells overexpressing size to the lymph nodes, at least one study did not detect
VEGF-C or VEGF-D induce peritumoral and in some cases tumor lymphangiogenesis in breast cancer [81].
T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563 555

6. VEGF-D mice die at E8.5 due to disorganization of blood vessels


and overgrowth of endothelial cells [3]. Although VEGFR-
Like VEGF-C, the human VEGF-D is processed in its N- 1 gene targeted mice do not survive, mice lacking only the
terminal and C-terminal ends; the mature form binds to and intracellular kinase (TK) domain are normal except for
activates VEGFR-2 and VEGFR-3, is mitogenic for EC and slightly impaired angiogenesis during pathologic condi-
angiogenic as well as lymphangiogenic in vivo [61]. Mouse tions [96,97]. Activation of the VEGFR-1 TK domain is a
VEGF-D binds only VEGFR-3, indicating a different role requisite for monocyte migration [98]. Moreover, VEGFR-
for VEGF-D in mice [82]. VEGF-D is present in most 1 mediated angiogenesis as well as arteriogenesis have
human tissues, most abundantly in the lung and skin during been shown to be dependent on monocytes [47,48], and
embryogenesis [83]. In experimental tumors VEGF-D angiogenesis during experimental tumor growth is at least
increases lymphatic vessel growth and lymphatic metastasis partially inhibited by anti-VEGFR-1 antibodies [47].
[84]. VEGF-D is expressed by melanoma cells and has been VEGFR-1 signaling is also involved in the recruitment
proposed to have a role in tumor angiogenesis and and survival of bone marrow derived progenitor cells
lymphangiogenesis in this tumor [85]. VEGF-D, along with [99,100].
VEGF-C, has recently been shown to be involved in A naturally occurring soluble form of VEGFR-1
lymphatic metastasis in experimental intrathoracic lung (sVEGFR-1), consisting of the extracellular part of
cancer [86]. In humans, VEGF-D has been shown to be of VEGFR-1, that is able to inhibit VEGF action has been
prognostic value for lymphatic vessel invasion and also described in humans and mice [3]. sVEGFR-1 has recently
survival in certain human cancers [80]. The fully processed been linked to the development of gestational toxemia and
form of VEGF-D is able to induce strong angiogenesis in preeclampsia [101]. Women with preeclampsia have ele-
addition to lymphangiogenesis in the rabbit hind limb vated circulating sVEGFR-1 levels and decreased circulat-
muscle [32,87]. ing levels of PlGF and VEGF possibly leading to EC
dysfunction in the maternal and placental vasculatures

Downloaded from by guest on December 22, 2014


[102].
7. VEGFR-1

VEGFR-1 (fms-like tyrosine kinase, Flt-1) is composed 8. VEGFR-2


of seven extracellular immunoglobulin (Ig) homology
domains, a single transmembrane region and an intra- The overall structure of VEGFR-2 (kinase-insert
cellular tyrosine kinase (TK) domain that is interrupted by domain receptor, KDR/fetal liver kinase, Flk-1) is similar
a kinase-insert domain (Fig. 1, Table 2) [88]. VEGFR-1 to that of VEGFR-1 (Table 2) [103,104]. VEGFR-2 binds
binds VEGF, VEGF-B and PlGF with high affinity. VEGF, VEGF-C and VEGF-D. Although the binding
VEGFR-1 by itself transmits only weak mitogenic signals affinity of VEGF towards VEGFR-2 is lower than that
in ECs [3], but it can heterodimerize with VEGFR-2 for VEGFR-1, selective activation of VEGFR-1 and
forming a complex with stronger signaling properties than VEGFR-2 has shown that VEGFR-2 is the primary
VEGFR-1 or VEGFR-2 homodimers [43,89] (Table 2). receptor transmitting VEGF signals in endothelial cells
VEGFR-1 is expressed in ECs as well as osteoblasts, [105–107]. The expression of VEGFR-2 is autoregulated:
monocytes/macrophages, pericytes, placental trophoblasts, VEGF, VEGF-C and VEGF-D up-regulate the expression
renal mesangial cells and also in some hematopoietic stem of VEGFR-2 [84,108]. In addition to the ECs, VEGFR-2 is
cells [3,90]. VEGFR-1 expression is up-regulated during also expressed on neuronal cells, osteoblasts, pancreatic
angiogenesis and also by hypoxia, unlike that of VEGFR-2 duct cells, retinal progenitor cells, megakaryocytes and
and VEGFR-3 [91,92]. hematopoietic stem cells [3,109]. During development
During development, VEGFR-1 is first expressed in VEGFR-2 is expressed by the primitive endoderm,
angioblasts and in the endothelium, although less strongly embryonic angioblasts and in the blood islands as well
than VEGFR-2; VEGFR-1 expression subsides during later as in angiogenic vessels [93]. VEGFR-2 gene targeted
embryonic development [93–95]. VEGFR-1 gene targeted mice die at E8.5-E9.5 due to lack of development of the
blood islands, embryonic vasculature and hematopoietic
cells [110,111]. VEGFR-2 expression is down-regulated in
Table 2 the adult blood vascular ECs, and is again up-regulated in
A table showing the chromosomal localization, major mRNA transcripts the endothelium of angiogenic blood vessels [92]. On the
and major protein sizes of VEGF receptors other hand, sequestration of VEGF results in down-
Gene Chromosomal Major mRNA Major protein regulation of VEGFR-2 and in apoptotic death of some
localization transcript size (kb) size (kDa) capillary endothelial cells in vivo [112]. Moreover,
VEGFR-1 13q12-q13 7.5, 8.0 180 VEGFR-2 may be associated with integrin-dependent
VEGFR-2 4q11-q12 5.8, 7.0 230 migration of ECs, as it forms a complex with integrin
VEGFR-3 5q33-qter 4.5, 5.8 195
aVh3 upon binding VEGF [113,114]. An interaction
556 T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563

between VEGFR-2 and VE-cadherin, a cell–cell adhesion arterial/venous identity of ECs [137]. Nrp-2 is expressed
molecule has also been described [115]. also on lymphatic ECs, and mutated Nrp-2 induces
abnormalities in the formation of small lymphatic vessels
and lymphatic capillaries in mice [138]. Nrp-1 gene
9. VEGFR-3 targeted mice die at E13 from vascular defects such as
insufficient development of yolk sac vascular networks,
VEGFR-3 (fms-like tyrosine kinase 4, Flt4) has only six deficient neural vascularization and transposition of large
Ig-homology domains as the fifth Ig-homology domain is vessels [139,140]. It is thought that Nrp-1 is required for
proteolytically cleaved soon after biosynthesis and the cardiovascular development because it regulates VEGF165
resulting polypeptide chains remain linked via a disulfide levels [139]. While Nrp-2 gene targeted mice have normal
bond (Table 2) [116,117]. In humans, alternative splicing blood vasculature, the compound Nrp-1/Nrp-2 knockout
of the VEGFR-3 gene generates two isoforms of VEGFR- mouse has a phenotype resembling that of the VEGF and
3 that differ in their C-termini [118]. VEGFR-3 binds VEGFR-2 gene targeted mice [141].
VEGF-C and VEGF-D [119,120]. VEGFR-3 is present on
all endothelia during development but in the adult it
becomes restricted to lymphatic ECs and certain fenes- 11. Disorders of the vascular system linked to the
trated blood vascular ECs [121,122]. VEGFR-3 is up- VEGFs
regulated on blood vascular ECs in pathologic conditions
such as in vascular tumors and in the periphery of solid 11.1. Therapeutic angiogenesis/arteriogenesis in ischemic
tumors [92,123]. Interestingly, EC contact with SMCs has heart disease and peripheral vascular disease
been shown to down-regulate VEGFR-3 in ECs [124],
suggesting that VEGFR-3 signaling is important in nascent Angiogenic growth factors may be useful for attempts
blood vessels, and it becomes redundant as the vessels to increase collateral vessel formation in ischemic heart

Downloaded from by guest on December 22, 2014


mature. Indeed, VEGFR-3 gene targeted mice exhibit a disease (IHD) or critical limb ischemia/claudication or in
dramatic blood vascular phenotype, with embryonic lethal- the treatment of diabetic neuropathy of the lower
ity at E9.5 from defective remodeling of the primary extremities [142]. Reduction of blood flow in thromboem-
vascular plexus and disturbed hematopoiesis [125,126]. bolic disease leads to collateral formation via arterio-
Transgenic mice overexpressing a soluble VEGFR-3-Ig genesis, a process during which preexisting arterioles
fusion protein in the skin lack dermal lymphatic vessels enlarge, leading to a 10- to 20-fold increase in blood flow
and have hypoplastic deeper lymphatic vessels, although [143]. Arteriogenic pathways such as the PlGF-VEGFR-1
the phenotype is less pronounced as the mice age [127]. pathway recruit monocytes and act directly on ECs and
Missense mutations in VEGFR-3 have been linked to SMCs by inducing the growth of these cell populations.
hereditary lymphedema in humans and also to a similar Recently, PlGF was reported to induce formation of vascular
condition in a mouse model of lymphedema [128–130]. collaterals in experimental models of myocardial infarction
More recently, VEGFR-3 has also been shown to modulate and lower limb ischemia in mice by amplification of VEGF
adaptive immunity in an experimental model of corneal signaling [40,43,47]. Thus, VEGFR-1 agonists are poten-
transplantation [131]. tially useful in restoration of blood perfusion in settings of
tissue ischemia.
Human trials utilizing proangiogenic therapy have
10. Neuropilins arrived at some successful endpoints [142]. Attempts at
treating inoperable IHD have been made with gene transfer
The neuropilins, Nrp-1 and Nrp-2, have roles in of VEGF by adenovirus and naked plasmid injections
immunology and neuronal development but they are also [144,145]. Preliminary reports targeting ischemic skeletal
involved in angiogenesis [132,133]. Neuropilins bind class muscle suggested that in patients with critical limb
3 semaphorins, which are secreted molecules that mediate ischemia/claudication, injection of VEGF plasmid may
repulsive signals during neuronal axon guidance [1,2,134]. induce the growth of functional collateral vessels [146]. In
Nrp-1 also binds VEGF, VEGF-B and PlGF while Nrp-2 the recent regional angiogenesis with VEGF (RAVE)
binds VEGF, VEGF-C and PlGF [132]. Nrp-1 acts as a randomized trial patients with claudication were treated
co-receptor enhancing VEGF-VEGFR-2 interactions, with adenoviral delivery of VEGF121 but the researchers
forming complexes with VEGFR-1 and augmenting tumor failed to see clinical benefits [147]. However, VEGF
angiogenesis in vivo [135]. Overexpression of Nrp-1 in driven angiogenesis in progressive ischemic disease may
chimeric mice leads to excessive formation of capillaries not be sufficient to restore the vascular perfusion because
and blood vessels and hemorrhages in addition to cardiac the formed vessels are not sufficiently functional, and even
malformations [136]. In chick embryos, endothelial Nrp-1 angiomas may develop [148]. Because of the permeability
expression is mostly restricted to arteries, whereas Nrp-2 side effects of VEGF, it has been suggested that a
primarily marks veins, indicating a role in mediating the combined approach with several growth factors such as
T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563 557

VEGF and angiopoietins, platelet-derived growth factors, ever, the capacity of bone marrow derived EPCs to
or the more arteriogenic PlGF may lead to better results incorporate into the vessel wall and transdifferentiate into
[42]. mature ECs in vivo has also been questioned [159–161].

11.2. The role of VEGF family growth factors in athero- 11.4. Tumor angiogenesis
sclerotic plaque formation
Blood vessels in tumors display several distinct features
Atherosclerotic plaque progression is associated with that have implications for tumor biology and treatment.
inflammatory angiogenesis through increased secretion of Blood vessels in tumors have a chaotic structure and are
angiogenic growth factors such as VEGF and basic leaky [162]. As a consequence, blood flow in the tumor is
fibroblast growth factor (bFGF) [149]. Plaque angiogenesis sluggish and the interstitial fluid pressure is high [162]. The
results in atherosclerotic plaque growth, increased plaque sluggish blood flow results in hypoxic regions and
instability and with the increase in the number of the perpetuates VEGF production while high interstitial fluid
adventitial vasa vasora, an increased risk of intraplaque pressure within the tumor hampers delivery of therapeutic
hemorrhage [150]. These factors contribute to the growing agents [162]. Since tumor growth is angiogenesis depend-
instability of the atherosclerotic plaque. ent, therapeutic targeting of the tumor vasculature is an
It is well known that plaque progression is associated attractive alternative or adjunct to conventional therapy. ECs
with inflammatory cell recruitment and deposition of in tumors express a number of molecules that are unique to
oxidized low-density lipoprotein (OxLDL). OxLDL blood vessels undergoing angiogenesis, such as VEGFR-2
increases VEGF production and VEGF secretion by and integrin aVh3 that can be targeted for therapeutic
inflammatory cells in vitro [151]. The number of VEGF purposes. Furthermore, work by Ruoslahti et al. has
positive cells has been shown to correlate with the degree of demonstrated that angiogenic blood vessel endothelium
atherosclerotic plaque vascularization [152]. Experimental expresses certain peptides/integrins on their cell surface and

Downloaded from by guest on December 22, 2014


atherosclerosis can be induced with VEGF and experimental these markers may serve as bzip codesQ for targeting
atherosclerotic changes can be reduced with antiangiogenic antiangiogenic molecules to tumor vessel endothelium
therapy [153,154]. In a model of experimental cardiac [163].
allografts, VEGF was found to mediate the progression of An important step in antiangiogenic cancer therapy was
arteriosclerosis [155]. However, recent results have sug- recently taken when the anti-VEGF blocking antibody
gested that the angiopoietins, another family of angiogenic bevacizumab (Genentech, USA) showed remarkable
molecules that bind and activate Tie2 tyrosine kinase results in the treatment of metastatic colorectal cancer,
receptors on ECs, could actually prevent some of the and it has recently been approved by the FDA for
inflammatory changes in the vessel wall [156]. treatment of metastatic colorectal cancer [27,164]. This
may only mark the beginning of antiangiogenic therapy
11.3. The role of endothelial progenitor cells in vascular for tumors, as at present bevacizumab is being evaluated
pathology in Phase III trials of metastatic breast cancer, nonsmall
cell lung cancer, pancreatic cancer and renal cell
The blood circulation has been suggested to contain carcinoma (see www.clinicaltrials.gov).
endothelial progenitor cells (EPCs) that incorporate into
newly formed vessels at sites of active angiogenesis [157]. 11.5. Some other human diseases involving angiogenesis
The EPCs are recruited to sites of angiogenesis where they
differentiate in situ into mature endothelial cells much like Excessive ocular neovascularization contributes to visual
during the embryonic vasculogenesis. EPCs express a loss in retinopathy of prematurity and diabetic retinopathy
variety of endothelial cell surface markers including platelet as well as in age-related macular degeneration. Retinal
endothelial cell adhesion molecule-1, von Willebrand factor hypoxia and excessive secretion of angiogenic factors like
(vWF) and VEGF receptors. EPCs are mobilized from the VEGF leads to inappropriate retinal neovascularization and
bone marrow by cytokines such as macrophage chemo- hemorrhages [165]. Experimental retinal neovascularization
attractant protein-1 (MCP-1), transforming growth factor- can be inhibited by antiangiogenic therapy [166–168].
beta (TGF-h), VEGF and PlGF [157]. Skeletal muscle has Phase III clinical trials are underway to treat wet age-related
also been suggested to contain cells with stem cell proper- macular degeneration with ranibizumab, an antibody frag-
ties that can contribute to angiogenesis, raising the ment to VEGF (Genentech USA). An aptamer inhibitor
possibility that tissues can contain resident angioblasts specific for the VEGF165 isoform (pegaptanib, EyeTech,
[158]. EPCs have been reported to incorporate into forming USA) has also entered phase II–III clinical trials with similar
blood vessels during physiological angiogenesis such as indications [169].
during the female reproductive cycle, during pathologic Psoriasis is a chronic inflammatory skin disorder
angiogenesis in experimental solid tumors or in the limb characterized by dermal angiogenesis and overexpression
muscles or the myocardium during ischemia [157]. How- of angiogenic factors such as VEGF [170]. In fact, trans-
558 T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563

genic mice with sustained overexpression of murine VEGF [178–180]. Several reports have documented a correlation
in the skin have many of the classical features of psoriasis, between expression levels of the lymphangiogenic factors
and treatment with VEGF Trap, a VEGF antagonist VEGF-C and VEGF-D and lymphatic metastasis in cancer
(Regeneron, USA), was shown to reverse this phenotype patients [80]. Thus the blocking of VEGF-C and VGEF-D
[23]. High VEGF levels are also involved in the pathologic signals with for example a soluble VEGFR-3-Ig fusion
angiogenesis observed in endometriosis and possibly also protein could be an attractive approach for inhibition of
other gynecological disorders such as uterine bleeding lymph node metastasis in human cancer patients, as already
disorders [171]. indicated by the results from preclinical studies [76,77].

11.6. Lymphedemas
12. Conclusion
In lymphedema, lymphatic drainage is impaired and
protein-rich fluid accumulates in the subcutaneous tissue The vascular endothelial growth factors and their recep-
leading to fibrosis, impaired immune responses and fatty tors play a paramount role in the development of the vascular
degeneration of the connective tissue. Lymphedemas are system, via vasculogenic and angiogenic mechanisms, as
classified as primary, congenital lymphedemas and secon- well as in the formation of the lymphatic vascular system.
dary or acquired lymphedemas. Secondary lymphedema is Later in life, these molecules are required for processes
usually caused by filariasis or by iatrogenic trauma such involving tissue repair, such as wound healing and the cyclic
as radiation therapy, surgery or infection [172]. Primary reconstitution of the female endometrium. Aberrant angio-
lymphedemas develop at birth or during adolescence. genesis is a key mechanism in the pathophysiology of, e.g.,
Congenital hereditary lymphedema, or Milroy disease, is atherosclerosis as well as tumor growth, while both angio-
characterized by hypoplastic superficial lymphatic vessels, genesis and lymphangiogenesis may contribute to tumor
while the lymphatics in Meige disease, or late-onset metastasis. Controlling these processes with targeted molec-

Downloaded from by guest on December 22, 2014


congenital lymphedema, appear enlarged and are dysfunc- ular therapies is therefore at the heart of research directed at
tional [173]. The genetic defects underlying several disease treatment. While proangiogenic therapies could be
primary lymphedemas have recently been characterized. employed in the treatment of various ischemic diseases,
In some patients with Milroy disease, missense mutations generation of new lymphatic vessels could help patients with
found in the TK domain of VEGFR-3 inhibit the function lymphedema. Meanwhile, a renaissance seems to be unrav-
of this receptor with resulting lymphedema [129]. In eling around the VEGF growth factors, as novel functions for
lymphedema distichiasis, mutations in the FOXC2 gene them continue to be discovered outside the scope of vascular
have been identified that lead to abnormal mural cell biology.
coating of lymphatic vessels and lack of lymphatic valves
[174,175]. Other human diseases involving lymphedema
with putative, as yet uncharacterized molecular mecha- Acknowledgements
nisms include Turner syndrome and cholestasis-lymphe-
dema. The authors wish to thank Dr. Marika K7rkk7inen for
helping with the graphics. The authors have been supported
11.7. Tumor lymphangiogenesis by the Human Frontier Science Program, EU (Lymphangio-
genomics LSHG-CT-2004-503573 and AngioNet QLG1-
The intravasation of tumor cells into lymphatic vessels is CT-2001-01172), the Novo Nordisk Foundation, the Finnish
one of the first steps in lymphatic metastasis. Lymphangio- Academy of Sciences, the Farmos Research Foundation, the
genesis around solid tumors could promote lymphatic Ida Montin Foundation, the Paulo Foundation, the Bio-
metastasis by providing a larger target for the intravasation medicum Helsinki Foundation and the Finnish Medical
of tumor cells [80]. Although enlarged lymphatic vessels at Foundation.
the tumor edge have occasionally been reported in human
cancers, functional intratumoral lymphatic vessels have not
been reported [79,176]. One explanation could be that the References
high interstitial fluid pressure in tumors compresses
[1] Kolodkin AL, Levengood DV, Rowe EG, Tai YT, Giger RJ, Ginty
lymphatic vessels. According to another hypothesis, com- DD. Neuropilin is a semaphorin III receptor. Cell 1997;90:753 – 62.
pressed lymphatic vessels seen in the tumor stroma did not [2] Chen H, Chedotal A, He Z, Goodman CS, Tessier-Lavigne M.
arise by lymphangiogenesis but are preexisting lymphatic Neuropilin-2, a novel member of the neuropilin family, is a high
vessels that were engulfed by the expanding tumor [177]. affinity receptor for the semaphorins Sema E and Sema IV but not
Although lymphatic metastasis is not necessarily dependent Sema III. Neuron 1997;19:547 – 59.
[3] Ferrara N, Gerber H-P, LeCouter J. The biology of VEGF and its
on lymphangiogenesis [176], tumor lymphangiogenesis has receptors. Nat Med 2003;9:669 – 76.
been reported to be a prognostic factor in cutaneous [4] Benjamin LE, Keshet E. Conditional switching of vascular endo-
melanoma and squamous cell cancers of the head and neck thelial growth factor (VEGF) expression in tumors: induction of
T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563 559

endothelial cell shedding and regression of hemangioblastoma-like vascular hyperpermeability and accelerated tumor development.
vessels by VEGF withdrawal. Proc Natl Acad Sci U S A Oncogene 1998;17:303 – 11.
1997;94:8761 – 6. [25] Rossiter H, Barresi C, Pammer J, Rendl M, Haigh J, Wagner EF, et
[5] Gerber H-P, Dixit V, Ferrara N. Vascular endothelial growth factor al. Loss of vascular endothelial growth factor a activity in murine
induces expression of the antiapoptotic proteins Bcl-2 and A1 in epidermal keratinocytes delays wound healing and inhibits tumor
vascular endothelial cells. J Biol Chem 1998;273:13313 – 6. formation. Cancer Res 2004;64:3508 – 16.
[6] Bates DO, Hillman NJ, Williams B, Neal CR, Pocock TM. [26] Sepp-Lorenzino L, Rands E, Mao X, Connolly B, Shipman J,
Regulation of microvascular permeability by vascular endothelial Antanavage J, et al. A novel orally bioavailable inhibitor of kinase
growth factors. J Anat 2002;200:581 – 97. insert domain-containing receptor induces antiangiogenic effects and
[7] Hood JD, Meininger CJ, Ziche M, Granger HJ. VEGF upregulates prevents tumor growth in vivo. Cancer Res 2004;64:751 – 6.
ecNOS message, protein, and NO production in human endothelial [27] Kabbinavar F, Hurwitz HI, Fehrenbacher L, Meropol NJ,
cells. Am J Physiol 1998;274:H1054 – 8. Novotny WF, Lieberman G, et al. Phase II, randomized trial
[8] Kroll J, Waltenberger J. A novel function of VEGF receptor-2 comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV)
(KDR): rapid release of nitric oxide in response to VEGF-A with FU/LV alone in patients with metastatic colorectal cancer. J
stimulation in endothelial cells. Biochem Biophys Res Commun Clin Oncol 2003;21:60 – 5 [comment].
1999;265:636 – 99. [28] Ferrara N. Role of vascular endothelial growth factor in physio-
[9] Storkebaum E, Lambrechts D, Carmeliet P. VEGF: once regarded as logical and pathological conditions: therapeutic implications. Semin
a specific angiogenic factor, now implicated in neuroprotection. Oncol 2002;29:10 – 4.
BioEssays 2004;26:943 – 54. [29] Nagy JA, Vasile E, Feng D, Sundberg C, Brown LF, Detmar MJ,
[10] Pepper MS, Ferrara N, Orci L, Montesano R. Vascular endothelial et al. Vascular permeability factor/vascular endothelial growth
growth factor (VEGF) induces plasminogen activators and plasmi- factor induces lymphangiogenesis as well as angiogenesis. J Exp
nogen activator inhibitor-1 in microvascular endothelial cells. Med 2002;196:1497 – 506.
Biochem Biophys Res Commun 1991;181:902 – 6. [30] Saaristo A, Veikkola T, Enholm B, Hytonen M, Arola J, Pajusola
[11] Unemori EN, Ferrara N, Bauer EA, Amento EP. Vascular endothelial K, et al. Adenoviral VEGF-C overexpression induces blood vessel
growth factor induces interstitial collagenase expression in human enlargement, tortuosity, and leakiness but no sprouting angio-
endothelial cells. J Cell Physiol 1992;153:557 – 62. genesis in the skin or mucous membranes. FASEB J 2002;16:
[12] Mandriota SJ, Seghezzi G, Vassalli J-D, Ferrara N, Wasi S, Mazzieri 1041 – 9.

Downloaded from by guest on December 22, 2014


R, et al. Vascular endothelial growth factor increases urokinase [31] Kubo H, Cao R, Brakenhielm E, Makinen T, Cao Y, Alitalo K.
receptor expression in vascular endothelial cells. J Biol Chem Blockade of vascular endothelial growth factor receptor-3 signaling
1995;270:9709 – 16. inhibits fibroblast growth factor-2-induced lymphangiogenesis in
[13] Ferrara N. Vascular endothelial growth factor: basic science and mouse cornea. Proc Natl Acad Sci U S A 2002;99:8868 – 73.
clinical progress. Endocr Rev 2004;25:581 – 611. [32] Rissanen TT, Markkanen JE, Gruchala M, Heikura T, Puranen A,
[14] Robinson CJ, Stringer SE. The splice variants of vascular Kettunen MI, et al. VEGF-D is the strongest angiogenic and
endothelial growth factor (VEGF) and their receptors. J Cell Sci lymphangiogenic effector among VEGFs delivered into skeletal
2001;114:853 – 65. muscle via adenoviruses. Circ Res 2003;92:1098 – 106.
[15] Hiratsuka S, Kataoka Y, Nakao K, Nakamura K, Morikawa S, [33] Cao R, Eriksson A, Kubo H, Alitalo K, Cao Y, Thyberg J.
Tanaka S, et al. VEGF-A is involved in guidance of VEGF-receptor- Comparative evaluation of FGF-2-, VEGF-A-, and VEGF-C-induced
positive cells to the anterior portion of early embryos. Mol Cell Biol angiogenesis, lymphangiogenesis, vascular fenestrations, and per-
2005;25:355 – 63. meability. Circ Res 2004;94:664 – 70.
[16] Weinstein BM. What guides early embryonic blood vessel forma- [34] Lee CG, Link H, Baluk P, Homer RJ, Chapoval S, Bhandari V, et al.
tion? Dev Dyn 1999;215:2 – 11. Vascular endothelial growth factor (VEGF) induces remodeling and
[17] Carmeliet P, Ng YS, Nuyens D, Theilmeier G, Brusselmans K, enhances T(H)2-mediated sensitization and inflammation in the lung.
Cornelissen I, et al. Impaired myocardial angiogenesis and ischemic Nat Med 2004;10:1095 – 103.
cardiomyopathy in mice lacking the vascular endothelial growth factor [35] Clauss M, Weich H, Breier G, Knies U, Rockl W, Waltenberger J,
isoforms VEGF164 and VEGF188. Nat Med 1999;5:495 – 502. et al. The vascular endothelial growth factor receptor Flt-1 mediates
[18] Stalmans I, Ng YS, Rohan R, Fruttiger M, Bouche A, Yuce A, et al. biological activities. Implications for a functional role of placenta
Arteriolar and venular patterning in retinas of mice selectively growth factor in monocyte activation and chemotaxis. J Biol Chem
expressing VEGF isoforms. J Clin Invest 2002;109:327 – 36. 1996;271:17629 – 34.
[19] Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role [36] Rafii S, Avecilla S, Shmelkov S, Shido K, Tejada R, Moore MA,
of the HIF system. Nat Med 2003;9:677 – 84. et al. Angiogenic factors reconstitute hematopoiesis by recruiting
[20] Luttun A, Brusselmans K, Fukao H, Tjwa M, Ueshima S, Herbert stem cells from bone marrow microenvironment. Ann N Y Acad
JM, et al. Loss of placental growth factor protects mice against Sci 2003;996:49 – 60.
vascular permeability in pathological conditions. Biochem Biophys [37] Cursiefen C, Chen L, Borges LP, Jackson D, Cao J, Radziejewski C,
Res Commun 2002;295:428 – 34. et al. VEGF-A stimulates lymphangiogenesis and hemangiogenesis
[21] Kempf VAJ, Lebiedziejewski M, Alitalo K, W7lzlein J-H, Ehehalt U, in inflammatory neovascularization via macrophage recruitment.
Fiebig J, et al. Activation of HIF-1 in bacillary angiomatosis: evidence J Clin Invest 2004;113:1040 – 50.
for a role of HIF-1 in bacterial infections. Circ Res 2005;3:623 – 32. [38] Karkkainen MJ, Haiko P, Sainio K, Partanen J, Taipale J, Petrova
[22] Oosthuyse B, Moons L, Storkebaum E, Beck H, Nuyens D, TV, et al. Vascular endothelial growth factor C is required for
Brusselmans K, et al. Deletion of the hypoxia-response element in sprouting of the first lymphatic vessels from embryonic veins. Nat
the vascular endothelial growth factor promoter causes motor neuron Immunol 2004;5:74 – 80.
degeneration. Nat Genet 2001;28:131 – 8. [39] Persico MG, Vincenti V, DiPalma T. Structure, expression and
[23] Xia YP, Li B, Hylton D, Detmar M, Yancopoulos GD, Rudge JS. receptor-binding properties of placenta growth factor (PlGF). In: L.
Transgenic delivery of VEGF to the mouse skin leads to an Claesson-Welsh editor. Vascular growth factors and angiogenesis.
inflammatory condition resembling human psoriasis. Blood 2003; Springer-Verlag, Berlin: Springer-Verlag, MG Persico, V Vincenti,
102:161 – 8. DiPalma T. 1999, p. 31-40.
[24] Larcher F, Murillas R, Bolontrade M, Conti CJ, Jorcano JL. VEGF/ [40] Autiero M, Waltenberger J, Communi D, Kranz A, Moons L,
VPF overexpression in skin of transgenic mice induces angiogenesis, Lambrechts D, et al. Role of PlGF in the intra- and intermolecular
560 T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563

cross talk between the VEGF receptors Flt1 and Flk1. Nat Med [59] Joukov V, Sorsa T, Kumar V, Jeltsch M, Claesson-Welsh L, Cao Y, et
2003;9:936 – 43. al. Proteolytic processing regulates receptor specificity and activity
[41] Park JE, Chen HH, Winer J, Houck KA, Ferrara N. Placenta growth of VEGF-C. EMBO J 1997;16:3898 – 911.
factor. Potentiation of vascular endothelial growth factor bioactivity, [60] Siegfried G, Basak A, Cromlish JA, Benjannet S, Marcinkiewicz J,
in vitro and in vivo, and high affinity binding to Flt-1 but not to Flk- Chretien M, et al. The secretory proprotein convertases furin, PC5,
1/KDR. J Biol Chem 1994;269:25646 – 54. and PC7 activate VEGF-C to induce tumorigenesis. J Clin Invest
[42] Nagy JA, Dvorak AM, Dvorak HF. VEGF-A(164/165) and PlGF: 2003;111:1723 – 32.
roles in angiogenesis and arteriogenesis. Trends Cardiovasc Med [61] Saharinen P, Tammela T, Karkkainen MJ, Alitalo K. Lymphatic
2003;13:169 – 75. vasculature: development, molecular regulation and role in tumor
[43] Carmeliet P, Moons L, Luttun A, Vincenti V, Compernolle V, De Mol metastasis and inflammation. Trends Immunol 2004;25:387 – 95.
M, et al. Synergism between vascular endothelial growth factor and [62] Kukk E, Lymboussaki A, Taira S, Kaipainen A, Jeltsch M, Joukov V,
placental growth factor contributes to angiogenesis and plasma et al. VEGF-C receptor binding and pattern of expression with
extravasation in pathological conditions. Nat Med 2001;7:575 – 83. VEGFR-3 suggests a role in lymphatic vascular development.
[44] Odorisio T, Schietroma C, Zaccaria ML, Cianfarani F, Tiveron C, Development 1996;122:3829 – 37.
Tatangelo L, et al. Mice overexpressing placenta growth factor [63] Lymboussaki A, Olofsson B, Eriksson U, Alitalo K. Vascular
exhibit increased vascularization and vessel permeability. J Cell Sci endothelial growth factor (VEGF) and VEGF-C show overlapping
2002;115:2559 – 67. binding sites in embryonic endothelia and distinct sites in differ-
[45] Oura H, Bertoncini J, Velasco P, Brown LF, Carmeliet P, Detmar M. entiated adult endothelia. Circ Res 1999;85:992 – 9.
A critical role of placental growth factor in the induction of [64] Oh SJ, Jeltsch MM, Birkenhager R, McCarthy JE, Weich HA, Christ
inflammation and edema formation. Blood 2003;101:560 – 7. B, et al. VEGF and VEGF-C: specific induction of angiogenesis and
[46] Carmeliet P. VEGF gene therapy: stimulating angiogenesis or lymphangiogenesis in the differentiated avian chorioallantoic mem-
angioma-genesis? Nat Med 2001;6:1102 – 3. brane. Dev Biol 1997;188:96 – 109.
[47] Luttun A, Tjwa M, Moons L, Wu Y, Angelillo-Scherrer A, Liao F, [65] Jeltsch M, Kaipainen A, Joukov V, Meng X, Lakso M, Rauvala H,
et al. Revascularization of ischemic tissues by PlGF treatment, and et al. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice.
inhibition of tumor angiogenesis, arthritis and atherosclerosis by Science 1997;276:1423 – 5.
anti-Flt1. Nat Med 2002;8:831 – 40. [66] Enholm B, Karpanen T, Jeltsch M, Kubo H, Stenback F, Prevo R,
[48] Pipp F, Heil M, Issbrucker K, Ziegelhoeffer T, Martin S, van den et al. Adenoviral expression of VEGF-C induces lymphangio-

Downloaded from by guest on December 22, 2014


Heuvel J, et al. VEGFR-1-selective VEGF homologue PlGF is genesis in the skin. Circ Res 2001;88:623 – 9.
arteriogenic: evidence for a monocyte-mediated mechanism. Circ [67] Veikkola T, Jussila L, Makinen T, Karpanen T, Jeltsch M, Petrova
Res 2003;92:378 – 85. TV, et al. Signalling via vascular endothelial growth factor receptor-3
[49] Olofsson B, Jeltsch M, Eriksson U, Alitalo K. Current biology of is sufficient for lymphangiogenesis in transgenic mice. EMBO J
VEGF-B and VEGF-C. Curr Opin Biotechnol 1999;10:528 – 35. 2001;20:1223 – 31.
[50] Olofsson B, Pajusola K, von Euler G, Chilov D, Alitalo K, Eriksson [68] Ristimaki A, Narko K, Enholm B, Joukov V, Alitalo K. Proin-
U. Genomic organization of the mouse and human genes for vascular flammatory cytokines regulate expression of the lymphatic endothe-
endothelial growth factor B (VEGF-B) and characterization of a lial mitogen vascular endothelial growth factor-C. J Biol Chem
second splice isoform. J Biol Chem 1996;271:19130 – 317. 1998;273:8413 – 8.
[51] Enholm B, Paavonen K, Ristimaki A, Kumar V, Gunji Y, Klefstrom [69] Chilov D, Kukk E, Taira S, Jeltsch MM, Kaukonen J, Palotie A, et al.
J, et al. Comparison of VEGF, VEGF-B, VEGF-C and Ang-1 mRNA Genomic organization of human and mouse genes for vascular
regulation by serum, growth factors, oncoproteins and hypoxia. endothelial growth factor C. J Biol Chem 1997;272:25176 – 83.
Oncogene 1997;14:2475 – 83. [70] Su JL, Shih JY, Yen ML, Jeng YM, Chang CC, Hsieh CY, et al.
[52] Salven P, Lymboussaki A, Heikkila P, Jaaskela-Saari H, Enholm B, Cyclooxygenase-2 induces EP1- and HER-2/Neu-dependent vascu-
Aase K, et al. Vascular endothelial growth factors VEGF-B and lar endothelial growth factor-C up-regulation: a novel mechanism of
VEGF-C are expressed in human tumors. Am J Pathol 1998;153: lymphangiogenesis in lung adenocarcinoma. Cancer Res 2004;64:
103 – 38. 554 – 64.
[53] Bellomo D, Headrick JP, Silins GU, Paterson CA, Thomas PS, [71] Kerjaschki D, Regele HM, Moosberger I, Nagy-Bojarski K,
Gartside M, et al. Mice lacking the vascular endothelial growth Watschinger B, Soleiman A, et al. Lymphatic neoangiogenesis in
factor-B gene (Vegfb) have smaller hearts, dysfunctional coronary human kidney transplants is associated with immunologically active
vasculature, and impaired recovery from cardiac ischemia. Circ Res lymphocytic infiltrates. J Am Soc Nephrol 2004;15:603 – 12.
2000;86:E29 – 35 [Online]. [72] Paavonen K, Mandelin J, Partanen T, Jussila L, Li T-F, Alitalo K,
[54] Aase K, von Euler G, Li X, Ponten A, Thoren P, Cao R, et al. et al. Vascular endothelial growth factors -C and -D and their
Vascular endothelial growth factor-B-deficient mice display an atrial VEGFR-2 and -3 receptors in blood and lymphatic vessels in
conduction defect. Circulation 2001;104:358 – 64. healthy and rheumatoid synovium. J Rheumatol 2002;29:39 – 45.
[55] Wanstall JC, Gambino A, Jeffery TK, Cahill MM, Bellomo D, [73] Skobe M, Hawighorst T, Jackson DG, Prevo R, Janes L, Velasco P,
Hayward NK, et al. Vascular endothelial growth factor-B-deficient et al. Induction of tumor lymphangiogenesis by VEGF-C promotes
mice show impaired development of hypoxic pulmonary hyper- breast cancer metastasis. Nat Med 2001;7:192 – 8.
tension. Cardiovasc Res 2002;55:361 – 8. [74] Karpanen T, Alitalo K. Lymphatic vessels as targets of tumor
[56] Louzier V, Raffestin B, Leroux A, Branellec D, Caillaud JM, Levame therapy? J Exp Med 2001;194:F37 – 42.
M, et al. Role of VEGF-B in the lung during development of chronic [75] Mandriota SJ, Jussila L, Jeltsch M, Compagni A, Baetens D,
hypoxic pulmonary hypertension. Am J Physiol, Lung Cell Mol Prevo R, et al. Vascular endothelial growth factor-C-mediated
Physiol 2003;284:L926 – 37. lymphangiogenesis promotes tumour metastasis. EMBO J
[57] Mould AW, Tonks ID, Cahill MM, Pettit AR, Thomas R, Hayward 2001;20:672 – 82.
NK, et al. Vegfb gene knockout mice display reduced pathology and [76] Karpanen T, Egeblad M, Karkkainen MJ, Kubo H, Yla-Herttuala S,
synovial angiogenesis in both antigen-induced and collagen-induced Jaattela M, et al. Vascular endothelial growth factor C promotes
models of arthritis. Arthritis Rheum 2003;48:2660 – 9. tumor lymphangiogenesis and intralymphatic tumor growth. Cancer
[58] Silvestre JS, Tamarat R, Ebrahimian TG, Le-Roux A, Clergue M, Res 2001;61:1786 – 90.
Emmanuel F, et al. Vascular endothelial growth factor-B promotes in [77] He Y, Kozaki K, Karpanen T, Koshikawa K, Yla-Herttuala S,
vivo angiogenesis. Circ Res 2003;93:114 – 23. Takahashi T, et al. Suppression of tumor lymphangiogenesis and
T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563 561

lymph node metastasis by blocking vascular endothelial growth [97] Hiratsuka S, Maru Y, Okada A, Seiki M, Noda T, Shibuya M.
factor receptor 3 signaling. J Natl Cancer Inst 2002;94:819 – 25. Involvement of Flt-1 tyrosine kinase (vascular endothelial growth
[78] He Y, Rajantie I, Ilmonen M, Makinen T, Karkkainen MJ, Haiko P, factor receptor-1) in pathological angiogenesis. Cancer Res 2001;
et al. Preexisting lymphatic endothelium but not endothelial 61:1207 – 13.
progenitor cells are essential for tumor lymphangiogenesis and [98] Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marme D.
lymphatic metastasis. Cancer Res 2004;64:3737 – 40. Migration of human monocytes in response to vascular endothelial
[79] Reis-Filho JS, Schmitt FC. Lymphangiogenesis in tumors: what do growth factor (VEGF) is mediated via the VEGF receptor flt-1.
we know? Microsc Res Tech 2003;60:171 – 80. Blood 1996;87:3336 – 43.
[80] He Y, Karpanen T, Alitalo K. Role of lymphangiogenic factors in [99] Carmeliet P, Luttun A. The emerging role of the bone marrow-
tumor metastasis. Biochim Biophys Acta 2004;1654:3 – 12. derived stem cells in (therapeutic) angiogenesis. Thromb Haemost
[81] Vleugel MM, Bos R, van der Groep P, Greijer AE, Shvarts A, Stel 2001;86:289 – 97.
HV, et al. Lack of lymphangiogenesis during breast carcinogenesis. [100] Hattori K, Heissig B, Wu Y, Dias S, Tejada R, Ferris B, et al.
J Clin Pathol 2004;57:746 – 51. Placental growth factor reconstitutes hematopoiesis by recruiting
[82] Baldwin ME, Catimel B, Nice EC, Roufail S, Hall NE, Stenvers KL, VEGFR1(+) stem cells from bone-marrow microenvironment. Nat
et al. The specificity of receptor binding by vascular endothelial Med 2002;8:841 – 9.
growth factor-d is different in mouse and man. J Biol Chem [101] Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, et al.
2001;276:19166 – 71. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may
[83] Farnebo F, Piehl F, Lagercrantz J. Restricted expression pattern of contribute to endothelial dysfunction, hypertension, and proteinuria
vegf-d in the adult and fetal mouse: high expression in the embyonic in preeclampsia. See editorial. J Clin Invest 2003;111:649 – 58.
lung. Biochem Biophys Res Commun 1999;257:891 – 4. [102] Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, et al.
[84] Stacker SA, Caesar C, Baldwin ME, Thornton GE, Williams RA, Circulating angiogenic factors and the risk of preeclampsia. New
Prevo R, et al. VEGF-D promotes the metastatic spread of tumor Engl J Med 2004;350:672 – 83.
cells via the lymphatics. Nat Med 2001;7:186 – 91. [103] Matthews W, Jordan CT, Gavin M, Jenkins NA, Copeland NG,
[85] Achen MG, Williams RA, Minekus MP, Thornton GE, Stenvers K, Lemischka IR. A receptor tyrosine kinase cDNA isolated from a
Rogers PA, et al. Localization of vascular endothelial growth factor- population of enriched primitive hematopoetic cells and exhibiting
D in malignant melanoma suggests a role in tumour angiogenesis. close genetic linkage to c-kit. Proc Natl Acad Sci U S A
J Pathol 2001;193:147 – 54. 1991;88:9026 – 30.

Downloaded from by guest on December 22, 2014


[86] Ishii H, Yazawa T, Sato H, Suzuki T, Ikeda M, Hayashi Y, et al. [104] Terman BI, Carrion ME, Kovacs E, Rasmussen BA, Eddy RL,
Enhancement of pleural dissemination and lymph node metastasis of Shows TB. Identification of a new endothelial cell growth factor
intrathoracic lung cancer cells by vascular endothelial growth factors receptor tyrosine kinase. Oncogene 1991;6:1677 – 83.
(VEGFs). Lung Cancer 2004;45:325 – 37. [105] Meyer M, Clauss M, Lepple-Wienhues A, Waltenberger J, Augustin
[87] Byzova TV, Goldman CK, Jankau J, Chen J, Cabrera G, Achen MG, M, Ziche M, et al. A novel vascular endothelial growth factor
et al. Adenovirus encoding vascular endothelial growth factor-D encoded by Orf virus, VEGF-E, mediates angiogenesis via signaling
induces tissue-specific vascular patterns in vivo. Blood 2002; through VEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine
99:4434 – 42. kinases. EMBO J 1999;18:363 – 74.
[88] Shibuya M, Yamaguchi S, Yamane A, Ikeda T, Tojo A, Matsushime [106] Wise LM, Veikkola T, Mercer AA, Savory LJ, Fleming SB, Caesar
H, et al. Nucleotide sequence and expression of a novel human C, et al. The vascular endothelial growth factor (VEGF)-like protein
receptor type tyrosine kinase gene (flt) closely related to the fms from orf virus NZ2 binds to VEGFR2 and neuropilin-1. Proc Natl
family. Oncogene 1990;5:519 – 24. Acad Sci U S A 1999;96:3071 – 6.
[89] Huang K, Andersson C, Roomans GM, Ito N, Claesson-Welsh L. [107] Gille H, Kowalski J, Li B, LeCouter J, Moffat B, Zioncheck TF, et al.
Signaling properties of VEGF receptor-1 and -2 homo- and Analysis of biological effects and signaling properties of Flt-1
heterodimers. Int J Biochem Cell Biol 2001;33:315 – 24. (VEGFR-1) and KDR (VEGFR-2). A reassessment using novel
[90] Zachary I, Gliki G. Signaling transduction mechanisms mediating receptor-specific vascular endothelial growth factor mutants. J Biol
biological actions of the vascular endothelial growth factor family. Chem 2001;276:3222 – 30.
Cardiovasc Res 2001;49:568 – 81. [108] Shima DT, Deutsch U, D’Amore PA. Hypoxic induction of vascular
[91] Gerber HP, Conorelli F, Park J, Ferrara N. Differential transcriptional endothelial growth factor (VEGF) in human epithelial cells is
regulation of the two vascular endothelial growth factor receptor mediated by increases in mRNA stability. FEBS Lett 1995;
genes. Flt-1, but not Flk-1/KDR, is up-regulated by hypoxia. J Biol 370:203 – 8.
Chem 1997;272:23659 – 67. [109] Matsumoto T, Claesson-Welsh L. VEGF receptor signal trans-
[92] Partanen TA, Alitalo K, Miettinen M. Lack of lymphatic vascular duction. Science’s STKE 2001;112:1 – 17.
specificity of vascular endothelial growth factor receptor 3 in 185 [110] Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF,
vascular tumors. Cancer 1999;86:2406 – 12. Breitman ML, et al. Failure of blood-island formation and vasculo-
[93] Kaipainen A, Korhonen J, Pajusola K, Aprelikova O, Persico MG, genesis in Flk-1-deficient mice. Nature 1995;376:62 – 6.
Terman BI, et al. The related FLT4, FLT1, and KDR receptor [111] Shalaby F, Ho J, Stanford WL, Fischer KD, Schuh AC, Schwartz L,
tyrosine kinases show distinct expression patterns in human fetal et al. A requirement for Flk1 in primitive and definitive hematopoi-
endothelial cells. J Exp Med 1993;178:2077 – 88. esis and vasculogenesis. Cell 1997;89:981 – 90.
[94] Peters KG, De Vries C, Williams LT. Vascular endothelial growth [112] Baffert F, Thurston G, Rochon-Duck M, Le T, Brekken R, McDonald
factor receptor expression during embryogenesis and tissue repair DM. Age-related changes in vascular endothelial growth factor
suggests a role in endothelial differentiation and blood vessel growth. dependency and angiopoietin-1-induced plasticity of adult blood
Proc Natl Acad Sci U S A 1993;90:8915 – 9. vessels. Circ Res 2004;94:984 – 92.
[95] Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role of the Flt-1 [113] Soldi R, Mitola S, Strasly M, Defilippi P, Tarone G, Bussolino F.
receptor tyrosine kinase in regulating the assembly of vascular Role of avh3 in the activation of vascular endothelial growth factor
endothelium. Nature 1995;376:66 – 70. receptor-2. EMBO J 1999;18:882 – 92.
[96] Hiratsuka S, Minowa O, Kuno J, Noda T, Shibuya M. Flt-1 lacking [114] Hutchings H, Ortega N, Plouet J. Extracellular matrix-bound
the tyrosine kinase domain is sufficient for normal development vascular endothelial growth factor promotes endothelial cell adhe-
and angiogenesis in mice. Proc Natl Acad Sci U S A 1998; sion, migration and survival through integrin ligation. FASEB J
4:9349 – 54. 2003;17:1520 – 2.
562 T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563

[115] Carmeliet P, Lampugnani MG, Moons L, Breviario F, Comper- [134] Kawasaki T, Bekku Y, Suto F, Kitsukawa T, Taniguchi M, Nagatsu I,
nolle V, Bono F, et al. Targeted deficiency or cytosolic truncation et al. Requirement of neuropilin 1-mediated Sema3A signals in
of the VE-cadherin gene in mice impairs VEGF-mediated patterning of the sympathetic nervous system. Development
endothelial survival and angiogenesis. Cell 1999;98:147 – 57. 2002;129:671 – 80.
[116] Pajusola K, Aprelikova O, Pelicci G, Weich H, Claesson-Welsh L, [135] M. Klagsbrun, J. Folkman, Angiogenesis. In: Sporn, M.B. Roberts,
Alitalo K. Signalling properties of FLT4, a proteolytically processed A.B. Peptide growth factors and their receptors. Springer-Verlag,
receptor tyrosine kinase related to two VEGF receptors. Oncogene New York: Springer-Verlag, M. Klagsbrun, Folkman J; 1991:549-86.
1994;9:3545 – 55. [136] Kitsukawa T, Shimono A, Kawakami A, Kondoh H, Hajime F. Over
[117] Lee J, Gray A, Yuan J, Louth S-M, Avraham H, Wood W. Vascular expression of a membrane protein, neuropilin, in chimeric mice
endothelial growth factor-related protein: a ligand and specific causes anomalities in the cardiovascular system, nervous system and
activator of the tyrosine kinase receptor Flt4. Proc Natl Acad Sci limbs. Development 1995;121:4309 – 18.
U S A 1996;93:1988 – 92. [137] Herzog Y, Kalcheim C, Kahane N, Reshef R, Neufeld G. Differential
[118] Hughes DC. Alternative splicing of the human VEGFGR-3/FLT4 expression of neuropilin-1 and neuropilin-2 in arteries and veins.
gene as a consequence of an integrated human endogenous Mech Dev 2001;109:115 – 9.
retrovirus. J Mol Evol 2001;53:77 – 9. [138] Yuan L, Moyon D, Pardanaud L, Breant C, Karkkainen MJ, Alitalo
[119] Joukov V, Pajusola K, Kaipainen A, Chilov D, Lahtinen I, Kukk E, K, et al. Abnormal lymphatic vessel development in neuropilin 2
et al. A novel vascular endothelial growth factor, VEGF-C, is a mutant mice. Development 2002;129:4797 – 806.
ligand for the Flt4 (VEGFR-3) and Kdr (VEGFR-2) receptor [139] Kawasaki T, Kitsukawa T, Bekku Y, Matsuda Y, Sanbo M, Yagi T,
tyrosine kinases. EMBO J 1996;15:290 – 8. et al. A requirement for neuropilin-1 in embryonic vessel
[120] Achen MG, Jeltsch M, Kukk E, Makinen T, Vitali A, Wilks AF, et al. formation. Development 1999;126:4895 – 902.
Vascular endothelial growth factor D (VEGF-D) is a ligand for the [140] Gu C, Rodriguez ER, Reimert DV, Shu T, Fritzsch B, Richards LJ,
tyrosine kinases VEGF receptor 2 (Flk1) and VEGF receptor 3 et al. Neuropilin-1 conveys semaphorin and VEGF signaling during
(Flt4). Proc Natl Acad Sci U S A 1998;95:548 – 53. neural and cardiovascular development. Dev Cell 2003;5:45 – 57.
[121] Kaipainen A, Korhonen J, Mustonen T, van Hinsbergh VM, Fang [141] Takashima S, Kitakaze M, Asakura M, Asanuma H, Sanada S,
G-H, Dumont D, et al. Expression of the fms-like tyrosine kinase Tashiro F, et al. Targeting of both mouse neuropilin-1 and neuropilin-
FLT4 gene becomes restricted to endothelium of lymphatic vessels 2 genes severely impairs developmental yolk sac and embryonic
during development. Proc Natl Acad Sci U S A 1995;92:3566 – 70. angiogenesis. Proc Natl Acad Sci U S A 2002;99:3657 – 62.

Downloaded from by guest on December 22, 2014


[122] Partanen T, Arola J, Saaristo A, Jussila L, Ora A, Miettinen M, et al. [142] Yla-Herttuala S, Alitalo K. Gene transfer as a tool to induce
VEGF-C and VEGF-D expression in neuroendocrine cells and their therapeutic vascular growth. Nat Med 2003;9:694 – 701.
receptor, VEGFR-3, in fenestrated blood vessels in human tissues. [143] Scholz D, Cai WJ, Schaper W. Arteriogenesis, a new concept of
FASEB J 2000;14:2087 – 96. vascular adaptation in occlusive disease. Angiogenesis 2001;4:
[123] Skobe M, Hamberg LM, Hawighorst T, Shirner M, Wolf GL, Alitalo 247 – 57.
K, et al. Concurrent induction of lymphangiogenesis, angiogenesis [144] Rosengart TK, Patel SR, Crystal RG. Therapeutic angiogenesis:
and macrophage recruitment by VEGF-C in melanoma. Am J Pathol protein and gene therapy delivery strategies. J Cardiovasc Risk
2001;159:893 – 903. 1999;6:29 – 40.
[124] Veikkola T, Lohela M, Ikenberg K, Makinen T, Korff T, Saaristo [145] Freedman SB, Vale P, Kalka C, Kearney M, Pieczek A, Symes J,
A,et al. Intrinsic versus microenvironmental regulation of lym- et al. Plasma vascular endothelial growth factor (VEGF) levels
phatic endothelial cell phenotype and function. FASEB J after intramuscular and intramyocardial gene transfer of VEGF-1
2003;17:2006 – 13. plasmid DNA. Hum Gene Ther 2002;13:1595 – 603.
[125] Dumont DJ, Jussila L, Taipale J, Mustonen T, Pajusola K, Breitman [146] Baumgartner I, Isner JM. Gene therapy for peripheral vascular
M, et al. Cardiovascular failure in mouse embryos deficient in VEGF disease. Israel Med Assoc J 2000;2:27 – 32.
receptor-3. Science 1998;282:946 – 9. [147] Rajagopalan S, Mohler ERr, Lederman RJ, Mendelsohn FO,
[126] Hamada K, Oike Y, Takakura N, Ito Y, Jussila L, Dumont DJ, et al. Saucedo JF, Goldman CK, et al. Regional angiogenesis with vascular
VEGF-C signaling pathways through VEGFR-2 and VEGFR-3 in endothelial growth factor in peripheral arterial disease: a phase II
vasculoangiogenesis and hematopoiesis. Blood 2000;96:3793 – 800. randomized, double-blind, controlled study of adenoviral delivery of
[127] Makinen T, Jussila L, Veikkola T, Karpanen T, Kettunen MI, vascular endothelial growth factor 121 in patients with disabling
Pulkkanen KJ, et al. Inhibition of lymphangiogenesis with resulting intermittent claudication. Circulation 2003;108:1933 – 8.
lymphedema in transgenic mice expressing soluble VEGF receptor- [148] Carmeliet P. VEGF gene therapy: stimulating angiogenesis or
3. Nat Med 2001;7:199 – 205. angioma-genesis? Nat Med 2000;6:1102 – 3.
[128] Irrthum A, Karkkainen MJ, Devriendt K, Alitalo K, Vikkula M. [149] Sueishi K, Yonemitsu Y, Nakagawa K, Kaneda Y, Kumamoto M,
Congenital hereditary lymphedema caused by a mutation that Nakashima Y. Atherosclerosis and angiogenesis. Its pathophysio-
inactivates VEGFR3 tyrosine kinase. Am J Hum Genet 2000;67: logical significance in humans as well as in an animal model induced
295 – 301. by the gene transfer of vascular endothelial growth factor. Ann N Y
[129] Karkkainen MJ, Ferrell RE, Lawrence EC, Kimak MA, Levinson Acad Sci 1997;811:311 – 24.
MA, McTigue MA, et al. Missense mutations interfere with VEGFR- [150] Hayden MR, Tyagi SC. Vasa vasorum in plaque angiogenesis,
3 signalling in primary lymphoedema. Nat Genet 2000;25:153 – 9. metabolic syndrome, type 2 diabetes mellitus, and atheroscleropathy:
[130] Karkkainen MJ, Saaristo A, Jussila L, Karila KA, Lawrence EC, a malignant transformation. Cardiovasc Diabetol 2004;3:1.
Pajusola K, et al. A model for gene therapy of human hereditary [151] Salomonsson L, Pettersson S, Englund MC, Wiklund O, Ohlsson
lymphedema. Proc Natl Acad Sci U S A 2001;98:12677 – 82. BG. Post-transcriptional regulation of VEGF expression by
[131] Chen L, Hamrah P, Cursiefen C, Zhang Q, Pytowski B, Streilein JW, oxidised LDL in human macrophages. Eur J Clin Invest 2002;32:
et al. Vascular endothelial growth factor receptor-3 mediates 767 – 74.
induction of corneal alloimmunity. Nat Med 2004;10:13 – 5. [152] Nakagawa K, Chen YX, Ishibashi H, Yonemitsu Y, Murata T, Hata
[132] Klagsbrun M, Takashima S, Mamluk R. The role of neuropilin in Y, et al. Angiogenesis and its regulation: roles of vascular endothelial
vascular and tumor biology. Adv Exp Med Biol 2002;515:33 – 48. cell growth factor. Semin Thromb Hemost 2000;26:61 – 6.
[133] Bagri A, Tessier-Lavigne M. Neuropilins as semaphorin receptors: in [153] Celletti FL, Waugh JM, Amabile PG, Brendolan A, Hilfiker PR,
vivo functions in neuronal cell migration and axon guidance. Adv Dake MD. Vascular endothelial growth factor enhances atheroscler-
Exp Med Biol 2002;515:13 – 31. otic plaque progression. Nat Med 2001;7:425 – 9.
T. Tammela et al. / Cardiovascular Research 65 (2005) 550–563 563

[154] Moulton KS, Vakili K, Zurakowski D, Soliman M, Butterfield C, model of proliferative retinopathy. Proc Natl Acad Sci U S A 1996;
Sylvin E, et al. Inhibition of plaque neovascularization reduces 93:4851 – 6.
macrophage accumulation and progression of advanced atheroscle- [168] Ozaki H, Seo MS, Ozaki K, Yamada H, Yamada E, Okamoto N,
rosis. Proc Natl Acad Sci U S A 2003;100:4736 – 41. et al. Blockade of vascular endothelial cell growth factor receptor
[155] Lemstrom KB, Krebs R, Nykanen AI, Tikkanen JM, Sihvola RK, signaling is sufficient to completely prevent retinal neovasculari-
Aaltola EM, et al. Vascular endothelial growth factor enhances zation. Am J Pathol 2000;156:697 – 707.
cardiac allograft arteriosclerosis. Circulation 2002;105:2524 – 30. [169] Ishida S, Usui T, Yamashiro K, Kaji Y, Amano S, Ogura Y, et al.
[156] Cho CH, Kammerer RA, Lee HJ, Yasunaga K, Kim KT, Choi HH, VEGF164-mediated inflammation is required for pathological, but
et al. Designed angiopoietin-1 variant, COMP-Ang1, protects not physiological, ischemia-induced retinal neovascularization. J Exp
against radiation-induced endothelial cell apoptosis. Proc Natl Med 2003;198:483 – 9.
Acad Sci U S A 2004;101:5553 – 8. [170] Detmar M. Evidence for vascular endothelial growth factor
[157] Rafii S, Lyden D. Therapeutic stem and progenitor cell trans- (VEGF) as a modifier gene in psoriasis. J Invest Dermatol
plantation for organ vascularization and regeneration. Nat Med 2004;122:xiv – v.
2003;9:702 – 12. [171] Reynolds LP, Grazul-Bilska AT, Redmer DA. Angiogenesis in the
[158] Majka SM, Jackson KA, Kienstra KA, Majesky MW, Goodell MA, female reproductive organs: pathological implications. Int J Exp
Hirschi KK. Distinct progenitor populations in skeletal muscle are Pathol 2003;83:151 – 63.
bone marrow derived and exhibit different cell fates during vascular [172] Rockson SG. Lymphedema. Am J Med 2001;110:288 – 95.
regeneration. J Clin Invest 2003;111:71 – 9. [173] Northup KA, Witte MH, Witte CL. Syndromic classification of
[159] Wagers AJ, Sherwood RI, Christensen JL, Weissman IL. Little hereditary lymphedema. Lymphology 2003;36:162 – 89.
evidence for developmental plasticity of adult hematopoietic stem [174] Fang J, Dagenais SL, Erickson RP, Arlt MF, Glynn MW, Gorski JL,
cells. Science 2002;297:2256 – 9. et al. Mutations in FOXC2 (MFH-1), a forkhead family transcription
[160] Ziegelhoeffer T, Fernandez B, Kostin S, Heil M, Voswinckel R, factor, are responsible for the hereditary lymphedema-distichiasis
Helisch A, et al. Bone marrow-derived cells do not incorporate into syndrome. Am J Hum Genet 2000;67:1382 – 8.
the adult growing vasculature. Circ Res 2004;94:230 – 8. [175] Petrova TV, Karpanen T, Norrmen C, Mellor R, Tamakoshi T,
[161] Rajantie I, Ilmonen M, Alminaite A, Ozerden U, Alitalo K, Salven P. Finegold D, et al. Defective valves and abnormal mural cell
Adult bone marrow-derived cells recruited during angiogenesis recruitment underlie lymphatic vascular failure in lymphedema
comprise precursors for periendothelial vascular mural cells. Blood distichiasis. Nat Med 2004;10:974 – 81.

Downloaded from by guest on December 22, 2014


2004;104:2084 – 6. [176] Padera TP, Kadambi A, di Tomaso E, Carreira CM, Brown EB,
[162] Jain RK. Molecular regulation of vessel maturation. Nat Med Boucher Y, et al. Lymphatic metastasis in the absence of functional
2003;9:685 – 93. intratumor lymphatics. Science 2002;296:1883 – 6.
[163] Ruoslahti E. Vascular zip codes in angiogenesis and metastasis. [177] Achen MG, Williams RA, Baldwin ME, Lai P, Roufail S, Alitalo K,
Biochem Soc Trans 2004;32:397 – 402. et al. The angiogenic and lymphangiogenic factor vascular endothe-
[164] Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, lial growth factor-D exhibits a paracrine mode of action in cancer.
Topalian SL, et al. A randomized trial of bevacizumab, an anti- Growth Factors 2002;20:99 – 107.
vascular endothelial growth factor antibody, for metastatic renal [178] Maula SM, Luukkaa M, Grenman R, Jackson D, Jalkanen S,
cancer. New Engl J Med 2003;349:427 – 34. Ristamaki R. Intratumoral lymphatics are essential for the metastatic
[165] Bainbridge JW, Mistry AR, Thrasher AJ, Ali RR. Gene therapy for spread and prognosis in squamous cell carcinomas of the head and
ocular angiogenesis. Clin Sci 2003;104:561 – 75. neck region. Cancer Res 2003;63:1920 – 6.
[166] Aiello LP, Pierce EA, Foley ED, Takagi H, Chen H, Riddle L, et al. [179] Dadras SS, Paul T, Bertoncini J, Brown LF, Muzikansky A, Jackson
Suppression of retinal neovascularization in vivo by inhibition of DG, et al. Tumor lymphangiogenesis: a novel prognostic indicator
vascular endothelial growth factor (VEGF) using soluble VEGF- for cutaneous melanoma metastasis and survival. Am J Pathol
receptor chimeric proteins. Proc Natl Acad Sci U S A 1995;92: 2003;162:1951 – 60.
10457 – 61. [180] Straume O, Jackson DG, Akslen LA. Independent prognostic impact
[167] Robinson GS, Pierce EA, Rook SL, Foley E, Webb R, Smith LES. of lymphatic vessel density and presence of low-grade lymphangio-
Oligodeoxynucleotides inhibit retinal neovascularization in a murine genesis in cutaneous melanoma. Clin Cancer Res 2003;9:250 – 6.

Das könnte Ihnen auch gefallen