vascular endothelial growth factors b and c: gene

60
Vascular Endothelial Growth Factors B and C: Gene Regulation and Gene Transfer In Vivo Berndt Enholm Molecular/Cancer Biology Laboratory Haartman Institute and Helsinki University Central Hospital Biomedicum Helsinki University of Helsinki Finland Academic Dissertation To be publicly discussed with the permission of the Faculty of Medicine of the University of Helsinki, in Auditorium 2, Biomedicum Helsinki, 6 th of August, 2004 at 12 noon

Upload: others

Post on 24-Jan-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Vascular Endothelial Growth Factors B and C: Gene

Vascular Endothelial Growth Factors B and C:

Gene Regulation and Gene Transfer In Vivo

Berndt Enholm

Molecular/Cancer Biology Laboratory

Haartman Institute and

Helsinki University Central Hospital

Biomedicum Helsinki

University of Helsinki

Finland

Academic Dissertation

To be publicly discussed with the permission of the Faculty of Medicine of the University

of Helsinki, in Auditorium 2, Biomedicum Helsinki, 6th of August, 2004 at 12 noon

Page 2: Vascular Endothelial Growth Factors B and C: Gene

2

Page 3: Vascular Endothelial Growth Factors B and C: Gene

3

Hic Labor,

Hoc Opus Est

Vergilius

Aeneas, song 6. verse 129

Page 4: Vascular Endothelial Growth Factors B and C: Gene

4

Supervised by:

Kari Alitalo, MD, Ph.D.

Research Professor of the Finnish Academy of Sciences

Molecular/Cancer Biology Laboratory

Biomedicum Helsinki

University of Helsinki

Reviewed by:

Ulf-Håkan Stenman, MD. Ph.D.

Department of Clinical Chemistry,

Helsinki University and Helsinki University Central Hospital

Biomedicum Helsinki

Olli Ritvos M.D., Ph.D.

Developmental and Reproductive Biology Program

Biomedicum Helsinki

University of Helsinki

Opponent:

Wolfgang Schaper MD, Ph.D.

Professor of Physiology

University of Giessen-Germany

Director of the Max Planck Institute for Physiological and

Clinical Research (W.G. Kerckhoff-Institut)

Bad Nauheim

Germany

Page 5: Vascular Endothelial Growth Factors B and C: Gene

5

TABLE OF CONTENTSTABLE OF CONTENTS .........................................................................................................................................5

ABBREVIATIONS...................................................................................................................................................7

ORIGINAL PUBLICATIONS................................................................................................................................9

ABSTRACT.............................................................................................................................................................10

REVIEW OF THE LITERATURE......................................................................................................................11

THE VASCULAR SYSTEM......................................................................................................................................11THE BLOOD VASCULAR SYSTEM .........................................................................................................................11

Embryonic Development, Structure and Function ........................................................................................11Angiogenesis in the Adult ...............................................................................................................................12

MOLECULAR REGULATORS OF THE BLOOD VASCULAR SYSTEM .......................................................................14VEGF...............................................................................................................................................................14VEGF-B...........................................................................................................................................................15PlGF ................................................................................................................................................................16VEGF Homologues Encoded by Orf and Parapox Viruses ..........................................................................17VEGFR-2 and VEGFR-1 ................................................................................................................................17Neuropilins......................................................................................................................................................18Ephs and Ephrins............................................................................................................................................18Platelet Derived Growth Factors (PDGFs)...................................................................................................18Angiopoietins and Tie Receptors....................................................................................................................19

MARKERS OF THE VASCULAR SYSTEM................................................................................................................19PECAM-1 ........................................................................................................................................................19vWF..................................................................................................................................................................20VE-cadherin ....................................................................................................................................................20

PATHOLOGIC ANGIOGENESIS AND ARTERIOGENESIS..........................................................................................21Angiogenesis in Ischemic Disease..................................................................................................................21Arteriogenesis .................................................................................................................................................21Tumor Angiogenesis .......................................................................................................................................22

THE LYMPHATIC SYSTEM ....................................................................................................................................23Embryonic Development, Structure and Function ........................................................................................23

MOLECULAR REGULATORS OF THE LYMPHATIC SYSTEM...................................................................................25VEGF-C and VEGF-D....................................................................................................................................25VEGFR-3.........................................................................................................................................................26

MARKERS OF THE LYMPHATIC SYSTEM ..............................................................................................................27Lymphatic Vessel Endothelial Hyaluronan Receptor-1 (LYVE-1)................................................................27Podoplanin ......................................................................................................................................................27Prox-1..............................................................................................................................................................27

PATHOLOGIC LYMPHANGIOGENESIS....................................................................................................................28Tumor Lymphangiogenesis.............................................................................................................................28Lymphedemas..................................................................................................................................................28

THERAPEUTIC APPROACHES ................................................................................................................................30Vascular Gene Therapy ..................................................................................................................................30Viral Vectors ...................................................................................................................................................30Proangiogenic Therapy ..................................................................................................................................31Lymphangiogenic Therapy .............................................................................................................................31

AIMS OF THE STUDY .........................................................................................................................................32

MATERIALS AND METHODS...........................................................................................................................33

RESULTS AND DISCUSSION.............................................................................................................................35

I. STUDIES ON THE REGULATION OF VEGF, VEGF-B, VEGF-C AND ANG-1 MRNA EXPRESSION..................35II. PROINFLAMMATORY CYTOKINES REGULATE EXPRESSION OF VEGF-C.......................................................36III. ADENOVIRAL EXPRESSION OF VEGF-C IN THE SKIN INDUCES LYMPHANGIOGENESIS...............................39IV. VEGF-B ACTIVATES VEGFR-1, BUT PROVIDES VERY LITTLE ANGIOGENIC OR ARTERIOGENICACTIVITY IN VIVO IN COMPARISON WITH PLGF..................................................................................................40

CONCLUDING REMARKS.................................................................................................................................42

Page 6: Vascular Endothelial Growth Factors B and C: Gene

6

ACKNOWLEDGEMENTS...................................................................................................................................43

REFERENCES........................................................................................................................................................44

Page 7: Vascular Endothelial Growth Factors B and C: Gene

7

ABBREVIATIONS

AAV Adeno associated virusAd AdenovirusAng AngiopoietinbFGF Basic fibroblast growth factorCAM Chorioallantoic membrane assayCEP Circulating endothelial progenitorEC Endothelial cellECM Extracellular matrixEGF Epidermal growth factorFlt-1 fms-like tyrosine kinase-1Flt-4 fms-like tyrosine kinase 4FOXC2 Forkhead box C2GM-CSF Granulocyte monocyte-colony stimulating factorHA HyaluronanHIF Hypoxia inducible factorHMW-MAA High molecular weight human melanoma associated antigenHRE Hypoxia regulated elementHSPG Heparan sulphate proteoglycanHUVEC Human umbilical vein endothelial cellIL InterleukinIg ImmunglobulinIGF Insulin like growth factorLacZ β-galactosidase geneLD Lymphedema distichiasisLYVE-1 Lymphatic vessel endothelial hyaluronan receptor-1MCP-1 Monocyte chemoattractant protein-1mRNA Messenger ribonucleic acidNrp NeuropilinORF Open reading framePDGF Platelet derived growth factorPDGFR Platelet derived growth factor receptorPECAM-1 Platelet endothelial cell adhesion molecule-1Prox-1 Prospero related homeobox protein-1RTK Receptor tyrosine kinaseSLC Secondary lymphoid chemokineSH Src homology domainTek Tunica interna endothelial cell kinase-1TGF-β Transforming growth factor-βTie Tyrosine kinase with Ig and EGF homology domainsTK Tyrosine kinaseTNF-α Tumor necrosis factor-αVEGF Vascular endothelial growth factorVEGFR Vascular endothelial growth factor receptorvWF Von Willebrand factor

Page 8: Vascular Endothelial Growth Factors B and C: Gene

8

Page 9: Vascular Endothelial Growth Factors B and C: Gene

9

ORIGINAL PUBLICATIONS

This thesis is based on the following articles, which will be referred to by their Roman

numerals.

I Enholm B, Paavonen K, Ristimäki A, Kumar V, Gunji Y, Klefström J, Kivinen L,

Laiho M, Olofsson B, Joukov V, Eriksson U, Alitalo K. Comparison of VEGF,

VEGF-B, VEGF-C and Ang-1 mRNA regulation by serum, growth factors,

oncoproteins and hypoxia. (1997) Oncogene, 14(20):2475-83.

II Ristimäki A, Narko K, Enholm B, Joukov V, Alitalo K. Proinflammatory cytokines

regulate expression of the lymphatic endothelial mitogen vascular endothelial

growth factor-C. (1998) J. Biol. Chem., 273(14):8413-8.

III Enholm B, Kärpänen T, Jeltsch M, Kubo H, Stenbäck F, Prevo R, Jackson DG,

Ylä-Herttuala S, Alitalo K. Adenoviral expression of vascular endothelial growth

factor-C induces lymphangiogenesis in the skin. (2001) Circ. Res., 88(6):623-9.

IV Enholm B, Tammela T, Paavonen K, Jeltsch M, Mustjoki S, Carmeliet P, Eriksson

U, Ylä-Herttuala S, Alitalo K. VEGF-B acitvates VEGFR-1, but provides very little

angiogenic or arteriogenic activity in vivo in comparison with PlGF. (2004).

Page 10: Vascular Endothelial Growth Factors B and C: Gene

10

ABSTRACT

The unfolding of the molecular biology of the vascular system has provided the scientific

community with an array of molecular tools for investigation of the circulatory system as

well as for therapeutic intervention in a number of disorders of the vasculature. The

circulatory system consists of the cardiovascular system and the lymphatic system that

consists of lymphatic capillaries, collecting ducts, the thoracic duct and lymph nodes.

While the cardiovascular system has been thoroughly studied and its role in disorders such

as ischemic heart disease, peripheral vascular disease and in the growth of solid tumors is

well known, the lymphatic system has until recently attracted very little attention from the

scientific community. The discovery of specific growth factors and their receptors and an

array of markers has enabled investigators to study the vascular system in detail. In

particular, the Vascular Endothelial Growth Factor (VEGF) family members VEGF,

VEGF-B, Placenta Growth Factor (PlGF), VEGF-C and VEGF-D and their receptors

VEGFR-1, -2 and -3 have facilitated understanding of the vasculature and provided tools

and targets for therapeutic intervention. Of the VEGF family members, VEGF induces

angiogenesis i.e. growth of new blood vessels from pre-existing ones, whereas PlGF

primarily mediates arteriogenesis, the formation of collateral arteries from pre-existing

arterioles. VEGF-C and VEGF-D have both angiogenic and lymphangiogenic effects.

This study examined aspects of the biology of VEGF-B and VEGF-C in vitro and in

vivo. We showed that VEGF-C mRNA expression is controlled by serum growth factors

and inflammatory mediators, suggesting a function for VEGF-C in inflammation. VEGF-B

mRNA levels did not respond to any of the stimuli tested. In vivo, expression of VEGF-C

in the skin by adenoviral gene transfer induced the formation of lymphatic vessels de novo,

or lymphangiogenesis. The lymphangiogenic effect of VEGF-C could be used in gene

therapy aimed at reconstructing lymphatic vessels in patients suffering from disorders of

the lymphatic system such as hereditary or acquired lymphedema. Over-expression of

VEGF-B in vivo had very little effects on the vasculature compared to the two other

VEGFR-1 agonists, VEGF and PlGF.

Page 11: Vascular Endothelial Growth Factors B and C: Gene

11

REVIEW OF THE LITERATURE

The Vascular System

The circulatory system consists of the heart and the blood circulation. The blood vascular

system is further divided into arterial and venous compartments. The blood vascular system

supplies the tissues with oxygen and nutrients and transports catabolic end products away

from tissues. The lymphatic system collects extravasated fluid, returns it to the blood, and

serves as an immunosurveillance organ. The growth and remodeling of the vascular system

is controlled in part through peptide growth factor signals transmitted via cell membrane

bound receptor tyrosine kinases (RTKs) that relay extracellular signals to the cytoplasm and

on to the nucleus where transcription of genes is readjusted. Perhaps the most important

groups of growth factors involved in the development and remodelling of the vascular

system are the VEGF family of growth factors (VEGF, VEGF-B, VEGF-C and VEGF-D

and their cognate receptors VEGFR1-3) and the Tie (tyrosine kinase with Ig and EGF

homology domains)-angiopoietin family (angiopoietins-1 to -4 and their cognate receptor

Tie-2, as well as Tie-1). These signalling systems are attractive targets for pro- or

antiangiogenic therapy aiming at vascular remodelling in various diseases ranging from

ischemic heart disease to suppression of angiogenesis in solid tumors.

The Blood Vascular System

Embryonic Development, Structure and Function

During embryogenesis, endothelial cells (ECs) develop from mesenchymal precursors

termed angioblasts and subsequently assemble into tubes that form a honeycomb-like

structure, the primary vascular plexus1,2(Figure 1). Recruitment of resident and circulating

angioblasts for formation of the primary vascular plexus is termed vasculogenesis.

Angioblasts originally develop from hemangioblasts, common precursors of hematopoietic

cells and ECs, that form aggregates called blood islands in the embryo (Figure 1).

Angioblasts express VEGFR-2 and angioblast differentiation is influenced by VEGF3-5 ,

however, VEGFR-1 signalling supresses angioblast differentiation6. After the formation of

the primary vascular plexus the development of the embryonic vasculature continues by

angiogenesis, the formation of new arteries and veins from the primary vascular plexus by

endothelial cell sprouting and splitting from pre-existing vessels (Figure 1). Embryonic

oxygenation initially depends on diffusion, but as tissues expand hypoxia sets in and

Page 12: Vascular Endothelial Growth Factors B and C: Gene

12

triggers signalling through hypoxia inducible transcription factors (HIFs)7. Downstream

effectors of HIFs include VEGF which mediates proliferation, sprouting and migration of

ECs8.

Structurally, the mature blood vessel wall consists of an inner layer of ECs

separated by a basal lamina from a perivascular coat formed by smooth muscle cells

(SMCs) and pericytes. The perivascular support structures have an important function in

providing vascular tone and they are required for the formation of mature, stable vessels.

Furthermore, vessel enlargement and endothelial cell sprouting in angiogenesis as well as

in formation of vascular collaterals are dependent on the disruption of this coat9.

Angiogenesis in the Adult

Angiogenesis involves a tightly regulated cascade of events12. ECs respond to the mitogenic

signals from angiogenic factors such as VEGF. Blood vessels lose their pericyte coverage,

the basement membrane and the extracellular matrix (ECM) are degraded by proteases and

formed EC tubuli are subsequently covered and stabilized by deposition of a basement

membrane and recruitment of perivascular pericytes and SMCs9. SMCs are important

regulators of vascular tone and provide support for the larger vessels. Angiogenesis in

healthy adults occurs only during certain processes such as in the ovary and endometrium

during the female reproductive cycle and the placenta during pregnancy14. Angiogenesis

also occurs during hair follicle development and exercise-induced remodelling of muscles11.

Angiogenesis also happens in a variety of pathophysiological conditions including wound

healing, solid tumor growth, ischemic disease such as ischemic heart and peripheral

Figure. 1 Embryonic development of the vasculature starts with the development of angioblasts fromhemangioblast precursors, subsequent vasculogenesis and angiogenesis and finally recruitment of mural cellsfor stabilisation of the newly formed vessels (Adapted from Carmeliet 2003 and Conway 200110,11).

Hemangioblastprecursor

Angioblasts VasculogenesisAngiogenesis

Mural cellrecruitment

Maturevasculature

Page 13: Vascular Endothelial Growth Factors B and C: Gene

13

disease, psoriasis, atherosclerosis, development of vascular tumors such as hemangiomas

and during pathological ocular neovascularisation15,16.

Recently the blood circulation has been suggested to contain circulating endothelial cell

progenitors (CEPs) which incorporate into newly formed vessels at sites of active

angiogenesis17-20. The contribution of CEPs to neovascularisation mimics embryonic

neovascularisation by differentiation of circulating angioblasts and is thus also termed

vasculogenesis. CEPs express a variety of endothelial cell surface markers including VEGF

receptors, platelet endothelial adhesion molecule-1 (PECAM-1) and von Willebrand factor

(vWF). CEPs are mobilised from the bone marrow by cytokines such as VEGF and

PlGF21,22. Skeletal muscle has also been suggested to contain cells with stem cell properties

that can contribute to angiogenesis, raising the possibility that tissues can contain resident

angioblasts23. CEPs are incorporated into forming blood vessels during physiological

angiogenesis such as during the female reproductive cycle24, during pathologic

angiogenesis in experimental solid tumors or in the limb muscles or the myocardium during

ischemia24-27. Furthermore, CEPs can develop a non-thrombogenic surface on endovascular

grafts after transplantation 28,29.

Figure 2. Vascular remodelling requires the interplay of different cell populations. Macrophages are recruitedto sites of active angiogenic cytokine production, followed by detachement of pericytes and SMCs andendothelial cell sprouting and/or vessel enlargenment. Remodelled blood vessels are stabilised by recruitedmural cells and failure to recruit these results in regression of the newly formed vessels (Adapted fromCarmeliet 2003 and Conway 200110,11).

Vasculogenesis

Macrophage recruitment

Rerecruitment of pericytes

= Macrophage = Hemangioblast = Endothelial cells = Mural cells

Angiogenesis Vessel regression

Remodeled, mature blood vessel

Page 14: Vascular Endothelial Growth Factors B and C: Gene

14

Molecular Regulators of the Blood Vascular System

Figure 3. The angiogenic and arteriogenic members of the VEGF family of growth factors and their cognatereceptors. Extracellular immunoglobulin homology domains are represented by white circles, theextracellular domains of Nrp-1 and Nrp-2 are shown in striped boxes and tyrosine kinase (TK) domains arerepresented by black boxes.

VEGF

VEGF is the ligand for VEGFR-1 and VEGFR-2 and one of the key players in the

induction of blood vessel formation30 (Figure 3). Human VEGF is expressed as seven

different splice isoforms consisting 121, 138, 145, 162, 165, 189 and 206 amino acid

residues respectively, these differ in their binding affinity for heparin and in their

interaction with neuropilins (Nrp) 1 and 2. VEGF is an EC mitogen, motogen,

chemoattractant and survival factor that increases the permeability of blood vessels31. The

importance of VEGF in the formation of the vascular- and hematopoietic systems is

exemplified by vegf gene haploinsuffiency in mice with one inactivated vegf allele5. Mice

deficient in VEGF die early in embryonic development due to defects in angiogenesis and

hematopoiesis4. Partial inactivation of VEGF function by deletion of isoforms 164 and 188

affects postnatal angiogenesis in the heart and kidney, retinal vascular patterning and

enchondral bone formation32-34. Loss of approximately 50% of progeny suggests that the

NRP-2 NRP-2

Page 15: Vascular Endothelial Growth Factors B and C: Gene

15

functions of VEGF isoforms are not redundant. Selective expression of VEGF120 resulted

in defective vascular outgrowth and patterning and selective expression of VEGF188 gave

rise to impaired arterial development35. Overexpression of VEGF in vivo induces blood

vessel formation as evidenced by adenoviral overexpression of VEGF in the skin of

mice36,37. Transcription of vegf is regulated by a variety of growth factors such as basic

Fibroblast Growth Factor (bFGF), Transforming Growth Factor-β (TGF-β) and Platelet

Derived Growth Factor (PDGF) as well as by oncogenes such as ras38. Importantly, VEGF

transcription is governed by hypoxia through interaction of hypoxia responsive elements

(HREs) in the VEGF gene promoter with HIF-1α, a transcription factor that mediates

intracellular hypoxic signaling39,40. Induction of VEGF expression by growth factors,

hypoxia and oncogenes mediates angiogenesis in solid tumors and in tissue ischemia7,41,42.

VEGF-B

VEGF-B is a ligand for VEGFR-1 and Nrp-1 (Figure 3). It can form heterodimers with

VEGF43-46(Table 1). VEGF-B is expressed as two different isoforms consisting of 167 or

186 amino acid residues47. VEGF-B167 binds heparan sulphate proteoglycans (HSPGs) and

is mostly sequestered in the ECM while VEGF-B186 is freely diffusible. In vivo, VEGF-

B167 is the predominant form and is abundantly expressed in brown fat, myocardium and

skeletal muscle48. VEGF-B may regulate the plasminogen activator activity in ECs45.

Deletion of the VEGF-B gene does not result in an obvious phenotype in mice49. These

mice do however have somewhat smaller hearts and a prolonged PQ interval in

electrocardiograms and they recover less well from myocardial ischemia than wild-type

mice4950. Furthermore, one report has claimed that mice lacking VEGF-B fail to develop

pulmonary hypertension in response to hypoxia, suggesting a role for this growth factor in

hypoxic pulmonary vascular remodelling51. Strikingly, VEGF-B was also reported to

protect mice lacking VEGF-B from the development of hypoxic pulmonary hypertension in

a manner comparable to VEGF52. Lack of VEGF-B does not affect development of the

retinal vasculature and neither does it affect development of hypoxia induced retinopathy of

the newborn53.

Page 16: Vascular Endothelial Growth Factors B and C: Gene

16

Table 1. Structure of VEGF family members.

Characteristic VEGF-A(Genatlasdatabase)

PlGF30,76 VEGF-B45 VEGF-C77 VEGF-D78

Gene 15.9 kb,7 exons

13.9 kb,7 exons

3.71 kb,7 exons

109.2 kb,7 exons

38.86 kb,7 exons

Protein spliceisoforms

VEGF-A121,

VEGF-A138,VEGF-A145,VEGF-A162,VEGF-A165,VEGF-A189,VEGF-A206

PlGF-1(PlGF131),PlGF-2(PlGF152),PlGF-3

VEGF-B167,VEGF-B186

PrecursorVEGF-C388

PrecursorsVEGF-D358,VEGF-D326

Chromosomelocation

6p12 14q24.3 11q13 4q34 Xp22.31

Sequencehomology

Isoformactivities aremodulated bytheir heparansulfate andneuropilinbinding;

42% sequenceidentity toVEGF-A

45% sequenceidentity toVEGF-A

30%homologousto VEGF-A165

61% sequenceidentity toVEGF-C;31% identicalto VEGF-A165

Dimerization Homodimers;heterodimerwith VEGF-Band PlGF

Homodimers,heterodimerswith VEGF-A

Homodimers;heterodimerswith VEGF-A

Homodimers;heterodimerswith VEGF-D

Homodimers;heterodimerswith VEGF-C

PlGF

PlGF and VEGF-B share properties such as VEGFR-1 and Nrp-1 binding and the capacity

to form heterodimers with VEGF (Table 1 and Figure 3). Of the three reported human

isoforms of PlGF (PlGF-1, -2 and -3)54-57 only PlGF-2 binds HSPGs58. Only the PlGF-2

isoform has been found in mice. PlGF/VEGF heterodimers bind VEGFR-2 and VEGFR-

1/VEGFR-2 heterodimers in vitro59,60. PlGF is predominantly expressed in the placenta,

heart and in the lungs61. Heterozygous deletion of the plgf gene does not result in any

apparent phenotypic change, these mice do however suffer from impaired recovery after

experimental myocardial infarcts and from impaired collateral formation during hind limb

ischemia62. Furthermore, administration of recombinant PlGF induced angiogenesis and

collateral formation after ligation of coronary or femoral arteries in wild-type mice63.

Overexpression of PlGF in the skin of transgenic mice resulted in a hypervascular

phenotype with increased inflammatory and permeability responses64,65 and local

administration of PlGF using recombinant adenoviruses or recobinant peptides induced the

Page 17: Vascular Endothelial Growth Factors B and C: Gene

17

formation of enlarged, leak-resistant blood vessels by arteriogenesis63 in a macrophage-

dependent manner66. Activation of VEGFR-1 by PlGF or VEGF was reported to induce

different gene expression profiles by phosphorylation of distinct tyrosine residues in the TK

domain of VEGFR-167 and combined administration of these factors enhanced VEGF

driven angiogenesis in vitro and in vivo62,68.

VEGF Homologues Encoded by Orf and Parapox Viruses

Genes with sequence homology to VEGF were discovered in two strains of orf viruses and

one strain of pseudocowpox viruses. These viruses denoted ORFVNZ2, ORFVNZ769 and

PCPV(VR634)70 , cause highly vascularised and edematous pustular dermatitis in sheep,

goats and occasionally in humans. Two of these viral VEGFs specifically bind and activate

VEGFR-2 and Nrp-171-73 whereas one binds only VEGFR-270. Consequently,

overexpression of ORFVNZ7 VEGF in the skin of transgenic mice results in a non-

edematous angiogenic phenotype77. Although viral VEGFs do not play a role in mammalian

vascular biology, they can be used as VEGFR-2 specific agonists in experimental models

of angiogenesis in vitro and in vivo.

VEGFR-2 and VEGFR-1

VEGFR-1 is a receptor for VEGF, VEGF-B and PlGF (Figure 3). VEGFR-1 by itself

transmits only weak mitogenic signals in ECs78 but it forms heterodimers with VEGFR-2

with stronger signalling properties than both VEGFR-1 and VEGFR-279. Targeted

disruption of vegfr1 results in embryonic lethality from an excess of ECs and disturbed

organisation of ECs into blood vessels6. The promoter of vegfr1 contains HREs and unlike

for VEGFR-2 and VEGFR-3, transcription of VEGFR-1 is induced by hypoxia41. In the

embryo, VEGFR-1 is expressed in angioblasts and ECs but is downregulated later in

development6,80,81. Chemotaxis of macrophages and macrophage mediated arteriogenesis is

relayed via VEGFR-1 signalling63,66,82. Furthermore, VEGFR-1 may mobilise bone-marrow

derived stem cells that incorporate into sites of active arteriogenesis62. A alternatively

spliced soluble form of VEGFR-1, sFlt-1, inhibits VEGF function and supresses tumor

angiogenesis83. This soluble form of VEGFR-1 has also been implicated in pre-eclampsia84.

VEGFR-2 binds VEGF, VEGF-C, VEGF-D and VEGF-E and relays important signals for

endothelial cell survival as well as in the migration and proliferation of ECs in VEGF

driven angiogenesis (Figure 3). Selective activation of VEGFR-1 and VEGFR-2 has shown

that VEGFR-2 is the primary receptor transmitting VEGF signals in ECs85. VEGFR-2 is

expressed by ECs and by embryonic angioblasts during development. In the adult, VEGFR-

Page 18: Vascular Endothelial Growth Factors B and C: Gene

18

2 is expressed by ECs during angiogenesis4,5,86. Disruption of the gene for VEGFR-2 in

transgenic mice results in embryonic lethality at an early developmental stage due to

defects in angiogenesis, vasculogenesis and hematopoiesis3. The expression of vegfr2 is

autoregulated: VEGF, VEGF-C and VEGF-D upregulate the expression of VEGFR-287,88

(III). Blockade of VEGFR-2 effectively blocks VEGF driven angiogenesis in experimental

tumors in mice89.

Neuropilins

The Neuropilins, Nrp-1 and Nrp-2 are receptors for semaphorins, molecules that govern

axonal guidance during neuronal development90,91. Nrp-1 and Nrp-2 are expressed by ECs

and VEGFs bind to them in an isoform specific fashion56,92,46,93,94 (Figure 3). Accordingly,

targeted disruption of Nrp1 and/or Nrp2 results in disturbed formation of the vasculature,

defects of the nervous system and, in the case of Nrp-1, lethality during embryonic

development95-98.

Ephs and Ephrins

The Ephs are transmembrane receptor TKs that direct axonal guidance during neural

development as well as migration and morphogenesis of other cells types such as ECs.

Ephs relay bidirectional signalling through interaction with ephrins, a class of cell

membrane bound ligands. In the vascular system ephrin-B2 and EphB4 form a ligand

receptor pair, whose components are specifically expressed in arteries and veins,

respectively. It has therefore been suggested that bidirectional signalling by this ligand

receptor pair relays repulsive signalling between the cell populations that eventually form

arteries and veins during embryonic develoment99. Targeted disruption of ephrin B2 or

EphB4 results in defective arterio-venous patterning of the primary vascular plexus and

consequent embryonic lethality100-102.

Platelet Derived Growth Factors (PDGFs)

The PDGFs function as paracrine growth factors during development and as mitogens for a

wide array of cells such as fibroblasts, SMCs and ECs. The eight cysteine residues of the

VEGF growth factors are conserved in the PDGFs103. Structurally, the PDGFs consist of

homo- and heterodimers formed by four different monomers: PDGF-A, PDGF-B, PDGF-C

and PDGF-D that bind their cognate RTKs PDGFR-α and PDGFR-β103. Receptor

specificity is determined by dimer composition, PDGF-AA, -AB, -BB and PDGF-CC can

Page 19: Vascular Endothelial Growth Factors B and C: Gene

19

induce αα-receptor homodimers, PDGF-AB and -BB can induce αβ-receptor heterodimers

and PDGF-BB and PDGF-DD can induce ββ-receptor dimers. PdgfB and Pdgfrβ are

essential for vascular development as the deletion of either gene results in embryonic

lethality due to hemorrhage caused by failure to assemble a mural-cell coating of blood

vessels104-106. Deficiency of PDGF-A and PDGFR-α causes embryonic lethality due to non-

vascular developmental defects107-110. In the adult, PDGF signalling occurs in wound

healing and is implicated in atherosclerosis, fibrotic diseases and cancer111,112.

Angiopoietins and Tie Receptors

The vascular structures are regulated in part by a family of receptors called the Tie

receptors and their ligands, the angiopoietins113. The Tie receptors, Tie-1 and Tie-2 have

divergent functions during development; Tie-1 is required for the structural integrity of

blood vessels during development, and a defiency in Tie-1 results in hemorraghe, edema

and embryonic death at E13.5114,115. Tie-2, or tunica interna endothelial cell kinase-1 (Tek),

mediates vascular-pericyte interactions in vessel remodeling and maturation114-117. While the

ligand for Tie-1 is not known, Tek has two ligands, Ang-1 and Ang-2, which have opposite

functions, Ang-1 is an agonist and Ang-2 is an antagonist of Tek. This theory is supported

by studies in transgenic mice in which overexpression of Ang-2 has a phenotype similar to

that of mice deficient in Ang-1118,119. The role of the Tie-Ang system in angiogenesis seems

to be the maturation of newly formed blood vessels by interactions with pericytes and

SMCs. Overexpression of Ang-1 in transgenic mice or by adenoviral gene transfer blocks

vessel permeability as induced by VEGF and co-expression of VEGF and Ang-1 induces

the formation of enlarged vessels that are leak-resistant120. On the other hand, it has been

suggested that Ang-2 induces detachment of pericytes from blood vessels and thereby

facilitates endothelial cell responses to EC mitogens121. Interestingly, Ang-2 knock-out

mice have defects in the lymphatic vasculature and chylous ascites, suggesting a role for

angiopoietin-2 in lymphatic development122. The lymphatic, but not the blood vascular

phenotype of these mice can be rescued by substitution of Ang-1 in place of Ang-2122.

Markers of the Vascular System

PECAM-1

PECAM-1 is a 130 kD glycoprotein member of the immunoglobulin (Ig) superfamily that

is abundantly expressed on the lateral junctions of endothelial cells as well as on the surface

of platelets and leukocytes123. PECAM-1 consists of six extracellular Ig domains, a

Page 20: Vascular Endothelial Growth Factors B and C: Gene

20

transmembrane domain and a cytoplasmic domain. The ligands for PECAM-1 include

itself, CD38 and αvβ3 integrin. Phosphorylation of intracytoplasmic tyrosyl residues of

PECAM-1 mediates the recruitment of proteintyrosine phosphatases and results in

supression of TK signalling124. In angiogenesis, PECAM-1 is involved in the formation and

stabilization of endothelial cell-cell contacts and maintenance of a vascular permeability

barrier125-127. PECAM-1 also mediates transendothelial migration of monocytes and

neutrophils128.

vWF

Factor VII related antigen (FVIIIRA) or vWF is a multimeric protein produced by

endothelial cells that mediates clotting on injured endothelial surfaces. vWF stains the

endothelium of especially larger vessels whereas capillaries stain more weakly and

lymphatic ECs are negative for vWF expression129,130. Thus, vWF expression demonstrates

specificity for certain EC populations.

VE-cadherin

Cell-cell and cell-matrix adhesions are mediated by integrins, selectins and cadherins. VE-

cadherin is an EC-specific adhesion molecule localised to the adherens junctions of most

ECs populations131. Targeted deletion of VE-cadherin is lethal as mice deficient in VE-

cadherin fail to develop a proper vascular architechture132. Among vascular markers, VE-

cadherin is unique in that it is not expressed in hematopoietic precursor cells or circulating

blood cells. VE-cadherin inhibits VEGFR-2 signalling and EC growth in a density

dependent manner in vitro133,134.

Pericyte Markers

Markers for the SMCs and pericytes that coat the endothelium in blood vessels and

lymphatic vessels include α-smooth muscle actin (α-SMA), PDGF-Rβ, desmin, human

melanoma associated antigen (HMV-MAA) and its murine counterpart NG2. α-SMA is

expressed in pericytes and SMCs covering arterioles, arteries and post-capillary venules in

normal human and murine tissues135. α-SMA expression in pericytes is altered in tumors136

and can be induced by TGF-β137. Smooth muscle actin is not expressed in the pericytes that

cover capillaries138. PDGFR-β is co-expressed with the HMW-MA on pericytes and

mediates recruitment of pericytes to form blood vessels139. HMV-MAA is a proteoglycan

originally described as a molecule expressed on pericytes in tumors and healing wounds,

Page 21: Vascular Endothelial Growth Factors B and C: Gene

21

and its murine counterpart NG2 is expressed by activated pericytes during both normal and

pathological neovascularisation140-142. Desmins are intermediate filaments that form part of

the cytoskeleton in striated and SMCs; mice deficient in desmin display defects in all types

of muscle tissue and die of cardiac failure143,144.

Pathologic Angiogenesis and Arteriogenesis

Blood vessel formation plays a critical role in a number of pathophysiological conditions,

most notably cardiovascular disease and cancer, the biggest causes of mortality in

developed countries. Many other disorders such as psoriasis, atherosclerosis, hemangiomas,

vascular malformations, Kaposi's sarcoma, diabetic retinopathy and arthritis are also

associated with disturbed blood vessels functions11. To date most attempts at interfering

with pathological angiogenesis have targeted solid tumors, peripheral tissue ischemia and

myocardial ischemia.

Angiogenesis in Ischemic Disease

Reduced blood perfusion of tissues results in hypoxia which triggers specific intracellular

signalling pathways and subsequent gene transcription7. Cellular hypoxia leads to inhibition

of prolyl hydroxylases that regulate the degradation of transcription factors HIF-1α to 3α7.

These factors form complexes with ARNT (HIF-1β) and bind HREs in the promoters of

hypoxia inducible genes involved in glucose transport, glycolysis, and angiogenesis.

Examples of hypoxia-regulated genes include Glut-1, Cox-2, VEGF and VEGFR-17.

Deletion of a HRE in the VEGF promoter in transgenic mice results in progressive motor

neuron degeneration presumably because of insufficient vascular perfusion of nervous

tissue and direct effects on motoneuron survival via loss of VEGF induction145. The

immature morphology of blood vessels generated in response to hypoxia-induced VEGF is

exemplified in the newborn by retinopathy and the chaotic vessel hierarchy and lack

integrity in tumor angiogenesis146. Thus, VEGF driven angiogenesis in progressive

ischemic disease may not be sufficient to restore perfusion because the formed vessels are

not functional.

Arteriogenesis

Reduction of blood flow in thromboembolic disease leads to collateral formation or

arteriogenesis, a process in which pre-existing arterioles enlarge, leading to a 10- to 20-fold

increase in blood flow147. Increased fluid shear stress induces expression of monocyte

Page 22: Vascular Endothelial Growth Factors B and C: Gene

22

chemotactic proteins and cell adhesion molecules, which recruit macrophages and initiate

arteriogenesis. The recruited macrophages release proteases that degrade the basal lamina

enabling the vessel to enlarge148. A new basal lamina is synthesized and mural cells such as

SMCs and pericytes are recruited to stabilize the remodelled vessel149. Enlargement of the

pre-existing collaterals is a process mediated by cytokines that enhance recruitment of

monocytes or prolong their life span such as monocyte chemoattractant protein-1 (MCP-1),

granulocyte monocyte-colony stimulating factor (GM-CSF), TGF-β or tumor necrosis

factor-α (TNF-α). Cytokines such as PlGF or FGFs not only recruit monocytes, but also act

directly on ECs and SMCs by inducing the growth of these cell populations. In general,

induction of vascular collaterals by cytokines in experimental animal models of myocardial

and hindlimb ischemia may restore perfusion and rescues tissue from

necrosis62,150,151.However, presumably because arteriogenesis is initiated by shear stress, the

arteriogenic process is prematurely terminated as shear stress decreases in the enlarging

vessel. Thus, compensatory conductance by arteriogenesis often remains at less than 50%

of the original conduction prior to occlusion.

Tumor Angiogenesis

Blood vessels in tumors display several distinct features that have implications for tumor

biology and treatment. Tumor angiogenesis is mediated mainly by VEGF expression in the

tumor cells152,153. Blood vessels in tumors have a chaotic structure and are leaky. As a

consequence, blood flow in the tumor is sluggish and the interstitial fluid pressure is

high154. The sluggish blood flow results in hypoxic necrosis of the inner parts of the tumor

and perpetuates VEGF production and the high interstitial fluid pressure hampers delivery

of therapeutic agents154. Since tumor growth is angiogenesis dependent, therapeutic

targeting of the tumor vasculature is an attractive alternative or adjunct to conventional

chemo- or radiotherapy155. ECs in tumors express a number of molecules that are unique to

tumor blood vessels such as VEGFR-2 and integrin αVβ3 that can be targeted for

therapeutic purposes. Furthermore, ECs are genetically stable and apparently cannot

develop drug resistance in the manner of tumor cells. To date, a number of angiogenesis

inhibitors have been tested in phase I-III clinical trials and one, the anti-VEGF blocking

antibody bevacizumab (Avastin, rhuMAb-VEGF) has shown efficacy in the treatment of

metastatic colorectal cancer and renal cancer156,157.

Page 23: Vascular Endothelial Growth Factors B and C: Gene

23

The Lymphatic System

Embryonic Development, Structure and Function

The lymphatic system is a network of vessels that maintains tissue homeostasis by

collecting extravasated plasma and returning it to the blood circulation via the lymph

nodes. The lymph also transports fatty acids absorbed in the gut and performs

immunosurveillance functions by directing leukocytes, especially antigen-presenting cells

such as Langerhans cells, and antigens to the lymph nodes. Accordingly, improper

functioning of the lymphatic system is implicated in a variety of diseases including

lymphedema, inflammation, infectious and immune disease. In cancer, malignant tumors

form distant metastases mainly by spreading through the lymphatics. The lymphatic system

was first described in the 17th century by Gasparo Aselli as he observed vessels filled with a

“milky fluid” in the stomach of a well-fed dog161. The lymphatic vessels consist of

capillaries that form collecting lymphatics which eventually empty into the venous system

Figure 4. According to the model proposed by Wigle et al158, venous ECs are initially pluripotent and committo the lymphatic phenotype after expression of Prox-1 begins. Committed venous ECs then express lymphaticmarkers such as VEGFR-3 and secondary lymphoid chemokine (SLC), and form lymphatic vessels bysprouting in response to VEGF-C159(adapted from Detmar and Oliver 2002160).

LYVE-1

Prox-1

LYVE-1 LYVE-1

Prox-1 SLC+

VEGF-C

VEGF-R3+

VEGF-C

Page 24: Vascular Endothelial Growth Factors B and C: Gene

24

Table 2. Morphological characteristics of blood and lymphatic vessel capillaries.Blood vessels Lymphatic vessels

Anchoring filaments Absent Present

Basement membrane Present Mostly absent

Endothelial cell cytoplasm Abundant Scant

Interendothelial tight junctions Common Infrequent

Lumen Regular Irregular

Overlapping endothelial cells Absent Present

Perivascular support structures Present Scant

(Based on data from Schmid-Schonbein 1990 and Nathanson, 2003)

via the thoracic duct. The lymphatic vessels themselves are thin tubes formed by ECs; the

larger lymphatics may contain a sparse covering of pericytes or SMCs and valves similiar

to those found in veins. Ultrastructurally, lymphatic vessels differs from blood vessels in

that that they have overlapping, valve-like endothelial cell junctions, a discontinous or

absent basement membrane and anchoring filaments in the vessel wall162 (Table 2). In

mouse embryos the first lymphatic outgrowths called lymphatic primordia can be identified

at embryonic day 10.5. The lymphatic primordia develop by budding from large central

veins in the perimesonephric and jugular regions. Subsequently, these lymphatic primordia

remodel into primitive lymphatic sacs from which the lymphatics develop by

sprouting163,164. An alternative hypothesis proposes that the lymphatics develop in situ from

mesenchymal precursors, or lymphangioblasts, or by a combination of sprouting and

development in situ165,166. The former theory is supported by studies demonstrating that

expression of VEGFR-3, which is initially expressed in veins, is later restricted to

lymphatic endothelia167-169. Furthermore, the prospero related homeobox protein-1 (Prox-1)

is initially expressed in a subset of ECs in the anterior cardinal vein which subsequently

buds to form the primitive lymph sacs (Figure 4). Deletion of Prox1 gene results in lack of

the lymphatics (see below). Support for the theory on the incorporation of

lymphangioblasts into developing lymphatics is obtained from studies in the avian

chorioallantoic membrane assay and in chick-quail chimeras170. In these studies, homotopic

transplantation of Prox-1 negative tissue resulted in lymphatics composed of cells from

both species171. Lymphangiogenesis in adults is assumed to occur in concert with

angiogenesis, as has been demonstrated during wound healing in pigs129.

Page 25: Vascular Endothelial Growth Factors B and C: Gene

25

Molecular Regulators of the Lymphatic System

Figure 5. Ligand-receptor interactions in the induction of lymphangiogenesis. Receptor extracellular Ighomology domains are represented by white circles, the extracellular domains of Nrp-1 and Nrp-2 are shownas striped boxes and the TK domains are represented by black boxes.

VEGF-C and VEGF-D

VEGF-C and VEGF-D (also called C-fos induced factor or figf) share sequence homology

with VEGF (Table 1) and bind VEGFR-2 and VEGFR-3172-175. In vivo, VEGF-C and

VEGF-D are potent inducers of lymphangiogenesis176.

The expression patterns of VEGF-C and VEGFR-3 during embryogenesis, suggests that

these molecules signal in a paracrine manner177. Expression of VEGF-C is first detected in

the head mesenchyme and the developing vertebrae at E8.5. Subsequently, VEGF-C

mRNA localises to the metanephric and jugular areas at sites where the primitive lymph

sacs develop, suggesting that VEGF-C has a role in the formation of these structures177. The

expression then decreases in several tissues, remaining high in the lymph nodes178. It may

be that in adults, VEGF-C expression is mainly regulated by proinflammatory cytokines

and growth factors, suggesting a role for lymphangiogenesis in inflammatory disease. In

contrast to VEGF, VEGF-C is not induced by hypoxia (I, II).

NRP-2NRP-2

Page 26: Vascular Endothelial Growth Factors B and C: Gene

26

In lymphatic ECs VEGF-C induces migration, proliferation and survival signals in

vitro179. In vivo, overexpression of VEGF-C or VEGF-D in the epidermis of transgenic

mice induces lymphatic vessel hyperplasia while local overexpression by adenoviral gene

transfer induces sprouting lymphangiogenesis176,180 (III). Transgenic mice lacking both

Vegfc alleles fail to develop lymphatic vessels and succumb to tissue edema at E15.5-

17.5159. Loss of one vegfc allele results in a phenotype characterised by hypoplasia of the

cutaneous lymphatic vessels and lymphedema159. Sprouting of endothelial cells committed

to the lymphatic endothelial lineage in vegfc-/- mice could be induced by recombinant

VEGF-C and to a lesser degree with VEGF-D, but not VEGF indicating that development

of the lymphatic vessels is dependent on VEGF-C in a non-redundant fashion159.

VEGF-C and VEGF-D have long N-terminal and C-terminal extensions that are

proteolytically cleaved upon secretion181,182. The receptor specificity of VEGF-C and

VEGF-D is regulated by this proteolytic processing as only the fully processed secreted

forms are capable of efficient binding of VEGFR-2181,182.

In vivo, VEGFR-3 activation is sufficient to induce lymphangiogenesis while the

vascular permeability induced by VEGF-C and VEGF-D is mediated by VEGFR-2182,183.

Furthermore, overexpression of a soluble form of the extracellular domain of VEGFR-3

blocks lymphangiogenesis in transgenic mice184. Interestingly, murine VEGF-D binds only

to VEGFR-3185. VEGF-D overexpression by adenoviral gene transfer in mouse skin or in

experimental tumors induces both angiogenesis and lymphangiogenesis88,176,186.

VEGFR-3

VEGFR-3 (fms-like tyrosine kinase 4, Flt4) is a transmembrane TK consisting of six

extracellular immunoglobulin like loops and an intracellular part containing a TK domain

for interaction with intracellular signal transduction molecules187,188. In humans, alternative

splicing of the VEGFR-3 gene generates two isoforms of VEGFR-3 that differ in their C-

termini and are designated VEGFR-3s (s=short) and VEGFR-3l (long)189. Expression of

VEGFR-3 is restricted to the lymphatic ECs in the adult whereas a blood vascular ECs also

express VEGFR-3 during embryonic develoment (Figure 4). VEGFR-3 defiency results in

embryonic lethality due to defects in cardiovascular remodelling167. In ECs, VEGFR-3

relays survival signalling by activation of Akt and p42/p44 MAPK. Migration of lymphatic

ECs is also stimulated by VEGFR-3 signals179. Intriguingly, a subpopulation of CD34

positive CEPs express VEGFR-3, and some of these CEPs differentiate in vitro and start to

express lymphatic endothelial cell markers suggesting that circulating lymphangioblasts

exist and could contribute to lymphangiogenesis in some conditions190.

Page 27: Vascular Endothelial Growth Factors B and C: Gene

27

Markers of the Lymphatic System

Lymphatic Vessel Endothelial Hyaluronan Receptor-1 (LYVE-1)

LYVE-1 is receptor for hyaluronan, an ECM glycosaminoglycan and a homologue of

CD44, a hyaluronan receptor expressed in lymphocytes191,192. The function of LYVE-1 in

lymphatic vessels may be to transport hyaluronan into the lymphatic vessels for transport

to, and degradation in regional lymph nodes192. However, LYVE-1 is not restricted to the

lymphatic ECs but is also expressed in normal liver blood sinusoids, activated macrophages

and capillaries in the lung193,194.

Podoplanin

Podoplanin is a 38 kD glomerular podocyte membrane mucoprotein expressed by the

lymphatic endothelium and glomerular epithelial cells. Podoplanin is expressed in all cells

in Kaposi’s sarcoma195 and angiosarcomas 196 and is co-expressed with VEGFR-3 in the

skin and kidney. During embryogenesis, podoplanin is first expressed between E10.5 and

11.5 in ECs of the cardinal vein and in budding Prox-1 positive lymphatic ECs197.

Podoplanin seems to be a Prox-1 regulated gene198,199. Homozygous deletion of podoplanin

in mice results in abnormal development of lymphatic vessels and lymphedema197.

Overexpression of podoplanin in vascular endothelial cell cultures promotes the formation

of elongated cell extensions and significantly increased endothelial cell adhesion, migration

and tube formation197.

Prox-1

Prox-1 is a transcription factor expressed in lymphatic ECs which has a similiar pattern of

expression to that of VEGFR-3. Knock-out studies have shown that Prox-1 is essential for

the formation of the lymphatic system during embryogenesis169 as well as for the

development of the liver 200 and the lens169. Prox-1 is expressed in the subpopulation of ECs

that give rise to the lymphatic sacs201 and sprouting and migration of ECs from these sacs is

impaired in the Prox1-/- mice158. Haploinsufficiency of Prox1 leads to intestinal chylous

fluid accumulation and death shortly after birth169. Evidence for the role of Prox-1 in the

determination of lymphatic endothelial cell fate is further demonstrated by the fact that its

overexpression in blood vascular ECs results in an alteration of their gene expression

profile towards that of the lymphatic endothelial cells198,199.

Page 28: Vascular Endothelial Growth Factors B and C: Gene

28

Pathologic Lymphangiogenesis

Tumor Lymphangiogenesis

The intravasation of tumor cells into lymphatic vessels is one of the first steps in lymphatic

metastasis. Consequently, lymphangiogenesis around a solid tumor should promote

lymphatic metastasis by providing a larger target for the intravasation of tumor cells.

Overexpression of VEGF-C in experimental tumors in mice induces growth of lymphatic

vessels into the tumor stroma indicating that expression of this growth factor in tumors can

induce lymphangiogenesis202,203. Furthermore, combined overexpression of VEGF-C and

the oncogenic SV40 virus large T antigen in the pancreatic islets of transgenic mice

induced lymphangiogenesis and promoted lymph node metastasis as compared to mice

expressing only the oncogene204. Overexpression of VEGF-D, a growth factor with both

lymphangiogenic and angiogenic properties, also promoted lymph node metastasis in

experimental tumors in mice88. Interestingly, neutralisation of VEGF-C and VEGF-D in an

experimental tumor by systemic overexpression of a recombinant molecule consisting of

the extracellular part of VEGFR-3 fused to the Fc domain of immunoglobulin G (VEGFR-3

Ig) reduced the number of lymph node metastases, indicating that inhibition of tumor

lymphangiogenesis could have therapeutic potential205. Although enlarged lymphatic

vessels at the tumor edge have been reported in human cancers, functional intratumoral

lymphatic vessels have not been reported206,207. One explanation could be that the high

interstitial fluid pressure in tumors compresses lymphatic vessels. According to another

hypothesis, compressed lymphatic vessels seen in the tumor stroma did not grow there by

lymphangiogenesis but were pre-existing lymphatic vessels that were engulfed by the

expanding tumor208. However, lymph node metastasis in human cancers correlates with

tumoral VEGF-C or VEGF-D expression209,210 and although lymphatic metastasis is not

necessarily dependent on lymphangiogenesis207, tumor lymphangiogenesis has been

reported to be a prognostic indicator for cutaneous melanoma211,212 and squamous cell

cancers of the head and neck213,214.

Lymphedemas

Lymphedemas are disorders of the lymphatic system in which lymphatic drainage is

impaired with resultant accumulation of fluid and swelling of the limbs or the scrotum. In

these disorders an exudate of protein rich fluid accumulates in the subcutis. This leads to

subcutaneous fibrosis, impaired immune responses and degeneration of the adipose tissue.

Page 29: Vascular Endothelial Growth Factors B and C: Gene

29

As a result lymphedema patients suffer from recurrent infections and impaired wound

healing.

Lymphedemas are classified as primary, congenital lymphedemas and secondary or

acquired lymphedemas. Primary lymphedemas develop at birth or during adolescence and

rarely occur in adults. The lymphatic vessel structure in individuals with primary

lymphedemas is disturbed. Congential hereditary lymphedema or Milroy’s disease is

characterised by hypoplastic superficial lymphatic vessels215 while the lymphatics in

Meige’s disease, or late-onset congenital lymphedema appear enlarged. Recently, the

genetic defects underlying two forms of primary lymphedema have been characterised. In

some patients with Milroy`s disease missense mutations in the TK domain of VEGFR-3

inhibit the function of this receptor with resultant lymphedema216. In lymphedema

distichiasis, (LD) mutations in the FOXC2 gene have been identified217. FOXC2 is a

member of the forkhead/winged helix family of transcription factors that have a variety of

functions during embryonic develoment218. Other hereditary congenital lymphedemas with

putative, as yet uncharacterised genetic defects, include Turner`s syndrome and cholestasis-

lymphedema.

Secondary lymphedema is caused by iatrogenic trauma such as irradiation,surgery

or infection. Iatrogenic trauma is exemplified by lumpectomy in breast cancer patients, of

which 6-30% suffer from postoperative lymphedema219. The most common cause of

secondary lymphedema is filariasis that is caused by the parasitic worms Wucheriya

bancroftii or Brugia malayi with a prevalence of over 120 million cases worldwide.

Page 30: Vascular Endothelial Growth Factors B and C: Gene

30

Therapeutic Approaches

Vascular Gene Therapy

The molecular networks that regulate the vascular system provide attractive targets for

therapy for various diseases. Most attempts at amplifying or inhibiting angiogenic

signalling utilises systemic administration of recombinant proteins or transfer of gene

constructs that overexpress a growth factor or an angiogenesis inhibitor. Generally, gene

transfer in vivo requires the use of vectors such as cationic liposomes or recombinant

viruses but naked plasmid DNA can also be taken up by muscle tissue with resultant

expression of the transferred gene85. Gene transfer by cationic liposomes is relatively

inefficient and results only in short term gene expression220. By contrast recombinant

viruses efficiently transfer genes which are then expressed for a prolonged period of time221.

Viral Vectors

Recombinant adenoviruses infect both dividing and non-dividing cells that express the

adenovirus vector receptors CAR, αVβ3 and αVβ5 integrin222,223. In immunocompetent

hosts, adenoviral gene transfer results in transient expression of the transferred gene for 1-2

weeks. The lack of chromosomal integration of the transferred DNA as well as the transient

nature of adenoviral gene expression make recombinant adenoviruses relatively safe and

they have therefore been widely used in clinical trials. Long term expression of transferred

genes can be achieved by recombinant retroviruses, adeno-associated (AAV) viruses and

lentiviruses. Retroviruses have mainly been used for ex-vivo gene therapy of bone marrow

or muscle cells in therapy of diseases such as severe combined immune deficiency and

hemophilia224,225. AAVs efficiently infect skeletal muscle, smooth muscle cells of blood

vessels and the myocardium, making it an attractive vector for gene therapy of

cardiovascular diseases226,227, both AAVs and lentiviruses readily infect the central nervous

system and the liver228. Vectors that integrate into the host genome raise a number of safety

issues, and cases of malignant transformation of target cells in humans have been

described229. Furthermore, prolonged expression of genes transferred via AAVs can

produce unwelcome effects such as hemangiomas after transfer of angiogenic growth

factors230. Thus, the clinical use of these vectors may require the development of vector

systems that can be regulated exogenously.

Page 31: Vascular Endothelial Growth Factors B and C: Gene

31

Proangiogenic Therapy

Although cells can respond to hypoxia by expressing angiogenic cytokines such as VEGF,

angiogenesis or collateral blood vessel formation is rarely sufficient to restore normal

perfusion. For this reason, attempts have been made to boost collateral vessel formation by

infusion or local expression in vivo of angiogenic cytokines231. The induction of

angiogenesis or arteriogenesis has been attempted in cardiovascular disease in man by

employing various growth factors administered either as recombinant proteins or by local

overexpression using gene therapy 232-236. Vascular growth factors that have proceeded to

phase II and phase III pro-angiogenic clinical trials include VEGF, FGF-2, GM-CSF and

FGF-4220. The efficacy of recombinant growth factor therapy for myocardial infarction or

peripheral vascular disease varies from neglible to improved function as measured by an

exercise tolerance test while adenoviral gene transfer of FGF-4 or VEGF showed efficacy

in improving myocardial and peripheral perfusion in patients with ischemic disease220.

In addition to adminstration of recombinant growth factor peptides or recombinant

viruses, the recent discovery of CEPs has raised the possibility of using these cells in the

treatment of disorders of the vascular system19. CEPs enriched ex vivo have been used to

treat patients suffering from acute myocardial infarction or lower limb ischemia resulting in

improved blood perfusion of the ischemic tissues237,238,239,240.

Lymphangiogenic Therapy

Lymphangiogenic gene therapy aims to correct congenital or acquired defects of the

lymphatic system by induction of lymphangiogenesis in vivo using lymphangiogenic

growth factors such as VEGF-C or VEGF-D241. Induction of lymphangiogenesis with

recombinant growth factors or by gene transfer of lymphangiogenic genes has restored

lymphatic function in experimental animal models of lymphedema. Mutations in the

VEGFR-3 gene cause the Chy phenotype in mice; this is characterised by an absence

subcutaneous lymphatic vessels and accumulation of chylous ascites after birth242. The Chy

genotype is analogous to that of Milroy`s disease in humans as mutations in the VEGFR3

gene cause both syndromes. Overexpression of VEGF-C in the skin of the Chy mice results

in the formation of functional lymphatic vessels, suggesting that high enough levels of

growth factor can compensate the impaired function of VEGFR-3 and that pre-existing

lymphatic vessels are not required for lymphangiogenesis in these mice243. Moreover,

administration of recombinant VEGF-C or VEGF-D protein can reconstitute the lymphatic

circulation in a rabbit ear model of lymphedema244,245. However, the efficacy of VEGF-C

and VEGF-D in the treatment of lymphedema in humans remains to be demonstrated.

Page 32: Vascular Endothelial Growth Factors B and C: Gene

32

AIMS OF THE STUDY

I. Study of the regulation of VEGF-C and VEGF-B mRNA by oncogenes, tumor promoter,

hypoxia and various cytokines compared to VEGF.

II. Characterisation of the induction of VEGF-C by pro-inflammatory cytokines

III. Experiments to explore if gene transfer and subsequent over-expression of VEGF-C can

induce lymphangiogenesis in vivo.

IV. Comparison of the effects of two VEGFR-1 agonists, VEGF-B and PlGF, on the mouse

vasculature in vivo.

Page 33: Vascular Endothelial Growth Factors B and C: Gene

33

MATERIALS AND METHODS

The materials and methods used in I-IV are accounted for below and described in detail in

the indicated publications.

Methods Used inAdenoviral infection of cell cultures III, IVCell culture I, II, III, IVDNA cloning and subcloning I, III, IVGeneration of recombinant adenoviruses III, IVGeneration of stable cell lines I, IVGeneration of transgenic mice IVImmunofluorescence IVImmunohistochemistry III, IVImmunoprecipitation II, III, IVMorphometry IIINorthern blotting I,II, III, IVNucear run-on IIPCR IVPreparation of mouse tissues III, IVReceptor stimulation and ligand binding III, IVSouthern blotting IVTransfection of cells I, II, III, IVWhole-mount immunohistochemistry IV

Cell line Description Source Used in293EBNA Epstein Barr (EBNA)

nuclear antigenATCC III,IV

Ba/F3

expressing humanembryonic kidney cellsMouse Pre-B lymphocytes Dr. T. Mäkinen IV

C6 Rat glioblastoma cells ATCC IHeLa Human adenocarcinoma ATTC IVHT-1080 Human fibrosarcoma ATCC IHUVEC Human umbilical vein EC Dr. A. Ristimäki I, IIIMR-90 Human fibroblast ATCC I, IINIH3T3 Mouse fibroblasts ATCC I

Page 34: Vascular Endothelial Growth Factors B and C: Gene

34

Growth factor Description Source Used inEGF Human epidermal growth factor R&D IIL-1β Human interleukin-1β R&D IIPDGF-B Human Platelet Derived Growth Factor-B R&D IPlGF Human Placenta Growth Factor-2 R&D IVPMA Phorbol 12-myristate 13-acetate Sigma I, IITGF-β Human transforming growth factor-β R&D ITNF-α Human tumor necrosis factor-α R&D ΙΙ

Antigen Antibodies Source Used inLYVE-1 Rabbit antiserum against mouse LYVE-1 Dr. D.Jackson III, IV

Rabbit antiserum against mouse LYVE-1 Dr. T.Petrova IVPCNA Mouse mAb against human PCNA ZYMED IIICD45 Rat mAb against mouse CD45 Pharmingen IVSMA Rat mAb against mouse SMA Sigma III, IVVEGF Rat mAb against mouse VEGF R&D III, IVVEGF-B Rabbit Ab against human VEGF-B Dr.B.Olofsson IVPlGF Rat mAb against human PlGF R&D IVPECAM-1 Mouse mAb against mouse PECAM-1 Pharmingen III, IVVEGF-C Rabbit Ab against human VEGF-C Dr. V.Joukov I, IVF4/80 Rat mAb against mouse F4/80 Serotec IV

Page 35: Vascular Endothelial Growth Factors B and C: Gene

35

RESULTS AND DISCUSSION

I. Studies on the regulation of VEGF, VEGF-B, VEGF-C and Ang-1 mRNA expression

The regulation of mRNA expression provides important clues to the biologic functions and

molecular regulators of angiogenesis. For this reason we compared the transcriptional

responses of VEGF-C, VEGF-B, Ang-1 and VEGF to a variety of stimuli such as serum

growth factors, hypoxia and oncogenes.

The mRNA levels of VEGF and VEGF-C in starved cultured cells increased in

response to FCS, and to a lesser degree in response to PDGF and TGF-β, both principal

component growth factors in FCS. As lymphangiogenesis has been described in wounds,

these data suggest that VEGF-C could be induced in response to serum growth factors in

wounds and subsequent lymphangiogenesis could clear extravasated plasma in granulation

tissue. Interestingly, Ang-1 mRNA levels decreased in response to FCS suggesting that

reduced levels of this cytokine may be required for vessel de-stabilisation and angiogenesis

in wound healing. However, according to recent reports on wound healing in mice, Ang-1

and Tie-1 expression is not altered during wound healing whereas Ang-2 and Tie-2

expression is strongly elevated246.

Activation of oncogenes increases the VEGF mRNA levels in cell culture providing

a link between tumor progession and tumor angiogenesis. Activation of an inducible

recombinant ras gene construct in cultured cells raised the VEGF mRNA levels while

VEGF-B and VEGF-C mRNA levels remained unaffected. Interestingly, several human

tumors express VEGF-C in vivo and show proliferation and growth of lymphatic vessels at

the tumor edge. VEGF-C in tumors could also be regulated in an indiarect manner by

inflammatory cells. Ras activation did not affect VEGF-C mRNA levels suggesting that the

Grb-Shc-Sos-Ras pathway is not involved in the regulation of VEGF-C expression.

However, phorbolester (PMA) induced an increase in VEGF-C mRNA levels presumably

through interaction with an AP-1 site in the VEGF-C gene promoter247. Induction of VEGF

mRNA levels by hypoxia is one of the key features of this gene as it governs the induction

of angiogenesis in tissue ischemia and during solid tumor growth30. VEGF responds to

hypoxia via HIF-1 induced activation of transcription and stabilisation of the mRNA`s 3´-

prime untranslated region248. In contrast, neither VEGF-C nor VEGF-B mRNA levels were

induced by hypoxia thus rendering VEGF the only hypoxia regulated growth factor in the

family as neither PlGF nor VEGF-D transcription is directly regulated by hypoxia249. Our

studies further indicated that VEGF-B mRNA has a very long half-life, over 8 hours,

VEGF-C mRNA has a half-life of approximately 3.5 hours and VEGF has a half-life of 1

Page 36: Vascular Endothelial Growth Factors B and C: Gene

36

hour. The mRNA half-lives of VEGF-C and VEGF were prolonged in the prescence of

serum, resulting in higher mRNA levels. The lack of induction of VEGF-B mRNA is

consistent with its very long half-life.

II. Proinflammatory Cytokines Regulate Expression of VEGF-C

The effect of the pro-inflammatory cytokines TNF-α and interleukin-1 (IL-1β) on VEGF,

VEGF-B, VEGF-C and Ang-1 mRNAs was studied in cultured human lung fibroblasts and

HUVECs in vitro. Incubation with recombinant IL-1β induced transcription of VEGF and

VEGF-C while Ang-1 transcription was decreased and VEGF-B transcription remained

unaffected. The level of secreted, partially processed VEGF-C also increased in resoponse

to TNF-α and IL-1β. The increased VEGF-C mRNA levels induced by TNF-α and IL-1β

were mainly the result of increased transcription as evidenced by nuclear run-on data. Thus,

transcriptional regulation of VEGF-C mRNA differs from that of VEGF in that VEGF

mRNA levels are regulated both by transcriptional activation and post-transcriptional

stabilisation mediated by RNA protein interactions in the 3`-UTR of the VEGF gene250. The

promoter of the VEGF-C gene contains a putative NF-κB binding sites247 that could

mediate the activation VEGF-C mRNA transcription as has been reported for

proinflammatory cytokines251. Induction of VEGF-C mRNA levels by proinflammatory

cytokines was inhibited by dexamethasone but not by indomethacine suggesting that

prostanoids do not mediate the effects of proinflammatory cytokines on VEGF-C mRNA

transcription. In contrast, induction of VEGF mRNA transcription by proinflammatory

cytokines is mediated indirectly by prostanoids252. However, a recent report demonstrated

that prostaglandin E2 mediated COX-2 activation increased VEGF-C mRNA transcription

indicating that prostanoids in some cells can induce VEGF-C driven lymphangiogenesis253.

Lymphangiogenesis in breast cancer was shown to correlate with COX-2 expression and

prostaglandin E2 mediated activation of COX-2 was shown to increase transcription of

VEGF-C mRNA in vitro253. Furthermore, both VEGF-C and COX-2 immunoreactivity in

clinical samples of breast cancer correlated with prognosis suggesting a role for prostanoid

driven lymphangiogenesis in mammary carcinomas253. VEGF-C mRNA transcription

wasalso induced in HUVECs in response to proinflammatory cytokines. In HUVECs,

VEGFR-2 transcription was increased by IL1-β. The regulation of VEGF-C mRNA

transcription by proinflammatory cytokines indicates that VEGF-C could regulate

lymphatic vessel function during inflammation, reflecting the role of the lymphatics in the

control of immune function and lymphocyte trafficking. Accordingly, VEGF-C is highly

expressed in arthritc joints in patients with rheumatoid arthritis in the absence of frank

lymphangiogenesis suggesting that either the lymphahangiogenic response is insufficient in

Page 37: Vascular Endothelial Growth Factors B and C: Gene

37

this disorder or that VEGF-C participates in other functions254,255. Recent reports have

shown that inflammatory reactions in the tracheal mucosa or in rejected human kidney

transplants are accompanied by abundant lymphangiogenesis (Tuomas Tammela, personal

communication) and that reactive lymphadenitis is mediated by VEGFR-3 signalling

(Peter Baluk, Donald McDonald, personal communication)256. In a rabbit cornea model of

inflammatory angiogenesis and lymphangiogenesis, depletion of the bone marrow or local,

selective depletion of macrophages blocked lymphangiogenesis, demonstrating that

inflammatory cells can mediate the formation of lymphatic vessels257. The angiogenic and

lymphangiogenic response was blocked by VEGF inhibitors indicating that VEGF induces

both angiogenesis and lymphangiogenesis in this model. However, recruited macrophages

expressed abundant amounts of VEGF-C and VEGF-D, suggesting that the

lymphangiogenic effect of VEGF is indirect and mediated by inflammatory cytokines that

induce the expression of lymphangiogenic cytokines in the inflammatory infiltrate.

Page 38: Vascular Endothelial Growth Factors B and C: Gene

38

Figure 6. Adenoviral overexpression of VEGFs or ß-galactosidase (LacZ) in vivo in the skin reveals the effectof these growth factors on the blood vessels and lymphatic vessels. While VEGF induces sproutingangiogenesis (A-C, arrows in C), VEGF-C is a selective lymphangiogenic growth factor (G-I). In contrast,VEGF-B has no apparent vascular effects in the skin (D-F, open arrowheads in G). Scalebar = 200 µm.

Page 39: Vascular Endothelial Growth Factors B and C: Gene

39

III. Adenoviral Expression of VEGF-C in the Skin Induces Lymphangiogenesis

The formation of lymphatic vessels or lymphangiogenesis occurs mainly during

embryogenesis and has been described in adults only during wound healing and

inflammation129,256,257. However, the induction of lymphangiogenesis could have therapeutic

value in the treatment of congenital, infectious, traumatic or iatrogenic lymphedema241.

Focal induction of lymphangiogenesis can be achieved by gene therapy and is a requisite

for successful growth factor therapy of lymphedemas. To explore, these therapeutic

concepts, recombinant adenoviruses encoding VEGF-C were constructed and used for

transfer of the VEGF-C gene into the skin of mice. Detection of lymphatic vessels by

immunohistochemistry for the lymphatic endothelial marker LYVE-1 combined with

detection of the proliferating cell nuclear antigen showed that VEGF-C induced abundant

lymphangiogenesis at two weeks after injection. Over-expression of VEGF-C induced the

expression of VEGFR-2 and VEGFR-3 in blood vessels raising the interesting possibility

that this growth factor could have vascular effects in vivo as well. However, VEGF-C did

not significantly affect the blood vessels as compared to VEGF, which induced intense

angiogenesis (Figure 6).

The partially overlapping receptor binding profiles of VEGF, VEGF-C and VEGF-

D have raised the question of how specific these factors are for lymphangiogenesis or

angiogenesis. Studies on the effect of overexpression of VEGF-C and VEGF-D in normal

tissues and in tumors indicate that VEGF-C is mainly lymphangiogenic while VEGF-D is

both angiogenic and lymphangiogenic180,258. Adenoviral overexpression of VEGF-C in the

skin of nude mice induced lymphangiogenesis and enlargement of blood vessel but no

sprouting angiogenesis180,258. Overexpression of VEGF-C in experimental tumors in mice

resulted in a robust lymphangiogenic, but only a slight angiogenic response259,204. The

angiogenic response in tumors overexpressing VEGF-C could be reduced by blocking

VEGF-R2260. However, while some authors report only lymphangiogenic effects of VEGF-

C in the chick chorioallantoic membrane assay (CAM) and in the mouse cornea assay,

others also detect angiogenesis in these assays, depending on the developmental stage of

the CAM261,262,263,264. Furthermore, VEGF-C exerts angiogenic effects after gene transfer

with recombinant plasmids in ischemic rabbit hindlimbs further indicating that VEGF-C

has angiogenic properties265. The receptor binding profiles of VEGF-C and VEGF-D are

determined by proteolytic processing as only the fully processed 21 kD forms of VEGF-C

and VEGF-D bind efficiently to VEGFR-2182. In vitro, VEGFR-2 relays the angiogenic

effects of VEGF and the mature, protelytically processed form of VEGF-C. Thus the

proteolytic processing of VEGF-C and VEGF-D as well as the upregulation of VEGFR-2

and VEGFR-3 on blood vessels in response to VEGF, VEGF-C and VEGF-D could explain

Page 40: Vascular Endothelial Growth Factors B and C: Gene

40

the angiogenic effects of VEGF-C and VEGF-D266. Plasmin proteolytically cleaves the N-

and C-terminal extensions of the immature forms of VEGF-C and VEGF-D thus regulating

their affinity for VEGFR-2 and VEGFR–3267. The presence of plasmin in the

microenvironment could determine the angiogenic and/or lymphangiogenic effects of

VEGF-C and VEGF-D in vivo. For specific, VEGFR-3 mediated lymphangiogenesis a

mutant form of VEGF-C that only activates VEGFR-3 can be used268. Selective activation

of VEGFR-3 circumvents potential side-effects of VEGFR-2 activation such as peripheral

edema269,270.

The lymphangiogenic effect of VEGF has been investigated in a number of studies.

Adenoviral overexpression of VEGF in the skin has yielded conflicting results as one study

has reported induction of giant lymphatic vessels in response to VEGF271 while others have

failed to detect any such effects180. Adenoviral overexpression of VEGF in muscle or

recombinant VEGF administered to the CAM or to the mouse cornea failed to induce

lymphangiogenesis176,263,272. Furthermore, studies in murine embryonic explants of VEGF-C

knock-out mice showed that VEGF-C, but not VEGF has the capacity to induce migration

of endothelial cells commited to the lymphatic endothelial lineage159. VEGF was reported

to induce the formation of “giant lymphatics” or "lymphangioma-like structures" after

adenoviral gene transfer of VEGF in the skin of nude mice271, an effect that we failed to

detect (Figure 6). However, the murine form of VEGF164 was used in the study by Nagy et

al and recent collaborative work between Dr. Nagy and Dr. Tuomas Tammela has shown

that the lymphangiogenic effect of murine VEGF164 and human VEGF165 differ in this

assay (Tuomas Tammela, personal communication).

IV. VEGF-B Activates VEGFR-1, But Provides Very Little Angiogenic or Arteriogenic

Activity In Vivo in Comparison with PlGF

The effect of VEGFR-1 agonists on the vasculature of mice was studied in vivo by

overexpression of two VEGFR-1 agonists, VEGF-B and PlGF by adenoviral gene transfer

in the skin of transgenic mice. While the effects of VEGF, VEGF-C and VEGF-D have

been extensively characterised in vivo, the functions PlGF and VEGF-B have remained

elusive. Recently, PlGF was reported to induce the formation of vascular collaterals in

experimental myocardial infarctions and lower limb ischemia in mice63 by amplification

VEGF signalling62,67. Thus, VEGFR-1 agonists are potentially useful in therapy aiming to

restore blood perfusion in tissue ischemia273. Overexpression of PlGF in the skin of

transgenic mice64,77 resulted in a vascular phenotype with enlarged tortous blood vessels,

morphologically similar to those seen after adenoviral overexpression of PlGF in the skin63.

In contrast, our findings demonstrate that over-expression of neither isoform of VEGF-B is

Page 41: Vascular Endothelial Growth Factors B and C: Gene

41

able to induce vascular remodelling in a manner similar to PlGF in the skin of transgenic or

nude mice (Figure 6). Whole mount immunohistochemistry to detect ECs, pericytes and

macrophages revealed arteriogenic and angiogenic effects of PlGF and VEGF respectively.

In contrast, none of the cell populations mentioned above responded to VEGF-B. As

vascular collateral formation, or arteriogenesis, is dependent on macrophage recruitment,

failure of VEGF-B to induce arteriogenesis in skeletal muscle could result from an inability

of VEGF-B to recruit macrophages when compared to PlGF or VEGF. The failure of

VEGF-B to induce remodelling of the vasculature is not dependent on prolonged exposure

of the growth factor as neither overexpression in vivo for six weeks, nor constitutive

overexpression in the skin or heart of transgenic mice resulted in a modified vascular

phenotype.

Clues to the biological function of VEGF-B may be provided by reports according

to which VEGF-B is highly expressed in striated and heart muscle and brown fat as well as

in numerous tumors274,275,276 (I). Thus, the biologic function of VEGF-B may be linked to

energy metabolism or tumor angiogenesis. Interestingly, transgenic mice deficient in

VEGF-B have subtle myocardial defects and impaired recovery after experimental

myocardial infarcts suggesting that the regeneration of the coronary circulation could at

least in part be dependent on VEGF-B50. According to one published study these mice

should also display reduced angiogenic responses in collagen induced arthritis, suggesting a

role for VEGF-B in inflammatory angiogenesis277. Finally, according to another isolated

study, overexpression of VEGF-B in vivo by injection of recombinant plasmids in a mouse

model of hindlimb ischemia induced angiogenesis and VEGF-B may thus have subtle

angiogenic effects in vivo278. However, our comparison of the vascular effects of VEGF-B

and PlGF overexpression in vivo indicates that the arteriogenic potential of VEGF-B is

modest at best.

The failure to detect a biological role for VEGF-B could depend on a number of

factors. One may be the species specificity of the factors and models used. For example,

human VEGF-B might not be optimal in mice. The study of cardiovascular physiology in

man requires animal models in which normal and pathological cardiovascular physiology

resembles that of humans as closely as possible. As the effects of VEGF-B in vivo have to

date been exclusively studied in mice using human recombinant VEGF-B, a proper study of

angiogenic growth factors in vivo should ultimately be conducted in relevant animal

models such as in pigs or dogs using corresponding recombinant human, porcine or canine

growth factors. Taken together, the above studies suggest that the angiogenic effect of

VEGF-B may be heart-specific.

Page 42: Vascular Endothelial Growth Factors B and C: Gene

42

CONCLUDING REMARKS

The discovery and characterisation of the molecular networks governing vascular

remodelling in adults has raised the possibility of using various components of these

networks for therapeutic purposes. Vascular growth factors such as the VEGF family and

their cognate receptors form a palette of tools that can be used for induction of blood- or

lymphatic vessel growth which could be useful for treatment of peripheral ischemic

disease, myocardial ischemia and disorders of the lymphatic system. Furthermore,

inhibition of angiogenesis and lymphangiogenesis may become incorporated into emerging

treatments for cancer.

Determination of the optimal therapeutic use of vascular cytokines requires

thorough understanding of their underlying biological functions. Intensive efforts over the

past ten years have clarified the function of a number of the cytokines that regulate the

vasculature. VEGF family members can now be used for induction of angiogenesis,

arteriogenesis and lymphangiogenesis while the angiopoietins could be necessary for

proper disassembly/assembly of the perivascular support structures. Many of the above

mentioned growth factors have shown promise in pre-clinical trials and a few, such as

VEGF have proceeded to bona fide clinical trials. However, successful use of vascular

cytokines for therapeutic purposes requires the development of refined gene transfer

techniques which allow for exogenous regulation and efficient over-expression of the

relevant genes at any site of an ongoing patho-physiological process.

Page 43: Vascular Endothelial Growth Factors B and C: Gene

43

ACKNOWLEDGEMENTS

This study was carried out at the Molecular/Cancer Biology Laboratory, the

Molecular/Cancer Biology Research Program at Biomedicum Helsinki and the Dept. of

Pathology, Haartman Institute, University of Helsinki.

I would like to thank professors Eero Saksela and Olli Jänne for providing excellent

research facilities in the Haartman Institute and Biomedicum Helsinki, respectively. My

sincere thanks go to my supervisor professor Kari Alitalo who gave me the opportunity to

work in his laboratory at the very forefront of international biomedical science. The

reviewers of this thesis professor Ulf-Håkan Stenman and docent Olli Ritvos are

acknowledged for their great insights and constructive criticisms of this thesis. I would like

to thank all past and present members of the Molecular/Cancer Biology Laboratory for

fruitful collaborations over the years. I would also like to thank all co-authors, especially

Ulf Eriksson, Seppo Ylä-Herttuala, Frej Stenbäck and Ari Ristimäki for their positive

attitude towards scientific collaboration with a junior scientist. Technical support at the

M/CBL is top-notch and Tapio Tainola, Sanna Lampi, Riikka Ekman, Alun Parsons,

Angela Flynt, Paula Hyvärinen and Mari Helanterä are kindly acknowledged for their help

in all matters technical over the years.

My parents Monica and Sten and my brother Robin have always provided generous

support of all kinds for which I am deeply thankful. Finally, I want to thank my wife Susa

and our son Oskar for those most important things in life.

Finska Läkare Sällskapet played a crucial role in the funding of this study and the support

of that society is greatly acknowledged. This work was also supported by the Finnish

Cancer Organizations, the Finnish Medical Foundation, the Helsinki University Central

Hospital (Helsinki and Uusimaa Hospital Group, Special State Grants), Helsinki University

Chancellor’s Office, the Paulo Foundation, the K. Albin Johansson Foundation, the Oskar

Öflund Foundation and the Maud Kuistila in Memoriam Foundation.

Page 44: Vascular Endothelial Growth Factors B and C: Gene

44

REFERENCES

1. Risau W. Mechanisms of angiogenesis. Nature. 1997;386:671-674.2. Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat. Med.

2000;6:389-395.3. Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML,

Schuh AC. Failure of blood-island formation and vasculogenesis in Flk-1-deficientmice. Nature. 1995;376:62-66.

4. Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O'Shea KS, Powell-BraxtonL, Hilan KJ, Moore MW. Heterozygous embryonic lethality induced by targetedinactivation of the VEGF gene. Nature. 1996;380:439-442.

5. Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, FahrigM, Vandenhoeck A, Harpal K, Ebenhardt C, Declercq C, Pawling J, Moons L,Collen D, Risau W, Nagy A. Abnormal blood vessel development and lethality inembryos lacking a single VEGF allele. Nature. 1996;380:435-439.

6. Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role of the Flt-1 receptortyrosine kinase in regulating the assembly of vascular endothelium. Nature.1995;376:66-70.

7. Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIFsystem. Nat. Med. 2003;9:677-84.

8. Schweiki D, Itin A, Soffer D, Keshet E. Vascular endothelial growth factor inducedby hypoxia may mediate hypoxia-initiated angiogenesis. Nature. 1992;359:843-848.

9. Jain RK. Molecular regulation of vessel maturation. Nat. Med. 2003;9:685-93.10. Conway EM, Collen D, Carmeliet P. Molecular mechanisms of blood vessel

growth. Cardiovasc. Res. 2001;49:507-521.11. Carmeliet P. Angiogenesis in health and disease. Nat. Med. 2003;9:653-660.12. Veikkola T, Karkkainen M, Claesson-Welsh L, Alitalo K. Regulation of

angiogenesis via vascular endothelial growth factor receptors. Cancer Res.2000;60:203-12.

13. Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenicswitch during tumorigenesis. Cell. 1996;86:353-364.

14. Findlay JK. Angiogenesis in reproductive tissues. J. Endocrinol. 1986;111:357-66.15. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature.

2000;407:249-255.16. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat.

Med. 1995;1:27-31.17. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler

B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells forangiogenesis. Science. 1997;275:964-967.

18. Rafii S, Meeus S, Dias S, Hattori K, Heissig B, Shmelkov S, Rafii D, Lyden D.Contribution of marrow-derived progenitors to vascular and cardiac regeneration.Semin. Cell Dev. Biol. 2002;13:61-67.

19. Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organvascularization and regeneration. Nat. Med. 2003;9:702-12.

20. Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M, Magner M, IsnerJM, Asahara T. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat. Med. 1999;5:434-8.

21. Rafii S, Avecilla S, Shmelkov S, Shido K, Tejada R, Moore MA, Heissig B, HattoriK. Angiogenic factors reconstitute hematopoiesis by recruiting stem cells from bonemarrow microenvironment. Ann. N. Y. Acad. Sci. 2003 May;996:49-60.

22. Gill M, Dias S, Hattori K, Rivera ML, Hicklin D, Witte L, Girardi L, Yurt R, HimelH, Rafii S. Vascular trauma induces rapid but transient mobilization ofVEGFR2(+)AC133(+) endothelial precursor cells. Circ. Res. 2001;88:167-74.

Page 45: Vascular Endothelial Growth Factors B and C: Gene

45

23. Majka SM, Jackson KA, Kienstra KA, Majesky MW, Goodell MA, Hirschi KK.Distinct progenitor populations in skeletal muscle are bone marrow derived andexhibit different cell fates during vascular regeneration. J. Clin. Invest.2003;111:71-9.

24. Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, Kearne M,Magner M, Isner JM. Bone marrow origin of endothelial progenitor cellsresponsible for postnatal vasculogenesis in physiological and pathologicalneovascularization. Circ. Res. 1999;85:221-228.

25. Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F, Nadal-Ginard B,Bodine DM, Leri A, Anversa P. Mobilized bone marrow cells repair the infarctedheart, improving function and survival. Proc. Natl. Acad. Sci. U.S.A.2001;98:10344-9.

26. Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, Kearne M,Magner M, Isner JM. Bone marrow origin of endothelial progenitor cellsresponsible for postnatal vasculogenesis in physiological and pathologicalneovascularization. Circ. Res. 1999;85:221-8.

27. Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, Chadburn A, Heissig B,Marks W, Witte L, Wu Y, Hicklin D, Zhu Z, Hackett NR, Crystal RG, Moore MA,Hajjar KA, Manova K, Benezra R, Rafii S. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesisand growth. Nat. Med. 2001;7:1194-201.

28. Bhattacharya V, McSweeney PA, Shi Q, Bruno B, Ishida A, Nash R, Storb RF,Sauvage LR, Hammond WP, Wu MH. Enhanced endothelialization and microvesselformation in polyester grafts seeded with CD34(+) bone marrow cells. Blood.2000;95:581-5.

29. Rafii S, Oz MC, Seldomridge JA, Ferris B, Asch AS, Nachman RL, Shapiro F,Rose EA, Levin HR. Characterization of hematopoietic cells arising on the texturedsurface of left ventricular assist devices. Ann. Thorac. Surg. 1995;60:1627-32.

30. Ferrara N, Gerber H-P, LeCouter J. The biology of VEGF and its receptors. Nat.Med. 2003;9:669-676.

31. Ferrara N, Davis-Smyth T. The biology of vascular endothelial growth factor.Endocr. Rev. 1997;18:4-25.

32. Maes C, Carmeliet P, Moermans K, Stockmans I, Smets N, Collen D, Bouillon R,Carmeliet G. Impaired angiogenesis and endochondral bone formation in micelacking the vascular endothelial growth factor isoforms VEGF164 and VEGF188.Mech. Dev. 2002;111:61-73.

33. Mattot V, Moons L, Lupu F, Chernavvsky D, Gomez RA, Collen D, Carmeliet P.Loss of the VEGF(164) and VEGF(188) isoforms impairs postnatal glomerularangiogenesis and renal arteriogenesis in mice. J. Am. Soc. Nephrol. 2002;13:1548-60.

34. Carmeliet P, Ng YS, Nuyens D, Theilmeier G, Brusselmans K, Cornelissen I, EhlerE, Kakkar VV, Stalmans I, Mattot V, Perriard JC, Dewerchin M, Flameng W, NagyA, Lupu F, Moons L, Collen D, D'Amore PA, Shima DT. Impaired myocardialangiogenesis and ischemic cardiomyopathy in mice lacking the vascular endothelialgrowth factor isoforms VEGF164 and VEGF188. Nat. Med. 1999;5:495-502.

35. Stalmans I, Ng YS, Rohan R, Fruttiger M, Bouche A, Yuce A, Fujisawa H,Hermans B, Shani M, Jansen S, Hicklin D, Anderson DJ, Gardiner T, Hammes HP,Moons L, Dewerchin M, Collen D, Carmeliet P, D'Amore PA. Arteriolar andvenular patterning in retinas of mice selectively expressing VEGF isoforms. J. Clin.Invest. 2002;109:327-336.

36. Sundberg C, Nagy JA, Brown LF, Feng D, Eckelhoefer IA, Manseau EJ, DvorakAM, Dvorak HF. Glomeruloid microvascular proliferation follows adenoviralvascular permeability factor/vascular endothelial growth factor-164 gene delivery.Am. J. Pat. 2001;158:1145-60.

37. Pettersson A, Nagy JA, Brown LF, Sundberg C, Morgan E, Jungles S, Carter R,Krieger JE, Manseau EJ, Harvey VS, Eckelhoefer IA, Feng D, Dvorak AM,

Page 46: Vascular Endothelial Growth Factors B and C: Gene

46

Mulligan RC, Dvorak HF. Heterogeneity of the angiogenic response induced indifferent normal adult tissues by vascular permeability factor/vascular endothelialgrowth factor. Lab. Invest. 2000;80:99-115.

38. Rak J, Mitsuhashi Y, Bayko L, Filmus J, Shirasawa S, Sasazuki T, Kerbel RS.Mutant Ras oncogenes upregulate VEGF/VPF expression - implications forinduction and inhibition of tumor angiogenesis. Cancer Res. 1995;55:4575-4580.

39. Liu Y, Cox SR, Morita T, Kourembanas S. Hypoxia regulates vascular endothelialgrowth factor gene expression in endothelial cells. Identifications of 5' enhancer.Circ. Res. 1995;77:638-643.

40. Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD, Semenza GL.Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol. Cell. Biol. 1996;16:4604-13.

41. Gerber HP, Conorelli F, Park J, Ferrara N. Differential transcriptional regulation ofthe two vascular endothelial growth factor receptor genes. Flt-1, but not Flk-1/KDR,is up-regulated by hypoxia. J. Biol. Chem. 1997;272:23659-23667.

42. Detmar M, Brown LF, Berse B, Jackman RW, Elicker BM, Dvorak HF, ClaffeyKP. Hypoxia regulates the expression of vascular permeability factor/vascularendothelial growth factor (VPF/VEGF) and its receptors in human skin. J. Invest.Dermatol. 1997;108:263-8.

43. Grimmond S, Lagercrantz J, Drinkwater C, Silins G, Townson S, Pollock P, GotleyD, Carson E, Rakar S, Nordenskjold M, Ward L, Hayward N, Weber G. Cloningand characterisation of a novel human gene related to vascular endothelial growthfactor. Genome Res. 1996;6:124-131.

44. Olofsson B, Pajusola K, Kaipainen A, von Euler G, Joukov V, Saksela O, OrpanaA, Pettersson RF, Alitalo K, Eriksson U. Vascular endothelial growth factor B, anovel growth factor for endothelial cells. Proc. Natl. Acad. Sci. USA. 1996;93:2576-2581.

45. Olofsson B, Korpelainen E, Mandriota S, Pepper MS, Aase K, Kumar V, Gunji Y,Jeltsch MM, Shibuya M, Alitalo K, Eriksson U. Vascular endothelial growth factor(VEGF) B binds to VEGF receptor-1 and regulates plasminogen activator activity inendothelial cells. Proc. Natl. Acad. Sci. USA. 1998;95:11709-11714.

46. Makinen T, Olofsson B, Karpanen T, Hellman U, Soker S, Klagsbrun M, ErikssonU, Alitalo K. Differential binding of vascular endothelial growth factor B splice andproteolytic isoforms to neuropilin-1. J. Biol. Chem. 1999;274:21217-21222.

47. Oloffson B, Pajusola K, von Euler G, Chilov D, Alitalo K, Eriksson U. Genomicorganization of the mouse and human genes for vascular endothelial growth factorB (VEGF-B) and characterization of a second splice isoform. J. Biol. Chem.1996;271:19130-19317.

48. Olofsson B, Jeltsch M, Eriksson U, Alitalo K. Current biology of VEGF-B andVEGF-C. Curr. Opin. Biotech. 1999;10:528-535.

49. Aase K, von Euler G, Li X, Ponten A, Thoren P, Cao R, Cao Y, Olofsson B, Gebre-Medhin S, Pekny M, Alitalo K, Betsholtz C, Eriksson U. Vascular endothelialgrowth factor-B-deficient mice display an atrial conduction defect. Circulation.2001;104:358-364.

50. Bellomo D, Headrick JP, Silins GU, Paterson CA, Thomas PS, Gartside M, MouldA, Cahill MM, Tonks ID, Grimmond SM, Townson S, Wells C, Little M,Cummings MC, Hayward NK, Kay GF. Mice lacking the vascular endothelialgrowth factor-B gene (Vegfb) have smaller hearts, dysfunctional coronaryvasculature, and impaired recovery from cardiac ischemia. Circ. Res. (Online).2000;86:E29-35.

51. Wanstall JC, Gambino A, Jeffery TK, Cahill MM, Bellomo D, Hayward NK, KayGF. Vascular endothelial growth factor-B-deficient mice show impaireddevelopment of hypoxic pulmonary hypertension. Cardiovasc. Res. 2002;55:361-368.

52. Louzier V, Raffestin B, Leroux A, Branellec D, Caillaud JM, Levame M, EddahibiS, Adnot S. Role of VEGF-B in the lung during development of chronic hypoxic

Page 47: Vascular Endothelial Growth Factors B and C: Gene

47

pulmonary hypertension. Am. J. Physiol. Lung Cell Mol. Physiol. 2003;284:L926-37.

53. Reichelt M, Shi S, Hayes M, Kay G, Batch J, Gole GA, Browning J. Vascularendothelial growth factor-B and retinal vascular development in the mouse. Clin.Experiment. Ophthalmol. 2003;31:61-5.

54. Maglione D, Guerriero V, Viglietto G, Delli-Bovi P, Persico MG. Isolation of ahuman placenta cDNA coding for a protein related to the vascular permeabilityfactor. Proc. Natl. Acad. Sci. USA. 1991;88:9267-9271.

55. Maglione D, Guerriero V, Viglietto G, Ferraro MG, Aprelikova O, Alitalo K, DelVecchio S, Lei K-J, Chou JY, Persico MG. Two alternative mRNAs coding for theangiogenic factor, placenta growth factor (PlGF), are transcribed from a single geneof chromosome 14. Oncogene. 1993;8:925-931.

56. Migdal M, Huppertz B, Tessler S, Comforti A, Shibuya M, Reich R, Baumann H,Neufeld G. Neuropilin-1 is a placenta growth factor-2 receptor. J. Biol. Chem.1998;273:22272-22278.

57. Cao Y, Ji WR, Qi P, Rosin A. Placenta growth factor: identification andcharacterization of a novel isoform generated by RNA alternative splicing.Biochem. Biophys. Res. Commun. 1997;235:493-8.

58. Hauser S, Weich HA. A heparin-binding form of placenta growth factor (PlGF-2) isexpressed in human umbilical vein endothelial cells and in placenta. GrowthFactors. 1993;9:259-268.

59. DiSalvo J, Bayne ML, Conn G, Kwok PW, Trivedi PG, Soderman DD, Palisi PM,Sullivan KA, Thomas KA. Purification and characterisation of a naturally occurringvascular endothelial growth factor. Placenta growth factor heterodimer. J. Biol.Chem. 1995;270:7717-7723.

60. Cao Y, Linden P, Shima D, Browne F, Folkman J. In vivo angiogenic activity andhypoxia induction of heterodimers of placenta growth factor/vascular endothelialgrowth factor. J. Clin. Invest. 1996;98:2507-2511.

61. Persico MG, Vincenti V, DiPalma T. Structure, expression and receptor-bindingproperties of placenta growth factor (PlGF). In: Claesson-Welsh L, ed. Vasculargrowth factors and angiogenesis. Berlin: Springer-Verlag; 1999:31-40.

62. Carmeliet P, Moons L, Luttun A, Vincenti V, Compernolle V, De Mol M, Wu Y,Bono F, Devy L, Beck H, Scholz D, Acker T, DiPalma T, Dewerchin M, Noel A,Stalmans I, Barra A, Blacher S, Vandendriessche T, Ponten A, Eriksson U, PlateKH, Foidart JM, Schaper W, Charnock-Jones DS, Hicklin DJ, Herbert JM, CollenD, Persico MG. Synergism between vascular endothelial growth factor andplacental growth factor contributes to angiogenesis and plasma extravasation inpathological conditions. Nat. Med. 2001;7:575-583.

63. Luttun A, Tjwa M, Moons L, Wu Y, Angelillo-Scherrer A, Liao F, Nagy JA,Hooper A, Priller J, De Klerck B, Compernolle V, Daci E, Bohlen P, Dewerchin M,Herbert JM, Fava R, Matthys P, Carmeliet G, Collen D, Dvorak HF, Hicklin DJ,Carmeliet P. Revascularization of ischemic tissues by PlGF treatment, andinhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1. Nat.Med. 2002;8:831-840.

64. Odorisio T, Schietroma C, Zaccaria ML, Cianfarani F, Tiveron C, Tatangelo L,Failla CM, Zambruno G. Mice overexpressing placenta growth factor exhibitincreased vascularization and vessel permeability. J. Cell Sci. 2002;115:2559-2567.

65. Oura H, Bertoncini J, Velasco P, Brown LF, Carmeliet P, Detmar M. A critical roleof placental growth factor in the induction of inflammation and edema formation.Blood. 2003;101:560-567.

66. Pipp F, Heil M, Issbrucker K, Ziegelhoeffer T, Martin S, van den Heuvel J, WeichH, Fernandez B, Golomb G, Carmeliet P, Schaper W, Clauss M. VEGFR-1-selective VEGF homologue PlGF is arteriogenic: evidence for a monocyte-mediatedmechanism. Circ. Res. 2003;92:378-385.

67. Autiero M, Waltenberger J, Communi D, Kranz A, Moons L, Lambrechts D, KrollJ, Plaisance S, De Mol M, Bono F, Kliche S, Fellbrich G, Ballmer-Hofer K,

Page 48: Vascular Endothelial Growth Factors B and C: Gene

48

Maglione D, Mayr-Beyrle U, Dewerchin M, Dombrowski S, Stanimirovic D, VanHummelen P, Dehio C, Hicklin DJ, Persico G, Herbert JM, Shibuya M, Collen D,Conway EM, Carmeliet P. Role of PlGF in the intra- and intermolecular cross talkbetween the VEGF receptors Flt1 and Flk1. Nat. Med. 2003;9:936-43.

68. Park JE, Chen HH, Winer J, Houck KA, Ferrara N. Placenta growth factor.Potentiation of vascular endothelial growth factor bioactivity, in vitro and in vivo,and high affinity binding to Flt-1 but not to Flk-1/KDR. J. Biol. Chem.1994;269:25646-54.

69. Lyttle DJ, Fraser KM, Fleming SB, Mercer AA, Robinson AJ. Homologs ofvascular endothelial growth factor are encoded by the poxvirus orf virus. J. Virol.1994;68:84-92.

70. Ueda N, Wise LM, Stacker SA, Fleming SB, Mercer AA. Pseudocowpox virusencodes a homolog of vascular endothelial growth factor. Virology. 2003;305:298-309.

71. Wise LM, Veikkola T, Mercer AA, Savory LJ, Fleming SB, Caesar C, Vitali A,Makinen T, Alitalo K, Stacker S. The vascular endothelial growth factor (VEGF)-like protein from orf virus NZ2 binds to VEGFR2 and neuropilin-1. Proc. Natl.Acad. Sci. USA. 1999;96:3071-3076.

72. Meyer M, Clauss M, Lepple-Wienhues A, Waltenberger J, Augustin HG, Ziche M,Lanz C, Böttner M, Rziha H-J, Dehio C. A novel vascular endothelial growth factorencoded by Orf virus, VEGF-E, mediates angiogenesis via signaling throughVEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine kinases. EMBO J.1999;18:363-374.

73. Ogawa S, Oku A, Sawano A, Yamaguchi S, Yazaki Y, Shibuya M. A novel type ofvascular endothelial growth factor, VEGF-E (NZ-7 VEGF), preferentially utilizesKDR/Flk-1 receptor and carries a potent mitotic activity without heparin-bindingdomain. J. Biol. Chem. 1998;273:31273-31282.

74. Dvorak HF. Vascular permeability factor/vascular endothelial growth factor: acritical cytokine in tumor angiogenesis and a potential target for diagnosis andtherapy. J. Clin. Oncol. 2002;20:4368-80.

75. Jussila L, Alitalo K. Vascular growth factors and lymphangiogenesis. Physiol. Rev.2002;82:673-700.

76. Baldwin ME, Stacker SA, Achen MG. Molecular control of lymphangiogenesis.Bioessays. 2002;24:1030-1040.

77. Kiba A, Sagara H, Hara T, Shibuya M. VEGFR-2-specific ligand VEGF-E inducesnon-edematous hyper-vascularization in mice. Biochem. Biophys. Res. Commun.2003;301:371-377.

78. Landgren E, Schiller P, Cao Y, Claesson-Welsh L. Placenta growth factorstimulates MAP kinase and mitogenicity but not phospholipase C- gamma andmigration of endothelial cells expressing Flt 1. Oncogene. 1998;16:359-367.

79. Huang K, Andersson C, Roomans GM, Ito N, Claesson-Welsh L. Signalingproperties of VEGF receptor-1 and -2 homo- and heterodimers. Int. J. Biochem. CellBiol. 2001;33:315-324.

80. Kaipainen A, Korhonen J, Pajusola K, Aprelikova O, Persico MG, Terman BI,Alitalo K. The related FLT4, FLT1, and KDR receptor tyrosine kinases showdistinct expression patterns in human fetal endothelial cells. J. Exp. Med.1993;178:2077-2088.

81. Peters KG, De Vries C, Williams LT. Vascular endothelial growth factor receptorexpression during embryogenesis and tissue repair suggests a role in endothelialdifferentiation and blood vessel growth. Proc. Natl. Acad. Sci. USA. 1993;90:8915-8919.

82. Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marme D. Migration ofhuman monocytes in response to vascular endothelial growth factor (VEGF) ismediated via the VEGF receptor flt-1. Blood. 1996;87:3336-3343.

83. Mahasreshti PJ, Navarro JG, Kataram M, Wang MH, Carey D, Siegal GP, BarnesMN, Nettelbeck DM, Alvarez RD, Hemminki A, Curiel DT. Adenovirus-mediated

Page 49: Vascular Endothelial Growth Factors B and C: Gene

49

soluble FLT-1 gene therapy for ovarian carcinoma. Clinical Cancer Res.2001;7:2057-66.

84. Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, Schisterman EF,Thadhani R, Sachs BP, Epstein FH, Sibai BM, Sukhatme VP, Karumanchi SA.Circulating angiogenic factors and the risk of preeclampsia. NEJM. 2004;350:672-83.

85. Gille H, Kowalski J, Li B, LeCouter J, Moffat B, Zioncheck TF, Pelletier N, FerraraN. Analysis of biological effects and signaling properties of Flt-1 (VEGFR-1) andKDR (VEGFR-2). A reassessment using novel receptor-specific vascularendothelial growth factor mutants. J. Biol. Chem. 2001;276:3222-3230.

86. Partanen TA, Alitalo K, Miettinen M. Lack of Lymphatic Vascular Specificity ofVascular Endothelial Growth Factor Receptor 3 in 185 vascular Tumors. Cancer.1999;86:2406-2412.

87. Shima D, Deutsch U, D`Amore P. Hypoxic induction of vascular endothelialgrowth factor(VEGF) in human epithelial cells is mediated by increases in mRNAstability. FEBS lett. 1995;370:203-208.

88. Stacker SA, Caesar C, Baldwin ME, Thornton GE, Williams RA, Prevo R, JacksonDG, Nishikawa S, Kubo H, Achen MG. VEGF-D promotes the metastatic spread oftumor cells via the lymphatics. Nat. Med. 2001;7:186-91.

89. Zhang W, Ran S, Sambade M, Huang X, Thorpe PE. A monoclonal antibody thatblocks VEGF binding to VEGFR2 (KDR/Flk-1) inhibits vascular expression of Flk-1 and tumor growth in an orthotopic human breast cancer model. Angiogenesis.2002;5:35-44.

90. Kolodkin AL, Levengood DV, Rowe EG, Tai YT, Giger RJ, Ginty DD. Neuropilinis a semaphorin III receptor. Cell. 1997;90:753-62.

91. Chen H, Chedotal A, He Z, Goodman CS, Tessier-Lavigne M. Neuropilin-2, anovel member of the neuropilin family, is a high affinity receptor for thesemaphorins Sema E and Sema IV but not Sema III. Neuron. 1997;19:547-59.

92. Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M. Neuropilin-1 isexpressed by endothelial and tumor cells as an isoform-specific receptor forvascular endothelial growth factor. Cell. 1998;92:735-745.

93. Karkkainen MJ, Saaristo A, Jussila L, Karila KA, Lawrence EC, Pajusola K, BuelerH, Eichmann A, Kauppinen R, Kettunen MI, Yla-Herttuala S, Finegold DN, FerrellRE, Alitalo K. A model for gene therapy of human hereditary lymphedema. Proc.Natl. Acad. Sci. USA. 2001;98:12677-82.

94. Gluzman-Poltorak Z, Cohen T, Herzog Y, Neufeld G. Neuropilin-2 is a receptor forthe vascular endothelial growth factor (VEGF) forms VEGF-145 and VEGF-165. J.Biol. Chem. 2000;275:18040-5.

95. Yuan L, Moyon D, Pardanaud L, Breant C, Karkkainen MJ, Alitalo K, Eichmann A.Abnormal lymphatic vessel development in neuropilin 2 mutant mice. Development.2002;129:4797-4806.

96. Takashima S, Kitakaze M, Asakura M, Asanuma H, Sanada S, Tashiro F, Niwa H,Miyazaki Ji J, Hirota S, Kitamura Y, Kitsukawa T, Fujisawa H, Klagsbrun M, HoriM. Targeting of both mouse neuropilin-1 and neuropilin-2 genes severely impairsdevelopmental yolk sac and embryonic angiogenesis. Proc. Natl. Acad. Sci. USA.2002;99:3657-3662.

97. Gu C, Rodriguez ER, Reimert DV, Shu T, Fritzsch B, Richards LJ, Kolodkin AL,Ginty DD. Neuropilin-1 conveys semaphorin and VEGF signaling during neuraland cardiovascular development. Dev. Cell. 2003;5:45-57.

98. Giger RJ, Cloutier JF, Sahay A, Prinjha RK, Levengood DV, Moore SE, PickeringS, Simmons D, Rastan S, Walsh FS, Kolodkin AL, Ginty DD, Geppert M.Neuropilin-2 is required in vivo for selective axon guidance responses to secretedsemaphorins. Neuron. 2000;25:29-41.

99. Lawson ND, Scheer N, Pham VN, Kim CH, Chitnis AB, Campos-Ortega JA,Weinstein BM. Notch signaling is required for arterial-venous differentiation duringembryonic vascular development. Development. 2001;128:3675-83.

Page 50: Vascular Endothelial Growth Factors B and C: Gene

50

100. Gerety SS, Wang HU, Chen ZF, Anderson DJ. Symmetrical mutant phenotypes ofthe receptor EphB4 and its specific transmembrane ligand ephrin-B2 incardiovascular development. Mol. Cell. 1999;4:403-14.

101. Wang HU, Chen ZF, Anderson DJ. Molecular distinction and angiogenic interactionbetween embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell. 1998;93:741-753.

102. Adams RH, Diella F, Hennig S, Helmbacher F, Deutsch U, Klein R. Thecytoplasmic domain of the ligand ephrinB2 is required for vascular morphogenesisbut not cranial neural crest migration. Cell. 2001;104:57-69.

103. Heldin CH, Eriksson U, Ostman A. New members of the platelet-derived growthfactor family of mitogens. Arch. Biochem. Biophys. 2002;398:284-290.

104. Lindahl P, Johansson BR, Leveen P, Betsholtz C. Pericyte loss and microaneurysmformation in PDGF-B-deficient mice. Science. 1997;277:242-5.

105. Leveen P, Pekny M, Gebre-Medhin S, Swolin B, Larsson E, Betsholtz C. Micedeficient for PDGF B show renal, cardiovascular, and hematological abnormalities.Genes Dev. 1994;8:1875-1887.

106. Lindahl P, Hellstrom M, Kalen M, Betsholtz C. Endothelial-perivascular cellsignaling in vascular development: lessons from knockout mice. Curr. Opin.Lipidol. 1998;9:407-11.

107. Bostrom H, Gritli-Linde A, Betsholtz C. PDGF-A/PDGF alpha-receptor signaling isrequired for lung growth and the formation of alveoli but not for early lungbranching morphogenesis. Dev. Dyn. 2002;223:155-62.

108. Karlsson L, Lindahl P, Heath JK, Betsholtz C. Abnormal gastrointestinaldevelopment in PDGF-A and PDGFR-(alpha) deficient mice implicates a novelmesenchymal structure with putative instructive properties in villus morphogenesis.Development. 2000;127:3457-66.

109. Gnessi L, Basciani S, Mariani S, Arizzi M, Spera G, Wang C, Bondjers C, KarlssonL, Betsholtz C. Leydig cell loss and spermatogenic arrest in platelet-derived growthfactor (PDGF)-A-deficient mice. JCB. 2000;149:1019-26.

110. Fruttiger M, Karlsson L, Hall AC, Abramsson A, Calver AR, Bostrom H, WillettsK, Bertold CH, Heath JK, Betsholtz C, Richardson WD. Defective oligodendrocytedevelopment and severe hypomyelination in PDGF-A knockout mice. Development.1999;126:457-67.

111. Cohen MA, Eaglstein WH. Recombinant human platelet-derived growth factor gelspeeds healing of acute full-thickness punch biopsy wounds. J. Am. Acad.Dermatol. 2001;45:857-62.

112. Yu J, Ustach C, Kim HR. Platelet-derived growth factor signaling and humancancer. J. Biochem. Mol. Biol. 2003;36:49-59.

113. Jones N, Iljin K, Dumont DJ, Alitalo K. Tie receptors: new modulators ofangiogenic and lymphangiogenic responses. Nat. Rev. Mol. Cell Biol. 2001;2:257-267.

114. Puri MC, Rossant J, Alitalo K, Bernstein A, Partanen J. The receptor tyrosinekinase TIE is required for integrity and survival of vascular endothelial cells. EMBOJ. 1995;14:5884-5891.

115. Dumont DJ, Gradwohl G, Fong GH, Puri MC, Gertsenstein M, Auerbach A,Breitman ML. Dominant-negative and targeted null mutations in the endothelialreceptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo.Genes Dev. 1994;8:1897-909.

116. Sato TN, Tozawa Y, Deutsch U, Wolburg-Buchholz K, Fujiwara Y, Gendron-Maguire M, Gridley T, Wolburg H, Risau W, Qin Y. Distinct roles of the receptortyrosine kinases Tie-1 and Tie-2 in blood vessel formation. Nature. 1995;376:70-74.

117. Dumont DJ, Fong G-H, Puri M, Gradwohl G, Alitalo K, Breitman ML.Vascularization of the mouse embryo: a study of flk-1, tek, tie and VEGF expressionduring development. Mech. Dev. 1995; 203: 80-92.

118. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C,Compton D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato TN,

Page 51: Vascular Endothelial Growth Factors B and C: Gene

51

Yancopoulos GD. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivoangiogenesis. Science. 1997;277:55-60.

119. Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, Sato TN,Yancopoulos GD. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor,during embryonic angiogenesis. Cell. 1996;87:1171-80.

120. Thurston G, Suri C, Smith K, McClain J, Sato TN, Yancopoulos GD, McDonaldDM. Leakage-resistant blood vessels in mice transgenically overexpressingangiopoietin-1. Science. 1999;286:2511-2514.

121. Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature. 2000;407:242-248.

122. Gale NW, Thurston G, Hackett SF, Renard R, Wang Q, McClain J, Martin C, WitteC, Witte MH, Jackson D, Suri C, Campochiaro PA, Wiegand SJ, Yancopoulos GD.Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, andonly the latter role is rescued by Angiopoietin-1. Dev. Cell. 2002;3:411-423.

123. Newman PJ. The biology of PECAM-1. J. Clin. Invest. 1997;100:S25-9.124. Jackson JR, Seed MP, Kircher CH, Willoughby DA, Winkler JD. The

codependance of angiogenesis and chronic inflammation. FASEB. 1997;11:457-465.

125. Ferrero E, Ferrero ME, Pardi R, Zocchi MR. The platelet endothelial cell adhesionmolecule-1 (PECAM1) contributes to endothelial barrier function. FEBS Letters.1995;374:323-6.

126. Schimmenti LA, Yan HC, Madri JA, Albelda SM. Platelet endothelial cell adhesionmolecule, PECAM-1, modulates cell migration. J. Cell. Physiol. 1992;153:417-28.

127. DeLisser HM, Christofidou-Solomidou M, Strieter RM, Burdick MD, Robinson CS,Wexler RS, Kerr JS, Garlanda C, Merwin JR, Madri JA, Albelda SM. Involvementof endothelial PECAM-1/CD31 in angiogenesis. Am. J. Pat. 1997;151:671-7.

128. Muller WA, Weigl SA, Deng X, Phillips DM. PECAM-1 is required fortransendothelial migration of leukocytes. J. Exp. Med. 1993;178:449-60.

129. Paavonen K, Puolakkainen P, Jussila L, Jahkola T, Alitalo K. Vascular endothelialgrowth factor receptor-3 in lymphangiogenesis in wound healing. Am. J. Pathol.2000;156:1499-504.

130. Yamamoto K, de Waard V, Fearns C, Loskutoff DJ. Tissue distribution andregulation of murine von Willebrand factor gene expression in vivo. Blood.1998;92:2791-801.

131. Breier G, Breviario F, Caveda L, Berthier R, Schnurch H, Gotsch U, Vestweber D,Risau W, Dejana E. Molecular cloning and expression of murine vascularendothelial-cadherin in early stage development of cardiovascular system. Blood.1996;87:630-41.

132. Carmeliet P, Lampugnani MG, Moons L, Breviario F, Compernolle V, Bono F,Balconi G, Spagnuolo R, Oostuyse B, Dewerchin M, Zanetti A, Angellilo A, MattotV, Nuyens D, Lutgens E, Clotman F, de Ruiter MC, Gittenberger-de Groot A,Poelmann R, Lupu F, Herbert JM, Collen D, Dejana E. Targeted deficiency orcytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediatedendothelial survival and angiogenesis. Cell. 1999;98:147-57.

133. Caveda L, Martin-Padura I, Navarro P, Breviario F, Corada M, Gulino D,Lampugnani MG, Dejana E. Inhibition of cultured cell growth by vascularendothelial cadherin (cadherin-5/VE-cadherin). J. Clin. Invest. 1996;98:886-93.

134. Rahimi N, Kazlauskas A. A role for cadherin-5 in regulation of vascular endothelialgrowth factor receptor 2 activity in endothelial cells. Mol. Biol. Cell. 1999;10:3401-7.

135. Sundberg C, Kowanetz M, Brown LF, Detmar M, Dvorak HF. Stable expression ofangiopoietin-1 and other markers by cultured pericytes: phenotypic similarities to asubpopulation of cells in maturing vessels during later stages of angiogenesis invivo. Lab. Invest. 2002;82:387-401.

136. Schlingemann RO, Rietveld FJ, Kwaspen F, van de Kerkhof PC, de Waal RM,Ruiter DJ. Differential expression of markers for endothelial cells, pericytes, and

Page 52: Vascular Endothelial Growth Factors B and C: Gene

52

basal lamina in the microvasculature of tumors and granulation tissue. Am. J. Pat.1991;138:1335-47.

137. Verbeek MM, Otte-Holler I, Wesseling P, Ruiter DJ, de Waal RM. Induction ofalpha-smooth muscle actin expression in cultured human brain pericytes bytransforming growth factor-beta 1. Am. J. Pat. 1994;144:372-82.

138. Nehls V, Drenckhahn D. The versatility of microvascular pericytes: frommesenchyme to smooth muscle? Histochemistry. 1993;99:1-12.

139. Sundberg C, Ljungstrom M, Lindmark G, Gerdin B, Rubin K. Microvascularpericytes express platelet-derived growth factor-beta receptors in human healingwounds and colorectal adenocarcinoma. Am. J. Pat. 1993;143:1377-88.

140. Schlingemann RO, Rietveld FJ, de Waal RM, Ferrone S, Ruiter DJ. Expression ofthe high molecular weight melanoma-associated antigen by pericytes duringangiogenesis in tumors and in healing wounds. Am. J. Pat. 1990;136:1393-405.

141. Ozerdem U, Grako KA, Dahlin-Huppe K, Monosov E, Stallcup WB. NG2proteoglycan is expressed exclusively by mural cells during vascularmorphogenesis. Dev. Dyn. 2001;222:218-27.

142. Ozerdem U, Monosov E, Stallcup WB. NG2 proteoglycan expression by pericytesin pathological microvasculature. Microvasc. Res. 2002;63:129-34.

143. Li Z, Colucci-Guyon E, Pincon-Raymond M, Mericskay M, Pournin S, Paulin D,Babinet C. Cardiovascular lesions and skeletal myopathy in mice lacking desmin.Dev. Biol. 1996;175:362-6.

144. Milner DJ, Weitzer G, Tran D, Bradley A, Capetanaki Y. Disruption of musclearchitecture and myocardial degeneration in mice lacking desmin. JCB.1996;134:1255-70.

145. Oosthuyse B, Moons L, Storkebaum E, Beck H, Nuyens D, Brusselmans K, VanDorpe J, Hellings P, Gorselink M, Heymans S, Theilmeier G, Dewerchin M,Laudenbach V, Vermylen P, Raat H, Acker T, Vleminckx V, Van Den Bosch L,Cashman N, Fujisawa H, Drost MR, Sciot R, Bruyninckx F, Hicklin DJ, Ince C,Gressens P, Lupu F, Plate KH, Robberecht W, Herbert JM, Collen D, Carmeliet P.Deletion of the hypoxia-response element in the vascular endothelial growth factorpromoter causes motor neuron degeneration. Nat. Genet. 2001;28:131-138.

146. Alon T, Hemo I, Itin A, Pe'er J, Stone J, Keshet E. Vascular endothelial growthfactor acts as a survival factor for newly formed retinal vessels and has implicationsfor retinopathy of prematurity. Nat. Med. 1995;1:1024-1028.

147. Scholz D, Cai WJ, Schaper W. Arteriogenesis, a new concept of vascular adaptationin occlusive disease. Angiogenesis. 2001;4:247-57.

148. Arras M, Ito WI, Scholz D, Winkler B, Schaper J, Schaper W. Monocyte Activationand Angiogenesis and Collateral Growth in the Rabbit Hindlimb. J. Clin. Invest.1998;101:40-50.

149. van Royen N, Piek JJ, Buschmann I, Hoefer I, Voskuil M, Schaper W. Stimulationof arteriogenesis; a new concept for the treatment of arterial occlusive disease.Cardiovasc. Res. 2001;49:543-53.

150. Luttun A, Tjwa M, Carmeliet P. Placental Growth Fator (PlGF) and Its ReceptorFlt1 (VEGFR-1): novel therapeutic targets for angiogenic disorders. Ann. N. Y.Acad. Sci. 2002;979:80-93.

151. Cao R, Brakenhielm E, Pawliuk R, Wariaro D, Post MJ, Wahlberg E, Leboulch P,Cao Y. Angiogenic synergism, vascular stability and improvement of hind-limbischemia by a combination of PDGF-BB and FGF-2. Nat. Med. 2003;9:604-13.

152. Benjamin LE, Keshet E. Conditional switching of vascular endothelial growthfactor (VEGF) expression in tumors: induction of endothelial cell shedding andregression of hemangioblastoma-like vessels by VEGF withdrawal. Proc. Natl.Acad. Sci. USA. 1997;94:8761-8766.

153. Benjamin LE, Golijanin D, Itin A, Pode D, Keshet E. Selective ablation ofimmature blood vessels in established human tumors follows vascular endothelialgrowth factor withdrawal. J. Clin. Invest. 1999;103:159-65.

Page 53: Vascular Endothelial Growth Factors B and C: Gene

53

154. Boucher Y, Leunig M, Jain RK. Tumor angiogenesis and interstitial hypertension.Cancer Res. 1996;56:4264-6.

155. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;1:57-70.156. Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL,

Steinberg SM, Chen HX, Rosenberg SA. A randomized trial of bevacizumab, ananti-vascular endothelial growth factor antibody, for metastatic renal cancer. NEJM.2003;349:427-34.

157. Kabbinavar F, Hurwitz HI, Fehrenbacher L, Meropol NJ, Novotny WF, LiebermanG, Griffing S, Bergsland E. Phase II, randomized trial comparing bevacizumab plusfluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastaticcolorectal cancer.J. Clin. Oncol. 2003;21:60-5.

158. Wigle JT, Harvey N, Detmar M, Lagutina I, Grosveld G, Gunn MD, Jackson DG,Oliver G. An essential role for Prox1 in the induction of the lymphatic endothelialcell phenotype. EMBO J. 2002;21:1505-1513.

159. Karkkainen MJ, Haiko P, Sainio K, Partanen J, Taipale J, Petrova TV, Jeltsch M,Jackson DG, Talikka M, Rauvala H, Betsholtz C, Alitalo K. Vascular endothelialgrowth factor C is required for sprouting of the first lymphatic vessels fromembryonic veins. Nat. Immunol. 2004;5:74-80.

160. Oliver G, Harvey N. A stepwise model of the development of lymphaticvasculature. Ann. N. Y. Acad. Sci. 2002;979:159-165.

161. Aselli G. De Lacticus Sive Lacteis Venis; 1627.162. Jeltsch M, Kaipainen A, Joukov V, Meng X, Lakso M, Rauvala H, Swartz M,

Fukumura D, Jain RK, Alitalo K. Hyperplasia of lymphatic vessels in VEGF-Ctransgenic mice. Science. 1997;276:1423-1425.

163. Sabin FR. The lymphatic system in human embryos, with a consideration of themorphology of the system as a whole. Am. J. Anat. 1909;9:43-91.

164. Sabin FR. On the origin of the abdominal lymphatics in mammals from the venacava and renal veins. Anat. Rec. 1912;6:335-343.

165. Huntington GS, McClure CFW. The anatomy and development of the jugularlymph sac in the domestic cat (Felis domestica). Anat. Rec. 1908;2:1-19.

166. van der Jagt ER. The origin and development of the anterior lymph sacs in the seaturtle (Thalassochelys caretta). Q. J. Microbiol. Sci. 1932;75:151-165.

167. Dumont DJ, Jussila L, Taipale J, Mustonen T, Pajusola K, Breitman M, Alitalo K.Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science.1998;282:946-949.

168. Kaipainen A, Korhonen J, Mustonen T, van Hinsbergh VM, Fang G-H, Dumont D,Breitman M, Alitalo K. Expression of the fms-like tyrosine kinase FLT4 genebecomes restricted to endothelium of lymphatic vessels during development. Proc.Natl. Acad. Sci. USA. 1995;92:3566-3570.

169. Wigle JT, Chowdhury K, Gruss P, Oliver G. Prox1 function is crucial for mouselens-fibre elongation. Nat. Gen. 1999;21:318-322.

170. Wilting J, Papoutsi M, Othman-Hassan K, Rodriguez-Niedenfuhr M, Prols F,Tomarev SI, Eichmann A. Development of the avian lymphatic system. Microsc.Res. Techniq. 2001;55:81-91.

171. Papoutsi M, Tomarev SI, Eichmann A, Prols F, Christ B, Wilting J. Endogenousorigin of the lymphatics in the avian chorioallantoic membrane. Dev. Dyn.2001;222:238-51.

172. Achen MG, Jeltsch M, Kukk E, Makinen T, Vitali A, Wilks AF, Alitalo K, StackerSA. Vascular endothelial growth factor D (VEGF-D) is a ligand for the tyrosinekinases VEGF receptor 2 (Flk1) and VEGF receptor 3 (Flt4). Proc. Natl. Acad. Sci.USA. 1998;95:548-553.

173. Joukov V, Pajusola K, Kaipainen A, Chilov D, Lahtinen I, Kukk E, Saksela O,Kalikkinen N, Alitalo K. A novel vascular endothelial growth factor, VEGF-C, is aligand for the Flt4 (VEGFR-3) and Kdr (VEGFR-2) receptor tyrosine kinases.EMBO J. 1996;15:290-298.

Page 54: Vascular Endothelial Growth Factors B and C: Gene

54

174. Lee J, Gray A, Yuan J, Louth S-M, Avraham H, Wood W. Vascular endothelialgrowth factor-related protein: A ligand and specific activator of the tyrosine kinasereceptor Flt4. Proc. Natl. Acad. Sci. USA. 1996;93:1988-1992.

175. Yamada Y, Nezu J, Shimane M, Hirata Y. Molecular cloning of a novel vascularendothelial growth factor, VEGF-D. Genomics. 1997;42:483-488.

176. Rissanen TT, Markkanen JE, Gruchala M, Heikura T, Puranen A, Kettunen MI,Kholova I, Kauppinen RA, Achen MG, Stacker SA, Alitalo K, Yla-Herttuala S.VEGF-D Is the Strongest Angiogenic and Lymphangiogenic Effector AmongVEGFs Delivered Into Skeletal Muscle via Adenoviruses. Circ. Res. 2003;92:1098-1106.

177. Kukk E, Lymboussaki A, Taira S, Kaipainen A, Jeltsch M, Joukov V, Alitalo K.VEGF-C receptor binding and pattern of expression with VEGFR-3 suggests a rolein lymphatic vascular development. Development. 1996;122:3829-3837.

178. Lymboussaki A, Olofsson B, Eriksson U, Alitalo K. Vascular endothelial growthfactor (VEGF) and VEGF-C show overlapping binding sites in embryonicendothelia and distinct sites in differentiated adult endothelia. Circ. Res.1999;85:992-9.

179. Makinen T, Veikkola T, Mustjoki S, Karpanen T, Catimel B, Nice EC, Wise L,Mercer A, Kowalski H, Kerjaschki D, Stacker SA, Achen MG, Alitalo K. Isolatedlymphatic endothelial cells transduce growth, survival and migratory signals via theVEGF-C/D receptor VEGFR-3. EMBO J. 2001;20:4762-4773.

180. Saaristo A, Veikkola T, Enholm B, Hytonen M, Arola J, Pajusola K, Turunen P,Jeltsch M, Karkkainen MJ, Kerjaschki D, Bueler H, Yla-Herttuala S, Alitalo K.Adenoviral VEGF-C overexpression induces blood vessel enlargement, tortuosity,and leakiness but no sprouting angiogenesis in the skin or mucous membranes.FASEB J. 2002;16:1041-1049.

181. Stacker SA, Stenvers K, Caesar C, Vitali A, Domagala T, Nice E, Roufail S,Simpson RJ, Moritz R, Karpanen T, Alitalo K, Achen MG. Biosynthesis of vascularendothelial growth factor-D involves proteolytic processing which generates non-covalent homodimers. J. Biol. Chem. 1999;274:32127-36.

182. Joukov V, Sorsa T, Kumar V, Jeltsch M, Claesson-Welsh L, Cao Y, Saksela O,Kalkkinen N, Alitalo K. Proteolytic processing regulates receptor specificity andactivity of VEGF-C. EMBO J. 1997;16:3898-3911.

183. Veikkola T, Jussila L, Makinen T, Karpanen T, Jeltsch M, Petrova TV, Kubo H,Thurston G, McDonald D, Achen MG, Stacker SA, Alitalo K. Signalling viavascular endothelial growth factor receptor -3 is sufficient for lymphangiogenesis intransgenic mice. EMBO J. 2001;20:1223-1231.

184. Makinen T, Jussila L, Veikkola T, Karpanen T, Kettunen MI, Pulkkanen KJ,Kauppinen R, Jackson DG, Kubo H, Nishikawa S, Yla-Herttuala S, K A. Inhibitionof lymphangiogenesis with resulting lymphedema in transgenic mice expressingsoluble VEGF receptor-3. Nat. Med. 2001;7:199-205.

185. Baldwin ME, Catimel B, Nice EC, Roufail S, Hall NE, Stenvers KL, KarkkainenMJ, Alitalo K, Stacker SA, Achen MG. The specificity of receptor binding byvascular endothelial growth factor-d is different in mouse and man. J. Biol. Chem.2001;276:19166-19171.

186. Byzova TV, Goldman CK, Jankau J, Chen J, Cabrera G, Achen MG, Stacker SA,Carnevale KA, Siemionow M, Deitcher SR, DiCorleto PE. Adenovirus encodingvascular endothelial growth factor-D induces tissue-specific vascular patterns invivo. Blood. 2002;99:4434-42.

187. Pajusola K, Aprelikova O, Korhonen J, Kaipainen A, Pertovaara L, Alitalo R,Alitalo K. FLT4 receptor tyrosine kinase cantains seven immunoglobulin-like loopsand is expressed in multiple human tissues and cell lines. Cancer Res.1992;52:5738-5743.

188. Galland F, Karamysheva A, Mattei MG, Rosnet O, Marchetto S, Birnbaum D.Chromosomal localization of FLT4, a novel receptor-type tyrosine kinase gene.Genomics. 1992;13:475-8.

Page 55: Vascular Endothelial Growth Factors B and C: Gene

55

189. Hughes DC. Alternative splicing of the human VEGFGR-3/FLT4 gene as aconsequence of an integrated human endogenous retrovirus. J. Mol. Evol.2001;53:77-9.

190. Salven P, Mustjoki S, Alitalo R, Alitalo K, Rafii S. VEGFR-3 and CD133 identify apopulation of CD34+ lymphatic/vascular endothelial precursor cells. Blood.2003;101:168-72.

191. Banerji S, Ni J, Wang SX, Clasper S, Su J, Tammi R, Jones M, Jackson DG.LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific receptorfor hyaluronan. JCB. 1999;144:789-801.

192. Prevo R, Banerji S, Ferguson DJ, Clasper S, Jackson DG. Mouse LYVE-1 is anendocytic receptor for hyaluronan in lymphatic endothelium. J. Biol. Chem.2001;276:19420-30.

193. Mouta Carreira C, Nasser SM, di Tomaso E, Padera TP, Boucher Y, Tomarev SI,Jain RK. LYVE-1 is not restricted to the lymph vessels: expression in normal liverblood sinusoids and down-regulation in human liver cancer and cirrhosis. CancerRes. 2001;61:8079-84.

194. Jackson KA, Majka SM, Wang H, Pocius J, Hartley CJ, Majesky MW, Entman ML,Michael LH, Hirschi KK, Goodell MA. Regeneration of ischemic cardiac muscleand vascular endothelium by adult stem cells. J. Clin. Invest. 2001;107:1395-402.

195. Weninger W, Partanen TA, Breiteneder-Geleff S, Mayer C, Kowalski H, MildnerM, Pammer J, Sturzl M, Kerjaschki D, Alitalo K, Tschachler E. Expression ofvascular endothelial growth factor receptor-3 and podoplanin suggests a lymphaticendothelial cell origin of Kaposi's sarcoma tumor cells. Lab. Invest. 1999;79:243-51.

196. Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E,Diem K, Weninger W, Tschachler E, Alitalo K, Kerjaschki D. Angiosarcomasexpress mixed endothelial phenotypes of blood and lymphatic capillaries:podoplanin as a specific marker for lymphatic endothelium. Am. J. Pat.1999;154:385-94.

197. Schacht V, Ramirez MI, Hong YK, Hirakawa S, Feng D, Harvey N, Williams M,Dvorak AM, Dvorak HF, Oliver G, Detmar M. T1alpha/podoplanin deficiencydisrupts normal lymphatic vasculature formation and causes lymphedema. EMBOJournal. 2003;22:3546-56.

198. Petrova TV, Makinen T, Makela TP, Saarela J, Virtanen I, Ferrell RE, FinegoldDN, Kerjaschki D, Yla-Herttuala S, Alitalo K. Lymphatic endothelialreprogramming of vascular endothelial cells by the Prox-1 homeobox transcriptionfactor. EMBO J. 2002;21:4593-4599.

199. Hong YK, Harvey N, Noh YH, Schacht V, Hirakawa S, Detmar M, Oliver G. Prox1is a master control gene in the program specifying lymphatic endothelial cell fate.Dev. Dyn. 2002;225:351-357.

200. Burke Z, Oliver G. Prox1 is an early specific marker for the developing liver andpancreas in the mammalian foregut endoderm. Mech. Dev. 2002;118:147-55.

201. Rodriguez-Niedenfuhr M, Papoutsi M, Christ B, Nicolaides KH, von KaisenbergCS, Tomarev SI, Wilting J. Prox1 is a marker of ectodermal placodes, endodermalcompartments, lymphatic endothelium and lymphangioblasts. Anatomy &Embryology. 2001;204:399-406.

202. Skobe M, Hawighorst T, Jackson DG, Prevo R, Janes L, Velasco P, Riccardi L,Alitalo K, Claffey K, Detmar M. Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat. Med. 2001;7:192-198.

203. Karpanen T, Alitalo K. Lymphatic vessels as targets of tumor therapy? J. Exp. Med.2001;194:F37-42.

204. Mandriota SJ, Jussila L, Jeltsch M, Compagni A, Baetens D, Prevo R, Banerji S,Huarte J, Montesano R, Jackson DG, Orci L, Alitalo K, Christofori G, Pepper MS.Vascular endothelial growth factor-C-mediated lymphangiogenesis promotestumour metastasis. EMBO J. 2001;20:672-682.

Page 56: Vascular Endothelial Growth Factors B and C: Gene

56

205. He Y, Kozaki K, Karpanen T, Koshikawa K, Yla-Herttuala S, Takahashi T, AlitaloK. Suppression of tumor lymphangiogenesis and lymph node metastasis byblocking vascular endothelial growth factor receptor 3 signaling. J. Natl. CancerInst. 2002;94:819-825.

206. Reis-Filho JS, Schmitt FC. Lymphangiogenesis in tumors: what do we know?Microscopy Research & Technique. 2003;60:171-80.

207. Padera TP, Kadambi A, di Tomaso E, Carreira CM, Brown EB, Boucher Y, ChoiNC, Mathisen D, Wain J, Mark EJ, Munn LL, Jain RK. Lymphatic metastasis in theabsence of functional intratumor lymphatics. Science. 2002;296:1883-6.

208. Achen MG, Williams RA, Baldwin ME, Lai P, Roufail S, Alitalo K, Stacker SA.The angiogenic and lymphangiogenic factor vascular endothelial growth factor-Dexhibits a paracrine mode of action in cancer. Growth Factors. 2002;20:99-107.

209. Duff SE, Li C, Jeziorska M, Kumar S, Saunders MP, Sherlock D, O'Dwyer ST,Jayson GC. Vascular endothelial growth factors C and D and lymphangiogenesis ingastrointestinal tract malignancy. Br. J. Cancer. 2003;89:426-30.

210. Karkkainen MJ, Alitalo K. Lymphatic endothelial regulation, lymphoedema, andlymph node metastasis. Seminars Cell Dev. Biol. 2002;13:9-18.

211. Dadras SS, Paul T, Bertoncini J, Brown LF, Muzikansky A, Jackson DG, EllwangerU, Garbe C, Mihm MC, Detmar M. Tumor lymphangiogenesis: a novel prognosticindicator for cutaneous melanoma metastasis and survival. Am. J. Pat.2003;162:1951-60.

212. Straume O, Jackson DG, Akslen LA. Independent prognostic impact of lymphaticvessel density and presence of low-grade lymphangiogenesis in cutaneousmelanoma. Clinical Cancer Res. 2003;9:250-6.

213. Maula SM, Luukkaa M, Grenman R, Jackson D, Jalkanen S, Ristamaki R.Intratumoral lymphatics are essential for the metastatic spread and prognosis insquamous cell carcinomas of the head and neck region. Cancer Res. 2003;63:1920-1926.

214. Beasley NJ, Prevo R, Banerji S, Leek RD, Moore J, van Trappen P, Cox G, HarrisAL, Jackson DG. Intratumoral lymphangiogenesis and lymph node metastasis inhead and neck cancer. Cancer Res. 2002;62:1315-20.

215. Witte L, Hicklin DJ, Zhu Z, Pytowski B, Kotanides H, Rockwell P, Bohlen P.Monoclonal antibodies targeting the VEGF receptor-2 (Flk1/KDR) as an anti-angiogenic therapeutic strategy. Cancer Metastasis Rev. 1998;17:155-61.

216. Karkkainen MJ, Ferrell RE, Lawrence EC, Kimak MA, Levinson KL, McTigueMA, Alitalo K, Finegold DN. Missense mutations interfere with VEGFR-3signalling in primary lymphoedema. Nat. Genet. 2000;25:153-159.

217. Fang J, Dagenais SL, Erickson RP, Arlt MF, Glynn MW, Gorski JL, Seaver LH,Glover TW. Mutations in FOXC2 (MFH-1), a forkhead family transcription factor,are responsible for the hereditary lymphedema-distichiasis syndrome. Am. J. Hum.Genet. 2000;67:1382-8.

218. Miura N, Wanaka A, Tohyama M, Tanaka K. MFH-1, a new member of the forkhead domain family, is expressed in developing mesenchyme. FEBS Letters.1993;326:171-6.

219. Petrek JA, Heelan MC. Incidence of breast carcinoma-related lymphedema. Cancer.1998;83:2776-81.

220. Yla-Herttuala S, Alitalo K. Gene transfer as a tool to induce therapeutic vasculargrowth. Nat. Med. 2003;9:694-701.

221. Kootstra NA, Verma IM. Gene therapy with viral vectors. Annu. Rev. Pharmacol.Toxicol. 2003:413-39.

222. Bergelson JM, Cunningham JA, Droguett G, Kurt-Jones EA, Krithivas A, Hong JS,Horwitz MS, Crowell RL, Finberg RW. Isolation of a common receptor forCoxsackie B viruses and adenoviruses 2 and 5. Science. 1997;275:1320-3.

223. Wickham TJ, Mathias P, Cheresh DA, Nemerow GR. Integrins alpha v beta 3 andalpha v beta 5 promote adenovirus internalization but not virus attachment. Cell.1993;73:309-19.

Page 57: Vascular Endothelial Growth Factors B and C: Gene

57

224. VandenDriessche T, Collen D, Chuah MK. Gene therapy for the hemophilias.Journal of Thrombosis & Haemostasis. 2003;1:1550-8.

225. Karlsson S, Ooka A, Woods NB. Development of gene therapy for blood disordersby gene transfer into haematopoietic stem cells. Haemophilia. 2002;8:255-60.

226. Monahan PE, Samulski RJ. Adeno-associated virus vectors for gene therapy: morepros than cons? Mol. Med. Today. 2000;6:433-40.

227. Svensson EC, Marshall DJ, Woodard K, Lin H, Jiang F, Chu L, Leiden JM.Efficient and stable transduction of cardiomyocytes after intramyocardial injectionor intracoronary perfusion with recombinant adeno-associated virus vectors.Circulation. 1999;99:201-5.

228. Naldini L, Blomer U, Gallay P, Ory D, Mulligan R, Gage FH, Verma IM, Trono D.In vivo gene delivery and stable transduction of nondividing cells by a lentiviralvector. Science. 1996;272:263-7.

229. Marshall E. Gene therapy. Second child in French trial is found to have leukemia.Science. 2003;299:320.

230. Carmeliet P. VEGF gene therapy: stimulating angiogenesis or angioma-genesis?Nat. Med. 2001;6:1102-1103.

231. Freedman SB, Isner JM. Therapeutic angiogenesis for coronary artery disease. Ann.Intern. Med. 2002;136:54-71.

232. Lederman RJ, Mendelsohn FO, Anderson RD, Saucedo JF, Tenaglia AN, HermillerJB, Hillegass WB, Rocha-Singh K, Moon TE, Whitehouse MJ, Annex BH,Investigators T. Therapeutic angiogenesis with recombinant fibroblast growthfactor-2 for intermittent claudication (the TRAFFIC study): a randomised trial.Lancet. 2002;359:2053-8.

233. Rajagopalan S, Shah M, Luciano A, Crystal R, Nabel EG. Adenovirus-mediatedgene transfer of VEGF(121) improves lower-extremity endothelial function andflow reserve. Circulation. 2001;104:753-5.

234. Losordo DW, Vale PR, Symes JF, Dunnington CH, Esakof DD, Maysky M, AshareAB, Lathi K, Isner JM. Gene therapy for myocardial angiogenesis: initial clinicalresults with direct myocardial injection of phVEGF165 as sole therapy formyocardial ischemia. Circulation. 1998;98:2800-4.

235. Losordo DW, Kawamoto A. Biological revascularization and the interventionalmolecular cardiologist: bypass for the next generation.Circulation. 2002;106:3002-5.

236. Grines CL, Watkins MW, Helmer G, Penny W, Brinker J, Marmur JD, West A,Rade JJ, Marrott P, Hammond HK, Engler RL. Angiogenic Gene Therapy(AGENT) trial in patients with stable angina pectoris.Circulation. 2002;105:1291-7.

237. Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, GrunwaldF, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S, Zeiher AM,Transplantation of Progenitor C, Regeneration Enhancement in Acute Myocardial I.Transplantation of Progenitor Cells and Regeneration Enhancement in AcuteMyocardial Infarction (TOPCARE-AMI). Circulation. 2002;106:3009-17.

238. Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, Lau CP. Angiogenesis in ischaemicmyocardium by intramyocardial autologous bone marrow mononuclear cellimplantation. Lancet. 2003;361:47-9.

239. Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV, Kogler G,Wernet P. Repair of infarcted myocardium by autologous intracoronarymononuclear bone marrow cell transplantation in humans. Circulation.2002;106:1913-8.

240. Kamihata H, Matsubara H, Nishiue T, Fujiyama S, Amano K, Iba O, Imada T,Iwasaka T. Improvement of collateral perfusion and regional function byimplantation of peripheral blood mononuclear cells into ischemic hibernatingmyocardium. Arterioscler. Thromb. Vasc. Biol. 2002;22:1804-10.

241. Saaristo A, Karkkainen MJ, Alitalo K. Insights into the molecular pathogenesis andtargeted treatment of lymphedema. Ann. N. Y. Acad. Sci. 2002;979:94-110.

Page 58: Vascular Endothelial Growth Factors B and C: Gene

58

242. Karkkainen MJ, Jussila L, Ferrell RE, Finegold DN, Alitalo K. Molecularregulation of lymphangiogenesis and targets for tissue oedema. Trends Mol. Med.2001;7:18-22.

243. Karkkainen MJ, Saaristo A, Jussila L, Karila KA, Lawrence EC, Pajusola K, BuelerH, Eichmann A, Kauppinen R, Kettunen MI, Yla-Herttuala S, Finegold DN, FerrellRE, Alitalo K. A model for gene therapy of human hereditary lymphedema. Proc.Natl. Acad. Sci. USA. 2001;98:12677-12682.

244. Szuba A, Skobe M, Karkkainen MJ, Shin WS, Beynet DP, Rockson NB, Dakhil N,Spilman S, Goris ML, Strauss HW, Quertermous T, Alitalo K, Rockson SG.Therapeutic lymphangiogenesis with human recombinant VEGF-C. FASEBJournal. 2002;16:1985-7.

245. Yoon YS, Murayama T, Gravereaux E, Tkebuchava T, Silver M, Curry C, WeckerA, Kirchmair R, Hu CS, Kearney M, Ashare A, Jackson DG, Kubo H, Isner JM,Losordo DW. VEGF-C gene therapy augments postnatal lymphangiogenesis andameliorates secondary lymphedema. J. Clin. Invest. 2003;111:717-725.

246. Kampfer H, Pfeilschifter J, Frank S. Expressional regulation of angiopoietin-1 and -2 and the tie-1 and -2 receptor tyrosine kinases during cutaneous wound healing: acomparative study of normal and impaired repair. Lab. Invest. 2001;81:361-73.

247. Chilov D, Kukk E, Taira S, Jeltsch MM, Kaukonen J, Palotie A, Joukov V, AlitaloK. Genomic organization of human and mouse genes for vascular endothelialgrowth factor C. J. Biol. Chem. 1997;272:25176-83.

248. Dor Y, Porat R, Keshet E. Vascular endothelial growth factor and vascularadjustments to perturbations in oxygen homeostasis. Am. J. Physiol., Cell Physiol.2001;280:C1367-74.

249. Gleadle J, Ebert B, Firth J, Rathcliffe P. Regulation of angiogenic growth factorexpression by hypoxia, transition metals, and chelating agents. Am. J. Physiol.1995;268:C1362-8.

250. Levy AP, Levy NS, Goldberg MA. Post-transcriptional regulation of vascularendothelial growth factor by hypoxia. J. Biol. Chem. 1996;271:2746-2753.

251. Baeuerle PA, Baltimore D. NF-kappa B: ten years after. Cell. 1996;87:13-20.252. Ben-Av P, Crofford LJ, Wilder RL, Hla T. Induction of vascular endothelial growth

factor expression in synovial fibroblasts by prostaglandin E and interleukin-1: apotential mechanism for inflammatory angiogenesis. FEBS Letters. 1995;372:83-7.

253. Su JL, Shih JY, Yen ML, Jeng YM, Chang CC, Hsieh CY, Wei LH, Yang PC, KuoML. Cyclooxygenase-2 induces EP1- and HER-2/Neu-dependent vascularendothelial growth factor-C up-regulation: a novel mechanism oflymphangiogenesis in lung adenocarcinoma. Cancer Res. 2004;64:554-64.

254. Paavonen K, Mandelin J, Partanen T, Jussila L, Li T-F, Alitalo K, Konttinen Y.Vascular Endothelial Growth Factors -C and -D and their VEGFR-2 and -3receptors in blood and lymphatic vessels in healthy and rheumatoid synovium. J.Rheumatol. 2002;29.

255. Wauke K, Nagashima M, Ishiwata T, Asano G, Yoshino S. Expression andlocalization of vascular endothelial growth factor-C in rheumatoid arthritis synovialtissue. J. Rheumatol. 2002;29:34-8.

256. Kerjaschki D, Regele HM, Moosberger I, Nagy-Bojarski K, Watschinger B,Soleiman A, Birner P, Krieger S, Hovorka A, Silberhumer G, Laakkonen P, PetrovaT, Langer B, Raab I. Lymphatic neoangiogenesis in human kidney transplants isassociated with immunologically active lymphocytic infiltrates. J. Am. Soc.Nephrol. 15(3):603-12, 2004 March.

257. Cursiefen C, Chen L, Borges LP, Jackson D, Cao J, Radziejewski C, D'Amore PA,Dana MR, Wiegand SJ, Streilein JW. VEGF-A stimulates lymphangiogenesis andhemangiogenesis in inflammatory neovascularization via macrophage recruitment.J. Clin. Invest. 113(7):1040-50, 2004 April.

258. Rissanen TT, Markkanen JE, Gruchala M, Heikura T, Puranen A, Kettunen MI,Kholova I, Kauppinen RA, Achen MG, Stacker SA, Alitalo K, Yla-Herttuala S.

Page 59: Vascular Endothelial Growth Factors B and C: Gene

59

VEGF-D is the strongest angiogenic and lymphangiogenic effector among VEGFsdelivered into skeletal muscle via adenoviruses. Circ. Res. 2003;92:1098-106.

259. Karpanen T, Egeblad M, Karkkainen MJ, Kubo H, Yla-Herttuala S, Jaattela M,Alitalo K. Vascular endothelial growth factor C promotes tumor lymphangiogenesisand intralymphatic tumor growth. Cancer Res. 2001;61:1786-90.

260. Kadambi A, Mouta Carreira C, Yun CO, Padera TP, Dolmans DE, Carmeliet P,Fukumura D, Jain RK. Vascular endothelial growth factor (VEGF)-C differentiallyaffects tumor vascular function and leukocyte recruitment: role of VEGF-receptor 2and host VEGF-A. Cancer Res. 2001;61:2404-8.

261. Cao R, Eriksson A, Kubo H, Alitalo K, Cao Y, Thyberg J. Comparative evaluationof FGF-2-, VEGF-A-, and VEGF-C-induced angiogenesis, lymphangiogenesis,vascular fenestrations, and permeability. Circ. Res. 94(5):664-70, March 19. 2004.

262. Kubo H, Cao R, Brakenhielm E, Makinen T, Cao Y, Alitalo K. Blockade ofvascular endothelial growth factor receptor-3 signaling inhibits fibroblast growthfactor-2-induced lymphangiogenesis in mouse cornea. Proc. Natl. Acad. Sci. USA.2002;99:8868-73.

263. Oh SJ, Jeltsch MM, Birkenhager R, McCarthy JE, Weich HA, Christ B, Alitalo K.VEGF and VEGF-C: specific induction of angiogenesis and lymphangiogenesis inthe differentiated avian chorioallantoic membrane. Dev. Biol. 1997;188:96-109.

264. Cao Y, Linden P, Farnebo J, Cao R, Eriksson A, Kumar V, Qi JH, Claesson-WelshL, Alitalo K. Vascular endothelial growth factor C induces angiogenesis in vivo.Proc. Natl. Acad. Sci. USA. 1998;95:14389-14394.

265. Witzenbichler B, Asahara T, Murohara T, Silver M, Spyridopoulos I, Magner M,Principe N, Kearney M, Hu JS, Isner JM. Vascular endothelial growth factor-C(VEGF-C/VEGF-2) promotes angiogenesis in the setting of tissue ischemia. Am. J.Pathol. 1998;153:381-394.

266. Tille JC, Wang X, Lipson KE, McMahon G, Ferrara N, Zhu Z, Hicklin DJ, SleemanJP, Eriksson U, Alitalo K, Pepper MS. Vascular endothelial growth factor (VEGF)receptor-2 signaling mediates VEGF-C(deltaNdeltaC)- and VEGF-A-inducedangiogenesis in vitro. Exp. Cell Res. 2003;285:286-98.

267. McColl BK, Baldwin ME, Roufail S, Freeman C, Moritz RL, Simpson RJ, AlitaloK, Stacker SA, Achen MG. Plasmin activates the lymphangiogenic growth factorsVEGF-C and VEGF-D. J. Exp. Med. 2003;198:863-8.

268. Saaristo A, Veikkola T, Tammela T, Enholm B, Karkkainen MJ, Pajusola K, BuelerH, Yla-Herttuala S, Alitalo K. Lymphangiogenic gene therapy with minimal bloodvascular side effects. J. Exp. Med. 2002;196:719-730.

269. Baumgartner I, Rauh G, Pieczek A, Wuensch D, Magner M, Kearney M, SchainfeldR, Isner JM. Lower-extremity edema associated with gene transfer of naked DNAencoding vascular endothelial growth factor. Ann. Intern. Med. 2000;132:880-884.

270. Vajanto I, Rissanen TT, Rutanen J, Hiltunen MO, Tuomisto TT, Arve K, NarvanenO, Manninen H, Rasanen H, Hippelainen M, Alhava E, Yla-Herttuala S. Evaluationof angiogenesis and side effects in ischemic rabbit hindlimbs after intramuscularinjection of adenoviral vectors encoding VEGF and LacZ. J. Gene Med.2002;4:371-80.

271. Nagy JA, Vasile E, Feng D, Sundberg C, Brown LF, Detmar MJ, Lawitts JA,Benjamin L, Tan X, Manseau EJ, Dvorak AM, Dvorak HF. Vascular permeabilityfactor/vascular endothelial growth factor induces lymphangiogenesis as well asangiogenesis. J. Exp. Med. 2002;196:1497-506.

272. Kubo H, Cao R, Brakenhielm E, Makinen T, Cao Y, Alitalo K. Blockade ofvascular endothelial growth factor receptor-3 signaling inhibits fibroblast growthfactor-2-induced lymphangiogenesis in mouse cornea. Proc. Natl. Acad. Sci. USA.2002;99:8868-8873.

273. Luttun A, Tjwa M, Carmeliet P. Placental growth factor (PlGF) and its receptor Flt-1 (VEGFR-1): novel therapeutic targets for angiogenic disorders. J. Exp. Med.2002;196:1497-506.

Page 60: Vascular Endothelial Growth Factors B and C: Gene

60

274. Salven P, Lymboussaki A, Heikkila P, Jaaskela-Saari H, Enholm B, Aase K, vonEuler G, Eriksson U, Alitalo K, Joensuu H. Vascular endothelial growth factorsVEGF-B and VEGF-C are expressed in human tumors. Am. J. Pat. 1998;153:103-8.

275. P Oc, Rhys-Evans P, Eccles SA. Expression of vascular endothelial growth factorfamily members in head and neck squamous cell carcinoma correlates with lymphnode metastasis. Cancer. 2001;92:556-68.

276. Kawakami M, Furuhata T, Kimura Y, Yamaguchi K, Hata F, Sasaki K, Hirata K.Expression analysis of vascular endothelial growth factors and their relationships tolymph node metastasis in human colorectal cancer. Exp. Clin. Cancer Res.2003;22:229-37.

277. Mould AW, Tonks ID, Cahill MM, Pettit AR, Thomas R, Hayward NK, Kay GF.Vegfb gene knockout mice display reduced pathology and synovial angiogenesis inboth antigen-induced and collagen-induced models of arthritis. Arthritis Rheum.2003;48:2660-9.

278. Silvestre JS, Tamarat R, Ebrahimian TG, Le-Roux A, Clergue M, Emmanuel F,Duriez M, Schwartz B, Branellec D, Levy BI. Vascular endothelial growth factor-Bpromotes in vivo angiogenesis. Circ. Res. 2003;93:114-23.