• Non ci sono risultati.

Vascular Endothelial Growth Factor Signaling

N/A
N/A
Protected

Academic year: 2022

Condividi "Vascular Endothelial Growth Factor Signaling"

Copied!
14
0
0

Testo completo

(1)

Vascular Endothelial Growth Factor Signaling

David Semela, Jean-François Dufour

8

8.1

Introduction

Vascular endothelial growth factor (VEGF) is the main driving force for angiogenesis and vasculo- genesis. Identified as a vascular endothelial cell mitogen and survival factor, it has been sequenced and cloned by Ferrara and Connolly in 1989 [74, 84].

Intense research over the past years has deciphered the gene, molecular pathways, receptors and func- tions of this angiogenic factor [35]. VEGF is thought to play an important role in liver regeneration, he- patic fibrogenesis, portal hypertension, hepatocar- cinogenesis and malignant ascites formation.

8.2 VEGF

8.2.1

Biological Functions of VEGF

Vascular endothelial growth factor is the key ang- iogenic factor of developmental, physiological and pathological angiogenesis, which is the formation of new microvessels from a pre-existing vascular bed.

VEGF is a glycoprotein that can be produced and secreted by most cells in mammals. The main tar- gets of VEGF are vascular endothelial cells. VEGF has been shown to promote proliferation [34] and survival [9, 45, 46, 175] in vascular endothelial cells in vivo and in vitro. It acts as an endothelial survival factor by inducing expression of the anti-apoptotic proteins Bcl-2 and A1 in vascular endothelial cells [45]. Furthermore, VEGF induces the expression of proteases such as collagenase [156], matrix met- alloproteinases [178], and urokinase- and tissue- type plasminogen activators [120], which enable endothelial cells to break down the surrounding basal membrane and extracellular matrix in order to migrate and form new blood vessels. Initially de-

scribed as vascular permeability factor (VPF) [139], VEGF increases the permeability of blood vessels up to 50,000 times more than histamine [28]. Trans- mission and scanning electron microscopy studies showed that VEGF regulates hepatic sinusoidal per- meability by inducing fenestration in hepatic sinu- soidal endothelial cells possibly through caveolin-1 protein [170].

During embryonic development, liver organo- genesis and vasculogenesis, which is defined as the de novo formation of blood vessels from heman- gioblasts, are regulated through the VEGF signal- ing system [15, 33, 100]. The importance of VEGF signaling during embryonic development is high- lighted by the fact that lack of a single VEGF gene allele results in abnormal blood vessel development and embryonic lethality in mice [15, 33]. Knockout of the genes for the VEGF receptors VEGFR-1 or -2 also results in embryonic lethality [38, 140]. Kidney development, skeletal growth, enchondral bone for- mation, wound healing and ovarian angiogenesis are further fundamental physiological processes regulated by VEGF [36].

Recent evidence suggests that elevated levels of systemically circulating VEGF lead to recruitment of hematopoietic stem cells and endothelial pro- genitor cells from bone marrow to sites of neovas- cularization (reviewed in [127]). These cells home to sites of neovascularization such as tumor microvas- culature and the regenerating liver, where they are incorporated and contribute to angiogenesis [23, 41, 94, 129].

The level of circulating VEGF in healthy individ-

uals is low and depends on the method and sample

used for measurement (free vs. total VEGF, plasma

vs. serum) [64]. Significant amounts of VEGF are

bound to plasma proteins such as α2-microglobulin

and are stored in platelets, which secrete VEGF from

their α granules upon activation during blood clot-

ting [162]. The complex biology of VEGF, its release

from platelets and the different isoforms (see below)

have to be considered when choosing a VEGF detec-

tion test [7, 64, 162].

(2)

8.2.2

VEGF Gene and Splice Variants

The human VEGF gene is localized on chromosome 6p21.3 [159] and contains eight exons (Fig. 8.1) [155].

VEGF is highly conserved across species with a ho-

mology of approximately 85% between human and rat VEGF [35]. Alternative exon splicing of the hu- man VEGF pre-mRNA produces six VEGF isoforms containing 121, 145, 165, 183, 189 or 206 amino acids [155]. All VEGF isoforms are secreted as covalently linked homodimers but display differences in the basic amino rich domains encoded by exons 6 and 7 [155, 132]. These domains are important sites for molecular interaction and mediate binding of VEGF to heparin, heparan sulfate proteoglycans and to el- ements of the extracellular matrix, which results in sequestration of certain VEGF isoforms in the ex- tracellular matrix or at the cell surface: VEGF

121

, an isoform that lacks the domains encoded by exons 6 and 7, does not bind to heparin and is therefore freely diffusible after secretion [62]. In contrast, the high- ly basic isoforms VEGF

189

and VEGF

206

containing exons 6 and 7 remain almost entirely bound to the extracellular matrix and cell surface [62]. VEGF

165

(exon 6 absent), the most abundant isoform (46 kDa as homodimer), displays an intermediate behav- ior with 50%–70% sequestration [62]. Sequestered VEGF can be released by heparin, heparan sulfate, and heparinase [62] and proteases such as plasmin or urokinase-type plasminogen activator [122].

8.2.3

VEGF Protein Family

Vascular endothelial growth factor is member of a gene family of growth factors consisting of VEGF it- self (also named VEGFA), VEGFB, VEGFC, VEGFD, VEGFE and placenta growth factor (PlGF, also known as PGF, consisting of three isoforms PlGF-1, -2 and -3). They share significant sequence homol- ogy and are ligands to the same receptors as VEGF (see below and Fig. 8.2) [132]. PlGF expression is not only restricted to the placenta [96] but can be up- regulated in different cell populations such as en- dothelial, smooth muscle, inflammatory and malig- nant cells [16]. VEGFB is mainly expressed in heart and skeletal muscle [115]. VEGFC, also referred to as VEGF-related protein [68, 82], and VEGFD [116]

are involved in lymphangiogenesis [73] and fetal lung development [30]. Overexpression of VEGFC induces selective proliferation of lymphatic, but not vascular, endothelial cells and lymphatic vessel en- largement [65]. However, recent evidence suggests

that VEGFC might play a role in angiogenesis by inducing fenestrations in vascular endothelial cells and by inducing blood capillaries under certain circumstances [14]. VEGFE has been found in the genome of orf virus, which affects sheep, goats and humans [95]. It is believed that the gene has been originally acquired from a mammalian host [95].

Interestingly, skin lesions induced by the orf virus show extensive dermal vascular endothelial cell proliferation and vasodilatation [95].

Fig. 8.1. The six different splice variants of the VEGF (VEGFA) gene ordered by length of the amino acid sequence (* trunca- tion in exon 6, ** additional sequence encoded by exon 6)

Fig. 8.2. VEGF receptors (VEGFR)-1, -2, -3 and the neuropilin (NRP) receptors 1 and 2 with their function in VEGF signaling

(3)

8.2.4

VEGF Gene Expression

The regulatory factors for VEGF expression can be divided into two main categories: hypoxia through hypoxia-inducible factor (HIF)-1 and a group of cy- tokines, growth factors and transcription factors other than HIF-1. Additionally, mutations in tumor suppressor genes and oncogenes such as p53, ras,

raf, VHL, myc, c-fos and others can induce and up-

regulate VEGF expression [49, 67, 76, 91, 109].

Hypoxia

Hypoxia is a potent stimulator of VEGF expression.

HIF-1 is the key transcription factor in hypoxic tis- sues and induces the expression of several hypoxia- response genes such as VEGF and VEGFR-1 [44, 57].

The detailed molecular mechanisms of HIF-1 and the regulation of VEGF expression by HIF-1 are re- viewed in Chap. 26.

Hypoxia not only induces and upregulates the expression of VEGF but also stabilizes the intrinsi- cally labile VEGF mRNA, which contains destabiliz- ing elements in its 3’ and 5’ untranslated and coding regions [25, 85, 90, 144]. The hypoxia-induced RNA- binding protein HuR binds with high affinity and specificity to regulatory elements of VEGF mRNA and prevents its degradation [86].

Cytokines and Growth Factors

Many cytokines such as interleukin (IL)-1 α, IL-1β, IL-6, nitric oxide (NO) and growth factors such as fibroblast growth factor (FGF), transforming growth factor (TGF)- α, TGF-β, tumor necrosis fac- tor (TNF)- α, platelet-derived growth factor (PDGF), epidermal growth factor (EGF), hepatocyte growth factor (HGF) and insulin-like growth factor-1 up- regulate VEGF, highlighting the complex regulation and redundancy of the angiogenesis network [36, 67, 132].

8.3

VEGF Receptors

The effects of VEGF are mediated mainly through two cell surface receptor tyrosine kinases, namely VEGFR-1 and VEGFR-2. Neuropilin-1 (NRP1) and neuropilin-2 (NRP2) are recently discovered VEGF receptors belonging to the semaphorin sub-family.

VEGFR-3 is a receptor for VEGFC and VEGFD but not for VEGF. VEGFR-1, -2 and -3 belong to the flt

sub-family of receptor tyrosine kinases [132] and consist of seven extracellular immunoglobulin- like domains, one transmembrane region and a conserved tyrosine kinase domain intracellularly, which is interrupted by a kinase insert domain [102, 142, 153]. The functions of the different VEGF recep- tors and their ligands are summarized in Fig. 8.2.

8.3.1 VEGFR-1

Vascular endothelial growth factor receptor-1 (also known as fms-like-tyrosine kinase [Flt]-1 or FLT-1 in humans [24]) is a 180-kDa glycoprotein bind- ing VEGF, VEGFB and PlGF [132, 137]. In contrast to VEGFR-2, VEGFR-1 is upregulated by hypoxia through HIF-1 (see Chap. 26) [44]. It is highly ex- pressed on the surface of the different endothelial cell populations (quiescent and cycling) such as en- dothelial cells of liver sinusoids and hepatic arteri- oles [81, 134, 168], but also on hepatocytes during liver regeneration [107, 134] and on a few other cells such as hepatic stellate cells [2, 99], monocyte-mac- rophages including Kupffer cells [22, 136], pericytes [114], smooth muscle cells [21], nerve cells [147] and hematopoietic cells [36]. The exact role of VEGFR-1 is still under debate and depends on the biological situation (i.e. physiological vs. pathological angio- genesis). Although a knockout of the VEGFR-1 gene results in the formation of abnormal vascular chan- nels, excessive angioblast proliferation and embry- onic lethality by day E8.5 in mice [38, 39], VEGFR-1 lacking only the tyrosine kinase domain is sufficient for normal development and angiogenesis [59]. In contrast, VEGFR-1 tyrosine kinase-deficient mice showed impaired angiogenesis during carcinoma growth [60]. No proliferative or migratory response in endothelial cells or hepatocytes has been attrib- uted to VEGFR-1 [118, 134, 160]. Signaling through VEGFR-1 was shown to contribute to the regulation of endothelial cell permeability [148]. Activation of VEGFR-1 by the VEGF homolog PlGF enhances VEGF-driven angiogenesis through VEGFR-2 [6, 16] (see below). Further, VEGFR-1 is involved in the recruitment of endothelial progenitor cells [94]

and bone marrow-derived myeloid progenitors [93]

and promotes survival of hematopoietic stem cells [47]. Matrix metalloproteinase 9 (MMP-9) has been shown to be induced specifically via VEGFR-1 in endothelial cells of the lung promoting pulmonary metastasis [61].

Recent evidence suggests an angiogenesis-in-

dependent endothelial protection of hepatocytes

through VEGFR-1 [81]. Selective activation of liver

sinusoidal endothelial cells via VEGFR-1 induces

(4)

paracrine secretion of the potent mitogens HGF and IL-6 (see Chap. 7), which promote hepatocyte prolif- eration and reduce liver damage in mice exposed to the hepatotoxin CCl

4

.

Besides a membrane-bound form of VEGFR-1, a soluble form of VEGFR-1 (sVEGFR-1, sFlt-1), which is produced by endothelial cells and monocytes by alternative splicing, has been identified [8, 75].

sVEGFR-1 has been detected in human serum and plasma of normal male and female donors [8]. By binding VEGF, sVEGFR-1 is a naturally occurring VEGF antagonist. The function of sVEGFR-1 in physiologic angiogenesis and malignant neovascu- larization is unclear.

8.3.2 VEGFR-2

Vascular endothelial growth factor receptor-2 (also known as mouse fetal liver kinase [Flk]-1 or kinase domain region [KDR] in humans [154]) is a 200–230- kDa VEGF receptor, which shares 85% of sequence identity with VEGFR-1 [102] but binds VEGFC, VEGFD and VEGFE instead of VEGFB or PlGF [132].

In contrast to the non-mitogenic and non-moto- genic VEGFR-1, VEGFR-2 induces endothelial cell proliferation, migration and survival [34, 46, 126].

VEGFR-2 is involved in the mechanotransduction of blood flow shear stress to the vascular endothelium by nuclear translocation of VEGFR-2 and consecu- tive binding to the cytoskeleton together with VE- cadherin and β-catenin [141]. VEGFR-2 is highly expressed on adult and embryonic endothelial cells, embryonic angioblasts and hematopoietic stem cells [36, 132]. In resting liver, VEGFR-2 expression is limited to endothelial cells of the larger hepatic vessels [134], although earlier reports described VEGFR-2 mRNA expression also in liver sinusoi- dal endothelial cells [168]. During liver regenera- tion, VEGFR-2 expression predominantly increases on endothelial cells of large vessels (portal venules, arterioles, central venules) and to a lesser extent on sinusoidal endothelial cells often in close proximity to large vessels [134]. Additionally, hepatic stellate cells have been found to express VEGFR-2 in vitro [99]. Homozygous loss of VEGFR-2 results in lack of endothelial cells, impaired liver organogenesis [100] and embryonic lethality at day E9.5 to E10.5 [140], whereas heterozygous mice are normal [140].

VEGFR-2 is phosphorylated in resting liver, but has been shown to increase in activation during liver re- generation [134].

8.3.3 VEGFR-3

Vascular endothelial growth factor receptor-3 (or fms-like-tyrosine kinase [Flt]-4) is a receptor for VEGFC and VEGFD and does not bind VEGF [37, 117]. VEGFR-3 is mainly expressed on lymphatic endothelium and is involved in lymphangiogenesis [65, 70]. VEGFR-3 could not be detected on endothe- lial cells in resting or regenerating liver [134].

8.3.4

Neuropilin-1 and -2

Neuropilin-1 (NRP1) and NRP2 are additional VEGF receptors unrelated to VEGFR-1, -2 and -3 [146]. They are isoform-specific receptors binding VEGF

165

but not VEGF

121

and are involved in neu- ronal guidance [112]. Coexpression of VEGFR-2 and NRP1 enhances binding of VEGF to VEGFR-2 [113].

Neuropilins can form complexes with VEGFR-1 and probably with VEGFR-2 [113]. No intrinsic recep- tor signaling has been shown after binding of VEGF to NRP1 or NRP2 [113]. To date, nothing is known about the presence or function of NRP1 and NRP2 in liver biology.

8.3.5

VEGF Receptor Signaling

After binding, soluble VEGF dimers induce VEGF receptor dimerization leading to homo- or het- erodimers [6]. The juxtaposed cytoplasmic tyrosine kinase domains of the VEGFR molecules transphos- phorylate several tyrosine residues in the neighbor molecule [6, 40, 101, 137]. Activated receptors in turn activate proteins of different signaling pathways by phosphorylation (reviewed in [101] and Chap. 20 in this book): phospholipase C (PLC), phosphatidyli- nositol 3’-kinase (PI-3 kinase)/Akt, Ras GTPase-ac- tivating protein (GAP), Src family kinases and Raf [1, 29, 46, 53].

A recent study showed that although VEGF and

PlGF both bind VEGFR-1, they activate this recep-

tor differently, leading to a distinct gene expression

profile, where only PlGF was capable of switch-

ing on downstream target genes [6]. Many of these

PlGF-regulated genes have a role in cell cycle (Ets2,

Map4k4, Fst, Jak2, Egr1), angiogenesis (Flt-1, NRP2, Angptl4, Dcn) and apoptosis (Birc2) [6]. The same

authors were able to show that there is an intra- and

intermolecular crosstalk between VEGFR-1 and

(5)

VEGFR-2: activation of VEGFR-1 by PlGF resulted in intermolecular transphosphorylation of VEGFR- 2, thereby amplifying VEGF-driven angiogenesis through VEGFR-2 [6]. Further, VEGF/PlGF het- erodimers activated intramolecular VEGF receptor crosstalk through formation of VEGFR-1/VEGFR-2 heterodimers [6].

8.4

VEGF Signaling in Specific Liver Conditions

8.4.1

Liver Organogenesis

During embryonic development of the liver, hepatic cells are induced within the endoderm by day E8.5 of mouse gestation [51]. Interaction between these cells and surrounding endothelial cells or angiob- lasts induces outgrowth of the liver bud into the mesenchyme [100]. This stage is then followed by the formation of a de novo local vascular network (vasculogenesis) and the recruitment of hematopoi- etic cells [100]. Mice with homozygous deficiency in

VEGFR-2 lacking mature endothelial cells and blood

vessels show normal thickening of the hepatic endo- derm but lack liver bud emergence [100]. Vasculo- genic endothelial cells with intact signaling through VEGFR-2 are therefore critical already in the earli- est stages of liver organogenesis, even prior to blood vessel function [100]. Similar interactions between endothelial cells and hepatocytes are at play in adult liver tissue, i.e. during liver regeneration.

8.4.2

Liver Regeneration

Liver regeneration is angiogenesis-dependent [27];

inhibition of angiogenesis by anti-angiogenic sub- stances such as angiostatin or TNP-470 impairs liver regeneration [27, 48]. In the course of liver regeneration after hepatectomy, there is an initial wave of hepatocyte proliferation with formation of avascular hepatocellular islands [134]. This stage is followed by endothelial cell proliferation [135] and consecutive penetration of these avascular islands by endothelial cells with the formation of new sinu- soids [97]. Recent evidence suggests that endothelial progenitor cells are mobilized from bone marrow and participate in this neovascularization by com- mitting to sinusoidal endothelial cells [41]. HIF-1 α and VEGF as key regulators have been shown to be upregulated during this process [97, 128]: hepatocel-

lular production of VEGF peaks 48–72 hours after hepatectomy and is detected mainly in periportal hepatocytes [97, 151]. VEGF production is accompa- nied by an increase in the expression of VEGFR-1 on hepatocytes and of VEGFR-1 and VEGFR-2 on sinusoidal endothelial cells [107, 128, 134, 135]. Ad- ministration of VEGF in hepatectomized rodents increases hepatocyte and sinusoidal endothelial cell proliferation [5, 151], accelerates gain in liver mass [81] and improves functional hepatic recovery [128]. Neutralizing antibodies against VEGF inhibit hepatocyte and endothelial cell proliferation after partial hepatectomy [151]. LeCouter and coworkers showed that this effect is likely due to a VEGF-in- duced release of HGF by sinusoidal endothelial cells [81, 168]. Additional effects of VEGF on functional hepatic recovery could be a stimulatory effect on the formation of new blood vessels and/or a direct ef- fect on the hepatocytes, which express VEGF recep- tors after partial hepatectomy [128, 134]. Transduc- tion of VEGF before hepatic resection also hastens functional hepatic recovery in mice with fatty liver, which is known for its impaired regenerative capac- ity [128].

8.4.3

Liver Fibrosis and Cirrhosis

Accumulating evidence suggests that VEGF and its receptors are also involved in fibrogenesis and cirrhotic remodeling. The concept of an interplay between fibrosis and angiogenesis probably medi- ated through the stimulus of hypoxia is supported by two facts. First, the deposited matrix proteins – a hallmark of fibrosis – contain and sequester differ- ent angiogenic factors such as VEGF, which are lib- erated during remodeling of the connective tissue framework by proteolytic enzymes such as matrix metalloproteinases [71, 72]. Second, an abundant vascular network develops in the fibrotic tissue around regenerative nodules [56, 167], leading to a remodeled and abnormal hepatic microcirculation in cirrhotic liver [63, 158].

Experimental liver fibrogenesis after common

bile duct ligation in rats induces VEGF expression

in hepatocytes and angiogenesis [133]. The authors

of this study showed that this process is hypoxia-

driven and that the percentage of VEGF-expressing

hepatocytes increases from 3% at the time of liga-

tion to >95% 2 weeks after ligation. The upregula-

tion of VEGF is followed by vascular endothelial cell

proliferation and angiogenesis in fibrotic areas by

week 3 [133]. A similar animal study with diethyl-

nitrosamine-induced cirrhosis confirmed the con-

cept that hepatocellular hypoxia and angiogenesis

(6)

progress together with fibrogenesis after liver inju- ry [20]. Using neutralizing monoclonal antibodies of VEGFR-1 and VEGFR-2 in murine CCl

4

-induced liver fibrosis, both significantly attenuated the de- velopment of fibrosis and suppressed neovasculari- zation in the liver [174]. Fibrosis markers (hepatic hydroxyproline, serum hyaluronic acid and procol- lagen III-N-peptide), the number of smooth muscle actin positive cells and procollagen mRNA expres- sion were also suppressed by this treatment [174].

The inhibitory effect of the anti-VEGFR-2 antibody was more potent than that of anti-VEGFR-1 and combination treatment with both almost completely attenuated fibrosis development [174].

Hypoxia directly contributes to the progression of liver fibrosis by inducing the expression of VEGF, VEGFR-1 and type I collagen in activated hepatic stellate cells, the key player cell in the pathogenesis of hepatic fibrosis [20, 161]. VEGF on the other hand increases α1(I)-procollagen mRNA expression and stimulates proliferation of activated hepatic stellate cells [174]. VEGF expression in hypoxic hepatic stel- late cells has been shown to be mediated by cyclo- oxygenase-2 (COX-2) protein and COX-2 inhibitors significantly blocked VEGF production via the HIF- 1 α pathway [161]. VEGF signaling through VEGFR- 1 was shown to inhibit hepatic stellate cell contrac- tion, probably through attenuation of smooth mus- cle α-actin expression [99].

8.4.4

Portal Hypertension

A major complication of chronic liver diseases is portal hypertension. The development of portal- systemic collateral vessels in portal hypertension is classically explained as a mechanical consequence of increased portal pressure with subsequent open- ing of collateral vessels [11]. Recent evidence sug- gests that active, VEGF-dependent angiogenesis is also involved in this process: VEGF, VEGFR-2 and CD31 (as specific endothelial cell marker) protein levels in splanchnic organs increased after partial portal vein ligation in mice in a time-dependent fashion during the evolution of portal hyperten- sion [32]. A monoclonal antibody against VEGFR-2 given to these animals after ligation decreased the expression of VEGFR-2 and CD31 significantly and inhibited the formation of portal-systemic collateral vessels measured with labeled microspheres [32].

Experiments using partial portal vein-ligated rats and a VEGFR-2-specific tyrosine kinase inhibitor (SU5416) confirmed that the formation of portal- systemic collateral vessels is an angiogenesis-de- pendent process, which can be inhibited by antago-

nization of the VEGF/VEGFR-2 signaling pathway [32].

8.4.5

Viral Hepatitis

Hepatitis B virus X protein (HBx) is a hepatitis B virus-encoded transcriptional activator, which is involved in hepatocarcinogenesis and hypoxia- induced angiogenesis [83]. Recent studies have shown that HBx protein stabilizes HIF-1 α and en- hances transcriptional activity of HIF-1 α through activation of the mitogen-activated protein kinase (MAPK) pathway under normoxic and hypoxic conditions [108, 171]. The expression of HIF-1 α and VEGF was increased in the liver of HBx-transgenic mice [171] and in HBx-transfected HCC cell lines [83, 152]. Immunohistochemical staining for VEGF in chronic hepatitis B correlated with the degree of injury (grade) and amount of fibrosis (stage) [169].

In chronic hepatitis C liver samples, an increase in VEGF expression and angiogenesis in portal tracts has been described [103]. Another study found no difference in VEGF mRNA expression ratios among steatohepatitis, chronic hepatitis C and hepatocel- lular carcinoma [145]. Hepatitis C virus (HCV) core protein has been found to activate expression of VEGF in HepG(2) cells [89].

8.4.6

Hepatocellular Carcinoma

Tumor growth beyond the size of 1–2 mm

3

requires

the formation of new blood vessels in order to supply

the malignant tissue with nutrients and oxygen [55,

138]. Central hypoxia is the main driving force of

tumor angiogenesis and upregulates proangiogenic

growth factors like VEGF (Fig. 8.3) [57]. Therefore it

is not surprising that VEGF is upregulated in most

human tumors and that direct correlation with in-

tratumoral microvessel density exists [34]. The

angiogenic switch, which describes the acquisition

of the capacity to stimulate angiogenesis by shift-

ing the balance between stimulatory and inhibi-

tory factors of angiogenesis towards proangiogenic

factors, is a rate-limiting step in tumoral develop-

ment [54]. VEGF and other growth factors promote

survival, proliferation and migration of endothelial

cells, which will finally result in the formation of

new tumoral blood vessels enhancing further tu-

mor growth. These growth factors are secreted by

neoplastic cells, adjacent stroma, hepatocytes, stel-

late cells and tumor-infiltrating inflammatory cells

[42, 54, 69, 119]. Besides hypoxia, mutations in tu-

(7)

mor suppressor genes and oncogenes and certain viral proteins are also involved in the upregulation of VEGF during hepatocarcinogenesis (see above).

In vitro studies have shown that different inflam- matory cytokines (IL-1 β, interferon-α, interferon-γ, TNF- α) and growth factors (EGF, PDGF, basic fi- broblast growth factor, TGF- α) increase the secre- tion of VEGF in HCC cell lines [165].

Hepatocellular carcinoma (HCC) is a hypervas- cular tumor [4, 78, 165] and arterial hypervascu- larization is included in the non-invasive criteria to establish HCC diagnosis [12]. Several studies report on the overexpression of VEGF in HCC [17, 19, 105, 106, 108, 110, 165, 166, 173, 177]. Grafting HCC tis- sue onto chick embryo chorioallantoic membrane, which is a classical angiogenesis assay, stimulates neovascularization [98]. It has been shown that dur- ing hepatocarcinogenesis, expression of VEGF in- creases gradually from low-grade dysplastic nodules to high-grade dysplastic nodules to early HCC [119].

The degree of VEGF expression during development of HCC correlates with the density of vessels, un- paired arteries (i.e. arteries not accompanied by bile ducts, indicative of angiogenesis), CD34 staining (as a marker of sinusoidal capillarization) and the pro- liferation of hepatocytes assessed by staining with PCNA [119]. Moreover, tumor expression of VEGF (mRNA and protein expression) significantly corre- lated with serum VEGF level per platelet in patients with HCC, providing the basis for using circulating VEGF as a prognostic marker [125]. Furthermore, hepatocytes and HCC cells adjacent to peliosis and fibrous septa showed stronger VEGF expression [119]. Small HCCs showed a higher status of neoan- giogenesis and cell proliferation activity than ad- vanced HCCs [119].

The circulating concentration of VEGF increases with the stage of HCC, the highest levels being in patients with metastasis [66]. A prospective study of 100 patients suffering from HCC found that high serum levels of VEGF significantly correlated with absence of tumor capsule, presence of intrahepatic metastasis, presence of microscopic venous inva- sion, advanced stage and postoperative recurrence [124]. Similar results have been found using serum VEGF per platelet count in 52 HCC patients [77].

In a recent study, preoperative serum VEGF in 98 patients with resectable HCC was a significant and independent predictor of tumor recurrence, dis- ease-free survival and overall survival [17]. In 80 patients with inoperable HCC undergoing transar- terial chemoembolization (TACE), Poon and cow- orkers evaluated the prognostic significance of pre- treatment serum VEGF levels prospectively [123]:

pretreatment serum VEGF levels were significantly higher in patients with progressive disease than in

those with stable or responsive disease. Patients with serum VEGF >240 pg/ml had significantly worse survival than those with serum VEGF <240 pg/ml (median survival 6.8 vs. 19.2 months, p=0.007). In a Cox multivariate analysis, serum VEGF >240 pg/

ml was an independent prognostic factor of survival [123]. Expression of VEGF in patients and animals with HCC increased significantly after TACE [87, 88, 150]. VEGF antisense oligodeoxynucleotides mixed with lipiodol inhibited HCC growth in rats signifi- cantly more than arterial embolization with lipiodol alone [163]. These studies suggest that the VEGF sig- naling pathway plays an important role in tumor re- sponse to hypoxia after arterial embolization treat- ment and that anti-VEGF strategies might enhance the efficacy of arterial embolization in HCC.

8.4.7

Malignant Ascites Formation

In 1983, Senger and coworkers reported that hepa- tocarcinoma cells secrete a vascular permeability factor (VPF, later named VEGF), which promotes accumulation of ascites fluid [139]. Ascites, which is defined as accumulation of excess fluid within the peritoneal cavity, is encountered in many patients

Fig. 8.3. VEGF signaling and its effect on tumoral endothelial cells and HCC growth

(8)

with cirrhosis, other forms of liver disease and ma- lignancies. The pathogenesis of ascites depends on the underlying disease. An important component in the formation of ascites in patients with malignan- cies of the liver and abdominal cavity is microvas- cular hyperpermeability of tumor vessels due to tu- mor-secreted VEGF with consecutive extravasation of plasma and plasma proteins. As a matter of fact, levels of biologically active VEGF in patients with malignant ascites are higher in comparison to pa- tients with ascites due to non-malignant or cirrhotic causes [26, 80, 157, 176].

Four pathways for macromolecular extravasation have been described: endothelial fenestrae [130, 131], interendothelial cell gaps [58], transendothelial cell pores [111] and vesiculo-vacuolar organelles [31, 79].

VEGF has been shown to induce fenestration and in- crease permeability in normal and tumoral microv- ascular endothelium [14, 50, 130, 131, 170]: within 10 minutes of VEGF application fenestrations appear even in vascular beds that do not have fenestrated endothelium under physiological circumstances [130]. Liver sinusoidal endothelial cells incubated with 100 ng VEGF/ml increased the number of fen- estrations and cell pores in vitro [43, 170]. Neutral- izing antibodies against VEGF or against VEGFR-2 significantly suppressed the volume of ascites, the number of tumor cells in ascites and the peritoneal capillary permeability and prolonged the survival of ascites-bearing mice suffering from HCC [172].

Soluble VEGFR-1 and VEGF-trap (both soluble de- coy receptors for endogenous VEGF), monoclonal antibody against VEGF or VEGFR-2 and the VEGF receptor tyrosine kinase inhibitor PTK 787 all pre- vented the formation of malignant ascites in animal tumor models [13, 104, 143, 149, 164]. These results suggest that the VEGF/VEGFR-2 interaction is a ma- jor regulator of malignant ascites formation.

Interestingly, ascites VEGF levels are higher in cirrhotic patients with spontaneous bacterial peri- tonitis, which is a frequent complication of cirrhotic patients with ascites, than in non-infected cirrhotic patients [121]. VEGF is thereby produced in perito- neal macrophages of cirrhotic patients and is mark- edly upregulated by bacterial lipopolysaccharide and cytokines such as IL-1 [121].

8.4.8

Liver Transplantation

Hepatocyte and endothelial cell damage due to ischemia/reperfusion injury in liver transplantation after cold preservation is an important determinant of graft function. Activation of sinusoidal endothe- lial cells by cold ischemia alters expression of differ-

ent adhesion molecules and sequesters leukocytes and platelets during reperfusion, leading to micro- circulatory disturbance and liver injury. VEGF ex- pression is upregulated in hepatocytes of rat livers preserved in University of Wisconsin (UW) solution for orthotopic liver transplantation probably due to hypoxic stress [3]. Another study showed that VEGF is expressed and released in a biphasic pattern by Kupffer cells and hepatocytes during the early post- operative period after transplantation in a syn- geneic rat orthotopic liver transplantation model [10]. Anti-VEGF antibody treatment, administered during reperfusion, decreased the degree of dam- age (measured as liver function tests, lipid peroxi- dation, and metalloproteinase activity), suggesting that VEGF may have a role in ischemia/reperfusion injury to liver grafts [10].

Cyclosporin A and sirolimus (also known as rapamycin) are potent immunosuppressive drugs used after liver transplantation. Both drugs have been shown to downregulate VEGF expression un- der certain circumstances [18, 52, 92].

8.5 Outlook

Years of intense basic and preclinical research and the recent FDA approval of the first angiogenesis in- hibitor drug bevacizumab have now opened a wide range of clinical applications for VEGF-based strat- egies in liver diseases. Anti-VEGF strategies might have a potential in the treatment of HCC, liver fibro- sis, portal hypertension and in the inhibition of ma- lignant ascites, whereas pro-VEGF strategies might improve outcome after liver resection and stimulate liver regeneration. VEGF expression levels may be useful as prognostic markers in HCC patients and as predictors of tumor response to treatment.

Selected Reading

Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its re- ceptors. Nat Med 2003;9:669–676. (This review [36] provides a detailed and comprehensive description of the different VEGF isoforms and receptors and discusses their role under physiological and pathological conditions.)

Matsumoto T, Claesson-Welsh L. VEGF receptor signal transduc- tion. Sci STKE 2001;2001:RE21. (This review [101] outlines the current knowledge about the signal transduction proper- ties of VEGF receptors, with focus on VEGF receptor-2.) Autiero M, Waltenberger J, Communi D et al. Role of PlGF in the

intra- and intermolecular cross talk between the VEGF re-

(9)

ceptors Flt1 and Flk1. Nat Med 2003;9:936–943. (This study [6] provides new insight into the poorly understood role of VEGFR-1 signaling and the interaction between VEGFR-1 and VEGFR-2.)

LeCouter J, Moritz DR, Li B et al. Angiogenesis-independent endothelial protection of liver: role of VEGFR-1. Science 2003;299:890–893. (This study [81] describes the paracrine crosstalk between hepatocytes and liver sinusoidal en- dothelial cells during hepatocyte growth.)

http://www.nature.com/focus/angiogenesis/ (This web focus on angiogenesis with a special section on VEGF signaling is a joint project of the journals Nature Medicine and Nature Reviews Cancer. The web site provides review articles and a selection of “classic” papers nominated by experts in the field of angiogenesis.)

References

1. Alavi A, Hood JD, Frausto R et al. Role of Raf in vascular pro- tection from distinct apoptotic stimuli. Science 2003;301:94–

96.

2. Ankoma-Sey V, Matli M, Chang KB et al. Coordinated induc- tion of VEGF receptors in mesenchymal cell types during rat hepatic wound healing. Oncogene 1998;17:115–121.

3. Arai S, Mochida S, Ohno A et al. Decreased expression of receptors for vascular endothelial growth factor and sinu- soidal endothelial cell damage in cold-preserved rat livers.

Transplant Proc 1999;31:2668–2672.

4. Arii S, Mise M, Harada T et al. Overexpression of matrix met- alloproteinase 9 gene in hepatocellular carcinoma with in- vasive potential. Hepatology 1996;24:316–322.

5. Assy N, Spira G, Paizi M et al. Effect of vascular endothelial growth factor on hepatic regenerative activity following partial hepatectomy in rats. J Hepatol 1999;30:911–915.

6. Autiero M, Waltenberger J, Communi D et al. Role of PlGF in the intra- and intermolecular cross talk between the VEGF receptors Flt1 and Flk1. Nat Med 2003;9:936–943.

7. Banks RE, Forbes MA, Kinsey SE et al. Release of the ang- iogenic cytokine vascular endothelial growth factor (VEGF) from platelets: significance for VEGF measurements and cancer biology. Br J Cancer 1998;77:956–964.

8. Barleon B, Reusch P, Totzke F et al. Soluble VEGFR-1 secreted by endothelial cells and monocytes is present in human serum and plasma from healthy donors. Angiogenesis 2001;4:143–154.

9. Benjamin LE, Golijanin D, Itin A et al. Selective ablation of im- mature blood vessels in established human tumors follows vascular endothelial growth factor withdrawal. J Clin Invest 1999;103:159–165.

10. Boros P, Tarcsafalvi A, Wang L et al. Intrahepatic expression and release of vascular endothelial growth factor following orthotopic liver transplantation in the rat. Transplantation 2001;72:805–811.

11. Bosch J, Pizcueta P, Feu F et al. Pathophysiology of portal hy- pertension. Gastroenterol Clin North Am 1992;21:1–14.

12. Bruix J, Sherman M, Llovet JM et al. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona- 2000 EASL conference. European Association for the Study of the Liver. J Hepatol 2001;35:421–430.

13. Byrne AT, Ross L, Holash J et al. Vascular endothelial growth factor-trap decreases tumor burden, inhibits ascites, and causes dramatic vascular remodeling in an ovarian cancer model. Clin Cancer Res 2003;9:5721–5728.

14. Cao R, Eriksson A, Kubo H et al. Comparative evaluation of FGF-2-, VEGF-A-, and VEGF-C-induced angiogenesis, lym- phangiogenesis, vascular fenestrations, and permeability.

Circ Res 2004;94:664–670.

15. Carmeliet P, Ferreira V, Breier G et al. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature 1996;380:435–439.

16. Carmeliet P, Moons L, Luttun A et al. Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasa- tion in pathological conditions. Nat Med 2001;7:575–583.

17. Chao Y, Li CP, Chau GY et al. Prognostic significance of vascu- lar endothelial growth factor, basic fibroblast growth factor, and angiogenin in patients with resectable hepatocellular carcinoma after surgery. Ann Surg Oncol 2003;10:355–362.

18. Cho ML, Cho CS, Min SY et al. Cyclosporine inhibition of vas- cular endothelial growth factor production in rheumatoid synovial fibroblasts. Arthritis Rheum 2002;46:1202–1209.

19. Chow NH, Hsu PI, Lin XZ et al. Expression of vascular en- dothelial growth factor in normal liver and hepatocellular carcinoma: an immunohistochemical study. Hum Pathol 1997;28:698–703.

20. Corpechot C, Barbu V, Wendum D et al. Hypoxia-induced VEGF and collagen I expressions are associated with angio- genesis and fibrogenesis in experimental cirrhosis. Hepatol- ogy 2002;35:1010–1021.

21. Couper LL, Bryant SR, Eldrup-Jorgensen J et al. Vascular en- dothelial growth factor increases the mitogenic response to fibroblast growth factor-2 in vascular smooth muscle cells in vivo via expression of fms-like tyrosine kinase-1. Circ Res 1997;81:932–939.

22. Davidson AJ, Zon LI. Biomedicine: love, honor, and protect (your liver). Science 2003;299:835–837.

23. de Bont ES, Guikema JE, Scherpen F et al. Mobilized human CD34+ hematopoietic stem cells enhance tumor growth in a non-obese diabetic/severe combined immunodeficient mouse model of human non-Hodgkin’s lymphoma. Cancer Res 2001;61:7654–7659.

24. de Vries C, Escobedo JA, Ueno H et al. The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Sci- ence 1992;255:989–991.

25. Dibbens JA, Miller DL, Damert A et al. Hypoxic regulation of vascular endothelial growth factor mRNA stability requires the cooperation of multiple RNA elements. Mol Biol Cell 1999;10:907–919.

(10)

26. Dong W, Sun X, Yu B et al. Role of VEGF and CD44v6 in dif- ferentiating benign from malignant ascites. World J Gastro- enterol 2003;9:2596–2600.

27. Drixler TA, Vogten MJ, Ritchie ED et al. Liver regeneration is an angiogenesis-associated phenomenon. Ann Surg 2002;236:703–712.

28. Dvorak HF, Brown LF, Detmar M, Dvorak AM. Vascular per- meability factor/vascular endothelial growth factor, micro- vascular hyperpermeability, and angiogenesis. Am J Pathol 1995;146:1029–1039.

29. Eliceiri BP, Paul R, Schwartzberg PL et al. Selective require- ment for Src kinases during VEGF-induced angiogenesis and vascular permeability. Mol Cell 1999;4:915–924.

30. Farnebo F, Piehl F, Lagercrantz J. Restricted expression pat- tern of vegf-d in the adult and fetal mouse: high expres- sion in the embryonic lung. Biochem Biophys Res Commun 1999;257:891–894.

31. Feng D, Nagy J, Dvorak A, Dvorak H. Different pathways of macromolecule extravasation from hyperpermeable tumor vessels. Microvasc Res 2000;59:24–37.

32. Fernandez M, Vizzutti F, Garcia-Pagan JC et al. Anti-VEGF receptor-2 monoclonal antibody prevents portal-systemic collateral vessel formation in portal hypertensive mice. Gas- troenterology 2004;126:886–894.

33. Ferrara N, Carver-Moore K, Chen H et al. Heterozygous em- bryonic lethality induced by targeted inactivation of the VEGF gene. Nature 1996;380:439–442.

34. Ferrara N, Davis-Smyth T. The biology of vascular endothe- lial growth factor. Endocr Rev 1997;18:4–25.

35. Ferrara N. VEGF and the quest for tumour angiogenesis fac- tors. Nat Rev Cancer 2002;2:795–803.

36. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003;9:669–676.

37. Finnerty H, Kelleher K, Morris GE et al. Molecular cloning of murine FLT and FLT4. Oncogene 1993;8:2293–2298.

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

39. Fong GH, Zhang L, Bryce DM, Peng J. Increased heman- gioblast commitment, not vascular disorganization, is the primary defect in flt-1 knock-out mice. Development 1999;126:3015–3025.

40. Fuh G, Li B, Crowley C et al. Requirements for binding and signaling of the kinase domain receptor for vascular en- dothelial growth factor. J Biol Chem 1998;273:11197–11204.

41. Fujii H, Hirose T, Oe S et al. Contribution of bone marrow cells to liver regeneration after partial hepatectomy in mice.

J Hepatol 2002;36:653–659.

42. Fukumura D, Xavier R, Sugiura T et al. Tumor induction of VEGF promoter activity in stromal cells. Cell 1998;94:715–

725.

43. Funyu J, Mochida S, Inao M et al. VEGF can act as vascular permeability factor in the hepatic sinusoids through upreg- ulation of porosity of endothelial cells. Biochem Biophys Res Commun 2001;280:481–485.

44. Gerber HP, Condorelli F, Park J, Ferrara N. Differential tran- scriptional regulation of the 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.

45. Gerber HP, Dixit V, Ferrara N. Vascular endothelial growth factor induces expression of the antiapoptotic proteins Bcl-2 and A1 in vascular endothelial cells. J Biol Chem 1998;273:13313–13316.

46. Gerber HP, McMurtrey A, Kowalski J et al. Vascular endothe- lial growth factor regulates endothelial cell survival through the phosphatidylinositol 3’-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation. J Biol Chem 1998;273:30336–30343.

47. Gerber HP, Malik AK, Solar GP et al. VEGF regulates haemat- opoietic stem cell survival by an internal autocrine loop mechanism. Nature 2002;417:954–958.

48. Greene AK, Wiener S, Puder M et al. Endothelial-directed hepatic regeneration after partial hepatectomy. Ann Surg 2003;237:530–535.

49. Grugel S, Finkenzeller G, Weindel K et al. Both v-Ha-Ras and v-Raf stimulate expression of the vascular endothelial growth factor in NIH 3T3 cells. J Biol Chem 1995;270:25915–

25919.

50. Grunstein J, Roberts WG, Mathieu-Costello O et al. Tumor- derived expression of vascular endothelial growth factor is a critical factor in tumor expansion and vascular function.

Cancer Res 1999;59:1592–1598.

51. Gualdi R, Bossard P, Zheng M et al. Hepatic specification of the gut endoderm in vitro: cell signaling and transcriptional control. Genes Dev 1996;10:1670–1682.

52. Guba M, von Breitenbuch P, Steinbauer M et al. Rapamycin inhibits primary and metastatic tumor growth by antiangio- genesis: involvement of vascular endothelial growth factor.

Nat Med 2002;8:128–135.

53. Guo D, Jia Q, Song HY et al. Vascular endothelial cell growth factor promotes tyrosine phosphorylation of me- diators of signal transduction that contain SH2 domains.

Association with endothelial cell proliferation. J Biol Chem 1995;270:6729–6733.

54. Hanahan D, Folkman J. Patterns and emerging mecha- nisms of the angiogenic switch during tumorigenesis. Cell 1996;86:353–364.

55. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57–70.

56. Haratake J, Hisaoka M, Yamamoto O, Horie A. Morphological changes of hepatic microcirculation in experimental rat cir- rhosis: a scanning electron microscopic study. Hepatology 1991;13:952–956.

57. Harris AL. Hypoxia – a key regulatory factor in tumour growth. Nat Rev Cancer 2002;2:38–47.

58. Hirata A, Baluk P, Fujiwara T, McDonald DM. Location of fo- cal silver staining at endothelial gaps in inflamed venules examined by scanning electron microscopy. Am J Physiol 1995;269:L403–L418.

59. Hiratsuka S, Minowa O, Kuno J et al. Flt-1 lacking the tyrosine kinase domain is sufficient for normal development and an-

(11)

giogenesis in mice. Proc Natl Acad Sci USA 1998;95:9349–

9354.

60. Hiratsuka S, Maru Y, Okada A et al. Involvement of Flt-1 tyro- sine kinase (vascular endothelial growth factor receptor-1) in pathological angiogenesis. Cancer Res 2001;61:1207–1213.

61. Hiratsuka S, Nakamura K, Iwai S et al. MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell 2002;2:289–300.

62. Houck KA, Leung DW, Rowland AM et al. Dual regulation of vascular endothelial growth factor bioavailability by genetic and proteolytic mechanisms. J Biol Chem 1992;267:26031–

26037.

63. Huet PM, Goresky CA, Villeneuve JP et al. Assessment of liver microcirculation in human cirrhosis. J Clin Invest 1982;70:1234–1244.

64. Jelkmann W. Pitfalls in the measurement of circulating vas- cular endothelial growth factor. Clin Chem 2001;47:617–

623.

65. Jeltsch M, Kaipainen A, Joukov V et al. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice. Science 1997;276:1423–1425.

66. Jinno K, Tanimizu M, Hyodo I et al. Circulating vascular en- dothelial growth factor (VEGF) is a possible tumor marker for metastasis in human hepatocellular carcinoma. J Gastro- enterol 1998;33:376–382.

67. Josko J, Mazurek M. Transcription factors having impact on vascular endothelial growth factor (VEGF) gene expression in angiogenesis. Med Sci Monit 2004;10:RA89–RA98.

68. Joukov V, Pajusola K, Kaipainen A et al. A novel vascular en- dothelial growth factor, VEGF-C, is a ligand for the Flt4 (VEG- FR-3) and KDR (VEGFR-2) receptor tyrosine kinases. EMBO J 1996;15:1751.

69. Jung JO, Gwak GY, Lim YS et al. [Role of hepatic stellate cells in the angiogenesis of hepatoma.] Korean J Gastroenterol 2003;42:142–148.

70. Kaipainen A, Korhonen J, Mustonen T et al. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lym- phatic endothelium during development. Proc Natl Acad Sci USA 1995;92:3566–3570.

71. Kalluri R, Sukhatme VP. Fibrosis and angiogenesis. Curr Opin Nephrol Hypertens 2000;9:413–418.

72. Kalluri R. Basement membranes: structure, assembly and role in tumour angiogenesis. Nat Rev Cancer 2003;3:422–

433.

73. Karkkainen MJ, Makinen T, Alitalo K. Lymphatic endothe- lium: a new frontier of metastasis research. Nat Cell Biol 2002;4:E2–E5.

74. Keck PJ, Hauser SD, Krivi G et al. Vascular permeability fac- tor, an endothelial cell mitogen related to PDGF. Science 1989;246:1309–1312.

75. Kendall RL, Wang G, Thomas KA. Identification of a natural soluble form of the vascular endothelial growth factor re- ceptor, FLT-1, and its heterodimerization with KDR. Biochem Biophys Res Commun 1996;226:324–328.

76. Kieser A, Weich HA, Brandner G et al. Mutant p53 potentiates protein kinase C induction of vascular endothelial growth factor expression. Oncogene 1994;9:963–969.

77. Kim SJ, Choi IK, Park KH et al. Serum vascular endothelial growth factor per platelet count in hepatocellular carcino- ma: correlations with clinical parameters and survival. Jpn J Clin Oncol 2004;34:184–190.

78. Koga H, Sakisaka S, Ohishi M et al. Expression of cyclooxy- genase-2 in human hepatocellular carcinoma: relevance to tumor dedifferentiation. Hepatology 1999;29:688–696.

79. Kohn S, Nagy JA, Dvorak HF, Dvorak AM. Pathways of mac- romolecular tracer transport across venules and small veins.

Structural basis for the hyperpermeability of tumor blood vessels. Lab Invest 1992;67:596–607.

80. Kraft A, Weindel K, Ochs A et al. Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer 1999;85:178–187.

81. LeCouter J, Moritz DR, Li B et al. Angiogenesis-independ- ent endothelial protection of liver: role of VEGFR-1. Science 2003;299:890–893.

82. Lee J, Gray A, Yuan J et al. Vascular endothelial growth factor-related protein: a ligand and specific activator of the tyrosine kinase receptor Flt4. Proc Natl Acad Sci USA 1996;93:1988–1992.

83. Lee SW, Lee YM, Bae SK et al. Human hepatitis B virus X pro- tein is a possible mediator of hypoxia-induced angiogenesis in hepatocarcinogenesis. Biochem Biophys Res Commun 2000;268:456–461.

84. Leung DW, Cachianes G, Kuang WJ et al. Vascular endothe- lial growth factor is a secreted angiogenic mitogen. Science 1989;246:1306–1309.

85. Levy AP, Levy NS, Goldberg MA. Post-transcriptional regula- tion of vascular endothelial growth factor by hypoxia. J Biol Chem 1996;271:2746–2753.

86. Levy NS, Chung S, Furneaux H, Levy AP. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA- binding protein HuR. J Biol Chem 1998;273:6417–6423.

87. Li X, Feng G, Zheng C et al. Influence of transarterial chem- oembolization on angiogenesis and expression of vascular endothelial growth factor and basic fibroblast growth fac- tor in rat with Walker-256 transplanted hepatoma: an experi- mental study. World J Gastroenterol 2003;9:2445–2449.

88. Liao X, Yi J, Li X et al. Expression of angiogenic factors in hepatocellular carcinoma after transcatheter arterial chem- oembolization. J Huazhong Univ Sci Technolog Med Sci 2003;23:280–282.

89. Liu C, Liu W, Yang J, Fang D. [HCV core protein activates ex- pression of vascular endothelial growth factor in HepG(2) cells.] Zhonghua Gan Zang Bing Za Zhi 2001;9:214–216.

90. Liu LX, Lu H, Luo Y et al. Stabilization of vascular endothelial growth factor mRNA by hypoxia-inducible factor 1. Biochem Biophys Res Commun 2002;291:908–914.

91. Longo R, Sarmiento R, Fanelli M et al. Anti-angiogenic ther- apy: rationale, challenges and clinical studies. Angiogenesis 2002;5:237–256.

(12)

92. Luan FL, Ding R, Sharma VK et al. Rapamycin is an effec- tive inhibitor of human renal cancer metastasis. Kidney Int 2003;63:917–926.

93. Luttun A, Tjwa M, Moons L et al. Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1. Nat Med 2002;8:831–840.

94. Lyden D, Hattori K, Dias S et al. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic pre- cursor cells blocks tumor angiogenesis and growth. Nat Med 2001;7:1194–1201.

95. Lyttle DJ, Fraser KM, Fleming SB et al. Homologs of vascular endothelial growth factor are encoded by the poxvirus orf virus. J Virol 1994;68:84–92.

96. Maglione D, Guerriero V, Viglietto G et al. Isolation of a human placenta cDNA coding for a protein related to the vascular permeability factor. Proc Natl Acad Sci USA 1991;88:9267–9271.

97. Martinez-Hernandez A, Amenta PS. The extracellular matrix in hepatic regeneration. FASEB J 1995;9:1401–1410.

98. Marzullo A, Vacca A, Roncali L et al. Angiogenesis in hepato- cellular carcinoma: an experimental study in the chick em- bryo chorioallantoic membrane. Int J Oncol 1998;13:17–21.

99. Mashiba S, Mochida S, Ishikawa K et al. Inhibition of hepatic stellate cell contraction during activation in vitro by vascu- lar endothelial growth factor in association with upregula- tion of FLT tyrosine kinase receptor family, FLT-1. Biochem Biophys Res Commun 1999;258:674–678.

100. Matsumoto K, Yoshitomi H, Rossant J, Zaret KS. Liver orga- nogenesis promoted by endothelial cells prior to vascular function. Science 2001;294:559–563.

101. Matsumoto T, Claesson-Welsh L. VEGF receptor signal trans- duction. Sci STKE 2001;2001:RE21.

102. Matthews W, Jordan CT, Gavin M et al. A receptor tyrosine kinase cDNA isolated from a population of enriched primi- tive hematopoietic cells and exhibiting close genetic link- age to c-kit. Proc Natl Acad Sci USA 1991;88:9026–9030.

103. Medina J, Caveda L, Sanz-Cameno P et al. Hepatocyte growth factor activates endothelial proangiogenic mecha- nisms relevant in chronic hepatitis C-associated neoangio- genesis. J Hepatol 2003;38:660–667.

104. Mesiano S, Ferrara N, Jaffe RB. Role of vascular endothelial growth factor in ovarian cancer: inhibition of ascites forma- tion by immunoneutralization. Am J Pathol 1998;153:1249–

1256.

105. Mise M, Arii S, Higashituji H et al. Clinical significance of vascular endothelial growth factor and basic fibroblast growth factor gene expression in liver tumor. Hepatology 1996;23:455–464.

106. Miura H, Miyazaki T, Kuroda M et al. Increased expression of vascular endothelial growth factor in human hepatocel- lular carcinoma. J Hepatol 1997;27:854–861.

107. Mochida S, Ishikawa K, Inao M et al. Increased expressions of vascular endothelial growth factor and its receptors, flt- 1 and KDR/flk-1, in regenerating rat liver. Biochem Biophys Res Commun 1996;226:176–179.

108. Moon E-J, Jeong C-H, Jeong J-W et al. Hepatitis B virus X protein induces angiogenesis by stabilizing hypoxia-induc- ible factor-1&alpha. FASEB J 2003:03–0153fje.

109. Mukhopadhyay D, Knebelmann B, Cohen HT et al. The von Hippel-Lindau tumor suppressor gene product interacts with Sp1 to repress vascular endothelial growth factor pro- moter activity. Mol Cell Biol 1997;17:5629–5639.

110. Nakashima Y, Nakashima O, Hsia CC et al. Vascularization of small hepatocellular carcinomas: correlation with differen- tiation. Liver 1999;19:12–18.

111. Neal CR, Michel CC. Transcellular gaps in microvascular walls of frog and rat when permeability is increased by perfusion with the ionophore A23187. J Physiol 1995;488(Pt 2):427–437.

112. Neufeld G, Cohen T, Shraga N et al. The neuropilins: multi- functional semaphorin and VEGF receptors that modulate axon guidance and angiogenesis. Trends Cardiovasc Med 2002;12:13–19.

113. Neufeld G, Kessler O, Herzog Y. The interaction of Neuropi- lin-1 and Neuropilin-2 with tyrosine-kinase receptors for VEGF. Adv Exp Med Biol 2002;515:81–90.

114. Nomura M, Yamagishi S, Harada S et al. Possible participa- tion of autocrine and paracrine vascular endothelial growth factors in hypoxia-induced proliferation of endothelial cells and pericytes. J Biol Chem 1995;270:28316–28324.

115. Olofsson B, Pajusola K, Kaipainen A et al. Vascular endothe- lial growth factor B, a novel growth factor for endothelial cells. Proc Natl Acad Sci USA 1996;93:2576–2581.

116. Orlandini M, Marconcini L, Ferruzzi R, Oliviero S. Identifica- tion of a c-fos-induced gene that is related to the platelet- derived growth factor/vascular endothelial growth factor family. Proc Natl Acad Sci USA 1996;93:11675–11680.

117. Pajusola K, Aprelikova O, Korhonen J et al. FLT4 receptor tyrosine kinase contains seven immunoglobulin-like loops and is expressed in multiple human tissues and cell lines.

Cancer Res 1992;52:5738–5743.

118. Park JE, Chen HH, Winer J et al. Placenta growth factor. Po- tentiation 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;269:25646–25654.

119. Park YN, Kim YB, Yang KM, Park C. Increased expression of vascular endothelial growth factor and angiogenesis in the early stage of multistep hepatocarcinogenesis. Arch Pathol Lab Med 2000;124:1061–1065.

120. Pepper MS, Ferrara N, Orci L, Montesano R. Vascular en- dothelial growth factor (VEGF) induces plasminogen ac- tivators and plasminogen activator inhibitor-1 in microv- ascular endothelial cells. Biochem Biophys Res Commun 1991;181:902–906.

121. Perez-Ruiz M, Ros J, Morales-Ruiz M et al. Vascular endothe- lial growth factor production in peritoneal macrophages of cirrhotic patients: regulation by cytokines and bacterial lipopolysaccharide. Hepatology 1999;29:1057–1063.

122. Plouet J, Moro F, Bertagnolli S et al. Extracellular cleavage of the vascular endothelial growth factor 189-amino acid

(13)

form by urokinase is required for its mitogenic effect. J Biol Chem 1997;272:13390–13396.

123. Poon R, Lau C, Yu W et al. High serum levels of vascular en- dothelial growth factor predict poor response to transar- terial chemoembolization in hepatocellular carcinoma: a prospective study. Oncol Rep 2004;11:1077–1084.

124. Poon RT, Ng IO, Lau C et al. Serum vascular endothelial growth factor predicts venous invasion in hepatocellular carcinoma: a prospective study. Ann Surg 2001;233:227–

235.

125. Poon RT-P, Lau CP-Y, Cheung S-T et al. Quantitative cor- relation of serum levels and tumor expression of vascular endothelial growth factor in patients with hepatocellular carcinoma. Cancer Res 2003;63:3121–3126.

126. Quinn TP, Peters KG, De Vries C et al. Fetal liver kinase 1 is a receptor for vascular endothelial growth factor and is se- lectively expressed in vascular endothelium. Proc Natl Acad Sci USA 1993;90:7533–7537.

127. Rafii S, Lyden D, Benezra R et al. Vascular and haematopoi- etic stem cells: novel targets for anti-angiogenesis therapy?

Nat Rev Cancer 2002;2:826–835.

128. Redaelli CA, Semela D, Carrick FE et al. Effect of vascular en- dothelial growth factor on functional recovery after hepa- tectomy in lean and obese mice. J Hepatol 2004;40:305–

312.

129. Reyes M, Dudek A, Jahagirdar B et al. Origin of endothelial progenitors in human postnatal bone marrow. J Clin Invest 2002;109:337–346.

130. Roberts WG, Palade GE. Increased microvascular perme- ability and endothelial fenestration induced by vascular endothelial growth factor. J Cell Sci 1995;108(Pt 6):2369–

2379.

131. Roberts WG, Palade GE. Neovasculature induced by vas- cular endothelial growth factor is fenestrated. Cancer Res 1997;57:765–772.

132. Robinson CJ, Stringer SE. The splice variants of vascular en- dothelial growth factor (VEGF) and their receptors. J Cell Sci 2001;114:853–865.

133. Rosmorduc O, Wendum D, Corpechot C et al. Hepatocellu- lar hypoxia-induced vascular endothelial growth factor ex- pression and angiogenesis in experimental biliary cirrhosis.

Am J Pathol 1999;155:1065–1073.

134. Ross MA, Sander CM, Kleeb TB et al. Spatiotemporal ex- pression of angiogenesis growth factor receptors during the revascularization of regenerating rat liver. Hepatology 2001;34:1135–1148.

135. Sato T, El-Assal ON, Ono T et al. Sinusoidal endothelial cell proliferation and expression of angiopoietin/Tie family in regenerating rat liver. J Hepatol 2001;34:690–698.

136. Sawano A, Iwai S, Sakurai Y et al. Flt-1, vascular endothelial growth factor receptor 1, is a novel cell surface marker for the lineage of monocyte-macrophages in humans. Blood 2001;97:785–791.

137. Seetharam L, Gotoh N, Maru Y et al. A unique signal trans- duction from FLT tyrosine kinase, a receptor for vascular en- dothelial growth factor VEGF. Oncogene 1995;10:135–147.

138. Semenza GL. Angiogenesis in ischemic and neoplastic dis- orders. Annu Rev Med 2003;54:17–28.

139. Senger DR, Galli SJ, Dvorak AM et al. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science 1983;219:983–985.

140. Shalaby F, Rossant J, Yamaguchi TP et al. Failure of blood-is- land formation and vasculogenesis in Flk-1-deficient mice.

Nature 1995;376:62–66.

141. Shay-Salit A, Shushy M, Wolfovitz E et al. VEGF recep- tor 2 and the adherens junction as a mechanical trans- ducer in vascular endothelial cells. Proc Natl Acad Sci USA 2002;99:9462–9467.

142. Shibuya M, Yamaguchi S, Yamane A et al. Nucleotide se- quence and expression of a novel human receptor-type tyrosine kinase gene (flt) closely related to the fms family.

Oncogene 1990;5:519–524.

143. Shibuya M, Luo J, Toyoda M, Yamaguchi S. Involvement of VEGF and its receptors in ascites tumor formation. Cancer Chemother Pharmacol 1999;43(Suppl):S72–S77.

144. Shima DT, Deutsch U, D’Amore PA. Hypoxic induction of vascular endothelial growth factor (VEGF) in human epi- thelial cells is mediated by increases in mRNA stability. FEBS Lett 1995;370:203–208.

145. Shimoda K, Mori M, Shibuta K et al. Vascular endothelial growth factor/vascular permeability factor mRNA expres- sion in patients with chronic hepatitis C and hepatocellular carcinoma. Int J Oncol 1999;14:353–359.

146. Soker S, Takashima S, Miao HQ et al. Neuropilin-1 is ex- pressed by endothelial and tumor cells as an isoform-spe- cific receptor for vascular endothelial growth factor. Cell 1998;92:735–745.

147. Sondell M, Lundborg G, Kanje M. Vascular endothelial growth factor has neurotrophic activity and stimulates ax- onal outgrowth, enhancing cell survival and Schwann cell proliferation in the peripheral nervous system. J Neurosci 1999;19:5731–5740.

148. Stacker SA, Vitali A, Caesar C et al. A mutant form of vascular endothelial growth factor (VEGF) that lacks VEGF receptor- 2 activation retains the ability to induce vascular perme- ability. J Biol Chem 1999;274:34884–34892.

149. Stoelcker B, Echtenacher B, Weich H et al. VEGF/Flk-1 inter- action, a requirement for malignant ascites recurrence. J Interferon Cytokine Res 2000;20:511–517.

150. Suzuki H, Mori M, Kawaguchi C et al. Serum vascular en- dothelial growth factor in the course of transcatheter arte- rial embolization of hepatocellular carcinoma. Int J Oncol 1999;14:1087–1090.

151. Taniguchi E, Sakisaka S, Matsuo K et al. Expression and role of vascular endothelial growth factor in liver regeneration after partial hepatectomy in rats. J Histochem Cytochem 2001;49:121–130.

152. Tao X, Shen D, Ren H et al. Hepatitis B virus X protein acti- vates expression of IGF-IR and VEGF in hepatocellular car- cinoma cells. Zhonghua Gan Zang Bing Za Zhi 2000;8:161–

163.

(14)

153. Terman BI, Carrion ME, Kovacs E et al. Identification of a new endothelial cell growth factor receptor tyrosine kinase. On- cogene 1991;6:1677–1683.

154. Terman BI, Dougher-Vermazen M, Carrion ME et al. Identifi- cation of the KDR tyrosine kinase as a receptor for vascular endothelial cell growth factor. Biochem Biophys Res Com- mun 1992;187:1579–1586.

155. Tischer E, Mitchell R, Hartman T et al. The human gene for vascular endothelial growth factor. Multiple protein forms are encoded through alternative exon splicing. J Biol Chem 1991;266:11947–11954.

156. Unemori EN, Ferrara N, Bauer EA, Amento EP. Vascular en- dothelial growth factor induces interstitial collagenase expression in human endothelial cells. J Cell Physiol 1992;153:557–562.

157. Verheul HMW, Hoekman K, Jorna AS et al. Targeting vascu- lar endothelial growth factor blockade: ascites and pleural effusion formation. Oncologist 2000;5:45–50.

158. Villeneuve JP, Dagenais M, Huet PM et al. The hepatic mi- crocirculation in the isolated perfused human liver. Hepa- tology 1996;23:24–31.

159. Vincenti V, Cassano C, Rocchi M, Persico G. Assignment of the vascular endothelial growth factor gene to human chromosome 6p21.3. Circulation 1996;93:1493–1495.

160. Waltenberger J, Claesson-Welsh L, Siegbahn A et al. Differ- ent signal transduction properties of KDR and Flt1, two re- ceptors for vascular endothelial growth factor. J Biol Chem 1994;269:26988–26995.

161. Wang Y, Luk J, Ikeda K et al. Regulatory role of vHL/HIF-1al- pha in hypoxia-induced VEGF production in hepatic stellate cells. Biochem Biophys Res Commun 2004;317:358–362.

162. Webb NJ, Bottomley MJ, Watson CJ, Brenchley PE. Vascular endothelial growth factor (VEGF) is released from platelets during blood clotting: implications for measurement of circulating VEGF levels in clinical disease. Clin Sci (Lond) 1998;94:395–404.

163. Wu H, Feng G, Liang H et al. Vascular endothelial growth factor antisense oligodeoxynucleotides with lipiodol in ar- terial embolization of liver cancer in rats. World J Gastroen- terol 2004;10:813–818.

164. Xu L, Yoneda J, Herrera C et al. Inhibition of malignant as- cites and growth of human ovarian carcinoma by oral ad- ministration of a potent inhibitor of the vascular endothe- lial growth factor receptor tyrosine kinases. Int J Oncol 2000;16:445–454.

165. Yamaguchi R, Yano H, Iemura A et al. Expression of vascular endothelial growth factor in human hepatocellular carci- noma. Hepatology 1998;28:68–77.

166. Yamaguchi R, Yano H, Nakashima Y et al. Expression and lo- calization of vascular endothelial growth factor receptors

in human hepatocellular carcinoma and non-HCC tissues.

Oncol Rep 2000;7:725–729.

167. Yamamoto T, Kobayashi T, Phillips MJ. Perinodular arteriolar plexus in liver cirrhosis. Scanning electron microscopy of microvascular casts. Liver 1984;4:50–54.

168. Yamane A, Seetharam L, Yamaguchi S et al. A new commu- nication system between hepatocytes and sinusoidal en- dothelial cells in liver through vascular endothelial growth factor and Flt tyrosine kinase receptor family (Flt-1 and KDR/Flk-1). Oncogene 1994;9:2683–2690.

169. Yan J, Chen W, Ma Y, Sun X. Expression of vascular endothe- lial growth factor in liver tissues of hepatitis B. Zhonghua Gan Zang Bing Za Zhi 2000;8:150–152.

170. Yokomori H, Oda M, Yoshimura K et al. Vascular endothelial growth factor increases fenestral permeability in hepatic sinusoidal endothelial cells. Liver Int 2003;23:467–475.

171. Yoo YG, Oh SH, Park ES et al. Hepatitis B virus X protein en- hances transcriptional activity of hypoxia-inducible factor- 1{alpha} through activation of mitogen-activated protein kinase pathway. J Biol Chem 2003;278:39076–39084.

172. Yoshiji H, Kuriyama S, Hicklin D et al. The vascular endothe- lial growth factor receptor KDR/Flk-1 is a major regulator of malignant ascites formation in the mouse hepatocellular carcinoma model. Hepatology 2001;33:841–847.

173. Yoshiji H, Kuriyama S, Yoshii J et al. Synergistic effect of basic fibroblast growth factor and vascular endothelial growth factor in murine hepatocellular carcinoma. Hepa- tology 2002;35:834–842.

174. Yoshiji H, Kuriyama S, Yoshii J et al. Vascular endothelial growth factor and receptor interaction is a prerequisite for murine hepatic fibrogenesis. Gut 2003;52:1347–1354.

175. Yuan F, Chen Y, Dellian M et al. Time-dependent vascular regression and permeability changes in established human tumor xenografts induced by an anti-vascular endothelial growth factor/vascular permeability factor antibody. Proc Natl Acad Sci USA 1996;93:14765–14770.

176. Zebrowski BK, Liu W, Ramirez K et al. Markedly elevated levels of vascular endothelial growth factor in malignant ascites. Ann Surg Oncol 1999;6:373–378.

177. Zhou XD. Recurrence and metastasis of hepatocellular car- cinoma: progress and prospects. Hepatobiliary Pancreat Dis Int 2002;1:35–41.

178. Zucker S, Mirza H, Conner CE et al. Vascular endothelial growth factor induces tissue factor and matrix metallopro- teinase production in endothelial cells: conversion of pro- thrombin to thrombin results in progelatinase A activation and cell proliferation. Int J Cancer 1998;75:780–786.

Riferimenti

Documenti correlati

The effect of TT temperature on CZTS thin film samples, obtained from 0.2 g/mL ink spread at a 6000 rpm spin rate, was studied in terms of its surface morphology, grain growth,

On the con- trary, currently, “young people are pushed into training and education that they feel they are not ready for; and young people at high risk but who are already in

Prevenzione e Gestione del rischio clinico, il progetto della ASL Roma B, TEME, 9.07 Roma. farmacia farmacia URP

LA DETERMINAZIONE DEL RELATIVO CANONE. Approvato con delibera C.C.. 2 - Disciplina per il rilascio della concessione e/o autorizzazione di occupazione _3 Art. 7 - Rinnovo

Conversely, non-classical HLA-E and HLA-G molecules are frequently overexpressed by cancer cells participating in tumor immune escape through the interaction with

OMC–2 FIR 4: a highly irradiated region In the previous section, we showed that in order to reproduce the temperature of the gas probed by the c- C 3 H 2 lines and its abundance the

1) Herschel images show the presence of 53 dense cores associated with the Trifid Nebula, mostly identified with the TC condensations detected at mm wavelength.. 2) Different stages

La trasformazione di Rohrwacher in Mark si inserisce su questa linea di sottrazione, ma il film di Laura Bispuri rifugge ogni enfasi o marcatura spettacolare: pur mettendo in