Next Article in Journal
Immune Response to Sipuleucel-T in Prostate Cancer
Next Article in Special Issue
P190B RhoGAP Regulates Chromosome Segregation in Cancer Cells
Previous Article in Journal
External Beam Radiotherapy of Recurrent Glioma: Radiation Tolerance of the Human Brain
Previous Article in Special Issue
Remodeling of Tumor Stroma and Response to Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Dynamics of Developmental and Tumor Angiogenesis—A Comparison

Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-17177, Sweden
*
Author to whom correspondence should be addressed.
Cancers 2012, 4(2), 400-419; https://doi.org/10.3390/cancers4020400
Submission received: 1 March 2012 / Revised: 3 April 2012 / Accepted: 4 April 2012 / Published: 11 April 2012
(This article belongs to the Special Issue Tumor Stroma)

Abstract

:
The blood vasculature in cancers has been the subject of intense interest during the past four decades. Since the original ideas of targeting angiogenesis to treat cancer were proposed in the 1970s, it has become evident that more knowledge about the role of vessels in tumor biology is needed to fully take advantage of such strategies. The vasculature serves the surrounding tissue in a multitude of ways that all must be taken into consideration in therapeutic manipulation. Aspects of delivery of conventional cytostatic drugs, induction of hypoxia affecting treatment by radiotherapy, changes in tumor cell metabolism, vascular leak and trafficking of leukocytes are affected by interventions on vascular function. Many tumors constitute a highly interchangeable milieu undergoing proliferation, apoptosis, and necrosis with abundance of growth factors, enzymes and metabolites. These aspects are reflected by the abnormal tortuous, leaky vascular bed with detached mural cells (pericytes). The vascular bed of tumors is known to be unstable and undergoing remodeling, but it is not until recently that this has been dynamically demonstrated at high resolution, facilitated by technical advances in intravital microscopy. In this review we discuss developmental genetic loss-of-function experiments in the light of tumor angiogenesis. We find this a valid comparison since many studies phenocopy the vasculature in development and tumors.

1. Introduction

1.1. The Tumor Vasculature

Tumor growth requires a supply of oxygen and nutrients, normally supported via blood vessels. Tumor cells positioned near a capillary are supported until the increasing volume limits sufficient diffusion. Cells that for genetic reasons can sustain cellular functions at low oxygen levels might be selected but in order to proliferate further, the supply of nutrients, hence the close vicinity to capillaries has to be maintained. When the oxygen level drops vascular endothelial growth factor-A (VEGFA, also denoted vascular permeability factor) will be secreted from the tumor cells (reviewed in [1]). This growth factor will induce endothelial cell (EC) proliferation, migration and vascular leakage that give rise to an expansion of the vascular bed. This expansion may occur through several conceptually different processes: sprouting angiogenesis (migration and proliferation of EC), intussusception (splitting of vessels by pillar formation), co-option (tumor cells adopt the preexisting vasculature), vascular mimicry (tumor cells and other stromal cells form channels) and possibly looping (new vascular loops are forming, driven by tension forces) and in situ differentiation of cancer stem-like cells [2,3,4,5,6]. Because of the technical challenges of studying these dynamic processes, we know little about their respective contribution to vascular growth in tumors [7]. Regardless of how vessels are formed, it is commonly accepted that the vasculature of tumors is different from that of the surrounding tissue. In comparison with normal well organized functional vessels they are often tortuous, pericyte-deficient and leaky, with irregular diameter and altered arterial to venous hierarchy [8]. Some of these properties (discussed below) are commonly seen in developmental angiogenesis—A process that has been intensely studied, providing deep knowledge on cellular behavior, especially in sprouting [9,10]. Because of these shared properties between the angiogenic vasculature in development and the tumor vasculature it might be informative to apply the knowledge gained from developmental studies on the process of tumor angiogenesis. Below we focus on molecules that have central roles during initiation, migration and elongation of new branches in the process of sprouting angiogenesis. It is important to point out that their requirement for guided sprouting does not exclude a role during the alternative modes of vascular expansion. Within this review we use detailed information from developmental gene loss-of-function (LOF) studies to further understand the process of tumor angiogenesis. We find this a valid comparison since many LOF studies phenocopy the vasculature in development and tumors (Table 1, and discussed below).
Table 1. Comparison of vascular phenotypes in development and cancer following gene modifications or drug interventions.
Table 1. Comparison of vascular phenotypes in development and cancer following gene modifications or drug interventions.
Genetic DeletionDrug/Ab Intervention
GeneDevelopmental phenotypeTumor phenotypeDevelopmental phenotypeTumor phenotype
VegfaVegfa−/−: lethal at E9.5 due to abnormal vascular development.
Vegfa+/−: lethal at E11.5 due to abnormal vascular development [11,12]
Vitreal injections of soluble VEGFR1 inhibited ischemia-induced neo-vascularization in rat retina [13]. Note:VEGFR1 also binds PlGFHuman melanoma cells stably expressing Vegf antisense RNA formed small and poorly vascularized tumors in mice [14]
Vegfr1Vegfr1−/−: lethal at E8.5 with excessive growth and disorganized vessels [15]. Reduced sprouting from embryonic stem cells [16,17]. VEGFR1 TK knockout: no effect on vascular development, but reduced macrophage migration [18]. Flt1 KD in zebrafish: increased intersegmental sprouting [19].Vegfr1 TK knockout: Impaired tumor metastasis by suppression of MMP-9 expression [20,21]. Blocking VEGFA and PlGF binding to VEGFR1: reduced tumor growth and vascularization [22,23]. Antisense mediated downregulation of VEGFR1 suppressed tumor growth in mice [24] Note that VEGFR1 is expressed by Leukocytes.
Vegfr2Vegfr2−/−: lethal at E8.5–9.5; defective hematopoietic and endothelial cell differentiation [25]. NTAb-mediated VEGFR2 Neutralization led to retraction of tip cell filopodia and inhibited angiogenesis in the postnatal retina [9,26].VEGFR2 block with small molecules or Abs suppressed endothelial growth and migration, induced apoptosis and regression of vessels in tumors [27,28,29]. Soluble VEGFR1 or neutralizing antibody: reduced tumor growth and metastasis [30,31].
Vegfr3Vegfr3−/−: lethal at E10.5, defective arterial-venous remodeling of primary vascular plexus [32]. EC specific deletion of VEGFR3 results in excessive angiogenesis [33].Excessive angiogenesis in tumor xenografts in endothelial specific VEGFR3 knockout mice [33]. VEGFR3 blocking Ab: reduced vascular density, number of branch points and sprouts in the postnatal retina [26].VEGFR3 blocking Ab: reduced sprouting angiogenesis in tumor xenografts in mice [26,34].
EphB4EphB4−/− mice: lethal at E10.5 as a consequence of perturbed arterial-venous differentiation; arrest in cardiac development [35,36].EphB4+/− mice: enhanced tumor growth in an intestinal tumor genesis model [37].Blocking EphB4/EphrinB2 signaling by soluble EphB4 inhibited angiogenesis in murine Matrigel and corneal pocket assays [38].Specific antibodies to EphB4, soluble EphB4 or small molecule inhibitors reduced tumor angiogenesis [38,39,40].
EphrinB2Efnb2−/−: lethal at E11.5, defective maturation of arteries and veins into capillaries [36,41]. EphrinB2 PDZ signaling-deficient mice displayed reduced number of tip cells, fewer filopodia at the vascular front in retina. Mural cell specific inactivation of EphrinB2 resulted in defective microvessel architecture [42].EphrinB2-PDZ signaling-deficient mice showed decreased tumor vascularization and reduced tumor growth [42].NTNT
Neuropilin-1Nrp1−/−: lethal around E13, loss of filopodia extension in tip cells and impaired microvessel branching [43,44,45].NTNrp-1 monoclonal Ab: reduced vascular sprouting and remodeling in the developing mouse retina [46].Anti-Nrp-1 in combination with anti-VEGF Ab: reduced vessel density in tumor and exhibited additive effect on tumor suppression [46].
Delta-like4Most Dll4+/− mice die around E10.5 (strain dependent) with defective arteriogenesis and vascular remodeling [47,48,49]. Dll4+/− or EC-specific ko display increased tip cell formation and branching [50,51,52,53]Dll4 targeting increased tumor vascular sprouting and branching, but with poor perfusion and therefor suppressed tumor growth [54,55,56].Antibody against Dll4 or soluble Dll4 promotes vascular sprouting and block artery development [56,57]. Dll4-Fc intraocular injections caused retinal hypersprouting [52]Antibody against Dll4, soluble Dll4 or Dll4 vaccination increased non-functional tumor vessel growth and inhibited tumor growth [56,57,58].
Delta-like1Dll1−/−:Lethal around E12 with sever hemorrhage [59]. Heterozygous Dll1 mice have impaired postnatal arteriogenesis [60].NTNTNT
Notch1Notch1−/−: lethal at E9.5 with disrupted vascular remodeling [61]. EC specific knockout of Notch1 increased tip cell formation [53].Induced ablation of Notch1 in adult mice facilitated tumorigenesis in the skin [62]Inhibition of Notch signaling by γ-secretase inhibitor increased tip cells and vessel branches [53].γ-secretase inhibitor suppressed tumor growth by directly targeting tumor cells and inhibition of angiogenesis [63].
Plexin D1plexinD1−/−:Perinatal lethality, abnormal cardiac development and defective intersomitic blood vessels and skeletal morphology [64,65].NTNTNT
Angiopoie-tinsAng1−/−: lethal at E12.5 with impaired vascular branching and remodeling [66].NTNTAntisense Ang1 RNA: Reduced xenograft tumor growth and angiogenesis [67]. Adenoviral mediated gene transfer of soluble Tie2 inhibited tumor growth and metastasis [68].
Tie1Tie1−/−: lethal between E13.5 and birth with edema and hemorrhage; defective structural integrity of ECs [69]. Ang1 and Tie1 double knockout failed to establish the right hand side venous system [70].NTNTNT
Semaph-orinsSema3a−/− (Nrp1 ligand) mice embryos showed disrupted vasculature [71]. Sema3C−/−: perinatal death, congenital cardiovascular defects with interruption of the aortic arch [72]. Sema3E−/−: disorganized embryonic intersomitic vessels [73].Impaired maturation of tumor vessels in Sema 4d−/− mice [74].NTSema 3A administration reduced tumor vascularization [75]
Unc5bUnc5b−/−: lethal from E12.5 (CD1 strain survives); enhanced filopodia formation and branching [76,77].NTUnc5b blocking Ab: Increased vascular density and sprouting of the postnatal retinal vasculature [77].NT
Some genes in the table have not been discussed in the text. E, embryonic day; KD, knock down; NT, not tested; TK, tyrosine kinase; MMP, matrix metallo-proteinase; Ab, antibody; EC, endothelial cell; ko, knock out; KD, knock down.

1.2. Players in the Dynamic Establishment of the Vasculature

Blood vessels are built of ECs, supportive mural cells (pericytes and smooth muscle cells), and their shared basement membrane. In the developing embryo the main vessels are assembled by in situ differentiation of precursor cells in a process denoted as vasculogenesis [78]. From this primary vascular network new branches are formed through coordinated events of EC proliferation and migration, termed sprouting angiogenesis [79]. A multitude of signaling pathways are at play to balance the frequency of sprouting events and to guide the extending branch (reviewed in [79]). VEGF-A and -C and their receptors VEGFR1-3, the neuropilins (Nrps), the semaphorins (Sema), the eph and ephrins [42], the angiopoietins (Ang) and endothelial TEK tyrosine kinase receptors (Tie), Jagged1 and Delta like ligands (Dll)-1 and -4 and Notch-1 and -2 and VE-Cadherin are all required for correct temporal and spatial vascular patterning [80,81,82]. Many of these molecules are differentially expressed by subsets of ECs within the vascular sprout, demarking the cellular heterogeneity of the growing vasculature. Cells at the very front of sprouts with extensive actin rich protrusions and a certain gene expression profile are termed tip cells whereas the cells located just behind are referred to as stalk cells [9,83,84]. The tip cells are morphologically very similar to the axon-guiding growth cone and have in part similar functions, to lead the way [9,84,85,86]. Cells at the very tip of the sprout frequently express relatively higher levels of several proteins such as VEGFR-2, -3, Dll4, neuropilin-1, platelet-derived growth factor (PDGF)-B, Unc5b, EphrinB2 and Cxcr4a than their neighbors [83,87,88,89]. The differential proteome of each cell will set its potential in acquiring the tip cell position. In part these relations are manifested by EC to EC signaling via the Dll4-Notch1 pathway and its ability to regulate expression of the VEGFRs. However, none of these proteins are exclusively expressed by the leading cell. In accordance we recently demonstrated that the tip cell phenotype is transient and that cells can be over-taken by previous stalk cells [10]. This indicates that the “tip cell gene expression profile” may be switched on even before the cell has acquired the tip position and its characteristic morphology. It is commonly stated in publications that the tip cells are highly motile whereas the following stalk cells are not. However, recent data demonstrate that also cells within the stalk might be very motile [10,90]. Also, not all cells within the sprout migrate in a determined direction but seem to be chemokinetic, patrolling along the length of the forming sprout. The tip cells and the following one or two stalk cells lack pericyte coverage but secrete PDGFB that attracts mural cells by activation of their PDGFRβ [91,92]. EC-pericyte connections are required for vascular patterning and function as demonstrated by abnormal angiogenesis in pericyte-deficient mouse models [91]. As new branches are formed the basement membrane is deposited by endothelial tip- and stalk cells [93]. Therefore the growing front has an immature basement membrane that matures over time. The emerging sprout is transient and usually does not extend beyond the length of 150 μm in vivo before it connects to another vessel or anastomoses with an adjacent sprout to subsequently open up. Dysfunctional branches are then pruned and degraded as observed by live imaging or by the presence of empty collagen IV sleeves [88,94,95]. This final selection of branches is regulated by several factors such as shear stress, VEGF levels, and activity within the Notch pathway [94,96]. For example VEGF inhibition led to increased intussusceptive pruning in the chicken chorioallantoic membrane [96]. These observations clearly demark an enormous flexibility of the developing vascular tree. The extent and functional relevance of cellular shuffling in angiogenesis is still unknown.

1.3. Vascular Dynamics in the Tumor Microenvironment, What Is Actually Known?

Current knowledge on the modes of angiogenesis in tumor models in mouse is limited and for human cancers essentially absent. It is evident however that the vasculature of human tumors has an abnormal architecture, similar to what is observed in mouse models. Furthermore, the developing sprouting vasculature and the tumor vasculature have common traits such as poorly defined arterial/venous identity [97], incomplete pericyte coverage [98,99], non-perfused regions [9] and abnormal basement membrane with non EC-associated collagen IV indicative of vessel pruning [8,100]. In tumors, vessels may form via different processes: sprouting, intussusception, co-option, vascular looping and cancer stem-like cell differentiation [2,3,4,5,6]. It is difficult to discriminate between these various processes by analysis of fixed tumor material. Intravital microscopy has been used since the 1930s to image the tumor vasculature and its dynamic nature [101]. However it is not until recently that improved microscopy techniques have allowed for more detailed information on vascular dynamics. Vajkoczy et al. [102] injected C6 glioma cell suspensions and multicellular spheroids into striated muscle and the cerebral cortex of mice. By in vivo imaging they described an initial migration of injected tumor cells along host vessels (not considered co-option), but as the tumor started to establish itself, sprouting angiogenesis was initiated. Continued observation revealed several dysfunctional vessels that over time were pruned and regressed. Kienast et al. [103] seeded human lung carcinoma or melanoma cells into the carotid artery and imaged metastasis growth using multiphoton laser scanning microscopy in a cranial window over several days. By expression of red fluorescent protein from tumor cells and intravenous injections of FITC-dextran they dynamically visualized the synchronous expansion of the tumor and its vasculature at high resolution. Whereas the vasculature of the lung-derived tumors was highly angiogenic already two days after injection, the melanoma-derived tumor vasculature displayed preferentially capillary loop formation, tortuous vascular structures and vasodilatation. In conclusion these studies demonstrate constant remodeling of the vasculature of injected tumors in mice. Since the vasculature was visualized by fluorescence in the blood stream, which does not allow visualization of EC protrusions, little information is given on the behavior at the cellular level. Experimental data indicate that mechanisms of tip/stalk selection are at play in at least certain tumor models. Noguera-Troise et al. demonstrated extensive filopodia rich sprouting blood vessels of subcutaneous Lewis lung tumors and C6 rat glioma tumors. These sprouts expressed high levels of Dll4, similar to sprouting vessels in development [54]. Furthermore detailed studies of pancreatic islet tumors in the RIP-Tag mouse model (Rat insulin promoter driving the large T antigen of the simian virus 40 giving rise to insulinomas) revealed extensive EC sprouts suggesting involvement of sprouting angiogenesis [8]. Also, ECs of orthotopic gliomas in mice have been shown to extend branches and filopodia [42].
The similarities between the angiogenic vasculature in development and in tumors suggest that cellular programs may be common. By comparing information on knockout phenotypes in development with the ones of tumors we likely improve our understanding of how the tumor vasculature takes shape. Below we describe molecular pathways primarily involved in sprouting angiogenesis and the process of tip/stalk selection.

2. Angiogenesis in Development and Cancer—Lessons from Gene and Drug Interventions

From genetic studies and drug intervention studies it is evident that drivers of angiogenesis are shared in development, physiology and cancer. As described above several receptor-ligand systems have been carefully analyzed with respect to sprouting angiogenesis in development. Comparing genetic and drug induced loss- and gain- of function of the developing versus the tumor vasculature provides potential information on the process of tumor angiogenesis [104].

2.1. The VEGFs and VEGFRs

To date, activation of VEGFR2 by the hypoxia inducible ligand VEGFA is considered the initiating and main driving event of angiogenesis (reviewed in [79,105,106,107]). The importance of this signaling system is demonstrated by the haploinsufficiency of VEGFA and the embryonic lethality as a consequence of either total VEGFR2 deletion or mutation of its tyrosine residue 1173 [11,12,25,108,109]. Over recent years its ability of inducing sprouting angiogenesis and EC migration has been shown in numerous studies. Conversely, inhibition of VEGFR2 phosphorylation by the tyrosine kinase inhibitor SU5416 reduced sprouting and filopodia formation of the intersomitic vessels in the zebra fish embryo [110]. Also, intraocular injections of either soluble VEGFR1, to sequester VEGFA, or a VEGFR2-blocking antibody, lead to reduced tip cell filopodia and vascular branching in the postnatal retina [9]. Naturally it has therefore been the main therapeutic target in disease, and blockage display dramatic effects on the morphology of the vasculature [104]. Studies of pancreatic islet tumors in the RIP-Tag mouse model indicate reduced endothelial processes and empty collagen “sleeves”, demarking a reduced angiogenic profile upon VEGF blockage [8]. In vivo imaging by Kienast et al. (see above) revealed inhibition of angiogenesis in lung metastasis in the brain by treatment with the VEGF-trap, bevacizumab [103]. Furthermore, the vasculature became “normalized” with increased pericyte coverage, reduced density and more regular sized vessels [111,112].
As discussed below VEGFR2 activity is modulated by several co-receptors, such as neuropilin1 and Heparan sulphate proteoglycans but also by the mode of internalization, in turn regulated by eph receptors and VE-Cadherins [42,113,114,115]. In addition to VEGFR2, ECs express VEGFR1 that binds VEGFA with high affinity. The extracellular domain of VEGFR1 is required for vascular development and patterning whereas signaling via its tyrosine kinases is not. VEGFR1 has a negative impact on sprout formation and reduced VEGFR1 levels potentiate the tip cell phenotype [10,19]. VEGFR1 is also positively regulated by notch signaling and possibly functions to shape gradients by sequestering VEGFA [116].
VEGFR3 is expressed by the developing blood vasculature but is almost restricted to the lymphatics after embryonic day (E)13.5 [117]. The receptor is however re-expressed in the blood vasculature of many tumors [26,118,119]. Apart from its crucial role in development of the lymphatic vasculature it is required for development of the blood vasculature [32]. In several publications VEGFR3 has been shown to be enriched in the sprouting tip region of the postnatal retina and also, like VEGFR2, to be negatively regulated by Notch signaling [50,120]. Inhibition by VEGFR3 blocking antibodies reduced filopodia and sprout formation in the developing retina and in tumors [33]. In accordance VEGFR3 has been shown to promote sprouting of the embryonic intersomitic vessels of the zebrafish [120]. However, EC specific deletion of VEGFR3 in mice leads to ectopic sprouting, excessive filopodia formation and increased vascular density of the hindbrain and retina [26]. Similarly the vasculature of syngeneic subcutaneously implanted Lewis lung carcinomas displayed extensive filopodia and branching upon EC-specific VEGFR3 deletion. Also, in a mosaic vasculature of wild type and VEGFR3+/− cells, VEGFR3+/− cells preferentially became tip cells. Thus VEGFR3 may act both in a pro- and anti-angiogenic fashion likely depending on the presence of VEGFR2 and balance of the ligands VEGF-A and -C [33,121].

2.2. Delta-Like, Jagged and Notches

As mentioned above the membrane bound ligand Dll4 and signaling by its receptor Notch1 restricts sprouting angiogenesis and filopodia formation in the developing zebrafish and in the postnatal retina [50,51,53,56,110,120]. Blockage of notch cleavage and signaling by the gamma secretase inhibitor DAPT or by administration of soluble Dll4 protein induced vascular branching in vitro and in vivo. Dll4 is thought to mainly signal to Notch located on adjacent cells and thereby regulates selection of stalk and tip cells. Clonal analysis of chimeric developing vasculature demonstrated that cells with relatively higher Dll4 expression than their neighbors acquired the tip position at high frequency [10]. Accordingly, cells with reduced Notch signaling capacity relative to their neighbor frequently adopted the tip cell position [10,53,120]. Another Notch ligand, Jagged-1, is preferentially expressed by stalk cells and suppresses the tip cell phenotype by inhibition of Dll4-mediated notch signaling. EC specific jagged-1 LOF leads to decreased vascular density and sprouting [50].
Interference with Dll4 in several tumor models recapitulates what has been reported in development [56,57,58]. Blocking Dll4 signaling by administration of Dll4-Fc protein gave rise to increased vascular density but reduced tumor growth. This controversy was explained by the low degree of perfusion, indicative of a dysfunctional hypersprouting vascular bed.

2.3. The Eph and Ephrins

Several studies have reported the importance of the Eph receptor tyrosine kinase receptors and their ligands, the ephrins, as guidance cues for the developing vasculature [35,36,87]. Mice lacking the intracellular PDZ domain of EphrinB2 display reduced number of tip cells and filopodia extensions of the post natal retinal vasculature. In the same study the authors demonstrate reduced vascular density and filopodia numbers in an orthotopic glioma tumor model. Mechanistically, loss of the PDZ domain reduces VEGFR2 internalization thereby reducing its activation [42]. These data suggest that EphrinB2-dependent tumor angiogenesis at least in part is mediated by sprouting angiogenesis. In an additional study by Wang et al. endothelial specific deletion of EphrinB2 leads to reduced retinal sprouting angiogenesis [41]. Here, the authors link reduced sprouting to reduced internalization and reduced signaling via VEGFR3 instead. In addition morpholino-induced knockdown of efnb2a in zebrafish inhibited intersomitic sprouting and endothelial filopodia extensions. The eph-ephrin-system is not exclusively involved in sprouting angiogenesis but also during angiogenesis in the yolk sac, and in arterial to venous specification in development, suggesting important roles in other modes of angiogenesis than sprouting [122].

2.4. The Angiopoietins and Ties

The angiopoietin/Tie2 signaling system is required for vascular remodeling, maturation and stabilization. Mice lacking Ang1 displayed abnormal vasculature with reduced branching and poor mural cell coverage [66]. Overexpression of Ang1 in the skin of mice results in hypervascularization [123]. Unlike the hyperpermeable vessels induced by VEGF, vessels induced by Ang1 overexpression are leakage-resistant [124]. Coexpression of Ang1 and VEGF led to abundant vessel growth without leakage [124]. These findings suggest synergic functions of Ang1 and VEGF in the development of mature and stable vessels. The mechanism may relate to the functions of Ang1 in formation of EC junctions, or its involvement in EC adhesion and mural cell recruitment. Studies on cultured ECs have shown that Ang1 decreases the basal phosphorylation level of VE-Cadherin and PECAM-1 and inhibits permeability [125]. Ang1 can also serve as an adhesive substrate and promote endothelial adhesion and migration on extracellular matrix in an integrin-dependent manner [126]. Mural cell recruitment induced by Ang1 is partially mediated by stimulation of monocyte chemotactic protein-1 expression [127]. Ang1 also up-regulates Dll4 expression thereby contributing to vascular quiescence [128]. Ang2, another ligand of the Tie2 receptor, can act as a natural antagonist for Ang1/Tie2 signaling. Overexpression of Ang2 inhibits angiogenesis in the mouse embryo [129]. Targeting of Ang2 in mouse blocked the natural hyaloid vessel regression, revealing its indispensable role in postnatal vascular remodeling and regression [130]. Ang2 is up-regulated in the tumor vessels and promotes vascular destabilization and regression, which facilitate VEGF-induced angiogenesis in the later stage [131]. However, both Ang1 and Ang2 can alter their role in angiogenesis. Ang1 suppresses angiogenesis in the mouse heart while Ang2 collaborates with VEGFA to promote angiogenesis [132]. It is reported that Tie2 can translocate to either cell-cell or cell-matrix contacts upon Ang1 stimulation and thereby induce different downstream pathways [133,134]. This could explain the two faces of Tie2 signaling; inducing vascular quiescent or angiogenesis. Tie2 signaling is maintained in a precise balance in normal tissue, which makes it a challenging target for anti-angiogenic therapy. However, it has been reported that inhibition of Tie2 signaling by antisense Ang1 RNA or soluble Tie2 can inhibit tumor growth by reducing tumor angiogenesis [67,68]. Drugs targeting Ang1/2 or Tie2 are currently in clinical cancer trials [135]. Tie1 is the homologue of Tie2, but remains an orphan receptor so far. Deletion of Tie1 results in lethality between E13.5 to birth depending on genetic background; edema and hemorrhage are observed in the Tie1−/− mice [69]. Double knockout of Ang1 and Tie1 generates a phenotype of right-hand side venous system loss [70]. Discovery of Tie1 ligands in the future will be helpful to elucidate its detailed function.

3. Conclusions

Almost all the proteins engaged in the regulation of developmental angiogenesis are likewise involved in tumor vascularization in mouse models (Table 1). Despite conserved EC expression and regulation of most of these molecules the resulting tumor vasculature is in part dysfunctional. This phenomenon should therefore be attributed differences in the non-vascular tissue of the developing organism and the tumor, respectively. In development tissue boundaries are well organized and patterning is strictly controlled. For example the intersomitic vessels are clearly guided by extracellular matrix proteins deposited in the intersegmental space and astrocytes and Müller glia form a scaffold on which the endothelial plexus can grow in the post natal retina of mice. In the tumor environment, tissue organization is often lost and production of growth factors and extracellular matrix components is abundant. Low oxygen levels, high enzymatic activity and altered pH create a highly reactive microenvironment prone to feed vascular malformations. As a consequence the vascular integrity is lost with detachment of pericytes and leakage of plasma proteins into the tumor stroma. Fibronectin deposition enhance migration of myofibroblasts and infiltrating leukocytes [136]. The altered flow dynamics of the irregular vasculature may in itself feedback to promote vascular remodeling. In development it was demonstrated that tip cell filopodia was instantly retracted upon initiation of flow in the sprouting central arteries of the zebrafish hindbrain, demonstrating the power of shear forces [88]. Furthermore, poor delivery of blood borne molecules (i.e., sphingosine-1 phosphate, Gaengel et al. unpublished) involved in mediating vascular quiescence could further add to the hyper active vascular phenotype. Within this review we have described that molecules known to regulate sprouting angiogenesis also have similar functions in the vasculature of tumors as judged from knockouts and drug targeting. Despite these observations very limited data on the actual dynamics of the tumor vasculature exist. It is not known what mode of vascular expansion i.e., sprouting, intussusception, vascular looping or vascular mimicry that is dominating in tumor growth. It is reasonable to believe that these various routes are closely linked and they may even occur simultaneously. Technical advances in the field of microscopy and imaging offer great potential in resolving many of these questions [137,138,139].

Acknowledgments

This work was supported by the Strategic Cardiovascular Program of KI and Stockholm County Council, the Swedish Cancer Society, and the Swedish Research Council (L.J.).

References

  1. Ferrara, N. Vascular endothelial growth factor: Basic science and clinical progress. Endocr. Rev. 2004, 25, 581–611. [Google Scholar]
  2. Soda, Y. Transdifferentiation of glioblastoma cells into vascular endothelial cells. Proc. Natl. Acad. Sci. USA 2011, 108, 4274–4280. [Google Scholar]
  3. Wang, R. Glioblastoma stem-like cells give rise to tumour endothelium. Nature 2010, 468, 829–833. [Google Scholar]
  4. Ricci-Vitiani, L.; Pallini, R.; Biffoni, M.; Todaro, M.; Invernici, G.; Cenci, T.; Maira, G.; Parati, E.A.; Stassi, G.; Larocca, L.M.; et al. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 2010, 468, 824–828. [Google Scholar]
  5. Ribatti, D.; Djonov, V. Intussusceptive microvascular growth in tumors. Cancer Lett. 2012, 316, 126–131. [Google Scholar]
  6. Kilarski, W.W.; Gerwins, P. A new mechanism of blood vessel growth—Hope for new treatment strategies. Discov. Med. 2009, 8, 23–27. [Google Scholar]
  7. Tozer, G.M.; Ameer-Beg, S.M.; Baker, J.; Barber, P.R.; Hill, S.A.; Hodgkiss, R.J.; Locke, R.; Prise, V.E.; Wilson, I.; Vojnovic, B. Intravital imaging of tumour vascular networks using multi-photon fluorescence microscopy. Adv. Drug Deliv. Rev. 2005, 57, 135–152. [Google Scholar]
  8. Baluk, P.; Morikawa, S.; Haskell, A.; Mancuso, M.; McDonald, D.M. Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am. J. Pathol. 2003, 163, 1801–1815. [Google Scholar]
  9. Gerhardt, H.; Golding, M.; Fruttiger, M.; Ruhrberg, C.; Lundkvist, A.; Abramsson, A.; Jeltsch, M.; Mitchell, C.; Alitalo, K.; Shima, D.; et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J. Cell Biol. 2003, 161, 1163–1177. [Google Scholar] [CrossRef]
  10. Jakobsson, L.; Franco, C.A.; Bentley, K.; Collins, R.T.; Ponsioen, B.; Aspalter, I.M.; Rosewell, I.; Busse, M.; Thurston, G.; Medvinsky, A.; et al. Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nat. Cell Biol. 2010, 12, 943–953. [Google Scholar] [CrossRef]
  11. Carmeliet, P.; Ferreira, V.; Breier, G.; Pollefeyt, S.; Kieckens, L.; Gertsenstein, M.; Fahrig, M.; Vandenhoeck, A.; Harpal, K.; Eberhardt, C.; et al. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature 1996, 380, 435–439. [Google Scholar]
  12. Ferrara, N.; Carver-Moore, K.; Chen, H.; Dowd, M.; Lu, L.; O'Shea, K.S.; Powell-Braxton, L.; Hillan, K.J.; Moore, M.W. Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature 1996, 380, 439–442. [Google Scholar]
  13. Rota, R.; Riccioni, T.; Zaccarini, M.; Lamartina, S.; Gallo, A.D.; Fusco, A.; Kovesdi, I.; Balestrazzi, E.; Abeni, D.C.; Ali, R.R.; et al. Marked inhibition of retinal neovascularization in rats following soluble-flt-1 gene transfer. J. Gene Med. 2004, 6, 992–1002. [Google Scholar] [CrossRef]
  14. Claffey, K.P.; Brown, L.F.; del Aguila, L.F.; Tognazzi, K.; Yeo, K.T.; Manseau, E.J.; Dvorak, H.F. Expression of vascular permeability factor/vascular endothelial growth factor by melanoma cells increases tumor growth, angiogenesis, and experimental metastasis. Cancer Res. 1996, 56, 172–181. [Google Scholar]
  15. Fong, G.H.; Rossant, J.; Gertsenstein, M.; Breitman, M.L. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 1995, 376, 66–70. [Google Scholar]
  16. Kearney, J.B.; Kappas, N.C.; Ellerstrom, C.; DiPaola, F.W.; Bautch, V.L. The VEGF receptor Flt-1 (VEGFR-1) is a positive modulator of vascular sprout formation and branching morphogenesis. Blood 2004, 103, 4527–4535. [Google Scholar]
  17. Chappell, J.C.; Taylor, S.M.; Ferrara, N.; Bautch, V.L. Local guidance of emerging vessel sprouts requires soluble Flt-1. Dev. Cell 2009, 17, 377–386. [Google Scholar]
  18. Hiratsuka, S.; Minowa, O.; Kuno, J.; Noda, T.; Shibuya, M. Flt-1 lacking the tyrosine kinase domain is sufficient for normal development and angiogenesis in mice. Proc. Natl. Acad. Sci. USA 1998, 95, 9349–9354. [Google Scholar]
  19. Krueger, J.; Liu, D.; Scholz, K.; Zimmer, A.; Shi, Y.; Klein, C.; Siekmann, A.; Schulte-Merker, S.; Cudmore, M.; Ahmed, A.; le Noble, F. Flt1 acts as a negative regulator of tip cell formation and branching morphogenesis in the zebrafish embryo. Development 2011, 138, 2111–2120. [Google Scholar]
  20. Hiratsuka, S.; Maru, Y.; Okada, A.; Seiki, M.; Noda, T.; Shibuya, M. Involvement of Flt-1 tyrosine kinase (vascular endothelial growth factor receptor-1) in pathological angiogenesis. Cancer Res. 2001, 61, 1207–1213. [Google Scholar]
  21. Hiratsuka, S.; Nakamura, K.; Iwai, S.; Murakami, M.; Itoh, T.; Kijima, H.; Shipley, J.M.; Senior, R.M.; Shibuya, M. MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell 2002, 2, 289–300. [Google Scholar]
  22. Fischer, C.; Jonckx, B.; Mazzone, M.; Zacchigna, S.; Loges, S.; Pattarini, L.; Chorianopoulos, E.; Liesenborghs, L.; Koch, M.; de Mol, M.; et al. Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell 2007, 131, 463–475. [Google Scholar] [CrossRef]
  23. Luttun, A.; Tjwa, M.; Moons, L.; Wu, Y.; Angelillo-Scherrer, A.; Liao, F.; Nagy, J.A.; Hooper, A.; Priller, J.; de Klerck, B.; 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. [Google Scholar]
  24. Weng, D.E.; Usman, N. Angiozyme: A novel angiogenesis inhibitor. Curr. Oncol. Rep. 2001, 3, 141–146. [Google Scholar]
  25. Shalaby, F.; Rossant, J.; Yamaguchi, T.P.; Gertsenstein, M.; Wu, X.F.; Breitman, M.L.; Schuh, A.C. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 1995, 376, 62–66. [Google Scholar]
  26. Tammela, T.; Zarkada, G.; Wallgard, E.; Murtomaki, A.; Suchting, S.; Wirzenius, M.; Waltari, M.; Hellstrom, M.; Schomber, T.; Peltonen, R.; et al. Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature 2008, 454, 656–660. [Google Scholar]
  27. McMahon, G. VEGF receptor signaling in tumor angiogenesis. Oncologist 2000, 5, S3–S10. [Google Scholar]
  28. Carmeliet, P.; Jain, R.K. Angiogenesis in cancer and other diseases. Nature 2000, 407, 249–257. [Google Scholar]
  29. Carmeliet, P. VEGF as a key mediator of angiogenesis in cancer. Oncology 2005, 69, S4–S10. [Google Scholar]
  30. Shibuya, M. Structure and dual function of vascular endothelial growth factor receptor-1 (Flt-1). Int. J. Biochem. Cell Biol. 2001, 33, 409–420. [Google Scholar]
  31. Stefanik, D.F.; Fellows, W.K.; Rizkalla, L.R.; Rizkalla, W.M.; Stefanik, P.P.; Deleo, A.B.; Welch, W.C. Monoclonal antibodies to vascular endothelial growth factor (VEGF) and the VEGF receptor, FLT-1, inhibit the growth of C6 glioma in a mouse xenograft. J. Neurooncol. 2001, 55, 91–100. [Google Scholar] [CrossRef]
  32. Dumont, D.J.; Jussila, L.; Taipale, J.; Lymboussaki, A.; Mustonen, T.; Pajusola, K.; Breitman, M.; Alitalo, K. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science 1998, 282, 946–949. [Google Scholar]
  33. Tammela, T.; Zarkada, G.; Nurmi, H.; Jakobsson, L.; Heinolainen, K.; Tvorogov, D.; Zheng, W.; Franco, C.A.; Murtomaki, A.; Aranda, E.; et al. VEGFR-3 controls tip to stalk conversion at vessel fusion sites by reinforcing Notch signalling. Nat. Cell Biol. 2011, 13, 1202–1213. [Google Scholar] [CrossRef]
  34. Laakkonen, P.; Waltari, M.; Holopainen, T.; Takahashi, T.; Pytowski, B.; Steiner, P.; Hicklin, D.; Persaud, K.; Tonra, J.R.; Witte, L.; et al. Vascular endothelial growth factor receptor 3 is involved in tumor angiogenesis and growth. Cancer Res. 2007, 67, 593–599. [Google Scholar]
  35. Adams, R.H.; Wilkinson, G.A.; Weiss, C.; Diella, F.; Gale, N.W.; Deutsch, U.; Risau, W.; Klein, R. Roles of ephrinB ligands and EphB receptors in cardiovascular development: Demarcation of arterial/venous domains, vascular morphogenesis, and sprouting angiogenesis. Genes Dev. 1999, 13, 295–306. [Google Scholar] [CrossRef]
  36. Heroult, M.; Schaffner, F.; Augustin, H.G. Eph receptor and ephrin ligand-mediated interactions during angiogenesis and tumor progression. Exp. Cell Res. 2006, 312, 642–650. [Google Scholar]
  37. Dopeso, H.; Mateo-Lozano, S.; Mazzolini, R.; Rodrigues, P.; Lagares-Tena, L.; Ceron, J.; Romero, J.; Esteves, M.; Landolfi, S.; Hernandez-Losa, J.; et al. The receptor tyrosine kinase EPHB4 has tumor suppressor activities in intestinal tumorigenesis. Cancer Res. 2009, 69, 7430–7438. [Google Scholar]
  38. Kertesz, N.; Krasnoperov, V.; Reddy, R.; Leshanski, L.; Kumar, S.R.; Zozulya, S.; Gill, P.S. The soluble extracellular domain of EphB4 (sEphB4) antagonizes EphB4-EphrinB2 interaction, modulates angiogenesis, and inhibits tumor growth. Blood 2006, 107, 2330–2338. [Google Scholar]
  39. Martiny-Baron, G.; Holzer, P.; Billy, E.; Schnell, C.; Brueggen, J.; Ferretti, M.; Schmiedeberg, N.; Wood, J.M.; Furet, P.; Imbach, P. The small molecule specific EphB4 kinase inhibitor NVP-BHG712 inhibits VEGF driven angiogenesis. Angiogenesis 2010, 13, 259–267. [Google Scholar]
  40. Krasnoperov, V.; Kumar, S.R.; Ley, E.; Li, X.; Scehnet, J.; Liu, R.; Zozulya, S.; Gill, P.S. Novel EphB4 monoclonal antibodies modulate angiogenesis and inhibit tumor growth. Am. J. Pathol. 2010, 176, 2029–2038. [Google Scholar]
  41. Wang, Y.; Nakayama, M.; Pitulescu, M.E.; Schmidt, T.S.; Bochenek, M.L.; Sakakibara, A.; Adams, S.; Davy, A.; Deutsch, U.; Luthi, U.; et al. Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature 2010, 465, 483–486. [Google Scholar]
  42. Sawamiphak, S.; Seidel, S.; Essmann, C.L.; Wilkinson, G.A.; Pitulescu, M.E.; Acker, T.; Acker-Palmer, A. Ephrin-B2 regulates VEGFR2 function in developmental and tumour angiogenesis. Nature 2010, 465, 487–491. [Google Scholar]
  43. Gerhardt, H.; Ruhrberg, C.; Abramsson, A.; Fujisawa, H.; Shima, D.; Betsholtz, C. Neuropilin-1 is required for endothelial tip cell guidance in the developing central nervous system. Dev. Dyn. 2004, 231, 503–509. [Google Scholar]
  44. Kawasaki, T.; Kitsukawa, T.; Bekku, Y.; Matsuda, Y.; Sanbo, M.; Yagi, T.; Fujisawa, H. A requirement for neuropilin-1 in embryonic vessel formation. Development 1999, 126, 4895–4902. [Google Scholar]
  45. Takashima, S.; Kitakaze, M.; Asakura, M.; Asanuma, H.; Sanada, S.; Tashiro, F.; Niwa, H.; Miyazaki Ji, J.; Hirota, S.; Kitamura, Y.; et al. Targeting of both mouse neuropilin-1 and neuropilin-2 genes severely impairs developmental yolk sac and embryonic angiogenesis. Proc. Natl. Acad. Sci. USA 2002, 99, 3657–3662. [Google Scholar]
  46. Pan, Q.; Chanthery, Y.; Liang, W.C.; Stawicki, S.; Mak, J.; Rathore, N.; Tong, R.K.; Kowalski, J.; Yee, S.F.; Pacheco, G.; et al. Blocking neuropilin-1 function has an additive effect with anti-VEGF to inhibit tumor growth. Cancer Cell 2007, 11, 53–67. [Google Scholar] [CrossRef]
  47. Gale, N.W.; Dominguez, M.G.; Noguera, I.; Pan, L.; Hughes, V.; Valenzuela, D.M.; Murphy, A.J.; Adams, N.C.; Lin, H.C.; Holash, J.; et al. Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc. Natl. Acad. Sci. USA 2004, 101, 15949–15954. [Google Scholar]
  48. Krebs, L.T.; Shutter, J.R.; Tanigaki, K.; Honjo, T.; Stark, K.L.; Gridley, T. Haploinsufficient lethality and formation of arteriovenous malformations in Notch pathway mutants. Genes Dev. 2004, 18, 2469–2473. [Google Scholar]
  49. Duarte, A.; Hirashima, M.; Benedito, R.; Trindade, A.; Diniz, P.; Bekman, E.; Costa, L.; Henrique, D.; Rossant, J. Dosage-sensitive requirement for mouse Dll4 in artery development. Genes Dev. 2004, 18, 2474–2478. [Google Scholar]
  50. Benedito, R.; Roca, C.; Sorensen, I.; Adams, S.; Gossler, A.; Fruttiger, M.; Adams, R.H. The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis. Cell 2009, 137, 1124–1135. [Google Scholar]
  51. Suchting, S.; Freitas, C.; le Noble, F.; Benedito, R.; Breant, C.; Duarte, A.; Eichmann, A. The Notch ligand Delta-like 4 negatively regulates endothelial tip cell formation and vessel branching. Proc. Natl. Acad. Sci. USA 2007, 104, 3225–3230. [Google Scholar]
  52. Lobov, I.B.; Renard, R.A.; Papadopoulos, N.; Gale, N.W.; Thurston, G.; Yancopoulos, G.D.; Wiegand, S.J. Delta-like ligand 4 (Dll4) is induced by VEGF as a negative regulator of angiogenic sprouting. Proc. Natl. Acad. Sci. USA 2007, 104, 3219–3224. [Google Scholar]
  53. Hellstrom, M.; Phng, L.K.; Hofmann, J.J.; Wallgard, E.; Coultas, L.; Lindblom, P.; Alva, J.; Nilsson, A.K.; Karlsson, L.; Gaiano, N.; et al. Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature 2007, 445, 776–780. [Google Scholar]
  54. Noguera-Troise, I.; Daly, C.; Papadopoulos, N.J.; Coetzee, S.; Boland, P.; Gale, N.W.; Lin, H.C.; Yancopoulos, G.D.; Thurston, G. Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis. Nature 2006, 444, 1032–1037. [Google Scholar]
  55. Li, J.L.; Sainson, R.C.; Shi, W.; Leek, R.; Harrington, L.S.; Preusser, M.; Biswas, S.; Turley, H.; Heikamp, E.; Hainfellner, J.A.; et al. Delta-like 4 Notch ligand regulates tumor angiogenesis, improves tumor vascular function, and promotes tumor growth in vivo. Cancer Res. 2007, 67, 11244–11253. [Google Scholar]
  56. Scehnet, J.S.; Jiang, W.; Kumar, S.R.; Krasnoperov, V.; Trindade, A.; Benedito, R.; Djokovic, D.; Borges, C.; Ley, E.J.; Duarte, A.; et al. Inhibition of Dll4-mediated signaling induces proliferation of immature vessels and results in poor tissue perfusion. Blood 2007, 109, 4753–4760. [Google Scholar]
  57. Ridgway, J.; Zhang, G.; Wu, Y.; Stawicki, S.; Liang, W.C.; Chanthery, Y.; Kowalski, J.; Watts, R.J.; Callahan, C.; Kasman, I.; et al. Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature 2006, 444, 1083–1087. [Google Scholar]
  58. Haller, B.K.; Brave, A.; Wallgard, E.; Roswall, P.; Sunkari, V.G.; Mattson, U.; Hallengard, D.; Catrina, S.B.; Hellstrom, M.; Pietras, K. Therapeutic efficacy of a DNA vaccine targeting the endothelial tip cell antigen delta-like ligand 4 in mammary carcinoma. Oncogene 2010, 29, 4276–4286. [Google Scholar]
  59. Hrabe de Angelis, M.; McIntyre, J., 2nd; Gossler, A. Maintenance of somite borders in mice requires the Delta homologue DII1. Nature 1997, 386, 717–721. [Google Scholar] [CrossRef]
  60. Limbourg, A.; Ploom, M.; Elligsen, D.; Sorensen, I.; Ziegelhoeffer, T.; Gossler, A.; Drexler, H.; Limbourg, F.P. Notch ligand Delta-like 1 is essential for postnatal arteriogenesis. Circ. Res. 2007, 100, 363–371. [Google Scholar]
  61. Krebs, L.T.; Xue, Y.; Norton, C.R.; Shutter, J.R.; Maguire, M.; Sundberg, J.P.; Gallahan, D.; Closson, V.; Kitajewski, J.; Callahan, R.; et al. Notch signaling is essential for vascular morphogenesis in mice. Genes Dev. 2000, 14, 1343–1352. [Google Scholar]
  62. Nicolas, M.; Wolfer, A.; Raj, K.; Kummer, J.A.; Mill, P.; van Noort, M.; Hui, C.C.; Clevers, H.; Dotto, G.P.; Radtke, F. Notch1 functions as a tumor suppressor in mouse skin. Nat. Genet. 2003, 33, 416–421. [Google Scholar]
  63. Kadoya, K.; Fukushi, J.; Matsumoto, Y.; Yamaguchi, Y.; Stallcup, W.B. NG2 proteoglycan expression in mouse skin: Altered postnatal skin development in the NG2 null mouse. J. Histochem. Cytochem. 2008, 56, 295–303. [Google Scholar]
  64. Gitler, A.D.; Lu, M.M.; Epstein, J.A. PlexinD1 and semaphorin signaling are required in endothelial cells for cardiovascular development. Dev. Cell 2004, 7, 107–116. [Google Scholar]
  65. Kanda, T.; Yoshida, Y.; Izu, Y.; Nifuji, A.; Ezura, Y.; Nakashima, K.; Noda, M. PlexinD1 deficiency induces defects in axial skeletal morphogenesis. J. Cell Biochem. 2007, 101, 1329–1337. [Google Scholar]
  66. Suri, C.; Jones, P.F.; Patan, S.; Bartunkova, S.; Maisonpierre, P.C.; Davis, S.; Sato, T.N.; Yancopoulos, G.D. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell 1996, 87, 1171–1180. [Google Scholar] [CrossRef]
  67. Shim, W.S.; Teh, M.; Mack, P.O.; Ge, R. Inhibition of angiopoietin-1 expression in tumor cells by an antisense RNA approach inhibited xenograft tumor growth in immunodeficient mice. Int. J. Cancer 2001, 94, 6–15. [Google Scholar]
  68. Lin, P.; Buxton, J.A.; Acheson, A.; Radziejewski, C.; Maisonpierre, P.C.; Yancopoulos, G.D.; Channon, K.M.; Hale, L.P.; Dewhirst, M.W.; George, S.E.; et al. Antiangiogenic gene therapy targeting the endothelium-specific receptor tyrosine kinase Tie2. Proc. Natl. Acad. Sci. USA 1998, 95, 8829–8834. [Google Scholar]
  69. Sato, T.N.; Tozawa, Y.; Deutsch, U.; Wolburg-Buchholz, K.; Fujiwara, Y.; Gendron-Maguire, M.; Gridley, T.; Wolburg, H.; Risau, W.; Qin, Y. Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation. Nature 1995, 376, 70–74. [Google Scholar]
  70. Loughna, S.; Sato, T.N. A combinatorial role of angiopoietin-1 and orphan receptor TIE1 pathways in establishing vascular polarity during angiogenesis. Mol. Cell 2001, 7, 233–239. [Google Scholar]
  71. Serini, G.; Valdembri, D.; Zanivan, S.; Morterra, G.; Burkhardt, C.; Caccavari, F.; Zammataro, L.; Primo, L.; Tamagnone, L.; Logan, M.; Tessier-Lavigne, M.; et al. Class 3 semaphorins control vascular morphogenesis by inhibiting integrin function. Nature 2003, 424, 391–397. [Google Scholar]
  72. Feiner, L.; Webber, A.L.; Brown, C.B.; Lu, M.M.; Jia, L.; Feinstein, P.; Mombaerts, P.; Epstein, J.A.; Raper, J.A. Targeted disruption of semaphorin 3C leads to persistent truncus arteriosus and aortic arch interruption. Development 2001, 128, 3061–3070. [Google Scholar]
  73. Gu, C.; Yoshida, Y.; Livet, J.; Reimert, D.V.; Mann, F.; Merte, J.; Henderson, C.E.; Jessell, T.M.; Kolodkin, A.L.; Ginty, D.D. Semaphorin 3E and plexin-D1 control vascular pattern independently of neuropilins. Science 2005, 307, 265–268. [Google Scholar]
  74. Sierra, J.R.; Corso, S.; Caione, L.; Cepero, V.; Conrotto, P.; Cignetti, A.; Piacibello, W.; Kumanogoh, A.; Kikutani, H.; Comoglio, P.M.; et al. Tumor angiogenesis and progression are enhanced by Sema4D produced by tumor-associated macrophages. J. Exp. Med. 2008, 205, 1673–1685. [Google Scholar] [CrossRef]
  75. Casazza, A.; Fu, X.; Johansson, I.; Capparuccia, L.; Andersson, F.; Giustacchini, A.; Squadrito, M.L.; Venneri, M.A.; Mazzone, M.; Larsson, E.; et al. Systemic and targeted delivery of semaphorin 3A inhibits tumor angiogenesis and progression in mouse tumor models. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 741–749. [Google Scholar] [CrossRef]
  76. Lu, X.; Le Noble, F.; Yuan, L.; Jiang, Q.; de Lafarge, B.; Sugiyama, D.; Breant, C.; Claes, F.; de Smet, F.; Thomas, J.L.; et al. The netrin receptor UNC5B mediates guidance events controlling morphogenesis of the vascular system. Nature 2004, 432, 179–186. [Google Scholar]
  77. Koch, A.W.; Mathivet, T.; Larrivee, B.; Tong, R.K.; Kowalski, J.; Pibouin-Fragner, L.; Bouvree, K.; Stawicki, S.; Nicholes, K.; Rathore, N.; et al. Robo4 maintains vessel integrity and inhibits angiogenesis by interacting with UNC5B. Dev. Cell 2011, 20, 33–46. [Google Scholar] [CrossRef]
  78. Risau, W.; Flamme, I. Vasculogenesis. Annu. Rev. Cell Dev. Biol. 1995, 11, 73–91. [Google Scholar]
  79. Herbert, S.P.; Stainier, D.Y. Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat. Rev. Mol. Cell Biol. 2011, 12, 551–564. [Google Scholar]
  80. Dejana, E. Endothelial cell-cell junctions: Happy together. Nat. Rev. Mol. Cell Biol. 2004, 5, 261–270. [Google Scholar]
  81. Montero-Balaguer, M.; Swirsding, K.; Orsenigo, F.; Cotelli, F.; Mione, M.; Dejana, E. Stable vascular connections and remodeling require full expression of VE-cadherin in zebrafish embryos. PLoS One 2009, 4, e5772. [Google Scholar]
  82. Abraham, S.; Yeo, M.; Montero-Balaguer, M.; Paterson, H.; Dejana, E.; Marshall, C.J.; Mavria, G. VE-Cadherin-mediated cell-cell interaction suppresses sprouting via signaling to MLC2 phosphorylation. Curr. Biol. 2009, 19, 668–674. [Google Scholar]
  83. Claxton, S.; Fruttiger, M. Periodic delta-like 4 expression in developing retinal arteries. Gene Expr. Patterns 2004, 5, 123–127. [Google Scholar]
  84. Dorrell, M.I.; Aguilar, E.; Friedlander, M. Retinal vascular development is mediated by endothelial filopodia, a preexisting astrocytic template and specific R-cadherin adhesion. Invest. Ophthalmol. Vis. Sci. 2002, 43, 3500–3510. [Google Scholar]
  85. Eichmann, A.; Makinen, T.; Alitalo, K. Neural guidance molecules regulate vascular remodeling and vessel navigation. Genes Dev. 2005, 19, 1013–1021. [Google Scholar]
  86. Ruhrberg, C.; Gerhardt, H.; Golding, M.; Watson, R.; Ioannidou, S.; Fujisawa, H.; Betsholtz, C.; Shima, D.T. Spatially restricted patterning cues provided by heparin-binding VEGF-A control blood vessel branching morphogenesis. Genes Dev. 2002, 16, 2684–2698. [Google Scholar]
  87. Pasquale, E.B. Eph receptors and ephrins in cancer: Bidirectional signalling and beyond. Nat. Rev. Cancer 2010, 10, 165–180. [Google Scholar]
  88. Bussmann, J.; Wolfe, S.A.; Siekmann, A.F. Arterial-venous network formation during brain vascularization involves hemodynamic regulation of chemokine signaling. Development 2011, 138, 1717–1726. [Google Scholar]
  89. Strasser, G.A.; Kaminker, J.S.; Tessier-Lavigne, M. Microarray analysis of retinal endothelial tip cells identifies CXCR4 as a mediator of tip cell morphology and branching. Blood 2010, 115, 5102–5110. [Google Scholar]
  90. Arima, S.; Nishiyama, K.; Ko, T.; Arima, Y.; Hakozaki, Y.; Sugihara, K.; Koseki, H.; Uchijima, Y.; Kurihara, Y.; Kurihara, H. Angiogenic morphogenesis driven by dynamic and heterogeneous collective endothelial cell movement. Development 2011, 138, 4763–4776. [Google Scholar]
  91. Armulik, A.; Genove, G.; Betsholtz, C. Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises. Dev. Cell 2011, 21, 193–215. [Google Scholar] [CrossRef]
  92. Gaengel, K.; Genove, G.; Armulik, A.; Betsholtz, C. Endothelial-mural cell signaling in vascular development and angiogenesis. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 630–638. [Google Scholar]
  93. Jakobsson, L.; Domogatskaya, A.; Tryggvason, K.; Edgar, D.; Claesson-Welsh, L. Laminin deposition is dispensable for vasculogenesis but regulates blood vessel diameter independent of flow. FASEB J. 2008, 22, 1530–1539. [Google Scholar]
  94. Phng, L.K.; Potente, M.; Leslie, J.D.; Babbage, J.; Nyqvist, D.; Lobov, I.; Ondr, J.K.; Rao, S.; Lang, R.A.; Thurston, G.; et al. Nrarp coordinates endothelial Notch and Wnt signaling to control vessel density in angiogenesis. Dev. Cell 2009, 16, 70–82. [Google Scholar] [CrossRef]
  95. Hughes, S.; Chang-Ling, T. Roles of endothelial cell migration and apoptosis in vascular remodeling during development of the central nervous system. Microcirculation 2000, 7, 317–333. [Google Scholar]
  96. Hlushchuk, R.; Ehrbar, M.; Reichmuth, P.; Heinimann, N.; Styp-Rekowska, B.; Escher, R.; Baum, O.; Lienemann, P.; Makanya, A.; Keshet, E.; et al. Decrease in VEGF expression induces intussusceptive vascular pruning. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2836–2844. [Google Scholar] [CrossRef]
  97. Fruttiger, M. Development of the retinal vasculature. Angiogenesis 2007, 10, 77–88. [Google Scholar]
  98. Gerhardt, H.; Betsholtz, C. Endothelial-pericyte interactions in angiogenesis. Cell Tissue Res. 2003, 314, 15–23. [Google Scholar]
  99. Abramsson, A.; Berlin, O.; Papayan, H.; Paulin, D.; Shani, M.; Betsholtz, C. Analysis of mural cell recruitment to tumor vessels. Circulation 2002, 105, 112–117. [Google Scholar]
  100. Fruttiger, M. Development of the mouse retinal vasculature: Angiogenesis versus vasculogenesis. Invest. Ophthalmol. Vis. Sci. 2002, 43, 522–527. [Google Scholar]
  101. Ide, A.G.; Baker, N.H.; Warren, S.L. Vascularization of the Brown-Pearce rabbit epithelioma transplant as seen in the transparent ear chamber. Am. J. Roentgenol. 1939, 42, 891–899. [Google Scholar]
  102. Vajkoczy, P.; Farhadi, M.; Gaumann, A.; Heidenreich, R.; Erber, R.; Wunder, A.; Tonn, J.C.; Menger, M.D.; Breier, G. Microtumor growth initiates angiogenic sprouting with simultaneous expression of VEGF, VEGF receptor-2, and angiopoietin-2. J. Clin. Invest. 2002, 109, 777–785. [Google Scholar]
  103. Kienast, Y.; von Baumgarten, L.; Fuhrmann, M.; Klinkert, W.E.; Goldbrunner, R.; Herms, J.; Winkler, F. Real-time imaging reveals the single steps of brain metastasis formation. Nat. Med. 2010, 16, 116–122. [Google Scholar]
  104. Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar]
  105. Olsson, A.K.; Dimberg, A.; Kreuger, J.; Claesson-Welsh, L. VEGF receptor signalling—In control of vascular function. Nat. Rev. Mol. Cell Biol. 2006, 7, 359–371. [Google Scholar]
  106. Ferrara, N. VEGF-A: A critical regulator of blood vessel growth. Eur. Cytokine Netw. 2009, 20, 158–163. [Google Scholar]
  107. Nagy, J.A.; Chang, S.H.; Dvorak, A.M.; Dvorak, H.F. Why are tumour blood vessels abnormal and why is it important to know? Br. J. Cancer 2009, 100, 865–869. [Google Scholar] [CrossRef]
  108. Sakurai, Y.; Ohgimoto, K.; Kataoka, Y.; Yoshida, N.; Shibuya, M. Essential role of Flk-1 (VEGF receptor 2) tyrosine residue 1173 in vasculogenesis in mice. Proc. Natl. Acad. Sci. USA 2005, 102, 1076–1081. [Google Scholar]
  109. Shalaby, F.; Ho, J.; Stanford, W.L.; Fischer, K.D.; Schuh, A.C.; Schwartz, L.; Bernstein, A.; Rossant, J. A requirement for Flk1 in primitive and definitive hematopoiesis and vasculogenesis. Cell 1997, 89, 981–990. [Google Scholar]
  110. Leslie, J.D.; Ariza-McNaughton, L.; Bermange, A.L.; McAdow, R.; Johnson, S.L.; Lewis, J. Endothelial signalling by the Notch ligand Delta-like 4 restricts angiogenesis. Development 2007, 134, 839–844. [Google Scholar]
  111. Crawford, Y.; Ferrara, N. VEGF inhibition: Insights from preclinical and clinical studies. Cell Tissue Res. 2009, 335, 261–269. [Google Scholar]
  112. Goel, S.; Duda, D.G.; Xu, L.; Munn, L.L.; Boucher, Y.; Fukumura, D.; Jain, R.K. Normalization of the vasculature for treatment of cancer and other diseases. Physiol. Rev. 2011, 91, 1071–1121. [Google Scholar]
  113. Jakobsson, L.; Kreuger, J.; Holmborn, K.; Lundin, L.; Eriksson, I.; Kjellen, L.; Claesson-Welsh, L. Heparan sulfate in trans potentiates VEGFR-mediated angiogenesis. Dev. Cell 2006, 10, 625–634. [Google Scholar]
  114. Lampugnani, M.G.; Orsenigo, F.; Gagliani, M.C.; Tacchetti, C.; Dejana, E. Vascular endothelial cadherin controls VEGFR-2 internalization and signaling from intracellular compartments. J. Cell Biol. 2006, 174, 593–604. [Google Scholar]
  115. Zachary, I.C. How neuropilin-1 regulates receptor tyrosine kinase signalling: The knowns and known unknowns. Biochem. Soc. Trans. 2011, 39, 1583–1591. [Google Scholar]
  116. Harrington, L.S.; Sainson, R.C.; Williams, C.K.; Taylor, J.M.; Shi, W.; Li, J.L.; Harris, A.L. Regulation of multiple angiogenic pathways by Dll4 and Notch in human umbilical vein endothelial cells. Microvasc. Res. 2008, 75, 144–154. [Google Scholar]
  117. Kaipainen, A.; Korhonen, J.; Mustonen, T.; van Hinsbergh, V.W.; Fang, G.H.; Dumont, D.; Breitman, M.; Alitalo, K. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc. Natl. Acad. Sci. USA 1995, 92, 3566–3570. [Google Scholar]
  118. Partanen, T.A.; Alitalo, K.; Miettinen, M. Lack of lymphatic vascular specificity of vascular endothelial growth factor receptor 3 in 185 vascular tumors. Cancer 1999, 86, 2406–2412. [Google Scholar]
  119. Valtola, R.; Salven, P.; Heikkila, P.; Taipale, J.; Joensuu, H.; Rehn, M.; Pihlajaniemi, T.; Weich, H.; deWaal, R.; Alitalo, K. VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Am. J. Pathol. 1999, 154, 1381–1390. [Google Scholar]
  120. Siekmann, A.F.; Lawson, N.D. Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries. Nature 2007, 445, 781–784. [Google Scholar]
  121. Hamada, K.; Oike, Y.; Takakura, N.; Ito, Y.; Jussila, L.; Dumont, D.J.; Alitalo, K.; Suda, T. VEGF-C signaling pathways through VEGFR-2 and VEGFR-3 in vasculoangiogenesis and hematopoiesis. Blood 2000, 96, 3793–3800. [Google Scholar]
  122. Wang, H.U.; Chen, Z.F.; Anderson, D.J. Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell 1998, 93, 741–753. [Google Scholar]
  123. Suri, C.; McClain, J.; Thurston, G.; McDonald, D.M.; Zhou, H.; Oldmixon, E.H.; Sato, T.N.; Yancopoulos, G.D. Increased vascularization in mice overexpressing angiopoietin-1. Science 1998, 282, 468–471. [Google Scholar]
  124. Thurston, G.; Suri, C.; Smith, K.; McClain, J.; Sato, T.N.; Yancopoulos, G.D.; McDonald, D.M. Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1. Science 1999, 286, 2511–2514. [Google Scholar]
  125. Gamble, J.R.; Drew, J.; Trezise, L.; Underwood, A.; Parsons, M.; Kasminkas, L.; Rudge, J.; Yancopoulos, G.; Vadas, M.A. Angiopoietin-1 is an antipermeability and anti-inflammatory agent in vitro and targets cell junctions. Circ. Res. 2000, 87, 603–607. [Google Scholar]
  126. Carlson, T.R.; Feng, Y.; Maisonpierre, P.C.; Mrksich, M.; Morla, A.O. Direct cell adhesion to the angiopoietins mediated by integrins. J. Biol. Chem. 2001, 276, 26516–26525. [Google Scholar]
  127. Aplin, A.C.; Fogel, E.; Nicosia, R.F. MCP-1 promotes mural cell recruitment during angiogenesis in the aortic ring model. Angiogenesis 2010, 13, 219–226. [Google Scholar]
  128. Zhang, J.; Fukuhara, S.; Sako, K.; Takenouchi, T.; Kitani, H.; Kume, T.; Koh, G.Y.; Mochizuki, N. Angiopoietin-1/Tie2 signal augments basal Notch signal controlling vascular quiescence by inducing delta-like 4 expression through AKT-mediated activation of beta-catenin. J. Biol. Chem. 2011, 286, 8055–8066. [Google Scholar]
  129. Maisonpierre, P.C.; Suri, C.; Jones, P.F.; Bartunkova, S.; Wiegand, S.J.; Radziejewski, C.; Compton, D.; McClain, J.; Aldrich, T.H.; Papadopoulos, N.; et al. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 1997, 277, 55–60. [Google Scholar]
  130. Gale, N.W.; Thurston, G.; Hackett, S.F.; Renard, R.; Wang, Q.; McClain, J.; Martin, C.; Witte, C.; Witte, M.H.; Jackson, D.; et al. Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1. Dev. Cell 2002, 3, 411–423. [Google Scholar] [CrossRef]
  131. Holash, J.; Maisonpierre, P.C.; Compton, D.; Boland, P.; Alexander, C.R.; Zagzag, D.; Yancopoulos, G.D.; Wiegand, S.J. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 1999, 284, 1994–1998. [Google Scholar]
  132. Visconti, R.P.; Richardson, C.D.; Sato, T.N. Orchestration of angiogenesis and arteriovenous contribution by angiopoietins and vascular endothelial growth factor (VEGF). Proc. Natl. Acad. Sci. USA 2002, 99, 8219–8224. [Google Scholar]
  133. Saharinen, P.; Eklund, L.; Miettinen, J.; Wirkkala, R.; Anisimov, A.; Winderlich, M.; Nottebaum, A.; Vestweber, D.; Deutsch, U.; Koh, G.Y.; et al. Angiopoietins assemble distinct Tie2 signalling complexes in endothelial cell-cell and cell-matrix contacts. Nat. Cell Biol. 2008, 10, 527–537. [Google Scholar] [CrossRef]
  134. Fukuhara, S.; Sako, K.; Minami, T.; Noda, K.; Kim, H.Z.; Kodama, T.; Shibuya, M.; Takakura, N.; Koh, G.Y.; Mochizuki, N. Differential function of Tie2 at cell-cell contacts and cell-substratum contacts regulated by angiopoietin-1. Nat. Cell Biol. 2008, 10, 513–526. [Google Scholar]
  135. Cascone, T.; Heymach, J.V. Targeting the angiopoietin/tie2 pathway: Cutting tumor vessels with a double-edged sword? J. Clin. Oncol. 2012, 30, 441–444. [Google Scholar] [CrossRef]
  136. Egeblad, M.; Nakasone, E.S.; Werb, Z. Tumors as organs: Complex tissues that interface with the entire organism. Dev. Cell 2010, 18, 884–901. [Google Scholar]
  137. Vakoc, B.J.; Lanning, R.M.; Tyrrell, J.A.; Padera, T.P.; Bartlett, L.A.; Stylianopoulos, T.; Munn, L.L.; Tearney, G.J.; Fukumura, D.; Jain, R.K.; et al. Three-dimensional microscopy of the tumor microenvironment in vivo using optical frequency domain imaging. Nat. Med. 2009, 15, 1219–1223. [Google Scholar]
  138. Pittet, M.J.; Weissleder, R. Intravital imaging. Cell 2011, 147, 983–991. [Google Scholar]
  139. Lohela, M.; Werb, Z. Intravital imaging of stromal cell dynamics in tumors. Curr. Opin. Genet. Dev. 2010, 20, 72–78. [Google Scholar]

Share and Cite

MDPI and ACS Style

Jin, Y.; Jakobsson, L. The Dynamics of Developmental and Tumor Angiogenesis—A Comparison. Cancers 2012, 4, 400-419. https://doi.org/10.3390/cancers4020400

AMA Style

Jin Y, Jakobsson L. The Dynamics of Developmental and Tumor Angiogenesis—A Comparison. Cancers. 2012; 4(2):400-419. https://doi.org/10.3390/cancers4020400

Chicago/Turabian Style

Jin, Yi, and Lars Jakobsson. 2012. "The Dynamics of Developmental and Tumor Angiogenesis—A Comparison" Cancers 4, no. 2: 400-419. https://doi.org/10.3390/cancers4020400

Article Metrics

Back to TopTop