·Review·  ·Current Issue·  ·Achieve·  ·Search Articles·  ·Online Submission·  ·About IJO·

 

Molecular underpinnings of corneal angiogenesis: advances over the past decade

 

Nizar Saleh Abdelfattah1, Mohamed Amgad2, Amira A. Zayed3, Heba Hussein4, Nawal Abd El-Baky5

 

1Doheny Image Reading Center, Doheny Eye Institute, Los Angeles, CA 90033, USA

2Faculty of Medicine, Cairo University, Cairo 11111, Egypt

3Department of Oncology, Mayo Clinic, Rochester, Minnesota 55904, USA

4Faculty of Oral and Dental Medicine, Cairo University, Cairo 11111, Egypt

5Antibody Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, Alexandria 22033, Egypt

Correspondence to: Nizar Saleh Abdelfattah. Doheny Image Reading Center, Doheny Eye Institute, 1355 San Pablo Street, Suite 100, Los Angeles, CA 90033, USA. nizar@ucla.edu  

Received: 2015-04-30             Accepted: 2016-01-19

 

Abstract

The cornea is maintained in an avascular state by maintaining an environment whereby anti-angiogenic factors take the upper hand over factors promoting angiogenesis. Many of the common pathologies affecting the cornea involve the disruption of such equilibrium and the shift towards new vessel formation, leading to corneal opacity and eventually-vision loss. Therefore it is of paramount importance that the molecular underpinnings of corneal neovascularization (CNV) be clearly understood, in order to develop better targeted treatments. This article is a review of the literature on the recent discoveries regarding pro-angiogenic factors of the cornea (such as vascular endothelial growth factors, fibroblast growth factor and matrix metalloproteinases) and anti-angiogenic factors of the cornea (such as endostatins and neostatins). Further, we review the molecular underpinnings of lymphangiogenesis, a process now known to be almost separate from (yet related to) hemangiogenesis.

KEYWORDS: cornea; neovascularization; angiogenesis; lymphangiogenesis; vascular endothelial growth factor

DOI:10.18240/ijo.2016.05.24

 

Citation: Abdelfattah NS, Amgad M, Zayed AA, Hussein H, Abd El-Baky N. Molecular underpinnings of corneal angiogenesis: advances over the past decade. Int J Ophthalmol 2016;9(5):768-779

 

Introduction

Corneal avascularity comprises its main functionality index as a lens. This article reviews the literature on corneal neovascularization (CNV) promoted by proteins such as vascular endothelial growth factors, fibroblast growth factor, and matrix metalloproteinases, and subsequently inhibited by endostatins, angiostatins, and related anti-angiogenic factors. It also reviews the normal versus pathogenic changes in corneal immunity leading to new vascular formation.

CNV is induced by various stimuli mainly associated with inflammation, trauma, transplantation, and infection of the ocular surface[1-2]. Both corneal hemangiogenesis and lymphangiogenesis are promoted or inhibited by a balance of mediators, including the dynamics between pro-angiogenic and anti-angiogenic substances. In corneas diseased by inflammation, infection, degeneration, transplantation, or trauma, the normal balance is shifted towards the pro-angiogenic status, leading to corneal hemangiogenesis and/or lymphangiogenesis.

Major Angiogenic Proteins of the Cornea

Vascular Endothelial Growth Factor (VEGF) VEGF-A is known to be linked to blood vessel formation in a wide range of events; including embryonic and physiologic growth, vascular pathologies and malignant tumor neovascularization. It acts directly on blood vessels by stimulating endothelial cell mitosis, migration, dissolution of original vessel membrane, and formation of new capillary tubes [3].

Following the discovery of VEGF, a series of subtypes were identified and named in alphabetical order: VEGF-A, VEGF-B, VEGF-C, and VEGF-D. Nevertheless, the chief VEGF was and still is VEGF-A. A wide range of heterogeneous cells were proven to secrete VEGF-A, including macrophages, pericytes, T-cells, astrocytes, fibroblasts, and retinal pigment epithelial cells [3]. 

VEGF has multiple isoforms, with its encoding gene comprised of eight exons. With differential pre-mRNA splicing, a single VEGF gene gives these different isoforms. Examples of these isoforms include VEGF121, VEGF165 and VEGF189. These numbers refer to number of amino acids composing the protein. These isoforms have different properties based on the presence of absence of the C-terminal protein domains encoded by exons 6 and 7. Also, these isoforms constitute a reservoir of growth factors acting without gene transcription. Moreover, interaction of VEGF165 isoform with heparan sulfate proteoglycan-Glypican-1 had been reported to play a role in extending the half-lives of the isoform in the process of hypoxia induced angiogenesis [4].

Alternative splicing of VEGF gene yields five isoforms of VEGF-A, including (VEGF115, VEGF 121, VEGF 165, VEGF 189, and VEGF 206) [5]. The shorter isoforms have more distinctive functions like the mitogenic activity of VEGF121 and VEGF165, and the more powerful angiogenic activity of VEGF121 than other longer isoforms [6]. VEGF proved its importance based on the inhibition of neovascularization in rat model following stromal implantation of an anti-VEGF-A blocking antibody [7].

VEGF binds to different surface receptor proteins (VEGFR). VEGFR-1 is a transmembrane receptor tyrosine kinase while VEGFR-2 is a major signaling receptor for VEGF. Additionally, heparan sulfate proteoglycan (HSPG) is low-affinity class of VEGF receptors that modulate the activities of wide range of heparin-binding growth factors, morphogens and chemokines [4].

Basic Fibroblast Growth Factor  Among the 23 heparin binding peptides fibroblast growth factor (FGF) family, basic fibroblast growth factor (bFGF) is a member that is hugely expressed in developing and adult tissues during cellular differentiation, angiogenesis, mitogenesis and wound repair. Moreover, it is upregulated after tissue injury and in stromal fibroblast/vascular endothelial cell co-cultures. FGFs mediate their action through interaction with peptide receptors (fibroblast growth factor receptors, FGFR), namely FGFR-1, -2, -3, and -4. Potential FGF- mediated intracellular signaling events are possibly present, also different FGFR isoforms show unique biological functions [8].

Besides, the diversity of FGFR biological response is manifested by its tissue-specific expression, which is regulated by differences in ligand function and specificity. The growth factor receptors being regulated are of great significance in the management of complex physiological processes [9-11].

FGFs share in a diverse set of actions modulated FGF-Receptor isoforms. FGF-1 is found in normal corneal epithelium, and differs from FGF-2 in that it is upregulated in injured cornea and in co-cultures of keratocytes and vascular endothelia. On the one hand, bFGF binds to Bowman’s membrane and Descement’s membrane in healthy corneas, yet on the other hand, it binds to vascular basement membranes of neovascularized corneas [12].

In fact, it is believed that bone marrow acts as a store of bFGF and VEGF, sequestering them in order to balance anti-angiogenesis [13]. Besides, the maturation of new blood vessels and the level of FGF binding are related.

Newly formed corneal vessels show similar binding capacities in comparison to normal limbal vessels. This has been linked to heparin sulfate proteoglycans and thus stresses on the role of extracellular matrix (ECM) components in the regulation of corneal angiogenesis [14].

The levels of FGF-1, 2, 3, 7, and 22 are found to be increased in alkali wounding model at 7 and 14d post-wounding [2]. It is thought that bFGF function in corneal angiogenesis is mediated through its effect on VEGF-A, -C and -D production. bFGF promotes angiogenesis greatly through this action.

Angiogenic Interaction Between Vascular Endothelial Growth Factor and Basic Fibroblast Growth Factor From the increased researches in the past decade on characterization of interaction between multiple membrane-bound receptors, a new hypothesis emerged. It stated that, membrane-anchored receptors associate and coordinate with each other to cooperatively induce an array of intracellular signaling cascades, instead of transmitting signals across the membrane individually. Instead of working individually it was observed that there is an interplay between FGF and VEGF signaling for the maintenance of endothelial junctions and vascular integrity during angiogenesis [15-16].

Also, a recent study about mustard intoxicated subjects who developed CNV found a significantly increased levels of growth factors, specifically VEGF-A165, bFGF and platelet derived growth factor-BB (PDGF-BB) [17].

FGF-VEGF signaling balance is assumed to lie at the center of the regulation of permeability and angiogenesis. They are both important angiogenic growth factors. As soon as VEGF activates VEGFR-2 (a.k.a. FLK-1/KDR), the receptor undergoes auto-phosphorylation on specific tyrosine residues, followed by the addition of Tyr(P) residues on signaling proteins and adapter that contain the Src homology domain 2 (SH2)[18].

As a result the receptor complexes and adapter activate multiple intracellular pathways through some effectors like focal adhesion kinase (FAK) and mitogen-activated protein kinases (MAPK) [19-20].

Flk-1/KDR can also trigger other cascades including PI3K-dependent AKT/PKB and phospholipase C-g (PLC-g)[21-22]. Flk-1/KDR-mediated intracellular signaling seems to be similar to bFGF signaling pathway; however, various lines of evidence suggest that bFGF-induced angiogenesis is independent of Src kinase activity unlike VEGF signaling [23].

In spite of the extensive research on bFGF- and VEGF induced angiogenesis, the complete intracellular signaling pathways that respond to each of them to induce angiogenesis are not fully understood. Specifically, how these pathways interact with molecular regulators is not well documented. It is suggested recently that membrane-type 1 metalloproteinase (MT1-MMP) may be one of many factors involved in connecting the two pathways of VEGF and FGF signaling [2]. It has been showed that MT1-MMP increased bFGF- induced VEGF upregulation and CNV in mice synergistically [24].

Besides, MT1-MMP raises bFGF-induced VEGF upregulation in enzymatically inactive MT1-MMP corneal stromal fibroblasts; which suggest that linking the VEGF and FGF signaling pathways may be in part due to MT1-MMP enzymatic activity.

Minor Angiogenic Proteins of the Cornea

Decorins  Decorins are members of the small leucine-rich proteoglycan (SLRP) family, which in turns belongs to a family of differently functioning molecules that are involved in the regulation of collagen fibrillogenesis, direct modulation of cell behavior and binding and inactivation of cytokines. Decorins consists of a protein core containing leucine repeats with a glycoseaminoglycan (GAG) chain of either dermatan sulfate or chondoroitin sulfate [25-27].

It has been demonstrated that decorins may regulate corneal angiogenesis [2,28]. Their effects on corneal angiogenesis in mice have been heavily studied, as well as the effects of biglycan and fibromodulin [29]. Using chemical cauterization, it was verified that in decorin-deficient mice (unlike biglycan and fibromodulin-deficient corneas), the growth of corneal vessels is significantly diminished compared to wild type (WT).

Recently, it was observed that bFGF induce MT1-MMP expression but diminish decorins expression [30]. Furthermore, it was demonstrated that MT1-MMP cleaves decorins in vitro, and that cell lysates from MT1-MMP-deficient keratocytes do not show decorins processing activity.

Ephrins and Eph Receptors  One of the largest known families of receptor tyrosine kinase (RTK) is the Eph/ephrin complex. It consists of 14 receptors and 8 ligands, and its family members are subcategorized to class A and class B depending on their structure and ligand binding receptor characteristics [31-32].

In several vascular endothelial cells, EphB1-B4 and ephrinB1 and B2 were found to be expressed [33-34]. In adult mice it was demonstrated that EphB1 and ephrinB2 induce corneal angiogenesis [35], and that ephrinB1 induces vascular endothelial cell migration, assembly, and adhesion [36].

Recently, immunohistochemical studies were used to demonstrate that ephrinB1 and EphB1 are expressed in bbFGF-induced vascular corneas [2], which proves that Eph and ephrin receptors play a role in corneal angiogenesis [37].

EphrinB1 is expressed in corneal-resident keratocytes and neutrophils. In order to test Eph and ephrin receptors’ role in angiogenesis, recombinant ephrinB1-Fc (which induces EphB receptor activation) was used. It was found to promote bFGF-induced tube formation in an in vitro aortic ring assay; as well as corneal angiogenesis in-vivo in a corneal pocket assay. These results suggest that ephrinB1 plays a synergistic role in CNV [2]. Ellenberg et al[2] also compared ephrinA/EphA expression to ephrinB/EphB expression in vascularized corneas. bFGF pellets were implanted to induce CNV. The eyes of WT, ephrinB2 tlacZ/+, and EphB4 tlacZ/+ heterozygous mice were harvested and sectioned 7d after pellet implantation. Confocal immunohistochemistry was performed to compare the expression of the Eph/ephrinA family and Eph/ephrinB family. EphA1, EphA3, ephrinA1, ephrinA2, EphB1, EphB4, ephrinB1, and ephrinB2 were detected in WT mouse corneal epithelial cells and keratocytes.

Using immunohistochemistry it was found that EphA2 was only located in the epithelial cells, while EphA3, ephrinA1, EphB1, EphB4, and ephrinB1 were localized in corneal epithelium and stroma. However, in neovascularized corneas; ephrinB1 was mainly localized to keratocytes around the vessels, and ephrinB2, EphB1, and EphB4 were mainly located simultaneously with CD31 in the vascular endothelial cells. These studies strengthen the suggestion that Eph/ephrin family of receptor tyrosine kinases and their ligands may play a role in the regulation of corneal angiogenesis [2].

Activin Receptor-like Kinase  Activin receptor-like kinase-1 (ALK-1) is one of the seven type I receptors recognizing transforming growth factor beta (TGF-β) family proteins [38]. It has been suggested that ALK-1 plays a role in the maturation phase of angiogenesis [39]. The transfection of a constitutively active form of ALK-1 inhibit not only, endothelial cell proliferation at the G1 phase of the cell cycle, but also endothelial cell migration through a modification of the dynamics of endothelial cell cytoskeleton [40].

Supporting these results is a zebrafish ALK-1 mutant, vgb, whose vessel dilation phenotype is reminiscent of ALK-1-/- mice. Its affected vessels showed an increased number of endothelial cells, supporting a role for ALK-1 in the inhibition of endothelial cell proliferation.

It have been demonstrated using the pellet induced CNV model that over expression of ALK-1 (using naked DNA injection) in mouse cornea does not induce CNV [2]. Besides, it can prevent growth of new bFGF-induced stromal vessels. All of that strengthen the possibility that ALK-1 plays an important role in angiogenesis.

Recently, Ellenberg et al [2] have described a proteomic approach to investigate the differential protein expression patterns and identify the physiologically relevant angiogenic and anti-angiogenic factors involved in the hyaloid vascular system regression. Differentially expressed proteins were identified using two-dimensional gel electrophoresis from the lens and vitreous of P1 and P16 mice followed by nanoflow chromatography coupled with tandem mass spectrometry [41]. Using this approach, the following factors expressed at P16 may be involved in angiogenesis: tumor necrosis factor-α (TNF-α), hepatoma-derived growth factor (HDGF), FGF-22, and kininogen. 

Integrins A major family of type I transmembrane cell surface receptors are the integrins. Currently, 18 individual α subunits and 8 β subunits have been identified. Integrins are heterodimers composed of one α and one β subunit [42-43].

It has been noticed that a significant upregulation of αvβ3 and α5β1 takes place on activated vascular endothelium during angiogenesis. It is suggested that α5 integrins play a key role during the development of the vascular system [44-45]. Testing that hypothesis with genetic ablation of integrin α5 leads to severe vascular abnormalities. α5β1 integrin as well as its extracellular ligand fibronectin which is able to provide proliferative signals to vascular cells both are upregulated in tumor new blood vessels and plays a role in tumor angiogenesis and growth. Besides, angiogenesis have been inhibited in-vivo and in-vitro using integrin αvβ3 and αvβ5 antagonists.

On the other hand, treating animals systemically with an α5β1-inhibiting small molecule showed significant inhibition of CNV. Combining them to integrin αv and α5 does not promote the anti-lymphangiogenic effect in vivo [46-47].

Matrix Metalloproteinases  Corneal extracellular matrix (ECM) remodeling by matrix metalloproteinases (MMPs) has also been implicated in corneal angiogenesis and in the maintenance of corneal avascularity. MMPs are described as a group of proteolytic enzymes that are zinc-binders, and participants in ECM remodeling, neovascularization, and lymphangiogenesis.

MMPs is a large family that involve 25 enzymes described so far, not less than 15 of which have been identified in the cornea; (MMP-1, -8 and -13) represent the collagenases,  MMP-2 and -9 represent the gelatinases A and B, MMP-3, -10 and -11 represent the stromelysins, MMP-7 is the matrilysin and MMP-12 is the macrophage metalloelastase while MMP-14, -15, -17, -24 and -25 all represent the membrane type of MMPs[48-52].

After several studies it became clear that MMP-mediated proteolysis induce several important biological functions including: 1) changing structural matrix proteins into signaling molecules (e.g. collagen XVIII that is present in the cornea and having an NC1 domain which is anti-angiogenic); 2) changing the structure of matrix proteins like cleaving perlecan and decorin-corneal ECM proteoglycans; 3) changing the architecture of the tissue (e.g. cleaving E-cadherin); 4) changes in chemotaxis; 5) inducing proliferation like its action through epidermal growth factor receptor ligand processing; 6) ensuring the cell survival (e.g. neuronal survival factor); 7) activating some latent signaling molecules (e.g. TNF-a shedding and IGF binding protein cleavage); 8) changing the range of signaling molecule action (e.g. changing the range of VEGF diffusion); 9) causing tissue differentiation (e.g. adipose tissue maturation)[52-55].

Increased expression of MMPs in corneas during angiogenesis has already been demonstrated [56-57]. However it is still vague what is their definitive role in regulation of angiogenesis because they can act as pro- and anti- angiogenic factors at the same time, which might be explained by their ability to degrade the ECM, allowing tissue invasion by endothelial cells bearing MMP, and to generate anti-angiogenic fragments from their precursors [52,58-59].

In the following sections, additional information on the roles of MMP-2, MMP-7, and MT1-MMP in corneal angiogenesis are highlighted.

Matrix Metalloproteinase-2-Gelatinase A  Gelatinase-A (MMP-2) has always been linked to angiogenesis. It was demonstrated that it is pro-angiogenic through facilitating vascular invasion by direct matrix degradation or through releasing matrix bound cytokines or growth factors [52,60-61].

MMP-2 expression by epithelial cells and stromal keratocytes has been confirmed by in situ hybridization [49]. Besides, its physiologic role in angiogenesis has been defined: when MMP-2 deficient mice were used to determine the role of MMP-2 in vascular endothelial cell migration and tube formation in vitro using aortic rings, it was demonstrated that bFGF mediated angiogenic response was diminished in mice lacking the functional MMP-2 gene compared to WT animals [50].

On the other hand, endothelial cells from MMP-2 lacking mice failed to display normal outgrowth after adding 5 ng/mL bFGF, which lead to the suggestion that the difference in bFGF-induced angiogenesis between MMP-2 lacking mice and WT mice may be due to the difference of vascular endothelial cells; as it could be inconvenient for endothelial cells lacking functional MMP-2 to traverse the basement membrane [2].

The MMP-2-null mice developed almost normally, and bFGF induced corneal angiogenesis even in the MMP-2-mutant mice, clearly indicating that the angiogenic process is not totally dependent on MMP-2. In another experiment, MT1-MMP null mice showed complete absence of corneal angiogenesis which lead to the suggestion that MT1-MMP by itself has an essential role in the process of angiogenesis [51]. Based on these data, further research is needed to explain the discrepancy between MMP-2 and MT1-MMP effect on angiogenesis.

Studies show that through intramolecular processing, MMPs can modulate the bioavailability of VEGF; a group of MMPs can cleave the matrix-bound isoforms of VEGF releasing soluble fragments to promote capillary dilatation of existent vessels [59,62-63].

In recent researches, MMP-2 could cleave connective tissue growth factor (CTGF) and heparin affin regulatory peptide (HARP) and inactivate them upon proteolysis. As these two are angiogenic and mitogenic cytokine inhibitors in complex with VEGF, cleaving those releases the VEGF. As a result, MMP-2 possesses potential pro-angiogenic activity by releasing intact VEGF from HARP or CTGF cytokine inhibitory complexes [62].

Matrix Metalloproteinase-7 (Matrilysin)  Matrilysin also called MMP-7 is expressed in basal epithelial cells in the migration and proliferation phases of corneal wound healing after excimer keratectomy [48-49]. Matrilysin has catalytic action against a wide range of ECM substrates e.g. gelatins (I, III, IV, and V), fibronectin, elastin, collagen IV, laminin and entactin-nidogen [64]. At the same time, it can cleave factors which modulate angiogenesis like CTGF, sVEGFR-1, plasminogen and collagen XVIII. It has been positively stained in basal epithelium of pterygium specimen suggesting its involvement in pathogenesis and angiogenesis in pterygium [65].

The anti-angiogenic role for MMP-7 in cornea is based on the fact that MMP-7 cleavage of corneal collagen XVIII yields a 28-kDa fragment which contains the endostatin domain of collagen XVIII that shows potent anti-angiogenic function [58]. Also in MMP-7 knock-out (KO) mice, in the keratectomy wounding model, a decrease in the levels of anti-angiogenic factors tilts the balance towards corneal angiogenesis [2].

Recent researches show that the induction of new vessel formation in diseased corneas involves not only upregulation and activation of angiogenic factors such as VEGF and bFGF but also suppression of anti-angiogenic factors.

Based on the observation that MMP-7 cleaves plasminogen and collagen XVIII in vitro to generate anti- angiogenic factors e.g. angiostatin and endostatin respectively suggest that the reduction of MMP-7 derived endostatin and/or angiostatin in the cornea may contribute to CNV after excimer keratectomy in MMP-7 KO animals [2].

Membrane-type 1 Metalloproteinase  MT1-MMP is the most important MMP in angiogenesis. MT1-MMP expression in the cornea has been detected in the epithelium and stromal keratocytes during wound healing [49]. Its importance becomes evident in that it is the only MMP that its absence is lethal demonstrated by genetic KO of MT1-MMP in mice lead to death within three to four weeks, unlike other MMPs who’s genetic KO might affect angiogenesis but is never lethal to the animal.

MT1-MMP proved to be important for angiogenesis and its absence causes delayed vascular development and impaired CNV by bFGF [51]. Further studies have been carried out to understand its role using genetic KO mice, antibodies against MT1-MMP, animal models and other methods[2]. It was noticed that there is an enhanced MT1-MMP expression in alkali wounded CNV. In addition using reverse transcription-polymerase chain reaction (RT-PCR) to evaluate the expression of growth factor receptors in WT, MT1-MMP KO, and MT1-MMP (knochin) KI mouse cornea stromal fibroblasts, it was found that no significant difference existed in the expression patterns of PDGFa, PDGFb, and VEGFR-1 in either type of cells.

On the other hand, endothelial growth factor receptor (EGFR) expression was decreased in MT1-MMP KO cells when compared to the WT and MT1-MMP KI cells, suggesting that MT1-MMP play a role in EGFR expression regulation. Since EGFR is also fibroblast proliferation and migration regulator, it may be responsible for some of MT1-MMP pro-angiogenic effects [66].

Angiogenic Interaction Between Membrane-type 1 Metalloproteinase and Vascular Endothelial Growth Factor/ Fibroblast Growth Factor  There have been new studies that elaborated the signal pathways involved in the interaction between these molecules and their role in CNV[67]. It was suggested that MT1-MMP may link the two signaling pathways of VEGF and FGF[24], but its specific role in linking them remains vague. The pro-angiogenic role of MT1-MMP has been reported to be in part mediated through the upregulation of both VEGF transcription and translation[68].

Immuno-histochemistry and RT-PCR analysis of human glioma tissue samples proved a functional link in tumor angiogenesis between MT1-MMP and VEGF by giving evidence to the link between their expressions[69].

Further evidence is the correlation between VEGF stimulation and hypoxia-induced upregulation of MT1-MMP in the murine bone marrow -derived stromal cells. Putting all these data together gives a strong evidence of linkage between signaling pathways of MT1-MMP and VEGF, which may play a role in regulating corneal angiogenesis[59]. The FGF and MT1-MMP interaction has been well documented: FGF-1 induction of MT1-MMP transcription in LNCaP prostate carcinoma cells has been reported. Besides, FGFR-1 and STAT3 involvement in FGF-1 mediated MT1-MMP expression has also been reported[70]. bFGF induced CNV increased when bFGF pellets have been used in combination with naked MT1-MMP DNA plasmid injection[2]. The interplay between MT1-MMP, VEGF, and bFGF has been demonstrated by experiments in which VEGF and MT1-MMP expression increased after implantation of bFGF- pellets in murine cornea.

Angiogenesis Inhibitors

Corneal angiogenesis privilege (CAP) is shown to be secondary to the interaction of multiple anti-angiogenic factors. Most notable of which are angiostatin, angiostatin-like fragments[71], and endostatin[72]. Other potent anti-angiogenic factors that modulate CAP include restin, arresten, canstatin, tumstatin, and pigment epithelial-derived factor (PEDF)[73].

They can be generally classified into, endostatin/endostatin analogues, and plasminogen/serine protease inhibitors.

Endostatin/Endostatin Analogues

Endostatin  Endostatin has been shown to inhibit in-vitro VEGF mediated endothelial migration and proliferation[74], as well as decrease tumor progression in in-vivo murine models. Moreover, endostatin has been successfully administered in corneal assays, with significant reduction in bFGF mediated angiogenesis[75].

The mechanism of action of Endostatin is relatively complex. It exerts its actions through primarily associating with tropomyosins, integrins, VEGF receptors, MMPs, and glypicans. Its action on VEGF is in the form of; Blockage of VEGF cell surface receptor KDR/FLK1, downstream inactivation of ERK, MAPK, and P125FAK, Ultimately, arrest of cell cycles in G1, with inhibition of mitogenic responses in vascular endothelial cells[76].

In addition to VEGF antagonism, endostatin enhances vascular endothelial apoptosis via increasing the activity caspase 3 [77].

Alongside angiogenesis inhibition, endostatin has been shown to affect lymphogenesis as well. Recombinant endostatin was shown to inhibit the proliferation and migration of lymphatic endothelial cells, in vitro[78]. One possible mechanism for this activity is the ability of endostatin to inhibit distribution of VEGF-C-producing tumor-associated inflammatory cells and to induce the apoptosis of VEGFR-3 expressing cells [79].

Neostatin  There are two major types of neostatins, both of which have been proven as potent angiogenesis inhibitors. Neostatin-7 (formed via MMP-7's cleavage of collagen XVIII), and neostatin-14 (formed via MT1-MMP-mediated cleavage of collagen XVIII) [79].

Both MMP-7 and MT1-MMP are expressed by corneal epithelial cells [48], where collagen XVIII is actively secreted as well[59]. Moreover, it was demonstrated that recombinant neostatin-7 blocks bFGF-induced corneal angiogenesis and lymphangiogenesis [80]. This strongly shows the important role of corneal epithelium in maintaining CAP.

Other Miscellaneous Molecules  Arresten, canstatin, and tumstatin are three type IV collagen-derived proteins that were shown to have potent anti-angiogenic activity[81-84].

Arresten actions are mediated via α1β1 integrin receptors. It successfully inhibits bFGF-induced proliferation, migration, and tube formation of cultured endothelial cells[82,85-86].

Another molecule, canstatin, acting via α3β1, αɣβ3, and αɣβ5 integrin receptors, has diverse functions. It causes suppression of tumor growth, inhibition of endothelial cell proliferation and migration, and induction of endothelial cell apoptosis [87-89].

Also, tumstatin which is a 28 kDa protein derived from type IV collagen α3 chain, exerts its action via αɣβ3 and α6β1 integrins receptors. It shares in suppression of tumor growth, inhibition of endothelial cell proliferation and migration, induction of endothelial cell apoptosis, and inhibition of protein synthesis [84,90-91].

Tumstatin can inhibit protein synthesis through the inhibition of phosphorylation of FAK, induced in endothelial cells via attachment to vitronectin, and by inhibiting the activation of PI3-kinase through αɣβ3 binding [84,92].

Plasminogen-derived and Serine Protease Inhibitors of Angiogenesis

Angiostatin  Angiostatin is a complex molecule that can inhibit primary and secondary tumor growth. One of the enzymes responsible for the generation of angiostatin in Lewis lung carcinoma has been identified as a macrophage derived metalloelastase (MMP-12)[93]. However, human matrilysin (MMP-7) and neutrophil gelatinase B (MMP-9) can also convert plasminogen to angiostatin fragments [94].

The suppressive action of recombinant angiostatin on in-vivo tumor growth and metastasis in animal models has also been demonstrated[95]. Moreover, angiostatin was proven to be a non-toxic inhibitor of neovascularization when injected to tumor-bearing mice[96].

One mechanism that explains these actions is down-regulation of endothelial cells migration and proliferation, through binding to ATP synthase and decreasing endothelial cell ATP production[97]. Another way is induction of endothelial cell apoptosis and arrest at the G2 to M transition phase [98].

Another possible mechanism for angiostatin is its binding to integrin αvβ3; thereby inhibiting its actions. Typically plasmin binds to αvβ3 through its kringle domains, promoting endothelial cell migration. This process can be disrupted through anti-integrin αvβ3 agents (i.e. angiostatin) and a serine protease inhibitor [99].

Angiostatin and angiostatin-like molecules play an important role in maintaining corneal avascualrity after injury, and it was shown that their expression in corneal epithelium increases remarkably at these events[100]. Moreover, corneal segmental neovascularization was demonstrated after excimer laser keratectomy followed by treatment with anti-angiostatin (anti-LBS or anti- K1e3) antibody injection, further affirming the role of angiostatin [2].

Pigment Epithelium Derived Factor  A member of the serine protease family, PEDF is a potent anti-angiogenic factor, expressed within endothelial and corneal epithelial cells[101]. It typically works through binding to surface receptors, such as glycosaminoglycans and collagen I[102].

Furthermore, the recombinant PEDF inhibited CNV as well[103]. These findings point towards an essential function of PEDF in maintaining avascular environment of the cornea. Other works established similar roles for PEDF in the vitreous, aqueous humor, and retina[2].

Given its effectiveness against multiple inducers of angiogenesis, as VEGF and interleukin-8 (IL-8), and its multiple sites of action, PEDF derivatives can prove highly effective in reversing pathological ocular angiogenesis processes.

Molecular underpinnings of lymphangiogenesis

The process of lymphangiogenesis was thought to have the same molecular underpinnings as hemangiogenesis for a long time. It is only relatively recently that lymphangiogenesis started to have a separate entity, especially after the discovery of the key lyphatic endothelial cells (LEC) marker, lymphatic vessel endothelial hyaluronan receptor (LYVE-1) [101]. The formation of lymphatics can be summarized in the following steps [104]:       

1) Endothelial cells differentiate from angioblasts into venous endothelial cells and arterial endothelial cells.

2) Venous endothelial cells highly express VEGFR-3 and a subset of them begins to express LYVE-1 as well. This subset represents the precursors of LEC's [105]. Bone marrow-derived cells, including macrophages, may also transdifferentiate into endothelial cells [106].

3) LEC precursors begin to express the transcription factor SOX18, which is present upstream of another transcription factor, Prox-1 [107]. SOX 18 induces the expression of Prox-a and triggers a set of incompletely-understood events that eventually determine the differentiated fate of LEC's [108]. In fact, Prox-1 is known to be expressed in a polarized manner in differentiating LEC's and is believed to be a "master switch" in LEC differentiation.

4) LEC precursors express Neuropilin-2 (NP-2)[109]. NP-2, while not triggering downstream signaling itself, sensitizes the LEC to VEGF-C stimulation and acts synergistically with VEGFR-3[109].

5) At this stage, the proliferating lymphatics begin to form lateral extensions from the veinules, known as "lymphatic sacs" [107].                                                                                

6) LEC's start expressing the transmembrane protein, podoplanin[110-111]. In turn, podoplanin binds to CLEC-2 receptors on platelets and activates SLP76 and Syk, leading to platelet aggregation [112].

7) The aggregated platelets block the connection between the developing lymphatic and the veinule from which it budded, eventually leading to physical separation of the two entities.

8) FoxC2 and NFACT-1 (nuclear factor of activated T cells), both of which are transcription factors downstream of VEGFR-3, cooperate in controlling genes that are important in further differentiation of the lymphatic tree, including lymphatic valves [113].

TGF-b and TNF-a both act to inhibit lymphangiogenesis [114-115]. On the other hand, VEGF-A, VEGF-C, FGF-2, IGF-1, IL-1B, HGF and PDGF have all been shown to simulate both hemangiogenesis and lymphangiogenesis (as has been discussed in detail earlier). It is noteworthy that the growth factors and cytokines that stimulate hemangiogenesis and lymphangiogenesis greatly overlap. Nonetheless, VEGF-C/VEGFR-3 signalling seems to be more specific to lymphangiogenesis and VEGF-3 ceases being expressed in adult blood vessels, with the exception of fenestrated vessels of endocrine glands [116-117]. Another factor that differentiates lymphangiogenesis from hemangiogenesis is the role of a5b1 integrins in lymphangiogenesis. An a5b1 antagonist results in a hemangiogenesis-dominant response to CNV induction [118]. Table 1 highlights some of the differences between hemangiogenesis and lymphangiogenesis [106,116-134].

Table 1 Differences between hemangiogenesis and lymphangiogenesis

Parameters

Hemangiogenesis

Lymphangiogenesis

Corneal privilege mechanism

Upregulation of anti-angiogenic factors and downregulation of angiogenic factors

Soluble VEGFR-1 and ectopic VEGFR-3 act as "VEGF traps" to prevent hemangiogenesis[117]

Soluble VEGFR-2 and VEGFR-3 act as "VEGF trap" and selectively suppresses physiologic growth of lymphatics[117,121]

Formation

Sprouting or budding from post-capillary veinules

LEC's arise from either:

Bone marrow-derived cells, such as transdifferentiated macrophages[106]

Primitive veinules and local lymphangioblasts (described in detail in the text)

Role of macrophages

Providing a temporary scaffold for the new vessels[122]

Provide paracrine support for vascular networks[123]

Interact physically with the blood vessels[124]

Transdifferentiate into endothelial cells, thus participate -structurally- in lymphatic vessels[122]

Secrete paracrine factors, most importantly VEGF-A[128]

Act as "guide cells" that guide tip cells into finding and anastomosing with tip cells from other sprouting lymphatics[125]

Expression of VEGFR-3

Expressed during early stages of endothelial development. Fully developed blood vessels seldom contain VEGFR-3, with the exception of fenestrated blood vessel of endocrine glands[116]

Highly expressed by LEC's

Role of VEGF-C and VEGF-D

Hemangiogenic properties through binding to VEGFR-2[126]

Both copies of the VEGF-C gene are needed (haploinsufficiency)[130]

Bind to VEGFR-3 and promote venous endothelial differentiation into lymphangioblasts[110,130]

VEGFR-3 blockade results in a hemangiogenesis-dominant cornea[131]

VEGF-C plays a key role in development of the lymphatic vascular tree, but is not needed to maintain lymphatics after that have already been developed[132]

Deletion of VEGF-D, in contrast, does not affect lymphatic vascular development[130]

Role VEGF-A[124-125]

Binds to VEGFR-1 and -2 to promote hemangiogenesis

Binds to VEGFR-1 and -2 to promote lymphangiogenesis

Also acts indirectly through recruitment of macrophages

Response to VEGF-A[118].

Early appearance of vessels in response to low VEGF-A levels

Delayed appearance of vessels and higher VEGF-A concentrations required1

Role of FGF-2

Stimulates angiogenesis only at high doses (80-100 ng)[133]

Low dose (12.5 ng) selectively stimulates lymphangiogenesis[134]

Regression after pathologic invasion of cornea[131]

Late

Early

Corneal transplant rejection[115]

Less important role

Important role

1The delayed appearance of lymphatics is attributed to the fact that sprouting blood vessels upregulate VEGFR-2, which traps VEGF-C and prevents it from forming new lymphatics at the early phases. This is hypothesized to allow more time for the immune cells to reach the inflammatory site before being cleared by lymphatics.

 

Conclusion

The cornea is maintained in an avascular state through a balance between naturally present pro-angiogenic and anti-angiogenic chemical mediators. CNV continues to be an incompletely understood process that requires further research and funding to reveal its molecular pathways.

While hemangiogenesis and lymphangiogenesis were seldom distinguished in the past decades, recent attention has been geared towards understanding the differences between these two different, yet interdependent processes.

We think that more attention is needed in the upcoming decade to address and enhance our understanding of the molecular and genetic processes hidden under the mere manifest blood vessel.

ACKNOWLEDGEMENTS

Conflicts of interest: Abdelfattah NS, None; Amgad M, None; Zayed AA, None; Hussein H, None; Abd El-Baky N, None.


References  [Top]

1 Abdelfattah NS, Amgad M, Salama AA, Israel ME, Elhawary GA, Radwan AE, Elgayar MM, El Nakhal TM, Elkhateb IT, Hashem HA, Embaby DK, Elabd AA, Elwy RK, Yacoub MS, Salem H, Abdel-Baqy M, Kassem A. Development of an Arabic version of the National Eye Institute Visual Function Questionnaire as a tool to study eye diseases patients in Egypt. Int J Ophthalmol  2014;7(5):891-897. [PMC free article] [PubMed]

2 Ellenberg D, Azar DT, Hallak JA, Tobaigy F, Han KY, Jain S, Zhou Z, Chang JH. Novel aspects of corneal angiogenic and lymphangiogenic privilege. Prog Retin Eye Res  2010;29(3):208-248. [CrossRef] [PubMed] [PMC free article]

3 Shibuya M. VEGF-VEGFR signals in health and disease. Biomol Ther(Seoul)  2014;22(1):1-9. [CrossRef] [PubMed] [PMC free article]

4 Zhang HT, Scott PA, Morbidelli L, Peak S, Moore J, Turley H, Harris AL, Ziche M, Bicknell R. The 121 amino acid isoform of vascular endothelial growth factor is more strongly tumorigenic than other splice variants in vivo. Br J Cancer  2000;83(1):63-68. [PMC free article] [PubMed]

5 Schlaeppi JM, Siemeister G, Weindel K, Schnell C, Wood J. Characterization of a new potent, in vivo neutralizing monoclonal antibody to human vascular endothelial growth factor. J Cancer Res Clin Oncol  1999;125 (6):336-342. [CrossRef]

6 Imamura T. Physiological functions and underlying mechanisms of fibroblast growth factor (FGF) family members: recent findings and implications for their pharmacological application. Biol Pharm Bull  2014;37 (7):1081-1089. [CrossRef]

7 Marie PJ, Miraoui H, Sévère N. FGF/FGFR signaling in bone formation: progress and perspectives. Growth Factors  2012;30 (2):117-123. [CrossRef] [PubMed]

8 Parker BC, Engels M, Annala M, Zhang W. Emergence of FGFR family gene fusions as therapeutic targets in a wide spectrum of solid tumours. J Pathol 2014;232(1):4-15. [CrossRef]

9 Raju R, Palapetta SM, Sandhya VK, Sahu A, Alipoor A, Balakrishnan L, Advani J, George B, Kini KR, Geetha NP, Prakash HS, Prasad TS, Chang YJ, Chen L, Pandey A, Gowda H. A Network Map of FGF-1/FGFR Signaling System. J Signal Transduct  2014;2014:962962. [CrossRef] [PubMed] [PMC free article]

10 Adamis AP, Meklir B, Joyce NC. In situ injury-induced release of basic-fibroblast growth factor from corneal epithelial cells. Am J Pathol  1991;139(5):961-967. [PMC free article] [PubMed]

11 Bikfalvi A, Klein S, Pintucci G, Rifkin DB. Biological roles of fibroblast growth factor-2. Endocr Rev  1997;18(1):26-45. [CrossRef]

12 Soubrane G, Jerdan J, Karpouzas I, Fayein NA, Glaser B, Coscas G, Courtois Y, Jeanny JC. Binding of basic fibroblast growth factor to normal and neovascularized rabbit cornea. Invest Ophthalmol Vis Sci 1990;31(2):323-333. [PubMed]

13 Eliceiri BP. Integrin and growth factor receptor crosstalk. Circ Res 2001;89(12):1104-1110. [CrossRef]

14 Komarova Y, Malik AB. FGF signaling preserves the integrity of endothelial adherens junctions. Dev Cell  2008;15(3):335-336. [CrossRef] [PubMed] [PMC free article]

15 Guo D, Jia Q, Song HY, Warren RS, Donner DB. Vascular endothelial cell growth factor promotes tyrosine phosphorylation of mediators of signal transduction that contain SH2 domains. Association with endothelial cell proliferation. J Biol Chem 1995;270(12):6729-6733. [CrossRef] [PubMed]

16 Abedi H, Zachary I. Vascular endothelial growth factor stimulates tyrosine phosphorylation and recruitment to new focal adhesions of focal adhesion kinase and paxillin in endothelial cells. J Biol Chem 1997;272(24):15442-15451. [CrossRef] [PubMed]

17 Abbaszadeh M, Aidenloo NS, Nematollahi MK, Motarjemizadeh Q. Investigating the association between angiogenic cytokines and corneal neovascularization in sulfur mustard intoxicated subjects 26 years after exposure. Toxicol Int  2014;21(3):300-306. [CrossRef] [PubMed] [PMC free article]

18 Wheeler-Jones C, Abu-Ghazaleh R, Cospedal R, Houliston RA, Martin J, Zachary I. Vascular endothelial growth factor stimulates prostacyclin production and activation of cytosolic phospholipase A2 in endothelial cells via p42/p44 mitogen-activated protein kinase. FEBS Lett 1997;420(1):28-32. [CrossRef]

19 Sun J, Wang DA, Jain RK, Carie A, Paquette S, Ennis E, Blaskovich MA, Baldini L, Coppola D, Hamilton AD, Sebti SM. Inhibiting angiogenesis and tumorigenesis by a synthetic molecule that blocks binding of both VEGF and PDGF to their receptors. Oncogene 2005;24(29):4701-4709. [CrossRef] [PubMed]

20 Zeng L, Xiao Q, Margariti A, Zhang Z, Zampetaki A, Patel S, Capogrossi MC, Hu Y, Xu Q. HDAC3 is crucial in shear- and VEGF-induced stem cell differentiation toward endothelial cells. J Cell Biol 2006;174(7):1059-1069. [CrossRef] [PubMed] [PMC free article]

21 Eliceiri BP, Puente XS, Hood JD, Stupack DG, Schlaepfer DD, Huang XZ, Sheppard D, Cheresh DA. Src-mediated coupling of focal adhesion kinase to integrin alpha(v)beta5 in vascular endothelial growth factor signaling. J Cell Biol 2002;157(1):149-160. [CrossRef] [PubMed] [PMC free article]

22 Onguchi T, Han KY, Chang JH, Azar DT. Membrane type-1 matrix metalloproteinase potentiates basic fibroblast growth factor-induced corneal neovascularization. Am J Pathol  2009;174(4):1564-1571. [CrossRef] [PubMed] [PMC free article]

23 Järveläinen H, Sainio A, Wight TN. Pivotal role for decorin in angiogenesis. Matrix Biol 2015;43:15-26. [CrossRef] [PubMed] [PMC free article]

24 Neill T, Schaefer L, Iozzo RV. Decorin: a guardian from the matrix. Am J Pathol 2012;181(2):380-387. [CrossRef] [PubMed] [PMC free article]

25 Seidler DG. The galactosaminoglycan-containing decorin and its impact on diseases. Curr Opin Struct Biol  2012;22(5):578-582. [CrossRef] [PubMed]

26 Mohan RR, Tovey JC, Gupta R, Sharma A, Tandon A. Decorin biology, expression, function and therapy in the cornea. Curr Mol Med  2011;11(2):110-128. [CrossRef]

27 Schönherr E, Sunderkötter C, Schaefer L, Thanos S, Grässel S, Oldberg A, Iozzo RV, Young MF, Kresse H. Decorin deficiency leads to impaired angiogenesis in injured mouse cornea. J Vasc Res 2004;41(6):499-508. [CrossRef] [PubMed]

28 Mimura T, Han KY, Onguchi T, Chang JH, Kim TI, Kojima T, Zhou Z, Azar DT. MT1-MMP-mediated cleavage of decorin in corneal angiogenesis. J Vasc Res  2009;46(6):541-550. [CrossRef] [PubMed] [PMC free article]

29 Lisabeth EM, Falivelli G, Pasquale EB. Eph receptor signaling and ephrins. Cold Spring Harb Perspect Biol 2013;5(9).pii:a009159. [CrossRef] [PubMed] [PMC free article]

30 Park I, Lee HS. EphB/ephrinB signaling in cell adhesion and migration. Mol Cells 2015;38(1):14-19. [CrossRef] [PubMed] [PMC free article]

31 Adams RH, Wilkinson GA, Weiss C, Diella F, Gale NW, 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(3):295-306. [CrossRef]

32 Salvucci O, Tosato G. Essential roles of EphB receptors and EphrinB ligands in endothelial cell function and angiogenesis. Adv Cancer Res  2012;114:21-57. [CrossRef] [PubMed] [PMC free article]

33 Huynh-Do U, Vindis C, Liu H, Cerretti DP, McGrew JT, Enriquez M, Chen J, Daniel TO. Ephrin-B1 transduces signals to activate integrin-mediated migration, attachment and angiogenesis. J Cell Sci 2002;115(Pt 15):3073-3081. [PubMed]

34 Nagashima K, Endo A, Ogita H, Kawana A, Yamagishi A, Kitabatake A, Matsuda M, Mochizuki N. Adaptor protein Crk is required for ephrin-B1-induced membrane ruffling and focal complex assembly of human aortic endothelial cells. Mol Biol Cell 2002;13(12):4231-4242. [CrossRef] [PubMed] [PMC free article]

35 Kojima T, Chang JH, Azar DT. Proangiogenic role of ephrinB1/EphB1 in basic fibroblast growth factor-induced corneal angiogenesis. Am J Pathol  2007;170(2):764-773. [CrossRef] [PubMed] [PMC free article]

36 Hinck AP. Structural studies of the TGF-βs and their receptors - Insights into evolution of the TGF-β superfamily. FEBS Lett 2012;586(14):1860-1870. [CrossRef] [PubMed]

37 Lamouille S, Mallet C, Feige JJ, Bailly S. Activin receptor-like kinase 1 is implicated in the maturation phase of angiogenesis. Blood 2002;100(13):4495-4501. [CrossRef] [PubMed]

38 Nespoli A, Vercillo R, di Nola L,Diani L, Giannattasio M, Plevani P, Muzi-Falconi M. Alk1 and Alk2 are two new cell cycle-regulated haspin-like proteins in budding yeast. Cell Cycle 2006;5(13):1464-1471. [CrossRef] [PubMed]

39 Alberts B, Johnson A, Lewis J, Raff M, Roberts K WP. The Adaptive Immune System in Molecular Biology of the Cell. Fourth. New York: Garland Science; 2002.

40 De Franceschi N, Hamidi H, Alanko J, Sahgal P, Ivaska J. Integrin traffic - the update. J Cell Sci  2015; 128(5):839-852. [CrossRef] [PubMed] [PMC free article]

41 Maartens AP, Brown NH. Anchors and signals: the diverse roles of integrins in development. Curr Top Dev Biol  2015;112:233-272. [CrossRef] [PubMed]

42 Rüegg C, Mariotti A. Vascular integrins: pleiotropic adhesion and signaling molecules in vascular homeostasis and angiogenesis. Cell Mol Life Sci 2003;60(6):1135-1157. [PubMed]

43 Plow EF, Meller J, Byzova TV. Integrin function in vascular biology: a view from 2013. Curr Opin Hematol 2014;21(3):241-247. [CrossRef] [PubMed] [PMC free article]

44 Lohela M, Saaristo A, Veikkola T, Alitalo K. Lymphangiogenic growth factors, receptors and therapies. Thromb Haemost  2003;90(2):167-184. [CrossRef]

45 Vlahakis NE, Young BA, Atakilit A, Hawkridge AE, Issaka RB, Boudreau N, Sheppard D. Integrin alpha9beta1 directly binds to vascular endothelial growth factor (VEGF)-A and contributes to VEGF-A-induced angiogenesis. J Biol Chem 2007;282(20):15187-15196. [CrossRef] [PubMed]

46 Lu PC, Ye H, Maeda M, Azar DT. Immunolocalization and gene expression of matrilysin during corneal wound healing. Invest Ophthalmol Vis Sci 1999;40(1):20-27. [PubMed]

47 Ye HQ, Azar DT. Expression of gelatinases A and B, and TIMPs 1 and 2 during corneal wound healing. Invest Ophthalmol Vis Sci  1998;39(6):913-921. [PubMed]

48 Kato T, Kure T, Chang JH, Gabison EE, Itoh T, Itohara S, Azar DT. Diminished corneal angiogenesis in gelatinase A-deficient mice. FEBS Lett 2001;508(2):187-190. [CrossRef]

49 Zhou Z, Apte SS, Soininen R, Cao R, Baaklini GY, Rauser RW, Wang J, Cao Y, Tryggvason K. Impaired endochondral ossification and angiogenesis in mice deficient in membrane-type matrix metalloproteinase I. Proc Natl Acad Sci U S A 2000;97(8):4052-4057. [CrossRef] [PubMed] [PMC free article]

50 Chen Q, Jin M, Yang F, Zhu J, Xiao Q, Zhang L. Matrix metalloproteinases: inflammatory regulators of cell behaviors in vascular formation and remodeling. Mediators Inflamm  2013;2013:928315.

51 Page-McCaw A. Remodeling the model organism: matrix metalloproteinase functions in invertebrates. Semin Cell Dev Biol 2008;19(1):14-23. [CrossRef] [PubMed] [PMC free article]

52 Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol 2007;8(3):221-233. [CrossRef] [PubMed] [PMC free article]

53 Han KY, Dugas-Ford J, Seiki M, Chang JH, Azar DT. Evidence for the involvement of MMP14 n MMP2 processing and recruitment in exosomes of corneal fibroblasts. Invest Ophthalmol Vis Sci 2015;56(9):5323-5329. [PMC free article] [PubMed]

54 Ma DH, Chen JK, Zhang F, Lin KY, Yao JY, Yu JS. Regulation of corneal angiogenesis in limbal stem cell deficiency. Prog Retin Eye Res 2006;25(6):563-590. [CrossRef] [PubMed]

55 Saika S, Yamanaka O, Okada Y, Miyamoto T, Kitano A, Flanders KC, Ohnishi Y, Nakajima Y, Kao WW, Ikeda K. Effect of overexpression of PPARgamma on the healing process of corneal alkali burn in mice. Am J Physiol Cell Physiol  2007;293(1):C75-C86. [CrossRef] [PubMed]

56 Lin HC, Chang JH, Jain S, Gabison EE, Kure T, Kato T, Fukai N, Azar DT. Matrilysin cleavage of corneal collagen type XVIII NC1 domain and generation of a 28-kDa fragment. Invest Ophthalmol Vis Sci 2001;42(11):2517-2524. [PubMed]

57 Vempati P, Popel AS, Mac Gabhann F. Extracellular regulation of VEGF: isoforms, proteolysis, and vascular patterning. Cytokine Growth Factor Rev 2014;25(1):1-19. [CrossRef] [PubMed] [PMC free article]

58 Azhagiya Singam ER, Rajapandian V, Subramanian V. Molecular dynamics simulation study on the interaction of collagen-like peptides with gelatinase-A (MMP-2). Biopolymers  2014;101(7):779-794. [CrossRef] [PubMed]

59 Gioia M, Monaco S, Van Den Steen PE, Sbardella D, Grasso G, Marini S, Overall CM, Opdenakker G, Coletta M. The collagen binding domain of gelatinase A modulates degradation of collagen IV by gelatinase B. J Mol Biol 2009;386(2):419-434. [CrossRef] [PubMed]

60 Dean RA, Butler GS, Hamma-Kourbali Y, Delbé J, Brigstock DR, Courty J, Overall CM. Identification of candidate angiogenic inhibitors processed by matrix metalloproteinase 2 (MMP-2) in cell-based proteomic screens: disruption of vascular endothelial growth factor (VEGF)/heparin affin regulatory peptide (pleiotrophin) and VEGF/Connective tissue growth factor angiogenic inhibitory complexes by MMP-2 proteolysis. Mol Cell Biol  2007;27(24):8454-8465. [CrossRef] [PubMed] [PMC free article]

61 Lee S, Jilani SM, Nikolova GV, Carpizo D, Iruela-Arispe ML. Processing of VEGF-A by matrix metalloproteinases regulates bioavailability and vascular patterning in tumors. J Cell Biol  2005;169(4):681-691. [CrossRef] [PubMed] [PMC free article]

62 Heinz A, Taddese S, Sippl W, Neubert RH, Schmelzer CE. Insights into the degradation of human elastin by matrilysin-1. Biochimie 2011;93(2):187-194. [CrossRef] [PubMed]

63 Di Girolamo N, Coroneo MT, Wakefield D. Active matrilysin (MMP-7) in human pterygia: potential role in angiogenesis. Invest Ophthalmol Vis Sci  2001;42(9):1963-1968. [PubMed]

64 Kajanne R, Miettinen P, Mehlem A, Leivonen SK, Birrer M, Foschi M, Kähäri VM, Leppä S. EGF-R regulates MMP function in fibroblasts through MAPK and AP-1 pathways. J Cell Physiol 2007;212(2):489-497. [CrossRef] [PubMed]

65 Sounni NE, Devy L, Hajitou A, Frankenne F, Munaut C, Gilles C, Deroanne C, Thompson EW, Foidart JM, Noel A. MT1-MMP expression promotes tumor growth and angiogenesis through an up-regulation of vascular endothelial growth factor expression. FASEB J 2002;16(6):555-564. [CrossRef]

66 Munaut C, Noël A, Hougrand O, Foidart JM, Boniver J, Deprez M. Vascular endothelial growth factor expression correlates with matrix metalloproteinases MT1-MMP, MMP-2 and MMP-9 in human glioblastomas. Int J Cancer 2003;106(6):848-855. [CrossRef] [PubMed]

67 Han KY, Fahd DC, Tshionyi M, Allemann N, Jain S, Chang JH, Azar DT. MT1-MMP modulates bFGF-induced VEGF-A expression in corneal fibroblasts. Protein Pept Lett 2012;19(12):1334-1339. [CrossRef]

68 Udayakumar TS, Nagle RB, Bowden GT. Fibroblast growth factor-1 transcriptionally induces membrane type-1 matrix metalloproteinase expression in prostate carcinoma cell line. Prostate  2004;58(1):66-75. [CrossRef] [PubMed]

69 O’Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, Lane WS, Cao Y, Sage EH, Folkman J. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 1994;79(2):315-328. [CrossRef]

70 Cao W, Tombran-Tink J, Chen W, Mrazek D, Elias R, McGinnis JF. Pigment epithelium-derived factor protects cultured retinal neurons against hydrogen peroxide-induced cell death. J Neurosci Res 1999;57(6):789-800. [CrossRef]

71 O’Reilly Ms, Boehm T, Shing Y, Fukai N, Vasios G, Lane Ws, Flynn E, Birkhead Jr, Olsen Br, Folkman J. Endostatin: An Endogenous Inhibitor Of Angiogenesis And Tumor Growth. Cell 1997;88(2):277-285. [Crossref]

72 Azar Dt. Corneal Angiogenic Privilege: Angiogenic And Antiangiogenic Factors In Corneal Avascularity, Vasculogenesis, And Wound Healing (An American Ophthalmological Society Thesis). Trans Am Ophthalmol Soc 2006;104:264-302.

73 Eriksson K, Magnusson P, Dixelius J, Claesson-Welsh L, Cross Mj. Angiostatin And Endostatin Inhibit Endothelial Cell Migration In Response To Fgf And Vegf Without Interfering With Specific Intracellular Signal Transduction Pathways. Febs Lett 2003;536(1-3):19-24. [Crossref]

74 Chang Jh, Javier Ja, Chang Gy, Oliveira Hb, Azar Dt. Functional Characterization Of Neostatins, The Mmp-Derived, Enzymatic Cleavage Products Of Type Xviii Collagen. Febs Lett  2005;579(17):3601-3606. [Crossref] [Pubmed]

75 Kim Ym, Hwang S, Kim Ym, Pyun Bj, Kim Ty, Lee St, Gho Ys, Kwon Yg. Endostatin Blocks Vascular Endothelial Growth Factor-Mediated Signaling Via Direct Interaction With Kdr/Flk-1. J Biol Chem 2002;277(31):27872-27879. [Crossref] [Pubmed]

76 Dhanabal M, Ramchandran R, Waterman Mj, Lu H, Knebelmann B, Segal M, Sukhatme Vp. Endostatin Induces Endothelial Cell Apoptosis. J Biol Chem 1999;274(17):11721-11726. [Crossref]

77 Shao Xj, Chi Xy. Influence Of Angiostatin And Thalidomide On Lymphangiogenesis. Lymphology 2005;38(3):146-155. [Pubmed]

78 Brideau G, Mäkinen Mj, Elamaa H, Tu H, Nilsson G, Alitalo K, Pihlajaniemi T, Heljasvaara R. Endostatin Overexpression Inhibits Lymphangiogenesis And Lymph Node Metastasis In Mice. Cancer Res  2007;67(24):11528-11535. [Crossref] [Pubmed]

79 Kojima T, Azar Dt, Chang Jh. Neostatin-7 Regulates Bfgf-Induced Corneal Lymphangiogenesis. Febs Lett 2008;582(17):2515-2520. [Crossref] [Pubmed] [Pmc Free Article]

80 Magnon C, Galaup A, Mullan B, Rouffiac V, Bidart Jm, Griscelli F, Opolon P, Perricaudet M. Canstatin Acts On Endothelial And Tumor Cells Via Mitochondrial Damage Initiated Through Interaction With Alphavbeta3 And Alphavbeta5 Integrins. Cancer Res  2005;65(10):4353-4361. [Crossref] [Pubmed]

81 Sudhakar A, Nyberg P, Keshamouni Vg, Mannam Ap, Li J, Sugimoto H, Cosgrove D, Kalluri R. Human Alpha1 Type Iv Collagen Nc1 Domain Exhibits Distinct Antiangiogenic Activity Mediated By Alpha1beta1 Integrin. J Clin Invest 2005;115(10):2801-2810. [Crossref] [Pubmed] [Pmc Free Article]

82 Sund M, Hamano Y, Sugimoto H, Sudhakar A, Soubasakos M, Yerramalla U, Benjamin Le, Lawler J, Kieran M, Shah A, Kalluri R. Function Of Endogenous Inhibitors Of Angiogenesis As Endothelium-Specific Tumor Suppressors. Proc Natl Acad Sci Usa 2005;102(8):2934-2939. [Crossref] [Pubmed] [Pmc Free Article]

83 Mundel Tm, Kalluri R. Type Iv Collagen-Derived Angiogenesis Inhibitors. Microvasc Res 2007;74(2-3):85-89. [Crossref] [Pubmed] [Pmc Free Article]

84 Colorado Pc, Torre A, Kamphaus G, Maeshima Y, Hopfer H, Takahashi K, Volk R, Zamborsky Ed, Herman S, Sarkar Pk, Ericksen Mb, Dhanabal M, Simons M, Post M, Kufe Dw, Weichselbaum Rr, Sukhatme Vp, Kalluri R. Anti-Angiogenic Cues From Vascular Basement Membrane Collagen. Cancer Res  2000;60(9):2520-2526. [Pubmed]

85 Boosani Cs, Nalabothula N, Sheibani N, Sudhakar A. Inhibitory Effects Of Arresten On Bfgf-Induced Proliferation, Migration, And Matrix Metalloproteinase-2 Activation In Mouse Retinal Endothelial Cells. Curr Eye Res  2010;35(1):45-55. [Crossref] [Pubmed] [Pmc Free Article]

86 Nyberg P, Xie L, Sugimoto H, Colorado P, Sund M, Holthaus K, Sudhakar A, Salo T, Kalluri R. Characterization Of The Anti-Angiogenic Properties Of Arresten, An Alpha1beta1 Integrin-Dependent Collagen-Derived Tumor Suppressor. Exp Cell Res  2008;314(18):3292-3305. [Crossref] [Pubmed] [Pmc Free Article]

87 Hwang-Bo J, Yoo Kh, Park Jh, Jeong Hs, Chung Is. Recombinant Canstatin Inhibits Angiopoietin-1-Induced Angiogenesis And Lymphangiogenesis. Int J Cancer  2012;131(2):298-309. [Crossref] [Pubmed]

88 Wang Y, Yin H, Chen P, Xie L, Wang Y. Inhibitory Effect Of Canstatin In Alkali Burn-Induced Corneal Neovascularization. Ophthalmic Res  2011;46(2):66-72. [Crossref] [Pubmed]

89 Esipov R, Beyrakhova K, Likhvantseva V, Stepanova E, Stepanenko V, Kostromina M, Abramchik Y, Miroshnikov A. Antiangiogenic And Antivascular Effects Of A Recombinant Tumstatin-Derived Peptide In A Corneal Neovascularization Model. Biochimie 2012;94(6):1368-1375. [Crossref] [Pubmed]

90 Ye Hx, Yao Y, Jiang Xj, Yuan Xr. Tumstatin Transfected Into Human Glioma Cell Line U251 Represses Tumor Growth By Inhibiting Angiogenesis. Chin Med J 2013;126(9):1720-1725.

91 Maeshima Y, Sudhakar A, Lively Jc, Ueki K, Kharbanda S, Kahn Cr, Sonenberg N, Hynes Ro, Kalluri R. Tumstatin, An Endothelial Cell-Specific Inhibitor Of Protein Synthesis. Science 2002;295(5552):140-143. [Crossref] [Pubmed]

92 Houghton Am, Grisolano Jl, Baumann Ml, Kobayashi Dk, Hautamaki Rd, Nehring Lc, Cornelius La, Shapiro Sd. Macrophage Elastase (Matrix Metalloproteinase-12) Suppresses Growth Of Lung Metastases. Cancer Res 2006;66(12):6149-6155. [Crossref] [Pubmed]

93 Patterson Bc, Sang Qa. Angiostatin-Converting Enzyme Activities Of Human Matrilysin (Mmp-7) And Gelatinase B/Type Iv Collagenase (Mmp-9) . J Biol Chem 1997;272(46):28823-28825. [Crossref] [Pubmed]

94 Dell’Eva R, Pfeffer U, Indraccolo S, Albini A, Noonan D. Inhibition Of Tumor Angiogenesis By Angiostatin: From Recombinant Protein To Gene Therapy. Endothelium  2002;9(1):3-10. [Crossref]

95 Shepard Sr, Boucher R, Johnston J, Boerner R, Koch G, Madsen Jw, Grella D, Sim Bk, Schrimsher Jl. Large-Scale Purification Of Recombinant Human Angiostatin. Protein Expr Purif  2000;20(2):216-227. [Crossref] [Pubmed]

96 Moser Tl, Stack Ms, Asplin I, Enghild Jj, Højrup P, Everitt L, Hubchak S, Schnaper Hw, Pizzo Sv. Angiostatin Binds Atp Synthase On The Surface Of Human Endothelial Cells. Proc Natl Acad Sci Usa 1999;96(6):2811-2816. [Crossref] [Pubmed]

97 Griscelli F, Li H, Bennaceur-Griscelli A, Soria J, Opolon P, Soria C, Perricaudet M, Yeh P, Lu H. Angiostatin Gene Transfer: Inhibition Of Tumor Growth In Vivo By Blockage Of Endothelial Cell Proliferation Associated With A Mitosis Arrest. Proc Natl Acad Sci Usa  1998;95(11):6367-6372. [Crossref]

98 Tarui T, Miles La, Takada Y. Specific Interaction Of Angiostatin With Integrin Alpha(V)Beta(3) In Endothelial Cells. J Biol Chem  2001;276(43):39562-39568. [Crossref] [Pubmed]

99 Gabison E, Chang Jh, Hernández-Quintela E, Javier J, Lu Pc, Ye H, Kure T, Kato T, Azar Dt. Anti-Angiogenic Role Of Angiostatin During Corneal Wound Healing. Exp Eye Res  2004;78(3):579-589. [Crossref]

100 Karakousis Pc, John Sk, Behling Kc, Surace Em, Smith Je, Hendrickson A, Tang Wx, Bennett J, Milam Ah. Localization Of Pigment Epithelium Derived Factor (Pedf) In Developing And Adult Human Ocular Tissues. Mol Vis 2001;7:154-163. [Pubmed]

101 Becerra Sp, Notario V. The Effects Of Pedf On Cancer Biology: Mechanisms Of Action And Therapeutic Potential. Nat Rev Cancer 2013;13(4):258-271. [Crossref] [Pubmed] [Pmc Free Article]

102 Kuerten D, Johnen S, Harmening N, Souteyrand G, Walter P, Thumann G. Transplantation Of Pedf-Transfected Pigment Epithelial Cells Inhibits Corneal Neovascularization In A Rabbit Model. Graefe’S Arch Clin Exp Ophthalmol  2015;253(7):1061-1069 [Crossref] [Pubmed]

103 Kaipainen A, Korhonen J, Mustonen T, Van Hinsbergh Vw, Fang Gh, 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(8):3566-3570. [Crossref]

104 Buttler K, Ezaki T, Wilting J. Proliferating Mesodermal Cells In Murine Embryos Exhibiting Macrophage And Lymphendothelial Characteristics. Bmc Dev Biol  2008;8:43. [Crossref] [Pubmed] [Pmc Free Article]

105 Tammela T, Alitalo K. Lymphangiogenesis: Molecular Mechanisms And Future Promise. Cell 2010;140(4):460-476. [Crossref] [Pubmed]

106 Hong Yk, Detmar M. Prox1, Master Regulator Of The Lymphatic Vasculature Phenotype. Cell Tissue Res  2003;314(1):85-92. [Crossref] [Pubmed]

107 François M, Caprini A, Hosking B, Orsenigo F, Wilhelm D, Browne C, Paavonen K, Karnezis T, Shayan R, Downes M, Davidson T, Tutt D, Cheah Ks, Stacker Sa, Muscat Ge, Achen Mg, Dejana E, Koopman P. Sox18 Induces Development Of The Lymphatic Vasculature In Mice. Nature 2008;456(7222):643-647. [Crossref] [Pubmed]

108 Xu Y, Yuan L, Mak J, Pardanaud L, Caunt M, Kasman I, Larrivée B, Del Toro R, Suchting S, Medvinsky A, Silva J, Yang J, Thomas Jl, Koch Aw, Alitalo K, Eichmann A, Bagri A. Neuropilin-2 Mediates Vegf-C-Induced Lymphatic Sprouting Together With Vegfr3. J Cell Biol  2010;188(1):115-130. [Crossref] [Pubmed] [Pmc Free Article]

109 Alitalo K, Tammela T, Petrova Tv. Lymphangiogenesis In Development And Human Disease. Nature 2005;438(7070):946-953. [Crossref] [Pubmed]

110 Pan Y, Wang W D, Yago T. Transcriptional Regulation Of Podoplanin Expression By Prox1 In Lymphatic Endothelial Cells. Microvasc Res 2014;94:96-102. [Crossref] [Pubmed]

111 Suzuki-Inoue K, Fuller Glj, García A, Eble Ja, Pöhlmann S, Inoue O, Gartner Tk, Hughan Sc, Pearce Ac, Laing Gd, Theakston Rd, Schweighoffer E, Zitzmann N, Morita T, Tybulewicz Vl, Ozaki Y, Watson Sp. A Novel Syk-Dependent Mechanism Of Platelet Activation By The C-Type Lectin Receptor Clec-2. Blood 2006;107(2):542-549. [Crossref] [Pubmed]

112 Norrmén C, Ivanov Ki, Cheng J, Zangger N, Delorenzi M, Jaquet M, Miura N, Puolakkainen P, Horsley V, Hu J, Augustin Hg, Ylä-Herttuala S, Alitalo K, Petrova Tv. Foxc2 Controls Formation And Maturation Of Lymphatic Collecting Vessels Through Cooperation With Nfatc1. J Cell Biol 2009;185(3):439-457. [Crossref] [Pubmed] [Pmc Free Article]

113 Oka M, Iwata C, Suzuki Hi, Kiyono K, Morishita Y, Watabe T, Komuro A, Kano Mr, Miyazono K. Inhibition Of Endogenous Tgf-Beta Signaling Enhances Lymphangiogenesis. Blood  2008;111(9):4571-4579. [Crossref] [Pubmed]

114 Chen Jx, Chen Y, Debusk L, Lin W, Lin Pc. Dual Functional Roles Of Tie-2/Angiopoietin In Tnf-Alpha-Mediated Angiogenesis. Am J Physiol Heart Circ Physiol  2004;287(1):H187-195. [Crossref] [Pubmed]

115 Singh N, Tiem M, Watkins R, Cho Yk, Wang Y, Olsen T, Uehara H, Mamalis C, Luo L, Oakey Z, Ambati Bk. Soluble Vascular Endothelial Growth Factor Receptor 3 Is Essential For Corneal Alymphaticity. Blood  2013;121(20):4242-4249. [Crossref] [Pubmed] [Pmc Free Article]

116 Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, Diem K, Weninger W, Tschachler E, Alitalo K, Kerjaschki D. Angiosarcomas Express Mixed Endothelial Phenotypes Of Blood And Lymphatic Capillaries: Podoplanin As A Specific Marker For Lymphatic Endothelium. Am J Pathol  1999;154(2):385-394. [Crossref]

117 Dietrich T, Bock F, Yuen D, Hos D, Bachmann Bo, Zahn G, Wiegand S, Chen L, Cursiefen C. Cutting Edge: Lymphatic Vessels, Not Blood Vessels, Primarily Mediate Immune Rejections After Transplantation. J Immunol  2010;184(2):535-539. [Crossref] [Pubmed] [Pmc Free Article]

118 Ambati Bk, Nozaki M, Singh N, Takeda A, Jani Pd, Suthar T, Albuquerque Rj, Richter E, Sakurai E, Newcomb Mt, Kleinman Me, Caldwell Rb, Lin Q, Ogura Y, Orecchia A, Samuelson Da, Agnew Dw, St Leger J, Green Wr, Mahasreshti Pj, Curiel Dt, Kwan D, Marsh H, Ikeda S, Leiper Lj, Collinson Jm, Bogdanovich S, Khurana Ts, Shibuya M, Baldwin Me, Ferrara N, Gerber Hp, De Falco S, Witta J, Baffi Jz, Raisler Bj, Ambati J. Corneal Avascularity Is Due To Soluble Vegf Receptor-1. Nature 2006;443(7114):993-997. [Crossref] [Pubmed] [Pmc Free Article]

119 Cursiefen C, Chen L, Saint-Geniez M, Hamrah P, Jin Y, Rashid S, Pytowski B, Persaud K, Wu Y, Streilein Jw, Dana R. Nonvascular Vegf Receptor 3 Expression By Corneal Epithelium Maintains Avascularity And Vision. Proc Natl Acad Sci U S A 2006;103(30):11405-11410. [Crossref] [Pubmed] [Pmc Free Article]

120 Nakao S, Zandi S, Hata Y, Kawahara S, Arita R, Schering A, Sun D, Melhorn Mi, Ito Y, Lara-Castillo N, Ishibashi T, Hafezi-Moghadam A. Blood Vessel Endothelial Vegfr-2 Delays Lymphangiogenesis: An Endogenous Trapping Mechanism Links Lymph- And Angiogenesis. Blood  2011;117(3):1081-1090. [Crossref] [Pubmed] [Pmc Free Article]

121 Moldovan Ni, Goldschmidt-Clermont Pj, Parker-Thornburg J, Shapiro Sd, Kolattukudy Pe. Contribution Of Monocytes/Macrophages To Compensatory Neovascularization: The Drilling Of Metalloelastase-Positive Tunnels In Ischemic Myocardium. Circ Res 2000;87(5):378-384. [Crossref]

122 Chung Es, Chauhan Sk, Jin Y, Nakao S, Hafezi-Moghadam A, Van Rooijen N, Zhang Q, Chen L, Dana R. Contribution Of Macrophages To Angiogenesis Induced By Vascular Endothelial Growth Factor Receptor-3-Specific Ligands. Am J Pathol 2009;175(5):1984-1992. [Crossref] [Pubmed] [Pmc Free Article]

123 Okuno Y, Nakamura-Ishizu A, Kishi K, Suda T, Kubota Y. Bone Marrow-Derived Cells Serve As Proangiogenic Macrophages But Not Endothelial Cells In Wound Healing. Blood 2011;117(19):5264-5272. [Crossref] [Pubmed] [Pmc Free Article]

124 Fantin A, Vieira Jm, Gestri G, Denti L, Schwarz Q, Prykhozhij S, Peri F, Wilson Sw, Ruhrberg C. Tissue Macrophages Act As Cellular Chaperones For Vascular Anastomosis Downstream Of Vegf-Mediated Endothelial Tip Cell Induction. Blood  2010;116(5):829-840. [Crossref] [Pubmed] [Pmc Free Article]

125 Handa O, Naito Y, Takagi T, Shimozawa M, Kokura S, Yoshida N, Matsui H, Cepinskas G, Kvietys Pr, Yoshikawa T. Tumor Necrosis Factor-Alpha-Induced Cytokine-Induced Neutrophil Chemoattractant-1 (Cinc-1) Production By Rat Gastric Epithelial Cells: Role Of Reactive Oxygen Species And Nuclear Factor-Kappab. J Pharmacol Exp Ther 2004;309(2):670-676. [Crossref] [Pubmed]

126 Hong Yk, Lange-Asschenfeldt B, Velasco P, Hirakawa S, Kunstfeld R, Brown Lf, Bohlen P, Senger Dr, Detmar M. Vegf-A Promotes Tissue Repair-Associated Lymphatic Vessel Formation Via Vegfr-2 And The Alpha1beta1 And Alpha2beta1 Integrins. Faseb J  2004;18(10):1111-1113. [Pubmed]

127 Witzenbichler B, Asahara T, Murohara T, Silver M, Spyridopoulos I, Magner M, Principe N, Kearney M, Hu Js, Isner Jm. Vascular Endothelial Growth Factor-C (Vegf-C/Vegf-2) Promotes Angiogenesis In The Setting Of Tissue Ischemia. Am J Pathol  1998;153(2):381-394. [Crossref]

128 Cursiefen C, Chen L, Borges Lp, Jackson D, Cao J, Radziejewski C, D'amore Pa, Dana Mr, Wiegand Sj, Streilein Jw. Vegf-A Stimulates Lymphangiogenesis And Hemangiogenesis In Inflammatory Neovascularization Via Macrophage Recruitment. J Clin Invest  2004;113(7)1040-1050. [Crossref] [Pubmed] [Pmc Free Article]

129 Karkkainen Mj, Haiko P, Sainio K, Partanen J, Taipale J, Petrova Tv, Jeltsch M, Jackson Dg, Talikka M, Rauvala H, Betsholtz C, Alitalo K. Vascular Endothelial Growth Factor C Is Required For Sprouting Of The First Lymphatic Vessels From Embryonic Veins. Nat Immunol  2004;5(1):74-80. [Crossref] [Pubmed]

130 Chung Es, Saban Dr, Chauhan Sk, Dana R. Regulation Of Blood Vessel Versus Lymphatic Vessel Growth In The Cornea. Invest Ophthalmol Vis Sci 2009;50(4):1613-1618. [Crossref] [Pubmed] [Pmc Free Article]

131 Lin J, Lalani As, Harding Tc, Gonzalez M, Wu Ww, Luan B, Tu Gh, Koprivnikar K, Vanroey Mj, He Y, Alitalo K, Jooss K. Inhibition Of Lymphogenous Metastasis Using Adeno-Associated Virus-Mediated Gene Transfer Of A Soluble Vegfr-3 Decoy Receptor. Cancer Res 2005;65(15):6901-6909. [Crossref] [Pubmed]

132 Kubo H, Cao R, Brakenhielm E, Mäkinen T, Cao Y, Alitalo K. Blockade Of Vascular Endothelial Growth Factor Receptor-3 Signaling Inhibits Fibroblast Growth Factor-2-Induced Lymphangiogenesis In Mouse Cornea. Proc Natl Acad Sci  2002;99(13):8868-8873. [Crossref] [Pubmed] [Pmc Free Article]

133 Chang Lk, Garcia-Cardeña G, Farnebo F, Fannon M, Chen Ej, Butterfield C, Moses Ma, Mulligan Rc, Folkman J, Kaipainen A. Dose-Dependent Response Of Fgf-2 For Lymphangiogenesis. Proc Natl Acad Sci U S A  2004;101(32):11658-11663. [Crossref] [Pubmed] [Pmc Free Article]

134 Cursiefen C, Maruyama K, Jackson Dg, Streilein Jw, Kruse Fe. Time Course Of Angiogenesis And Lymphangiogenesis After Brief Corneal Inflammation. Cornea  2006;25(4):443-447.<Bb> [Crossref] [Pubmed]

[Top]