Survey
Extracellular regulation of VEGF: Isoforms, proteolysis, and vascular patterning

https://doi.org/10.1016/j.cytogfr.2013.11.002Get rights and content

Abstract

The regulation of vascular endothelial growth factor A (VEGF) is critical to neovascularization in numerous tissues under physiological and pathological conditions. VEGF has multiple isoforms, created by alternative splicing or proteolytic cleavage, and characterized by different receptor-binding and matrix-binding properties. These isoforms are known to give rise to a spectrum of angiogenesis patterns marked by differences in branching, which has functional implications for tissues. In this review, we detail the extensive extracellular regulation of VEGF and the ability of VEGF to dictate the vascular phenotype. We explore the role of VEGF-releasing proteases and soluble carrier molecules on VEGF activity. While proteases such as MMP9 can ‘release’ matrix-bound VEGF and promote angiogenesis, for example as a key step in carcinogenesis, proteases can also suppress VEGF's angiogenic effects. We explore what dictates pro- or anti-angiogenic behavior. We also seek to understand the phenomenon of VEGF gradient formation. Strong VEGF gradients are thought to be due to decreased rates of diffusion from reversible matrix binding, however theoretical studies show that this scenario cannot give rise to lasting VEGF gradients in vivo. We propose that gradients are formed through degradation of sequestered VEGF. Finally, we review how different aspects of the VEGF signal, such as its concentration, gradient, matrix-binding, and NRP1-binding can differentially affect angiogenesis. We explore how this allows VEGF to regulate the formation of vascular networks across a spectrum of high to low branching densities, and from normal to pathological angiogenesis. A better understanding of the control of angiogenesis is necessary to improve upon limitations of current angiogenic therapies.

Introduction

VEGF-A is a key member of the VEGF family of cytokines, along with VEGF-B, -C, -D, and PlGF [1], [2]. VEGF-A mediates angiogenesis, the expansion of an existing vascular bed by sprouting of new blood vessels [3]. Angiogenesis typically occurs as a response to a stimulus such as tissue hypoxia, and results in improved perfusion and increased oxygen delivery. Other stimuli can induce angiogenesis, including shear stress [4] and genetic transformation in tumor cells [3]. Angiogenesis is important for organ development [5] as well as for physiological processes including wound closure and exercise training [6], [7]. It is upregulated but disorganized in pathological processes such as diabetic retinopathy and solid organ tumorigenesis [8], [9], [10], where vasculature is needed to supply the tumor's rapid consumption of glucose and oxygen beyond the limits of diffusion.

The vegfa gene is translated into a number of splice isoforms, the most notable in humans being VEGF121, VEGF165, and VEGF189 (Fig. 1). These isoforms have differences in biochemical properties such as their affinities for VEGF receptors and heparan sulfate proteoglycans (HSPGs), resulting in strikingly different effects on vessel growth. A major focus of the current review is the extracellular regulation of VEGF (Sections 3 Local availability and activity of VEGF: ECM, proteases and inhibitors, 4 ). In normal healthy situations, VEGF isoforms are differentially sequestered by heparan sulfate proteoglycans (HSPGs) in the ECM (Section 3.1) and are subject to various VEGF inhibitors (Section 3.2), e.g. sVEGFR1, a secreted isoform of the membrane VEGF receptor VEGFR1 [11]; these inhibitors are involved in establishing vascular quiescence [12]. During inflammation and tumorigenesis, sequestered VEGF can be released by proteases, such as the zinc-dependent matrix metalloproteinases (MMPs). Extracellular proteases can act on VEGF in several ways (Section 3.3) including cleavage of the ECM, cleavage of VEGF generating new isoforms such as VEGF114, and also cleavage of the soluble inhibitors of VEGF. These can lead to different biological outcomes. Proteases such as MMP9 are typically thought to release VEGF and induce angiogenesis, but in other situations may reduce angiogenesis activity, e.g. by cleavage of VEGF [13]. We will explore what dictates whether proteolytic release of VEGF is pro- or anti-angiogenic, and the roles of specific proteases.

The spatial distribution of VEGF is a key regulator of angiogenesis and is itself regulated by both matrix binding and proteolytic release (Section 4). For example, VEGF isoforms that bind strongly to the ECM, such as VEGF165 and VEGF189, have a steep gradient [14], [15] and tight pericellular sequestration [15], [16], [17], [18]. Gradient formation has been commonly thought to be due to a restriction of the rate of diffusion by ECM binding (Section 4.2). However, using computational modeling, we have shown that HSPG binding alone cannot explain most aspects of VEGF gradients [19]. This and other differences between experimental and theoretical results require us to revisit the underlying mechanics of VEGF transport in vivo (Sections 4.3 Combined sequestration and degradation can explain VEGF gradient formation, 4.4 Proteolytic release of VEGF increases VEGF spatial range by reducing degradation). Recent advances have indicated that soluble VEGF inhibitors also play an important role in VEGF patterning [20], [21], [22].

Different tissues express different ratios of the VEGF isoforms (Fig. 2) and this may serve to produce vascular networks that match the specific needs of each tissue [23]. Mice expressing only VEGF120 instead of the full range of VEGF isoforms have significant defects in cardiac and pulmonary development due to defective angiogenesis [24], [25]. On the other hand, tumor growth appears to be most rapid in tumors that express VEGF164 [16], [26]. We review how VEGF, via its spatial distribution and receptor signaling, regulates angiogenesis. Heparin-binding VEGF isoforms produce a branching network with narrow vessels, while VEGF120 (the murine equivalent of VEGF121) results in poorly branching, tortuous, leaky vessels [14], [15], [27], [28] (Section 5.2). We explore the specific mechanisms by which VEGF isoforms can cause these different vascularization states (Section 5.3). VEGF is a mediator of sprouting angiogenesis, but in some situations high levels of VEGF can result in a highly proliferative, dysregulated state and lack of sprouting [14], [29]. We explore how these pathological angiogenesis states can arise (Section 5.4).

This review aims to provide a comprehensive overview of the biochemistry, physical transport, and biology of the splice and proteolytic isoforms of VEGF. We highlight several uncertainties in our understanding of VEGF, which may be avenues for future research.

Section snippets

Splice isoforms and proteolytic isoforms of VEGF

The vegfa gene encodes several splice isoforms of VEGF-A, each of which may be processed by a variety of proteases to produce yet more isoforms (Fig. 1). Detailed reviews of VEGF splicing, VEGF receptor binding and intracellular signaling are available [1], [30], [31], [32]. Here, we discuss how the structure of the native and proteolytically-processed isoforms determine binding to receptors, co-receptors, and extracellular matrix proteoglycans.

Local availability and activity of VEGF: ECM, proteases and inhibitors

The rate of VEGF secretion is a key driver of VEGF-induced angiogenesis [80]. However, once secreted, numerous processes regulate VEGF activity in vivo; for example, in the cornea, the activity of secreted VEGF is repressed by co-secretion of sVEGFR1 [12]. Along with interstitial diffusion and convection, several distinct processes affect local VEGF availability and activity: sequestration of VEGF by stationary molecules in the ECM or on cell surfaces; VEGF inhibition or activation by other

In vivo spatial patterning of VEGF isoform gradients

Spatial gradients of VEGF regulate vessel activation and sprout guidance [14], [86], and may be shaped by numerous mechanisms in vivo including diffusion, matrix sequestration, competitive binding, and proteolytic release [121]. Heavier VEGF isoforms show increased matrix sequestration and steeper spatial gradients, suggesting that heparin binding, by slowing diffusion, directly leads to sharper gradients. However, theoretical models show that the heparin binding alone is not the source of

VEGF isoform control of vascular patterning

VEGF isoforms, with their differences in biotransport, sequestration, and NRP-1 binding, induce a spectrum of vascular phenotypes, from the malformed, edematous, hypovascular networks of VEGF120, to the stable, thin, and branching vessels of VEGF188 (Fig. 4). While in normal tissues vascular networks are organized hierarchically and adequately meet the needs of tissues, numerous disease states are characterized by exuberant, highly disturbed phenotypes, the result of pathological angiogenesis.

Perspective

Modulating angiogenesis via control of the VEGF family is a promising therapeutic approach for numerous diseases. In this review, we have detailed important aspects of VEGF extracellular regulation, including the effects of proteases and the formation of VEGF gradients. The different VEGF isoforms play key roles in the resultant vascular morphology, and there is still much that remains to be learned, especially regarding the control of alternative splicing, the role of soluble VEGF inhibitors

Acknowledgments

This work was supported by the National Institutes of Health (NIH) grants R01 HL101200 and R01 CA138264 (ASP) and R00 HL093219 (FMG). The authors thank Dr. David Noren, Dr. Elena Rosca, and Dr. Marianne O. Engel-Stefanini and other members of the Popel laboratory for useful discussions and critical comments.

Prakash Vempati, M.Sc., is a medical student at the Vanderbilt University School of Medicine in Nashville, TN. He completed a master's degree under the direction of Dr. Aleksander S. Popel in biomedical engineering at The Johns Hopkins University in 2009 studying the extracellular regulation of VEGF and matrix metalloproteinases in angiogenesis. He is pursuing a career in internal medicine and is interested in the application of plasma biomarkers and pharmacogenetic information towards clinical

References (223)

  • G. Fuh et al.

    The interaction of neuropilin-1 with vascular endothelial growth factor and its receptor flt-1

    J Biol Chem

    (2000)
  • Q. Pan et al.

    Neuropilin-1 binds to VEGF121 and regulates endothelial cell migration and sprouting

    J Biol Chem

    (2007)
  • H. Gitay-Goren et al.

    The binding of vascular endothelial growth factor to its receptors is dependent on cell surface-associated heparin-like molecules

    J Biol Chem

    (1992)
  • Z. Gluzman-Poltorak et al.

    Neuropilin-2 is a receptor for the vascular endothelial growth factor (VEGF) forms VEGF-145 and VEGF-165 [corrected]

    J Biol Chem

    (2000)
  • B. Favier et al.

    Neuropilin-2 interacts with VEGFR-2 and VEGFR-3 and promotes human endothelial cell survival and migration

    Blood

    (2006)
  • H. Jia et al.

    Peptides encoded by exon 6 of VEGF inhibit endothelial cell biological responses and angiogenesis induced by VEGF

    Biochem Biophys Res Commun

    (2001)
  • J. Plouet et al.

    Extracellular cleavage of the vascular endothelial growth factor 189-amino acid form by urokinase is required for its mitogenic effect

    J Biol Chem

    (1997)
  • Z. Poltorak et al.

    VEGF145, a secreted vascular endothelial growth factor isoform that binds to extracellular matrix

    J Biol Chem

    (1997)
  • M. Ancelin et al.

    Vascular endothelial growth factor VEGF189 induces human neutrophil chemotaxis in extravascular tissue via an autocrine amplification mechanism

    Lab Invest

    (2004)
  • M.A. Herve et al.

    VEGF189 stimulates endothelial cells proliferation and migration in vitro and up-regulates the expression of Flk-1/KDR mRNA

    Exp Cell Res

    (2005)
  • M.A. Herve et al.

    Overexpression of vascular endothelial growth factor 189 in breast cancer cells leads to delayed tumor uptake with dilated intratumoral vessels

    Am J Pathol

    (2008)
  • K.A. Houck et al.

    Dual regulation of vascular endothelial growth factor bioavailability by genetic and proteolytic mechanisms

    J Biol Chem

    (1992)
  • G. Hashimoto et al.

    Matrix metalloproteinases cleave connective tissue growth factor and reactivate angiogenic activity of vascular endothelial growth factor 165

    J Biol Chem

    (2002)
  • L.J. Hawinkels et al.

    VEGF release by MMP-9 mediated heparan sulphate cleavage induces colorectal cancer angiogenesis

    Eur J Cancer

    (2008)
  • T.K. Ito et al.

    Degradation of soluble VEGF receptor-1 by MMP-7 allows VEGF access to endothelial cells

    Blood

    (2009)
  • S. Lee et al.

    Autocrine VEGF signaling is required for vascular homeostasis

    Cell

    (2007)
  • A. Sahni et al.

    Vascular endothelial growth factor binds to fibrinogen and fibrin and stimulates endothelial cell proliferation

    Blood

    (2000)
  • E.S. Wijelath et al.

    Fibronectin promotes VEGF-induced CD34 cell differentiation into endothelial cells

    J Vasc Surg

    (2004)
  • A. Luque et al.

    ADAMTS1/METH1 inhibits endothelial cell proliferation by direct binding and sequestration of VEGF165

    J Biol Chem

    (2003)
  • G. Bhattacharjee et al.

    The conformation-dependent interaction of alpha 2-macroglobulin with vascular endothelial growth factor. A novel mechanism of alpha 2-macroglobulin/growth factor binding

    J Biol Chem

    (2000)
  • S. Kaur et al.

    Thrombospondin-1 inhibits VEGF receptor-2 signaling by disrupting its association with CD47

    J Biol Chem

    (2010)
  • A. Purushothaman et al.

    Heparanase-enhanced shedding of syndecan-1 by myeloma cells promotes endothelial invasion and angiogenesis

    Blood

    (2010)
  • S. Koch et al.

    Signal transduction by vascular endothelial growth factor receptors

    Cold Spring Harb Perspect Med

    (2012)
  • F. Mac Gabhann et al.

    Systems biology of vascular endothelial growth factors

    Microcirculation

    (2008)
  • P. Carmeliet et al.

    Molecular mechanisms and clinical applications of angiogenesis

    Nature

    (2011)
  • S. Egginton

    In vivo shear stress response

    Biochem Soc Trans

    (2011)
  • A.S. Chung et al.

    Developmental and pathological angiogenesis

    Annu Rev Cell Dev Biol

    (2011)
  • G. Liu et al.

    Module-based multiscale simulation of angiogenesis in skeletal muscle

    Theor Biol Med Model

    (2011)
  • T. Gustafsson

    Vascular remodelling in human skeletal muscle

    Biochem Soc Trans

    (2011)
  • P. Carmeliet et al.

    Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases

    Nat Rev Drug Discov

    (2011)
  • L. Claesson-Welsh et al.

    VEGFA and tumour angiogenesis

    J Intern Med

    (2013)
  • K. Kinnunen et al.

    Vascular endothelial growth factors in retinal and choroidal neovascular diseases

    Ann Med

    (2012)
  • F.T. Wu et al.

    A systems biology perspective on sVEGFR1: its biological function, pathogenic role and therapeutic use

    J Cell Mol Med

    (2010)
  • B.K. Ambati et al.

    Corneal avascularity is due to soluble VEGF receptor-1

    Nature

    (2006)
  • S. Lee et al.

    Processing of VEGF-A by matrix metalloproteinases regulates bioavailability and vascular patterning in tumors

    J Cell Biol

    (2005)
  • H. Gerhardt et al.

    VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia

    J Cell Biol

    (2003)
  • C. Ruhrberg et al.

    Spatially restricted patterning cues provided by heparin-binding VEGF-A control blood vessel branching morphogenesis

    Genes Dev

    (2002)
  • J. Grunstein et al.

    Isoforms of vascular endothelial growth factor act in a coordinate fashion To recruit and expand tumor vasculature

    Mol Cell Biol

    (2000)
  • J.E. Park et al.

    The vascular endothelial growth factor (VEGF) isoforms: differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF

    Mol Biol Cell

    (1993)
  • C. Ruiz de Almodovar et al.

    Matrix-binding vascular endothelial growth factor (VEGF) isoforms guide granule cell migration in the cerebellum via VEGF receptor Flk1

    J Neurosci

    (2010)
  • Cited by (234)

    View all citing articles on Scopus

    Prakash Vempati, M.Sc., is a medical student at the Vanderbilt University School of Medicine in Nashville, TN. He completed a master's degree under the direction of Dr. Aleksander S. Popel in biomedical engineering at The Johns Hopkins University in 2009 studying the extracellular regulation of VEGF and matrix metalloproteinases in angiogenesis. He is pursuing a career in internal medicine and is interested in the application of plasma biomarkers and pharmacogenetic information towards clinical practice in vascular diseases, hemostasis, and oncology.

    Aleksander S. Popel, Ph.D., is a Professor of Biomedical Engineering at the Johns Hopkins University School of Medicine. He holds joint appointments as Professor of Oncology in the School of Medicine, and Professor of Chemical & Biomolecular Engineering in the Johns Hopkins Whiting School of Engineering. He is a member of the Institute for Nanobiotechnology, In Vivo Cellular Molecular Imaging Center, and the Sydney Kimmel Comprehensive Cancer Center. He has published over 250 scientific papers in the areas of angiogenesis and microcirculation, systems biology, computational medicine & biology. He is the recipient of the Eugene M. Landis Award from the Microcirculatory Society. He is a Fellow of the American Institute of Medical and Biological Engineering, American Heart Association, American Physiological Society, and American Society of Mechanical Engineers, and an Inaugural Fellow of the Biomedical Engineering Society. He has been a member of editorial boards of biological and biomedical engineering journals, and has served in an advisory role to biotech and pharmaceutical companies. He regularly serves on grant review boards and advisory panels at the National Institutes of Health, National Science Foundation, and other US and international funding agencies.

    Feilim Mac Gabhann, Ph.D., joined Johns Hopkins University as an Assistant Professor in 2009, with an appointment in Biomedical Engineering and in the Institute for Computational Medicine. He completed his PhD in Biomedical Engineering in 2007, also at Johns Hopkins University, working with Aleksander S. Popel to create mathematical models of growth factor networks in peripheral artery disease and cancer. During postdoctoral work with Shayn M. Peirce and Thomas C. Skalak at the University of Virginia, he conducted experimental research on microvascular remodeling in mouse skeletal muscle. The Mac Gabhann lab creates molecularly-detailed mathematical models of human physiology and disease, including peripheral artery disease, cancer, ALS, pre-eclampsia and HIV. The models have a particular focus on the development and testing of therapeutics. Dr. Mac Gabhann is a Sloan Research Fellow and recipient of a K99/R00 NIH Pathway to Independence Award, the 2010 August Krogh Young Investigator Award from the Microcirculatory Society, and the 2012 Arthur C. Guyton Award for Excellence in Integrative Physiology from the American Physiology Society. He is the author of 44 peer-reviewed papers, and is an Associate Editor for PLoS Computational Biology and BMC Physiology.

    View full text