Elsevier

Survey of Ophthalmology

Volume 61, Issue 4, July–August 2016, Pages 478-497
Survey of Ophthalmology

Major review
Matrix metalloproteinase 14 modulates signal transduction and angiogenesis in the cornea

https://doi.org/10.1016/j.survophthal.2015.11.006Get rights and content

Abstract

The cornea is transparent and avascular, and retention of these characteristics is critical to maintaining vision clarity. Under normal conditions, wound healing in response to corneal injury occurs without the formation of new blood vessels; however, neovascularization may be induced during corneal wound healing when the balance between proangiogenic and antiangiogenic mediators is disrupted to favor angiogenesis. Matrix metalloproteinases (MMPs), which are key factors in extracellular matrix remodeling and angiogenesis, contribute to the maintenance of this balance, and in pathologic instances, can contribute to its disruption. Here, we elaborate on the facilitative role of MMPs, specifically MMP-14, in corneal neovascularization. MMP-14 is a transmembrane MMP that is critically involved in extracellular matrix proteolysis, exosome transport, and cellular migration and invasion, processes that are critical for angiogenesis. To aid in developing efficacious therapies that promote healing without neovascularization, it is important to understand and further investigate the complex pathways related to MMP-14 signaling, which can also involve vascular endothelial growth factor, basic fibroblast growth factor, Wnt/β-catenin, transforming growth factor, platelet-derived growth factor, hepatocyte growth factor or chemokines, epidermal growth factor, prostaglandin E2, thrombin, integrins, Notch, Toll-like receptors, PI3k/Akt, Src, RhoA/RhoA kinase, and extracellular signal-related kinase. The involvement and potential contribution of these signaling molecules or proteins in neovascularization are the focus of the present review.

Introduction

Neovascularization (NV) is the term used to describe the local formation of new vascular structures at previously avascular sites. Several models of neovascular processes have been proposed10, 14, 18, 24, 25, 43, 81, 104, 117 including 1) vasculogenesis, which is the formation of new blood vessels from bone marrow-derived angioblasts, predominantly during embryogenesis; 2) local recruitment of endothelial progenitor cells; and 3) angiogenesis, which is the formation of new vessels from preexisting vascular structures.31, 170 Angiogenesis is a common feature of corneal and retinal disorders as well as cancer metastasis and is usually stimulated by changes in the endothelial cell microenvironment (e.g., trauma, hypoxia, oxidative stress, mechanical strain, and genetic changes).46 Physical changes that occur during NV include extracellular matrix (ECM) degradation, ECM remodeling, cellular migration, and cellular invasion. The pathologies associated with NV in the normally avascular cornea include herpetic stromal keratitis, diverse inflammatory disorders, systemic or autoimmune diseases, corneal graft rejection, infectious keratitis (and other corneal infection/inflammation), contact lens–related hypoxia, alkali burns, stromal ulceration, corneal epithelium weakness, recurrent erosion syndrome, diabetes mellitus–related epithelial weakening, and limbal stem cell deficiency.171 Corneal NV is also seen in some congenital disorders such as aniridia, which involves complete or partial absence of the iris. In such conditions, the balance between proangiogenic and antiangiogenic factors favors NV, with both upregulation of proangiogenic factors, such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), and downregulation of antiangiogenic factors, such as endostatin and thrombospondin-1.43, 47, 176

The cornea is avascular under normal conditions and, even when damaged, typically heals without NV.24 This persistent avascularity of the cornea, which is necessary for vision clarity, may be facilitated by: 1) the tightly organized packing of collagen fibrils, 2) the angiostatic nature of corneal epithelial cells, 3) the immune privilege of the cornea, mediated by factors such as transforming growth factor-β (TGF-β) in tears, 4) the comparatively hypothermic nature of the cornea, 5) extensive neuronal innervation, 6) the movement of the aqueous humor on the endothelium, 7) low levels of proangiogenic matrix metalloproteinases (MMPs), 8) active production of antiangiogenic factors after corneal injury, and 9) the barrier function of the limbus.14, 43

To maintain corneal clarity, proper corneal wound healing is critical. Corneal stromal wound healing occurs in 4 phases. In the first phase the keratocytes adjacent to the area of the epithelial defect undergo apoptosis, leaving a central zone devoid of cells. This cell death has been suggested to initiate the healing response.111 In the second phase the keratocytes immediately adjacent to the area of cell death proliferate to repopulate the wound area. For example, in rat corneas, proliferation occurs 24 to 48 hours postwound.34 Within this phase the keratocytes transform into fibroblasts that migrate into the wound area, a process which may take up to 1 week.34 This transformation is evident at the molecular level, with reorganization of the actin cytoskeleton in the development of stress fibers and focal adhesion structures and activation of new genes encoding ECM components, such as fibronectin, cell adhesion molecule, α5 integrin, ECM-degrading MMPs, and cytokines.14, 15, 16 The same transition can be seen in vitro. When keratocytes are isolated from the corneal stroma and subcultured in serum-containing medium, they acquire the fibroblast phenotype.104 The migratory repair fibroblasts contain filamentous-actin and are elongated, spindle shaped, and highly reflective. These fibroblasts induce the synthesis of the α5 integrin chain, which results in the formation of the α5β1 integrin heterodimer, the classic fibronectin receptor. This occurs concomitant with a reduction in fibronectin content in the wound area. In addition to forming the ECM, these repair fibroblasts synthesize several MMPs, including MMP-1, -2, -3, -9, and -14.104

In the third phase of stromal wound healing, transformation of fibroblasts into myofibroblasts may occur and can be observed via α-smooth muscle actin staining. Myofibroblasts appear as stellate cells and are highly reflective, but are limited to within the wound area. The extent of fibroblast transformation into myofibroblasts seems to be dependent on the type of wound and the integrity of the Bowman membrane. In general, gaping wounds and wounds in which the Bowman membrane is removed result in greater myofibroblast generation than wounds that do not penetrate the Bowman membrane. This process, which may take up to 1 month to become histologically apparent, can lead to a decrease in corneal clarity and vision deterioration.

The fourth and final phase of stromal wound healing involves stromal remodeling and is largely dependent on the characteristics of the original wound. Within this setting, intricate, but incompletely understood, relationships among keratocytes, fibroblasts, and myofibroblasts play key roles.45 It is theorized that TGF-β and fibroblast-like synoviocytes are key intermediaries in the process of remodeling.34 Wounds that have completely healed contain few, if any, myofibroblasts, presumably because these cells revert to the fibroblast phenotype or undergo apoptosis during wound healing.69 The entire process of corneal healing after an injury may take more than 1 year.34

In the setting of certain inflammatory, infectious, degenerative, and traumatic states, corneal NV may be induced during wound healing.24, 183 When NV does occur, blood vessel invasion into the cornea is associated with significant visual impairment, which can ultimately progress to blindness. Three distinct morphologies of corneal NV are most commonly diagnosed: 1) deep NV overlying Descemet membrane (Fig. 1A), 2) stromal NV observed in interstitial keratitis (Fig. 1B), and 3) superficial vascular pannus (Fig. 1C).

NV occurs when there is a disturbance in the balance between proangiogenic and antiangiogenic factors. Proangiogenic factors include VEGF, bFGF (also referred to as FGF-2), and platelet-derived growth factor (PDGF). Antiangiogenic factors include angiostatin, endostatin, pigment epithelium-derived factor (PEDF), thrombospondin-1, and soluble VEGF receptor 1 (VEGFR1). A striking indication of this delicate balancing act is that many of the antiangiogenic factors are proteolytic degradation products derived from ECM fragments formed during the initial invasion of cells into the ECM during angiogenesis.7, 14 MMPs have been implicated as both proangiogenic and antiangiogenic molecules that are, in part, responsible for orchestrating the delicate balance between corneal angiogenesis and avascularity.14, 63 In this review, we present evidence for the facilitative role of MMPs, specifically MMP-14, in corneal NV.

Section snippets

MMP members

Among the 25 MMPs identified to date, at least 16 have been found in the cornea, including collagenases (MMP-1, -8, and -13), gelatinases A and B (MMP-2 and -9), stromelysins (MMP-3, -10, and -11), matrilysin (MMP-7), macrophage metalloelastase (MMP-12), and the membrane type MMPs (MMP-14, -15, -16, -17, -24, -25).6, 9, 19, 24, 38, 39, 41, 65, 81, 98, 99, 114, 130, 134, 160, 168 MMPs display dual functions during angiogenesis based on their ability to 1) degrade ECM, which allows tissue

MMP-14 processing and regulation

The precise nature of MMP-14 processing, shedding, and endocytosis conveys unique regulatory qualities with inherent complexity. MMP-14 is produced as an inactive zymogen and then cleaved into a 57-kDa peptide that inserts into the plasma membrane. MMP-14 can undergo autocatalysis to generate a 44-kDa peptide that is an inactive degradation product. Osenkowski and colleagues demonstrated that the hinge region of MMP-14 is the “hot spot” for autocatalytic activity, which produces the 44-kDa

MMP-14 mechanisms of action

MMP-14 is the most prevalent MMP involved in angiogenesis and ECM remodeling.75, 120 It leads to the disruption of endothelial tight junctions, reorganization of the actin cytoskeleton, and proteolysis of the basement membrane and interstitial matrix. MMP-14 exerts its effects via 4 key mechanisms: 1) cleavage of ECM molecules, such as type I collagen,72, 156 2) upregulation of angiogenic factors, such as VEGF,156 3) interactions with the cellular adhesion molecules expressed on the cell

Pathways of MMP-14 signaling

MMP-14 expression is regulated through a multitude of signaling pathways, and its expression leads to the production of multiple factors that support or inhibit angiogenesis and ECM remodeling. Furthermore, MMP-14 expression is associated with various disease processes, such as NV and cancer metastasis, through its ability to degrade ECM components and participate in multiple signaling pathways. Such interactions illustrate the complexity of MMP-14 signaling and demonstrate the delicate balance

Conclusions

Corneal NV is a pathologic condition characterized by angiogenesis and ECM remodeling within the cornea, a physiologically avascular tissue. Proangiogenic factors such as MMP-14 play important roles in inducing corneal NV. The signaling pathways by which MMP-14 induces angiogenic processes and ECM degradation in conjunction with other proangiogenic factors such as VEGF, FGF, HGF, and Src have been summarized here.

Currently, anti-MMP14 (9EB and DX2400) antibodies have been successfully used to

Disclosure

No conflicts of interest, financial or otherwise, are declared by the authors.

Acknowledgments

This study was supported by grants from the National Institutes of Health EY10101 (Dimitri T. Azar), EY023691, EY021886, I01 BX002386 (Jin-Hong Chang), and EY01792, and an unrestricted grant from Research to Prevent Blindness, New York, NY.

References (183)

  • D. Ellenberg et al.

    Novel aspects of corneal angiogenic and lymphangiogenic privilege

    Prog Retin Eye Res

    (2010)
  • M.E. Fini

    Keratocyte and fibroblast phenotypes in the repairing cornea

    Prog Retin Eye Res

    (1999)
  • J. Folkman

    Tumor angiogenesis

    Adv Cancer Res

    (1985)
  • J. Folkman et al.

    Angiogenesis

    J Biol Chem

    (1992)
  • V.S. Golubkov et al.

    Intradomain cleavage of inhibitory prodomain is essential to protumorigenic function of membrane type-1 matrix metalloproteinase (MT1-MMP) in vivo

    J Biol Chem

    (2011)
  • K.Y. Han et al.

    Involvement of lysosomal degradation in VEGF-C-induced down-regulation of VEGFR-3

    FEBS Lett

    (2014)
  • S. Hernandez-Barrantes et al.

    Differential roles of TIMP-4 and TIMP-2 in pro-MMP-2 activation by MT1-MMP

    Biochem Biophys Res Commun

    (2001)
  • S. Hernandez-Barrantes et al.

    Binding of active (57 kDa) membrane type 1-matrix metalloproteinase (MT1-MMP) to tissue inhibitor of metalloproteinase (TIMP)-2 regulates MT1-MMP processing and pro-MMP-2 activation

    J Biol Chem

    (2000)
  • S. Ingvarsen et al.

    Targeting a single function of the multifunctional matrix metalloprotease MT1-MMP: impact on lymphangiogenesis

    J Biol Chem

    (2013)
  • K. Kessenbrock et al.

    Matrix metalloproteinases: regulators of the tumor microenvironment

    Cell

    (2010)
  • S. Kim et al.

    Posttranslational regulation of membrane type 1-matrix metalloproteinase (MT1-MMP) in mouse PTEN null prostate cancer cells: Enhanced surface expression and differential O-glycosylation of MT1-MMP

    Biochim Biophys Acta

    (2010)
  • K. Lehti et al.

    Oligomerization through hemopexin and cytoplasmic domains regulates the activity and turnover of membrane-type 1 matrix metalloproteinase

    J Biol Chem

    (2002)
  • K. Lehti et al.

    Regulation of membrane-type-1 matrix metalloproteinase activity by its cytoplasmic domain

    J Biol Chem

    (2000)
  • X. Li et al.

    Novel VEGF family members: VEGF-B, VEGF-C and VEGF-D

    Int J Biochem Cell Biol

    (2001)
  • A.M. Limaye et al.

    Regulation of mRNAs encoding MMP-9 and MMP-2, and their inhibitors TIMP-1 and TIMP-2 by androgens in the rat ventral prostate

    Mol Cell Endocrinol

    (2008)
  • K.V. Lu et al.

    Upregulation of tissue inhibitor of metalloproteinases (TIMP)-2 promotes matrix metalloproteinase (MMP)-2 activation and cell invasion in a human glioblastoma cell line

    Lab Invest

    (2004)
  • Y. Maeshima et al.

    Two RGD-independent alpha vbeta 3 integrin binding sites on tumstatin regulate distinct anti-tumor properties

    J Biol Chem

    (2000)
  • Y. Maeshima et al.

    Identification of the anti-angiogenic site within vascular basement membrane-derived tumstatin

    J Biol Chem

    (2001)
  • Y. Maeshima et al.

    Extracellular matrix-derived peptide binds to alpha (v) beta (3) integrin and inhibits angiogenesis

    J Biol Chem

    (2001)
  • A.P. Adamis et al.

    In situ injury-induced release of basic-fibroblast growth factor from corneal epithelial cells

    Am J Pathol

    (1991)
  • E.I. Ager et al.

    Blockade of MMP14 activity in murine breast carcinomas: implications for macrophages, vessels, and radiotherapy

    J Natl Cancer Inst

    (2015)
  • D. Al-Raawi et al.

    Membrane type-1 matrix metalloproteinase (MT1-MMP) correlates with the expression and activation of matrix metalloproteinase-2 (MMP-2) in inflammatory breast cancer

    Int J Clin Exp Med

    (2011)
  • R.J. Albuquerque et al.

    Alternatively spliced vascular endothelial growth factor receptor-2 is an essential endogenous inhibitor of lymphatic vessel growth

    Nat Med

    (2009)
  • S. Amano et al.

    Requirement for vascular endothelial growth factor in wound- and inflammation-related corneal neovascularization

    Invest Ophthalmol Vis Sci

    (1998)
  • B.K. Ambati et al.

    Corneal avascularity is due to soluble VEGF receptor-1

    Nature

    (2006)
  • K. Ando et al.

    Crucial role of membrane type 1 matrix metalloproteinase (MT1-MMP) in RhoA/Rac1-dependent signaling pathways in thrombin-stimulated endothelial cells

    J Atheroscler Thromb

    (2011)
  • B. Annabi et al.

    Hypoxia promotes murine bone-marrow-derived stromal cell migration and tube formation

    Stem Cells

    (2003)
  • T. Asahara et al.

    VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells

    Embo J

    (1999)
  • D. Assent et al.

    A membrane-type-1 matrix metalloproteinase (MT1-MMP)—discoidin domain receptor 1 axis regulates collagen-induced apoptosis in breast cancer cells

    PLoS One

    (2015)
  • P. Auguste et al.

    Inhibition of fibroblast growth factor/fibroblast growth factor receptor activity in glioma cells impedes tumor growth by both angiogenesis-dependent and -independent mechanisms

    Cancer Res

    (2001)
  • D.T. Azar

    Corneal angiogenic privilege: angiogenic and antiangiogenic factors in corneal avascularity, vasculogenesis, and wound healing (an American Ophthalmological Society thesis)

    Trans Am Ophthalmol Soc

    (2006)
  • D.T. Azar et al.

    Effect of MT1-MMP deficiency and overexpression in corneal keratocytes on vascular endothelial cell migration and proliferation

    Curr Eye Res

    (2008)
  • D.T. Azar et al.

    Corneal epithelialMT1-MMP inhibits vascular endothelial cell proliferation and migration

    Cornea

    (2010)
  • D.T. Azar et al.

    Matrix metalloproteinases are expressed during wound healing after excimer laser keratectomy

    Cornea

    (1996)
  • L. Beck et al.

    Vascular development: cellular and molecular regulation

    FASEB J

    (1997)
  • M.B. Berman

    Regulation of corneal fibroblast MMP-1 secretion by cytochalasins

    Cornea

    (1994)
  • C.E. Brinckerhoff et al.

    Interstitial collagenases as markers of tumor progression

    Clin Cancer Res

    (2000)
  • S. Chakraborti et al.

    Regulation of matrix metalloproteinases: an overview

    Mol Cell Biochem

    (2003)
  • J.H. Chang et al.

    Corneal neovascularization

    Curr Opin Ophthalmol

    (2001)
  • J.C. Clark et al.

    RECK—a newly discovered inhibitor of metastasis with prognostic significance in multiple forms of cancer

    Cancer Metastasis Rev

    (2007)
  • Cited by (0)

    View full text