Next Article in Journal
miRNAs Potentially Involved in Post Lung Transplant-Obliterative Bronchiolitis: The Role of miR-21-5p
Next Article in Special Issue
The Role of Heparan Sulfate and Neuropilin 2 in VEGFA Signaling in Human Endothelial Tip Cells and Non-Tip Cells during Angiogenesis In Vitro
Previous Article in Journal
Microglial Pruning: Relevance for Synaptic Dysfunction in Multiple Sclerosis and Related Experimental Models
Previous Article in Special Issue
Immune-Mediated Retinal Vasculitis in Posterior Uveitis and Experimental Models: The Leukotriene (LT)B4-VEGF Axis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell–Matrix Interactions in the Eye: From Cornea to Choroid

1
Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
2
Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
*
Author to whom correspondence should be addressed.
Cells 2021, 10(3), 687; https://doi.org/10.3390/cells10030687
Submission received: 14 January 2021 / Revised: 3 March 2021 / Accepted: 16 March 2021 / Published: 20 March 2021
(This article belongs to the Special Issue Vascular Signalling)

Abstract

:
The extracellular matrix (ECM) plays a crucial role in all parts of the eye, from maintaining clarity and hydration of the cornea and vitreous to regulating angiogenesis, intraocular pressure maintenance, and vascular signaling. This review focuses on the interactions of the ECM for homeostasis of normal physiologic functions of the cornea, vitreous, retina, retinal pigment epithelium, Bruch’s membrane, and choroid as well as trabecular meshwork, optic nerve, conjunctiva and tenon’s layer as it relates to glaucoma. A variety of pathways and key factors related to ECM in the eye are discussed, including but not limited to those related to transforming growth factor-β, vascular endothelial growth factor, basic-fibroblastic growth factor, connective tissue growth factor, matrix metalloproteinases (including MMP-2 and MMP-9, and MMP-14), collagen IV, fibronectin, elastin, canonical signaling, integrins, and endothelial morphogenesis consistent of cellular activation-tubulogenesis and cellular differentiation-stabilization. Alterations contributing to disease states such as wound healing, diabetes-related complications, Fuchs endothelial corneal dystrophy, angiogenesis, fibrosis, age-related macular degeneration, retinal detachment, and posteriorly inserted vitreous base are also reviewed.

Graphical Abstract

1. Introduction

The extracellular matrix (ECM) is an essential and major component of the ocular microenvironment. It forms a complex but organized meshwork surrounding cells and confers not only cellular structural and mechanical support, but also regulates cellular homeostasis and signaling [1]. Proteoglycans (including heparan sulfate, chondroitin sulfate, and keratin sulfate), hyaluronic acid, collagen, elastin, laminin, fibronectin, and fibrillin represent major components of the ECM [2,3]. Other components include extracellular proteases (such as matrix metalloproteinases, aka MMPs), immune mediators and growth factors [4].
The first portion of this manuscript is a review loosely organized from the front to the back of the eye, starting with the cornea, then addressing parts of the eye involved in intraocular pressure maintenance and glaucoma (i.e., trabecular meshwork, optic nerve, conjunctiva and tenon’s layer), vitreous, retina, retinal pigment epithelium, Bruch’s membrane, and choroid. Each section describes how ECM components are involved in homeostasis but also details its alterations resulting in disease states such as wound healing, diabetes-related complications, Fuchs endothelial corneal dystrophy, angiogenesis, fibrosis, age-related macular degeneration, choroidal neovascularization, retinal detachment, and posteriorly inserted vitreous base.

2. Cornea

Under normal conditions, the cornea is able to support visual transparency by remaining avascular. A complex counterbalance of homeostatic mechanisms exist to maintain corneal avascularity. In the most anterior layers of the cornea, these mechanisms include soluble factors in the precorneal tear film that mediate corneal immune privilege, such as transforming growth factor-β (TGF-β); the limbal stem cell niche, which prevents conjunctivalization of the epithelium; and structural factors in the stroma that prevent vascular ingrowth mechanically, such as tight packing of the collagen lamellae and dense corneal innervation [5]. Perturbations of these homeostatic conditions, such as those occurring with trauma, aging and various infectious and inflammatory diseases, can result in degeneration of these functional barriers against opacification due to vascular infiltration and fibrosis of the cornea [6]. Corneal opacification due to scarring and vascularization may require optical rehabilitation with contact lenses or surgical rehabilitation with keratoplasty and can lead to blindness.
In addition to corneal avascularity, the cornea under normal conditions is able to support visual clarity by maintaining appropriate hydration. The corneal endothelium-Descemet membrane (EDM) complex—the functional unit that comprises the two innermost layers of the cornea—is the primary regulator of corneal hydration. Within this complex, the corneal endothelial cell (CEC) monolayer functions as the primary boundary between the corneal stroma and the anterior chamber. CECs maintain the clarity of the cornea by regulating corneal stroma hydration through barrier and pump functions. However, the EDM undergoes changes with aging. There is a high density of endothelial cells at birth, but these cells are arrested in G1 phase and therefore do not repopulate themselves following death of adjacent cells. As a result, human corneal endothelial cell density decreases at an average rate of approximately 0.6% per year in normal corneas throughout adult life, with gradual increases in polymegathism and pleomorphism to maintain a continuous endothelial cell layer [7]. Descemet membrane, the basement membrane of the corneal endothelium, also undergoes age-related changes. This specialized extracellular matrix is comprised of a fetal anterior banded layer that is present at birth and a posterior non-banded layer that thickens with age as CECs continue to secrete extracellular matrix proteins. Reductions in endothelial cell density and alterations to Descemet membrane occur together in aging and in pathological conditions affecting the posterior cornea. As CECs do not divide readily, sodium-potassium adenosine triphosphatase pump sites and cell-cell junctions also decline when the endothelial cell density falls below a critical level. When the EDM undergoes cell death that exceed age-predicted changes, premature corneal edema and vision loss can occur, and keratoplasty using donor CECs may be indicated.
We will review the effects of damaging conditions on the anterior corneal layers, and the interplay between corneal neovascularization and extracellular matrix alterations after epithelial and stromal wounding. With the wide variety of disease conditions that can lead to the development of corneal neovascularization (e.g., infection, inflammation, trauma, degeneration), it is important to understand the mechanisms by which neovascularization can lead to loss of stromal clarity. We will also review the effects of two common diseases affecting cell–matrix interactions in the EDM complex—Fuchs endothelial corneal dystrophy and diabetes mellitus—and the impact of these pathologies on posterior corneal health and function over time. With the emergence of single-layer endothelial keratoplasty techniques such as Descemet membrane endothelial keratoplasty (DMEK) and Descemet stripping automated endothelial keratoplasty (DSAEK), it is imperative to understand cell–matrix interactions in the posterior cornea in more detail, particularly because posterior lamellar donor tissues may be altered by disease states such as diabetes [8,9,10].

2.1. Stromal Extracellular Matrix Alterations and Corneal Neovascularization

The cornea is able to maintain clarity and avascularity, even in most cases after sustaining injury to the epithelium and stroma, by a variety of mechanisms that preserve the homeostatic balance between pro-angiogenic and anti-angiogenic factors. However, corneal neovascularization can occur in a variety of conditions—including microbial keratitis, autoimmune and systemic inflammatory conditions, corneal graft rejection, neurotrophic keratitis, chemical injury, contact lens overwear, and limbal stem cell deficiency—where angiogenesis is initiated despite the presence of homeostatic anti-angiogenic regulatory mechanisms [6]. Most often, when corneal neovascularization does occur, it involves the anterior two-thirds of the stroma (89%) and is frequently associated with corneal edema and/or inflammatory cell recruitment [11].
Corneal stromal wound healing occurs in 4 phases [5]. In the first phase, keratocytes at the area of wounding undergo apoptosis, which initiates a healing response and leaves a central acellular zone [12]. In the second phase, adjacent keratocytes immediately begin to proliferate to repopulate the wound area and transform into fibroblasts that migrate into the wound area, a process that can take days. In the third phase, transformation of fibroblasts into myofibroblasts occurs, and in the fourth phase cell-mediated remodeling of the stroma occurs, which can take more than 1 year. The process of stromal wound healing is mediated by signaling of transforming growth factor-beta (TGF-β) [13], matrix metalloproteinases (MMPs), and a balance between pro-angiogenic factors (vascular endothelial growth factor [VEGF], basic fibroblastic growth factor [bFGF], also referred to as FGF-2], and platelet-derived growth factor [PDGF]) and anti-angiogenic factors (angiostatin, endostatin, pigment epithelium-derived factor [PEDF], thrombospondin-1, and soluble VEGF receptor 1 [VEGFR1]) [5,14] (Figure 1).
When the balance between pro-angiogenesis and anti-angiogenesis is not maintained, corneal neovascularization can occur (Figure 2). As a result of corneal epithelial and stromal injury, bFGF becomes upregulated and mediates fibroblast activation, whereas stromal fibroblast MMP-14 initiates enzymatic activity [5]. bFGF-mediated fibroblasts and stromal fibroblasts show upregulation of VEGF, and MMP-14 potentiates bFGF-induced corneal NV [15]. In addition, MMP-14 also mediates the degradation of ECM. Both VEGF upregulation and ECM degradation enhance vascular endothelial cell proliferation, migration, and tube formation. In addition to MMP-14 and VEGF, vascular growth in the corneal stroma is also associated with MMP-2, tissue inhibitors of metalloproteinases-2 (TIMP-2), and Src [5,16], and requires both cell proliferation/migration and extracellular matrix turnover.
Of note, MMP-14 is the most prevalent MMP involved in angiogenesis and ECM remodeling in the cornea [17,18], and induces angiogenesis and ECM degradation by a variety of other signaling pathways in addition to the bFGF pathway [5]. MMP-14 activity leads to the disruption of endothelial tight junctions, reorganization of the actin cytoskeleton, and proteolysis of the basement membrane and interstitial matrix. Furthermore, MMP-14 cleaves ECM molecules such as type I collagen, degrading the ECM as well as stimulating migration, organization, and guidance of vascular endothelial cells to form new blood vessels [19].

2.2. Fuchs Endothelial Corneal Dystrophy (FECD) and EDM Pathology

FECD, the most prevalent indication for endothelial keratoplasty in the United States [20], results in both CEC dysfunction and abnormal extracellular matrix deposition that are observable clinically as corneal edema and characteristic excrescences on the posterior cornea (guttae) [19]. Endothelial cell characteristics of FECD include channel protein dysfunction, mitochondrial dysfunction, reactive oxygen species accumulation, endoplasmic reticulum stress, DNA alterations, unfolded protein response, and cell apoptosis and dropout [21,22].
Experimental data using tissues from patients with FECD as well as expanded, immortalized cell cultures that model FECD demonstrate the overexpression of collagen 1, fibronectin, and collagen 4 in the EDM complex [23]. These protein expression changes are the result of zinc finger E-box binding homeobox 1 (ZEB1) and Snail1 activation in FECD, which can be stimulated and/or augmented with TGF-β exposure. The result of this disease-mediated overexpression is both thickening of Descemet membrane and buildup of matrix proteins that result in corneal guttae formation.
Corneal guttae indicate regions of endothelial cell dropout in FECD. Until recently, it was not clear whether extracellular matrix buildup caused cellular dropout or whether cell loss resulted in matrix changes and guttae formation. Importantly, Kocaba et al. have tested the impact that Descemet membrane tissues with guttae have on normal corneal endothelial cell health. After seeding healthy immortalized CECs on decellularized Descemet membrane explants from FECD patients undergoing surgery, they observed a decrease in the coverage area and number of normal cells seeded onto these abnormal membranes, and an increase in apoptosis surrounding large diameter guttae, compared to decellularized membranes from healthy guttae-free control donor corneas [24]. The same group also noted an increase in the expression of alpha-smooth muscle actin, N-cadherin, Snail1, and NOX4 in normal CECs that were grown in the presence of large diameter guttae. These expression footprints indicate that endothelial-mesenchymal transition (EndoMT) with loss of cell phenotype, as well as increased extracellular matrix component transcription, occur in conjunction with apoptotic cell death in FECD. Taken together, these findings indicate that the presence of extracellular matrix alterations alone are capable of causing altered expression of EndoMT proteins and additional matrix proteins, yielding a vicious cycle of aberrant cell and matrix changes that over time can result in Descemet membrane thickening, guttae formation, and ultimately endothelial cell phenotype loss and cell death (Figure 3).

2.3. Diabetes and EDM Pathology

Type II diabetes mellitus (T2DM) causes dramatic pathological effects on the corneal EDM complex and can result in vision loss due to corneal edema. Characteristic disease-related changes include lower than average CEC densities [25], greater than average postoperative cell loss [9], altered matrix properties altering EDM biomechanical stiffness [24], and reduced mitochondrial quality. Endothelial cell dropout in patients with diabetes may result in greater than average need for keratoplasty. Importantly, changes in both cell health and Descemet membrane that occur in tandem due to diabetes are important indicators for keratoplasty surgical outcomes, particularly as they relate to donor tissue health [8].
T2DM CECs have several functional changes that may explain the greater amount of cell loss observed in donor corneal tissues. As described by Aldrich et al., CECs from donors with a history of advanced diabetes have lower mitochondrial respiratory capacity than control cells, as measured using the Seahorse Extracellular Flux Analyzer [26]. Looking closely at diabetic CEC mitochondrial morphology using transmission electron microscopy, there are increases in cristae dropout, inclusion body formation, and average surface area in donor tissues with T2DM. Taken with the functional data, these findings demonstrate that the mitochondria in CECs with T2DM have a decreased functional capacity despite a larger surface area, and indicate an imbalance in mitochondrial dynamics related to mitochondrial fission and mitophagy (mitochondrial autophagy). Without proper clearance of dysfunctional mitochondria, CECs may become taxed to the point of cell death, leading to the higher amounts of dropout observed. Additional studies are indicated in this area, particularly given recent prospective randomized clinical trial data documenting increased graft failure rates and greater CEC loss three years following DSAEK surgery using diabetic graft tissues [9,10].
In addition to these functional changes, T2DM CECs have several structural matrix properties that may explain their tendency to tear during surgical preparation. Mechanical peel testing during controlled separation of donor corneal EDM tissues from stroma showed higher mean values for elastic peel tension (TE), average delamination tension (TD), and maximum tension (TMAX) in advanced diabetic donor corneas compared to non-diabetic donor corneas [27]. The region being peeled, between Descemet membrane and the stroma, is known as the interfacial matrix. Alterations occurring in diabetes to this region may be responsible for the higher mechanical peel test results, but further research is required. Using transmission electron microscopy (TEM), Rehany et al. found there were abnormal 120-nm wide-spaced collagen fibril bundles within both Descemet membrane and the stroma of noninsulin dependent diabetic patients (n = 16) compared to nondiabetic controls (n = 16) [28]. The authors hypothesized that the wide spacing was due to excessive glycosylation products. Together, the mechanical abnormalities and structural abnormalities in Descemet membrane, the interfacial matrix, and the stroma may explain the increased risk of tearing diabetic EDM grafts during donor tissue preparation for Descemet membrane endothelial keratoplasty [8].

3. Glaucoma

Abnormalities of the ECM have been implicated in some models of glaucomatous optic neuropathy. Cellular and extracellular matrix (ECM) interactions contribute to the resistance at the trabecular meshwork to aqueous outflow [29]. Increased deposition or impaired remodeling of extracellular matrix, changes in actin fiber contractility and arrangement, and regulatory derangements of cell adhesion all appear to play a role in the pathophysiology of glaucoma [30]. Vascular signaling also appears to play a role, as recent studies of nitric oxide and cyclic guanosine monophosphate signaling show alterations in vascular tone in many anterior segment structures as well as improved outflow facility [31,32]. This has been adapted for therapeutic use [33,34]. However, it is still unclear whether this signaling pathway involves any mediation or interaction with extracellular matrix, therefore the rest of this sub-section will focus on pathways that do.

3.1. Trabecular Meshwork

The ECM is a vital component of all three segments of the trabecular meshwork: the corneoscleral, uveoscleral, and juxtacanalicular layers. The trabecular meshwork ECM is comprised of numerous glycosaminoglycans and proteoglycans, collagens, elastic fibrils, basement membrane, and matrix proteins [29]. In the corneoscleral and uveoscleral layers, the trabecular meshwork cells wrap around these components to form the trabecular beams, between which are relatively large intertrabecular pores (Figure 4) [30]. In the juxtacanalicular layer, which is the site of highest resistance to aqueous outflow, trabecular meshwork cells have a more interwoven and irregular spatial relationship with ECM fibrils [35].
The components of the trabecular meshwork ECM are dynamic in that their expression or function can be induced by interaction with other components via bidirectional signaling, or as responses to environmental stimuli, such as mechanical stretch [30,36,37,38,39,40]. As one example, the matrix metalloproteinases (MMPs) can be induced by mechanical stretch, glucocorticoid steroids such as dexamethasone, laser trabeculoplasty, and the inflammatory cytokines TNF-α and TGF-β, leading to ECM remodeling and subsequent alterations to outflow facility [41,42,43]. Processes that impede MMP function and ECM remodeling may therefore decrease outflow facility [36,38,40,44,45]. Similarly, the expression and/or induction of numerous other proteoglycans and matricellular proteins, such as tenascin C, thrombospondin-1 and -2, SPARC (secreted protein, acidic and rich in cysteine), connective tissue growth factor (CTGF), fibronectin, various integrins, and periostin, have also been associated with changes in intraocular pressure [30,41,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67]. These mechanisms are not limited to ECM turnover, and also include modulation of such processes as cellular contraction, adhesion and migration, proliferation, and phagocytosis [60,68,69,70,71] (Figure 5). It has been suggested that cell–matrix interactions like these are mechanisms by which the trabecular meshwork can regulate intraocular pressure homeostasis [30], and elements in these interactive pathways, such as Rho kinase inhibitors, have shown promise as therapeutic targets [71,72,73].

3.2. Optic Nerve

Optic nerve head (ONH) remodeling in the glaucomatous excavation process also appears to be modulated by cellular and ECM interactions. The lamina cribrosa is a porous support structure for retinal ganglion cell axons passing through the scleral canal and out of the eye. The lamina cribrosa contains three cell types, including astrocytes, lamina cribrosa cells, and microglia [74,75,76], all of which are interspersed with the ECM. The ECM of the lamina cribrosa is comprised of proteoglycans as well as the relatively stiff collagens and relatively flexible elastin [77]. The last two mediate the distensibility of the lamina cribrosa, which overall becomes less mechanically compliant with age that is further accentuated in glaucoma [37,77,78]. In glaucomatous eyes, astrocytes at the prelaminar region have relatively enhanced expression of collagen type IV mRNA, while lamina cribrosa astrocytes have de novo expression of elastin mRNA. Animal models of glaucoma have more elastotic fibers at the lamina cribrosa compared to models of nerve transection or fellow eyes, suggesting that the increased elastin synthesis is a response to increased intraocular pressure [74]. Stress–strain models and analyses further support the notion that ECM deposition and dysregulation of ECM remodeling are eventual responses to mechanical stretch [37,79,80]. TGF-β1 and MMPs appear to have a role in mediating this pathway, and many other genes, including elastin, collagens IV, VI, VIII and IX, thrombospondin, perlecan, and lysl oxidase, show increased expression in response to 24 h cyclical stretch [42,75,81]. It has been postulated that the intraocular pressure-induced changes in the laminar ECM may even impede axonal nutrition despite stable laminar capillary flow [79].

3.3. Conjunctival and Tenon’s Layer

Subconjunctival fibrosis is also a process involving overproduction of ECM and is of particular interest for subconjunctival glaucoma surgeries such as trabeculectomy. After a filtration procedure, fibroblasts of Tenon’s layer (human Tenon’s fibroblasts, HTFs) may be chemoattracted to the surgical area by such factors as fibronectin in the aqueous [82]. after which there is increased ECM synthesis and collagen contraction [82,83,84]. Component proteins of the subconjunctival ECM include fibronectin and collagen type I, which are further induced by TGF-β1 [85]. One matricellular protein, SPARC, has been noted to be significantly increased in HTFs after exposure to TGF-β1 or TGF-β2, and relatively more SPARC is found in scarred blebs compared to normal Tenon’s. Additionally, SPARC-null knockout mice have HTFs that do not respond to TGF-β1, and filtration surgery in these mice functioned longer and with more expansive blebs than compared to wild type mice [86,87]. Another matricellular protein, CTGF, seems to promote bleb scarring and has been found to be overexpressed in filtration blebs [88,89]. Subconjunctival injection of a CTGF antibody after filtration surgery in rabbits also led to relatively larger blebs and lower intraocular pressure [90]. These matricellular proteins may hold promise as therapeutic targets for enhancing filtration surgery efficacy.

4. Vitreous

The vitreous humor is unique in that it is comprised almost entirely (>98%) of water but has various ECM components that give its gel-like consistency at birth and liquefies with age. Multiple blinding disorders can result from pathologic changes at the vitreoretinal interface. Normal vitreous is nearly completely acellular except for some macrophage-like hyalocytes [91]. The vitreous contains a network of glycosaminoglycans (GAGs), primarily hyaluronan, that supports a scaffold of collagen fibrils allowing a swelling osmotic gradient to inflate the gel [92].
The most prevalent form of collagen found in the vitreous is type II, which is secreted as procollagen prior to cleavage. Alternative splicing of exon 2 pre-mRNA can yield two forms: if exon 2 is expressed it is called procollagen IIA (which more prevalent in the vitreous); if exon 2 is excluded it is procollagen IIB [93,94]. Other collagens that are less prevalent but have a relatively prominent role in the vitreous include types V/XI, [95] IX, [93] XVIII, and VI [96].
Orientation of collagen fibrils varies in different regions of the eye. The central vitreous fibrils tend to course parallel in an anterior-posterior direction as opposed to the vitreous base where the fibrils insert directly into the internal limiting membrane (ILM) perpendicularly [97,98]. The precise mechanisms for adhesion of the vitreous to the ILM are not fully understood but differs at the vitreous base compared to the rest of the eye.

4.1. Age-Induced ECM Changes in the Vitreous Causing Retinal Detachment

Aging results in vitreous liquefaction and weakening of vitreoretinal adhesion that is associated with loss of type IX collagen and its chondroitin sulfate side-chains, and a four-fold increase in ‘sticky’ type II collagen predisposing to fibril fusion [99,100]. Opticin on the surface of cortical vitreous collagen fibrils may bind heparan sulphate proteoglycan chains on the ILM, including type XVIII collagen, which can also mediate vitreoretinal adhesion [101,102]. The role of opticin in angiogenesis from proliferative diabetic retinopathy (PDR) is discussed further in Section 5.1 but it is noteworthy that complete posterior vitreous detachment is protective against PDR as the collagenenous scaffold network for neovascularization is no longer present.
Vitreoretinal adhesion is critical in formation of retinal breaks and ensuing rhegmatogenous retinal detachment during the process of posterior vitreous detachment [103,104]. Lattice degeneration increases risk for retinal tears as there is increased vitreoretinal adhesion in these lesions with overlying vitreous liquefaction as well as alterations in the ILM, absence of basement membranes over the lattice, and increased presence of astrocytes [105,106].

4.2. Posteriorly Inserted Vitreous Base

The vitreous base can migrate posteriorly with advanced age that could be due to synthesis of new collagen by retinal cells [107]. Posteriorly inserted vitreous base (PIVB), generally defined as a wider than average vitreous base that straddles the ora serrata, has been observed in human donor eyes [108,109,110]. We defined vitreous base as posteriorly inserted if the posterior hyaloid membrane could not be elevated during pars plana vitrectomy anterior to the vortex veins, which approximates the equator of the eye that averages a distance of 7.6 mm posterior to the ora serrata in human donor eyes (Figure 6). PIVB can present challenges to eyes undergoing vitrectomy due to the increased number retinal tears (average 3.1) pre-operatively, high incidence of new breaks occurring during vitrectomy (30%), and increased risk for proliferative vitreoretinopathy needing re-operation [111]. Primary scleral buckle can be used for some of these cases but if vitrectomy is employed for a retinal detachment with PIVB, use of a wider buckle, meticulous shaving of the vitreous base, 360 degree laser, longer-acting tamponade agents, and potentially removal of crystalline lens [112] may help reduce rates of re-operation and vision loss [113].

5. Retina and Retinal Pigment Epithelium (RPE)

In the retina, ECM is organized into the interphotoreceptor matrix (IPM) and the retinal ECM (RECM) [4]. The IPM (Figure 7) represents the meshwork occupying the subretinal space between the photoreceptor cells and the retinal pigmented epithelium (RPE), and is comprised of a unique array of glycoproteins, while the RECM represents ECM outside the IPM. Structurally, ECM is found in basement membranes including the inner limiting membrane, the vasculature and Bruch’s membrane (BM) [1]. The major source of RECM are the Müller cells, intraretinal and migrating glial cells [114] while most of the IPM components, of which hyaluronan (HA) forms the basic scaffold, are synthesized by either the RPE or photoreceptors [115].
Within a given tissue, the ECM is a milieu in constant evolution and could show variation in its composition and organization over time [4]. For instance, age-related posterior vitreous detachment [116]. A well-known physiologic phenomenon, and ILM increasing in thickness and stiffness [117]. Both ECM-driven processes are mediated by changes in retinal cellular differentiation, migration, and adhesion [1,118,119,120]. Other physiologic processes related to ECM functionality and breakdown include tissue wound healing, innate immune defense, and angiogenesis [121,122,123,124].

5.1. Retinal Endothelial Cells and Angiogenesis

The pathophysiology of angiogenesis with relation to ECM are complex and not fully understood yet. Retinal angiogenesis is vitally important in age-related macular degeneration (AMD; discussed in Section 5.3) and blinding retinal vascular diseases such as proliferative diabetic retinopathy (PDR) and retinal vein occlusion, where preretinal neovascularization could result in massive pre-retinal hemorrhage, contractile fibrovascular membranes and tractional retinal detachment. Before detailing the role of ECM in vessel formation, it is useful to understand two concepts of angiogenesis: sprouting and intussusceptive [125]. In sprouting angiogenesis, new vessels are formed after an initial endothelial “tip cell” degrades the pre-existing vessel basement membrane, migrates into the surrounding ECM, proliferates and directs the remaining “stalk cells” in the formation of a cord [125]. This process is dependent on ECM-based growth factor signaling including vascular endothelial growth factor (VEGF) [125]. Intussusceptive angiogenesis, which is the basis for preretinal neovascularization in PDR [126] is different as the newly formed vessel emerges from the splitting of pre-existing blood vessels [125].
The ECM plays a critical role in most, if not all, aspects of vascular biology. ECM vasculogenic functionality includes the following: (1) supporting key signaling events in endothelial cell adhesion, proliferation and survival, (2) providing a scaffold and organizational cues for endothelial cells, (3) control and orchestration of the endothelial cells’ cytoskeleton via integrin-dependent signal transduction pathways, (4) tubulogenesis and three-dimensional remodeling of endothelial cell sheets, and (5) vessel maturation and stabilization [127].
Endothelial cell morphogenesis follows a programmed stepwise chain of events that starts with basement membrane breakdown, followed by cellular migration and proliferation, and ending with lumen formation and stabilization [128,129]. The concept of “fire and ice” was introduced to describe the role of endothelial ECM in vascular biosynthesis, remodeling, morphogenesis, and stabilization [127]. In fact, there is an ECM-based signaling balance which dictates when endothelial cells are activated or stabilized [127]. The process of activation will eventually lead to basement membrane degradation, cellular invasion, proliferation, migration and lumen formation. Both collagen (via interaction with α1β1, α2β1 integrins), fibrin, and fibronectin (via interaction with α5β1 and αVβ3 integrins) are essential for the “fire” chain of events, in conjunction with VEGF (Figure 8) [127].
During new blood vessel sprouting from pre-existing vasculature, membrane-type matrix metalloproteinases (MT-MMPs) are important regulators of cellular invasion into adjacent collagen or fibrin matrices due to their role in degrading ECM proteins at the cell surface–ECM interface [128,130,131,132]. MMPs themselves are regulated by tissue inhibitors of metalloproteinases (TIMPs), including TIMP-1, TIMP-2, TIMP-3, and TIMP-4, and it is the balance between MMPs and TIMPs that controls membrane degradation [132,133]. For instance, the “tip cell” utilizes MT1-MMP to degrade the surrounding ECM [134]. TIMP-2 and TIMP-3 are then subsequently produced when “stalk cells” contact pericytes in an attempt to halt the MT1-MMP induced ECM degradation [135].
Endothelial cells must then structurally adhere to the adjacent ECM in order to migrate [136]. The process of adhesion to ECM is mediated via specific surface integrins and ensures endothelial cell proliferation, survival and directional motility [137,138,139,140]. Endothelial cell proliferation is potentiated by activation of the p44/p42 (Erk1/Erk2) mitogen-activated protein kinase (MAPK) signal transduction pathway, which is itself activated by adhesion to fibronectin, a key ECM component [141,142,143]. Endothelial cell survival is also ensured by their adhesion to the ECM, which is a powerful regulator of Fas-induced apoptosis [144]. When attached to the ECM, endothelial cells are protected from apoptosis [144]. ECM modulates Fas-mediated apoptosis by altering the expression of Fas and c-Flip, an endogenous antagonist of caspase-8, which is a proteolytic enzyme involved in programmed cell death [144,145,146].
ECM also regulates endothelial cell morphogenesis and contractility. The matrix-integrin-cytoskeletal signaling axis results in both sprouting (= cord formation) and luminal vacuolization, which ultimately connect endothelial cells tubular structure together [127,128]. Collagen I promotes shape changes that lead to precapillary cord formation witnessed during angiogenesis [147,148,149]. Endothelial cells cytoskeletal contractility, which drive cord assembly, is potentiated by collagen I interaction with (1) integrins α1β1 and α2β1, which suppress the cAMP-dependent protein kinase A [150], and (2) integrin β1, which activates Src kinase and the GTPase Rho [151].
The interaction between laminin and each integrin α1β1, α2β1, α3β1 and α6β1 in conjunction with TGF-β are essential for “ice” chain of events which result in endothelial cell differentiation and stabilization via the cessation of cellular proliferation and related morphogenic sequelae [125,127,146].
It is important to note that most of the angiogenic pathways described above are highly conserved and are not unique to human species and their retina. Specifically, many genes necessary for animal multicellularity including fibronectin [152], cadherin [153], integrins [154], extracellular matrix domain [155,156], and VEGF machinery [157,158] code for ancient highly conserved proteins. Interestingly, many of these proteins were conserved all the way to the ‘‘Urmetazoan” [156] and multiple phyla [159]. These demonstrate the importance of these canonical pathways in vascular biology and, consequently, mammalian and non-mammalian species development [160,161,162].

5.2. Angiogenesis and Fibrosis in Proliferative Diabetic Retinopathy

As mentioned above, there is a substantial role for ECM in controlling intussusceptive angiogenesis, which is the basis for pre-retinal neovascularization seen in PDR [125,126]. This pathologic venous-based angiogenesis is an attempt to re-vascularize the ischemic retinal areas. In such cases, the cortical vitreous serves as a structural scaffold for pre-retinal neovascularization, specifically using vitreous ECM as a primary substrate for their formation [125,126]. Initially, the provisional vascular matrix was found to contain fibronectin and vitronectin [163], followed by collagen types I and III, which are deposited by fibroblast-like cells [125,164].
Interestingly, the vitreous normally hosts an anti-angiogenic milieu thanks to ECM components such as opticin, thrombospondins and endostatin (a fragment of type XVIII collagen) [125], which are well-known anti-angiogenic mediators [165,166,167]. In fact, opticin, which belongs to a family of ECM leucine-rich repeat proteoglycans [168], is a powerful dose-dependent inhibitor of preretinal neovascularization [169]. Hence, there must be a pro-angiogenic signaling shift on the collagen fibril surfaces within the vitreous ECM of PDR eyes. Opticin and other anti-angiogenic mediators could be affected by the enzymatic degradation of MMPs (1, 2, 3, 7, 8 and 9) and ADAMTS-4 and -5 (a family of extracellular protease enzymes, short for multi-domain extracellular protease enzyme) [170].
Research in PDR brought up the idea of an ‘angiofibrotic switch’ as a shift in balance between VEGF and CTGF mediating angiogenesis and fibrosis, respectively [171,172]. The basis for this work was done primarily on clinical grading and aqueous fluid extracted from patients. However, membranes removed from patients in a reverse translational, randomized controlled trial using VEGF inhibition for end-stage diabetic fibrovascular membranes demonstrated that VEGF and CTGF were not significantly different between intervention groups despite suppression of VEGF fluid levels in those that received bevacizumab (Figure 9) [173,174]. CTGF levels in the vitreous and aqueous were also unchanged in controls and those receiving bevacizumab, but a fair number of these patients had severely fibrotic, end-stage membranes where a change in CTGF would not have been as likely [174]. This study as well as others found that eyes receiving bevacizumab may have higher levels of apoptosis [174,175], supporting the notion that VEGF inhibition induces contraction of blood vessels rather than obliteration of them [176,177]. Endothelial-to-mesenchymal transition may be involved in diabetic membrane formation as there is evidence that endothelin-1, a potential vasoconstrictor that promotes fibrosis, is present at higher levels in diabetic compared to non-diabetic epiretinal membranes [178].

5.3. Choroidal Neovascularization, Autosomal Dominant Radial Drusen and AMD

In degenerative retinal diseases (whether acquired or inherited), there can be a tipping point at which the degenerative process is accelerated, leading to phenotypic manifestations of the disease [4]. This could be represented by the loss of a critical mass of retinal ECM [4]. Evidence supporting this hypothesis stems from numerous studies conducted in age-related macular degeneration (AMD), the most common cause of irreversible blindness in the developed world [179]. Drusen, which are extracellular lipid filled deposits between the RPE and the choriocapillaris, are the earliest hallmarks of AMD and tend to form over ECM areas with low density or absent choriocapillaris [180,181,182]. Additionally, drusen in AMD do not express collagen type IV in contrast to drusen to patients with genetic mutation in epidermal growth factor–containing fibrillin-like extracellular matrix protein 1 (EFEMP-1) causing autosomal dominant radial drusen (ADRD, aka Malattia Leventinese and Doyne Honeycomb Retinal Dystrophy) (Figure 10) [183]. Furthermore, the density of drusen correlates well with the density of “ghost” choriocapillaris vessels that is independent of RPE cell height [181,184,185].
The initial insult driving pathologic changes in AMD is not well understood. Early pathophysiological changes can be localized to the choriocapillaris, where an abundance of membrane attack complex (MAC) resulting in aberrant complement activation has been reported [182,186,187,188,189]. MAC based complement injury to choriocapillaries could be irreversible that leads to uncontrolled angiogenic drive and the formation of choroidal neovascular membranes (CNV) [185] and geographic atrophy [190]. Acute complement injuries have been associated with higher levels of MMP-3 and -9 [182].
The pathogenesis of CNV in AMD is complex and several interconnected pathways including genetic predisposition, oxidative stress, inflammatory/immune mechanism, and angiogenesis play a role [191,192]. Monogenic inherited retinal diseases directly affecting extracellular matrix such as ADRD from EFEMP-1 mutation results in early geographic atrophy and CNV, which is responsive to anti-VEGF therapy [193]. Despite the overwhelming success of anti-VEGF intravitreal injections (IVI) in treating active CNV due to exudative AMD [194,195,196], there is still a subset of incomplete respondents (~15%) who have persistent sub-retinal fluid (with or without intra-retinal fluid) despite chronic continued treatment [197,198,199]. Interestingly, some incomplete responders may initially show a good response to anti-VEGF IVI but then become treatment resistant and lose significant vision over time [200,201,202].
The mechanism of resistance to anti-VEGF IVI treatment is unknown. Tachyphylaxis was previously proposed as a possible explanation, especially in those eyes that show initial improvement [203,204,205]. There may also be a special role of MMP-mediated immune response in angiogenesis and CNV formation [206,207,208,209]. Of note, MMPs are essential in the degradation of ECM and basement membrane [210,211] and thus are crucial in tissue remodeling and repair [212,213]. Their major targets are elastin, fibrinogen, gelatin and various types of collagen molecules, including I, IV and V [214].
Most targets of MMP-9 are structural components of BM, which forms a major angiogenic barrier to CNV-based insult in exudative AMD [183,215,216]. There is mounting evidence of the causal relationship between MMPs, BM pathological remodeling, and CNV in AMD [215,217,218,219,220]. MMPs were reported to be increasingly expressed in pathologically stressed tissue, such as BM of eyes with AMD [221,222]. Breakdown of BM structural molecules (specifically elastin and collagen IV) allows for the migration of endothelial cells during angiogenesis [16,223,224]. Both MMP-9 enzymatic activity and the incidence of exudative AMD increase with age, suggesting a correlative risk [183,215,216]. MMP-9 is expressed in choroidal macrophages [225] and has also been found near BM the margins of CNV membranes [207]. Additionally, MMP-9 was found to increase the RPE VEGF levels by decreasing levels of pigment epithelium-derived factor (PEDF), which is the main antagonist of VEGF in the RPE [226,227,228]. Both VEGF and PEDF are highly expressed in AMD and their interplay serves as a mediator in the development of CNV [229,230,231,232,233]. A significant reduction in CNV incidence and severity was reported in MMP-9 knockout mice artificially subjected to laser injury [234,235]. In addition, inhibition of MMP-9 was experimentally found to block CNV development [235,236]. In humans, exogenous MMP-9 upregulated the gene expression of VEGF in human RPE cells [237,238]. In their study, Liutkevicien et al. showed a significant association between MMP-9 specific single nucleotide polymorphism and the incidence of AMD at a younger age (<65-year-old) [238]. Chau et al. also reported three folds higher plasma [239] and aqueous humour [240] levels of MMP-9 in AMD patients compared to healthy controls.
Furthermore, MMP-9 is known to interact with TIMP-3, mutations of which cause Sorsby fundus dystrophy in which CNV invariably develops patients by their 4th-5th decade of life [241,242,243]. Additionally, AMD eyes with marked choroidal thinning due to geographic atrophy have been reported to have a marked increase in TIMP-3 activity [244,245]. This is could then hinder the normal choroidal physiological angiogenic repair and contribute to the observed choroidal thinning [244]. In addition, there is a genetic association of the MMP-9 locus with exudative AMD, which was found in the International AMD Genetics Consortium in a large genome wide association study [245] and independently confirmed recently in an Iowa cohort of patients with AMD [246].

6. Bruch’s Membrane and Choroid

Bruch’s membrane is a complex, multilayered extracellular matrix compartment. It is comprised of two layers with salient features of fibrillar collagen surrounding a central layer of elastin and related molecules (Figure 11). While the basal laminae of the RPE and choriocapillaris are sometimes considered the inner and outer boundaries of Bruch’s membrane, increasingly investigators find it useful to consider these separately [247]. Both the structure and pathology of Bruch’s membrane are reminiscent of the arterial wall in atherosclerotic disease [248].
Bruch’s membrane itself has numerous functions: it serves as a barrier to abnormal neovascularization from the choroid, occupying the interface between the abundant vasculature of the choriocapillaris and the avascular outer retina. It contains both structural proteins and matricellular proteins. An abbreviated list of constituents of this unusual ECM compartment includes collagens I, III, IV, V, VI, VIII, fibrillin-1, fibulins 3 and 5, TIMP3, MMPs, and antiangiogenic effectors [249,250,251,252,253,254]. Several ECM constituents of Bruch’s membrane become greatly reduced during aging and macular disease, concomitant with the accumulation of lipidic debris [255,256], including the anti-angiogenic matricellular protein thrombospondin [257]. Age-related structural and biochemical changes in Bruch’s membrane are mirrored by an age-related decrease in hydraulic conductivity [258]. The elastic layer of Bruch’s membrane further becomes fragmented in AMD, with a loss of elastin integrity quantifiable both by histochemical staining and ultrastructural appearance [259]. Elastin degradation results in the liberation of elastin derived peptides (EDPs) which have been found to be elevated in the circulation of patients with neovascular AMD [260]. Liberated elastin fragments activate choroidal endothelial cells to migrate toward the source of the peptides, which ECs detect using a heterotrimeric cell surface elastin receptor.
The loss of elastin in Bruch’s membrane (whether due to increased metalloproteinase activity, macrophage extravasation, increased brittleness due to calcification, and/or other events) has the potential to both create a physical opening for growing vascular tubes as well as signaling choroidal endothelial cell migration. Moreover, human primary choroidal ECs stimulated with elastin fragments increase MMP-9 expression, potentially promoting further loss of Bruch’s membrane elastin and further amplification of angiogenesis [261]. The reader is referred to several reviews for more information (Figure 11) [216,262,263].
Outside of Bruch’s membrane, the choroidal ECM has been relatively understudied. The choroid is comprised of several cell types with their own basal laminae, including endothelial cells (which differ in composition between cells at different positions in the vascular tree and between the choriocapillaris and RPE) [253], pericytes/smooth muscle cells surrounding capillaries and large vessels respectively, and Schwann cells [225]. Other abundant cells such as melanocytes and especially fibroblasts contribute to ECM synthesis. The choroidal stroma, which occupies the space between and around vascular lumens, includes abundant fibrillar collagens with a ground substance containing a complex array of glycosaminoglycans including heparan sulfate and chondroitin sulfate proteoglycans [264]. Choroidal thinning, observed in normal aging and in geographic atrophy, is characterized by persistence of collagen fibrils and loss of ground substance, with a shift in the balance of serine protease inhibitors and metalloproteinase inhibitors as described above [244]. Other clinically meaningful aspects of choroidal thickness, such as the changes that occur in pachychoroid spectrum diseases like central serous chorioretinopathy, remain to be elucidated.

Author Contributions

Conceptualization R.F.M. and E.H.S.; methodology C.J., R.F.M. and E.H.S.; investigation, C.J., R.F.M. and E.H.S.; resources J.H.F., R.F.M., M.A.G. and E.H.S.; writing—original draft preparation, all authors; writing—review and editing, all authors; supervision, E.H.S.; project administration E.H.S.; funding acquisition J.H.F., R.F.M., M.A.G. and E.H.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by NEI R01 EY026547, P30 EY025580.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest related to this work. E.H.S. has received research funding from Oxford BioMedica. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Reinhard, J.; Joachim, S.C.; Faissner, A. Extracellular matrix remodeling during retinal development. Exp. Eye Res. 2015, 133, 132–140. [Google Scholar] [CrossRef]
  2. Yue, B. Biology of the extracellular matrix: An overview. J. Glaucoma 2014, 23, S20–S23. [Google Scholar] [CrossRef]
  3. Hubmacher, D.; Apte, S.S. The biology of the extracellular matrix: Novel insights. Curr. Opin. Rheumatol. 2013, 25, 65–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Al-Ubaidi, M.R.; Naash, M.I.; Conley, S.M. A perspective on the role of the extracellular matrix in progressive retinal degenerative disorders. Investig. Ophthalmol. Vis. Sci. 2013, 54, 8119–8124. [Google Scholar] [CrossRef] [Green Version]
  5. Chang, J.H.; Huang, Y.H.; Cunningham, C.M.; Han, K.Y.; Chang, M.; Seiki, M.; Zhou, Z.; Azar, D.T. Matrix metalloproteinase 14 modulates signal transduction and angiogenesis in the cornea. Surv. Ophthalmol. 2016, 61, 478–497. [Google Scholar] [CrossRef]
  6. Tshionyi, M.; Shay, E.; Lunde, E.; Lin, A.; Han, K.Y.; Jain, S.; Chang, J.H.; Azar, D.T. Hemangiogenesis and lymphangiogenesis in corneal pathology. Cornea 2012, 31, 74–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Bourne, W.M.; Nelson, L.R.; Hodge, D.O. Central corneal endothelial cell changes over a ten-year period. Investig. Ophthalmol. Vis. Sci. 1997, 38, 779–782. [Google Scholar]
  8. Greiner, M.A.; Rixen, J.J.; Wagoner, M.D.; Schmidt, G.A.; Stoeger, C.G.; Straiko, M.D.; Zimmerman, M.B.; Kitzmann, A.S.; Goins, K.M. Diabetes mellitus increases risk of unsuccessful graft preparation in Descemet membrane endothelial keratoplasty: A multicenter study. Cornea 2014, 33, 1129–1133. [Google Scholar] [CrossRef]
  9. Lass, J.H.; Benetz, B.A.; Patel, S.V.; Szczotka-Flynn, L.B.; O’Brien, R.; Ayala, A.R.; Maguire, M.G.; Daoud, Y.J.; Greiner, M.A.; Hannush, S.B.; et al. Donor, Recipient, and Operative Factors Associated With Increased Endothelial Cell Loss in the Cornea Preservation Time Study. JAMA Ophthalmol. 2019, 137, 185–193. [Google Scholar] [CrossRef] [PubMed]
  10. Terry, M.A.; Aldave, A.J.; Szczotka-Flynn, L.B.; Liang, W.; Ayala, A.R.; Maguire, M.G.; Croasdale, C.; Daoud, Y.J.; Dunn, S.P.; Hoover, C.K.; et al. Donor, Recipient, and Operative Factors Associated with Graft Success in the Cornea Preservation Time Study. Ophthalmology 2018, 125, 1700–1709. [Google Scholar] [CrossRef]
  11. Cursiefen, C.; Küchle, M.; Naumann, G.O. Angiogenesis in corneal diseases: Histopathologic evaluation of 254 human corneal buttons with neovascularization. Cornea 1998, 17, 611–613. [Google Scholar] [CrossRef]
  12. Netto, M.V.; Mohan, R.R.; Ambrosio, R., Jr.; Hutcheon, A.E.; Zieske, J.D.; Wilson, S.E. Wound healing in the cornea: A review of refractive surgery complications and new prospects for therapy. Cornea 2005, 24, 509–522. [Google Scholar] [CrossRef] [PubMed]
  13. Anitua, E.; de la Fuente, M.; Muruzabal, F.; Riestra, A.; Merayo-Lloves, J.; Orive, G. Plasma rich in growth factors (PRGF) eye drops stimulates scarless regeneration compared to autologous serum in the ocular surface stromal fibroblasts. Exp. Eye Res. 2015, 135, 118–126. [Google Scholar] [CrossRef]
  14. Warejcka, D.J.; Vaughan, K.A.; Bernstein, A.M.; Twining, S.S. Differential conversion of plasminogen to angiostatin by human corneal cell populations. Mol. Vis. 2005, 11, 859–868. [Google Scholar] [PubMed]
  15. Onguchi, T.; Han, K.Y.; Chang, J.H.; Azar, D.T. Membrane type-1 matrix metalloproteinase potentiates basic fibroblast growth factor-induced corneal neovascularization. Am. J. Pathol. 2009, 174, 1564–1571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kvanta, A.; Sarman, S.; Fagerholm, P.; Seregard, S.; Steen, B. Expression of matrix metalloproteinase-2 (MMP-2) and vascular endothelial growth factor (VEGF) in inflammation-associated corneal neovascularization. Exp. Eye Res. 2000, 70, 419–428. [Google Scholar] [CrossRef]
  17. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [CrossRef] [Green Version]
  18. Page-McCaw, A.; Ewald, A.J.; Werb, Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat. Rev. Mol. Cell Biol. 2007, 8, 221–233. [Google Scholar] [CrossRef] [PubMed]
  19. Sounni, N.E.; Paye, A.; Host, L.; Noel, A. MT-MMPS as Regulators of Vessel Stability Associated with Angiogenesis. Front. Pharm. 2011, 2, 111. [Google Scholar] [CrossRef] [Green Version]
  20. Sarnicola, C.; Farooq, A.V.; Colby, K. Fuchs Endothelial Corneal Dystrophy: Update on Pathogenesis and Future Directions. Eye Contact Lens 2019, 45, 1–10. [Google Scholar] [CrossRef] [PubMed]
  21. Vedana, G.; Villarreal, G., Jr.; Jun, A.S. Fuchs endothelial corneal dystrophy: Current perspectives. Clin. Ophthalmol. 2016, 10, 321–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Nanda, G.G.; Alone, D.P. REVIEW: Current understanding of the pathogenesis of Fuchs’ endothelial corneal dystrophy. Mol. Vis. 2019, 25, 295–310. [Google Scholar]
  23. Okumura, N.; Minamiyama, R.; Ho, L.T.; Kay, E.P.; Kawasaki, S.; Tourtas, T.; Schlotzer-Schrehardt, U.; Kruse, F.E.; Young, R.D.; Quantock, A.J.; et al. Involvement of ZEB1 and Snail1 in excessive production of extracellular matrix in Fuchs endothelial corneal dystrophy. Lab. Investig. 2015, 95, 1291–1304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Kocaba, V.; Katikireddy, K.R.; Gipson, I.; Price, M.O.; Price, F.W.; Jurkunas, U.V. Association of the Gutta-Induced Microenvironment With Corneal Endothelial Cell Behavior and Demise in Fuchs Endothelial Corneal Dystrophy. JAMA Ophthalmol. 2018, 136, 886–892. [Google Scholar] [CrossRef] [Green Version]
  25. Liaboe, C.A.; Aldrich, B.T.; Carter, P.C.; Skeie, J.M.; Burckart, K.A.; Schmidt, G.A.; Reed, C.R.; Zimmerman, M.B.; Greiner, M.A. Assessing the Impact of Diabetes Mellitus on Donor Corneal Endothelial Cell Density. Cornea 2017, 36, 561–566. [Google Scholar] [CrossRef] [PubMed]
  26. Aldrich, B.T.; Schlötzer-Schrehardt, U.; Skeie, J.M.; Burckart, K.A.; Schmidt, G.A.; Reed, C.R.; Zimmerman, M.B.; Kruse, F.E.; Greiner, M.A. Mitochondrial and Morphologic Alterations in Native Human Corneal Endothelial Cells Associated With Diabetes Mellitus. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2130–2138. [Google Scholar] [CrossRef]
  27. Schwarz, C.; Aldrich, B.T.; Burckart, K.A.; Schmidt, G.A.; Zimmerman, M.B.; Reed, C.R.; Greiner, M.A.; Sander, E.A. Descemet membrane adhesion strength is greater in diabetics with advanced disease compared to healthy donor corneas. Exp. Eye Res. 2016, 153, 152–158. [Google Scholar] [CrossRef] [PubMed]
  28. Rehany, U.; Ishii, Y.; Lahav, M.; Rumelt, S. Ultrastructural changes in corneas of diabetic patients: An electron-microscopy study. Cornea 2000, 19, 534–538. [Google Scholar] [CrossRef]
  29. Acott, T.S.; Kelley, M.J. Extracellular matrix in the trabecular meshwork. Exp. Eye Res. 2008, 86, 543–561. [Google Scholar] [CrossRef] [Green Version]
  30. Vranka, J.A.; Kelley, M.J.; Acott, T.S.; Keller, K.E. Extracellular matrix in the trabecular meshwork: Intraocular pressure regulation and dysregulation in glaucoma. Exp. Eye Res. 2015, 133, 112–125. [Google Scholar] [CrossRef] [Green Version]
  31. Pasquale, L.R. Vascular and autonomic dysregulation in primary open-angle glaucoma. Curr. Opin. Ophthalmol. 2016, 27, 94–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Buys, E.S.; Ko, Y.C.; Alt, C.; Hayton, S.R.; Jones, A.; Tainsh, L.T.; Ren, R.; Giani, A.; Clerté, M.; Abernathy, E.; et al. Soluble guanylate cyclase α1-deficient mice: A novel murine model for primary open angle glaucoma. PLoS ONE 2013, 8, e60156. [Google Scholar] [CrossRef] [PubMed]
  33. Cavet, M.E.; Vittitow, J.L.; Impagnatiello, F.; Ongini, E.; Bastia, E. Nitric oxide (NO): An emerging target for the treatment of glaucoma. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5005–5015. [Google Scholar] [CrossRef] [Green Version]
  34. Weinreb, R.N.; Ong, T.; Scassellati Sforzolini, B.; Vittitow, J.L.; Singh, K.; Kaufman, P.L. A randomised, controlled comparison of latanoprostene bunod and latanoprost 0.005% in the treatment of ocular hypertension and open angle glaucoma: The VOYAGER study. Br. J. Ophthalmol. 2015, 99, 738–745. [Google Scholar] [CrossRef] [Green Version]
  35. Tamm, E.R. The trabecular meshwork outflow pathways: Structural and functional aspects. Exp. Eye Res. 2009, 88, 648–655. [Google Scholar] [CrossRef] [PubMed]
  36. Bradley, J.M.; Vranka, J.; Colvis, C.M.; Conger, D.M.; Alexander, J.P.; Fisk, A.S.; Samples, J.R.; Acott, T.S. Effect of matrix metalloproteinases activity on outflow in perfused human organ culture. Investig. Ophthalmol. Vis. Sci. 1998, 39, 2649–2658. [Google Scholar]
  37. Hopkins, A.A.; Murphy, R.; Irnaten, M.; Wallace, D.M.; Quill, B.; O’Brien, C. The role of lamina cribrosa tissue stiffness and fibrosis as fundamental biomechanical drivers of pathological glaucoma cupping. Am. J. Physiol. Cell Physiol. 2020, 319, C611–C623. [Google Scholar] [CrossRef] [PubMed]
  38. Bradley, J.M.; Kelley, M.J.; Zhu, X.; Anderssohn, A.M.; Alexander, J.P.; Acott, T.S. Effects of mechanical stretching on trabecular matrix metalloproteinases. Investig. Ophthalmol. Vis. Sci. 2001, 42, 1505–1513. [Google Scholar]
  39. Chudgar, S.M.; Deng, P.; Maddala, R.; Epstein, D.L.; Rao, P.V. Regulation of connective tissue growth factor expression in the aqueous humor outflow pathway. Mol. Vis. 2006, 12, 1117–1126. [Google Scholar]
  40. Bradley, J.M.; Kelley, M.J.; Rose, A.; Acott, T.S. Signaling pathways used in trabecular matrix metalloproteinase response to mechanical stretch. Investig. Ophthalmol. Vis. Sci. 2003, 44, 5174–5181. [Google Scholar] [CrossRef] [Green Version]
  41. Vittal, V.; Rose, A.; Gregory, K.E.; Kelley, M.J.; Acott, T.S. Changes in Gene Expression by Trabecular Meshwork Cells in Response to Mechanical Stretching. Investig. Opthalmol. Vis. Sci. 2005, 46, 2857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kirwan, R.P.; Fenerty, C.H.; Crean, J.; Wordinger, R.J.; Clark, A.F.; O’Brien, C.J. Influence of cyclical mechanical strain on extracellular matrix gene expression in human lamina cribrosa cells in vitro. Mol. Vis. 2005, 11, 798–810. [Google Scholar]
  43. Parshley, D.E.; Bradley, J.M.; Samples, J.R.; Van Buskirk, E.M.; Acott, T.S. Early changes in matrix metalloproteinases and inhibitors after in vitro laser treatment to the trabecular meshwork. Curr. Eye Res. 1995, 14, 537–544. [Google Scholar] [CrossRef]
  44. Alexander, J.P.; Samples, J.R.; Van Buskirk, E.M.; Acott, T.S. Expression of matrix metalloproteinases and inhibitor by human trabecular meshwork. Investig. Ophthalmol. Vis. Sci. 1991, 32, 172–180. [Google Scholar]
  45. Alexander, J.P.; Samples, J.R.; Acott, T.S. Growth factor and cytokine modulation of trabecular meshwork matrix metalloproteinase and TIMP expression. Curr. Eye Res. 1998, 17, 276–285. [Google Scholar] [CrossRef] [PubMed]
  46. Keller, K.E.; Vranka, J.A.; Haddadin, R.I.; Kang, M.H.; Oh, D.J.; Rhee, D.J.; Yang, Y.F.; Sun, Y.Y.; Kelley, M.J.; Acott, T.S. The effects of tenascin C knockdown on trabecular meshwork outflow resistance. Investig. Ophthalmol. Vis. Sci. 2013, 54, 5613–5623. [Google Scholar] [CrossRef] [PubMed]
  47. Haddadin, R.I.; Oh, D.J.; Kang, M.H.; Filippopoulos, T.; Gupta, M.; Hart, L.; Sage, E.H.; Rhee, D.J. SPARC-null mice exhibit lower intraocular pressures. Investig. Ophthalmol. Vis. Sci. 2009, 50, 3771–3777. [Google Scholar] [CrossRef] [Green Version]
  48. Haddadin, R.I.; Oh, D.-J.; Kang, M.H.; Villarreal, G.; Kang, J.-H.; Jin, R.; Gong, H.; Rhee, D.J. Thrombospondin-1 (TSP1)–Null and TSP2-Null Mice Exhibit Lower Intraocular Pressures. Investig. Opthalmol. Vis. Sci. 2012, 53, 6708. [Google Scholar] [CrossRef]
  49. Yan, Q.; Clark, J.I.; Sage, E.H. Expression and characterization of SPARC in human lens and in the aqueous and vitreous humors. Exp. Eye Res. 2000, 71, 81–90. [Google Scholar] [CrossRef] [PubMed]
  50. Rhee, D.J.; Fariss, R.N.; Brekken, R.; Helene Sage, E.; Russell, P. The matricellular protein SPARC is expressed in human trabecular meshwork. Exp. Eye Res. 2003, 77, 601–607. [Google Scholar] [CrossRef]
  51. Kang, M.H.; Oh, D.-J.; Kang, J.-H.; Rhee, D.J. Regulation of SPARC by Transforming Growth Factor β2 in Human Trabecular Meshwork. Investig. Opthalmol. Vis. Sci. 2013, 54, 2523. [Google Scholar] [CrossRef] [Green Version]
  52. Bollinger, K.E.; Crabb, J.S.; Yuan, X.; Putliwala, T.; Clark, A.F.; Crabb, J.W. Quantitative Proteomics: TGFβ2Signaling in Trabecular Meshwork Cells. Investig. Opthalmol. Vis. Sci. 2011, 52, 8287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Tomarev, S.I.; Wistow, G.; Raymond, V.; Dubois, S.P.; Malyukova, I. Gene Expression Profile of the Human Trabecular Meshwork: NEIBank Sequence Tag Analysis. Investig. Opthalmol. Vis. Sci. 2003, 44, 2588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Junglas, B.; Yu, A.H.; Welge-Lüssen, U.; Tamm, E.R.; Fuchshofer, R. Connective tissue growth factor induces extracellular matrix deposition in human trabecular meshwork cells. Exp. Eye Res. 2009, 88, 1065–1075. [Google Scholar] [CrossRef] [PubMed]
  55. Wallace, D.M.; Clark, A.F.; Lipson, K.E.; Andrews, D.; Crean, J.K.; O’Brien, C.J. Anti-Connective Tissue Growth Factor Antibody Treatment Reduces Extracellular Matrix Production in Trabecular Meshwork and Lamina Cribrosa Cells. Investig. Opthalmol. Vis. Sci. 2013, 54, 7836. [Google Scholar] [CrossRef] [Green Version]
  56. Tripathi, B.J.; Tripathi, R.C.; Yang, C.; Millard, C.B.; Dixit, V.M. Synthesis of a thrombospondin-like cytoadhesion molecule by cells of the trabecular meshwork. Investig. Ophthalmol. Vis. Sci. 1991, 32, 181–188. [Google Scholar]
  57. Hiscott, P.; Schlötzer-Schrehardt, U.; Naumann, G.O.H. Unexpected expression of thrombospondin 1 by corneal and iris fibroblasts in the pseudoexfoliation syndrome. Hum. Pathol. 1996, 27, 1255–1258. [Google Scholar] [CrossRef]
  58. Faralli, J.A.; Filla, M.S.; Peters, D.M. Role of Fibronectin in Primary Open Angle Glaucoma. Cells 2019, 8, 1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Faralli, J.A.; Schwinn, M.K.; Gonzalez, J.M., Jr.; Filla, M.S.; Peters, D.M. Functional properties of fibronectin in the trabecular meshwork. Exp. Eye Res. 2009, 88, 689–693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Heimark, R.L.; Kaochar, S.; Stamer, W.D. Human Schlemm’s canal cells express the endothelial adherens proteins, VE-cadherin and PECAM-1. Curr. Eye Res. 2002, 25, 299–308. [Google Scholar] [CrossRef]
  61. Filla, M.S.; Faralli, J.A.; Peotter, J.L.; Peters, D.M. The role of integrins in glaucoma. Exp. Eye Res. 2017, 158, 124–136. [Google Scholar] [CrossRef] [Green Version]
  62. Murphy-Ullrich, J.E.; Downs, J.C. The Thrombospondin1-TGF-beta Pathway and Glaucoma. J. Ocul Pharm. Ther. 2015, 31, 371–375. [Google Scholar] [CrossRef]
  63. Nikhalashree, S.; Karthikkeyan, G.; George, R.; Shantha, B.; Vijaya, L.; Ratra, V.; Sulochana, K.N.; Coral, K. Lowered Decorin With Aberrant Extracellular Matrix Remodeling in Aqueous Humor and Tenon’s Tissue From Primary Glaucoma Patients. Investig. Opthalmol. Vis. Sci. 2019, 60, 4661. [Google Scholar] [CrossRef] [Green Version]
  64. Overby, D.R.; Bertrand, J.; Tektas, O.Y.; Boussommier-Calleja, A.; Schicht, M.; Ethier, C.R.; Woodward, D.F.; Stamer, W.D.; Lütjen-Drecoll, E. Ultrastructural changes associated with dexamethasone-induced ocular hypertension in mice. Investig. Ophthalmol. Vis. Sci. 2014, 55, 4922–4933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Li, G.; Lee, C.; Agrahari, V.; Wang, K.; Navarro, I.; Sherwood, J.M.; Crews, K.; Farsiu, S.; Gonzalez, P.; Lin, C.W.; et al. In vivo measurement of trabecular meshwork stiffness in a corticosteroid-induced ocular hypertensive mouse model. Proc. Natl. Acad. Sci. USA 2019, 116, 1714–1722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Mohd Nasir, N.A.; Agarwal, R.; Krasilnikova, A.; Sheikh Abdul Kadir, S.H.; Iezhitsa, I. Effect of dexamethasone on the expression of MMPs, adenosine A1 receptors and NFKB by human trabecular meshwork cells. J. Basic Clin. Physiol. Pharm. 2020. [Google Scholar] [CrossRef]
  67. Wallace, D.M.; Murphy-Ullrich, J.E.; Downs, J.C.; O’Brien, C.J. The role of matricellular proteins in glaucoma. Matrix Biol. 2014, 37, 174–182. [Google Scholar] [CrossRef] [Green Version]
  68. Buller, C.; Johnson, D.H.; Tschumper, R.C. Human trabecular meshwork phagocytosis. Observations in an organ culture system. Investig. Ophthalmol. Vis. Sci. 1990, 31, 2156–2163. [Google Scholar]
  69. Porter, K.M.; Epstein, D.L.; Liton, P.B. Up-Regulated Expression of Extracellular Matrix Remodeling Genes in Phagocytically Challenged Trabecular Meshwork Cells. PLoS ONE 2012, 7, e34792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Porter, K.; Lin, Y.; Liton, P.B. Cathepsin B Is Up-Regulated and Mediates Extracellular Matrix Degradation in Trabecular Meshwork Cells Following Phagocytic Challenge. PLoS ONE 2013, 8, e68668. [Google Scholar] [CrossRef] [Green Version]
  71. Chen, W.; Yang, X.; Fang, J.; Zhang, Y.; Zhu, W.; Yang, X. Rho-Associated Protein Kinase Inhibitor Treatment Promotes Proliferation and Phagocytosis in Trabecular Meshwork Cells. Front. Pharmacol. 2020, 11, 302. [Google Scholar] [CrossRef]
  72. Tian, B.; Kaufman, P.L. Effects of the Rho kinase inhibitor Y-27632 and the phosphatase inhibitor calyculin A on outflow facility in monkeys. Exp. Eye Res. 2005, 80, 215–225. [Google Scholar] [CrossRef]
  73. Yu, M.; Chen, X.; Wang, N.; Cai, S.; Li, N.; Qiu, J.; Brandt, C.R.; Kaufman, P.L.; Liu, X. H-1152 effects on intraocular pressure and trabecular meshwork morphology of rat eyes. J. Ocul. Pharm. Ther. 2008, 24, 373–379. [Google Scholar] [CrossRef]
  74. Hernandez, M.R. The optic nerve head in glaucoma: Role of astrocytes in tissue remodeling. Prog. Retin. Eye Res. 2000, 19, 297–321. [Google Scholar] [CrossRef]
  75. Yuan, L.; Neufeld, A.H. Activated microglia in the human glaucomatous optic nerve head. J. Neurosci. Res. 2001, 64, 523–532. [Google Scholar] [CrossRef]
  76. Wallace, D.M.; O’Brien, C.J. The role of lamina cribrosa cells in optic nerve head fibrosis in glaucoma. Exp. Eye Res. 2016, 142, 102–109. [Google Scholar] [CrossRef] [PubMed]
  77. Albon, J. Age related compliance of the lamina cribrosa in human eyes. Br. J. Ophthalmol. 2000, 84, 318–323. [Google Scholar] [CrossRef] [Green Version]
  78. Vogel, H. Influence of maturation and aging on mechanical and biochemical properties of connective tissue in rats. Mech. Ageing Dev. 1980, 14, 283–292. [Google Scholar] [CrossRef]
  79. Burgoyne, C.F.; Downs, J.C.; Bellezza, A.J.; Suh, J.K.; Hart, R.T. The optic nerve head as a biomechanical structure: A new paradigm for understanding the role of IOP-related stress and strain in the pathophysiology of glaucomatous optic nerve head damage. Prog. Retin. Eye Res. 2005, 24, 39–73. [Google Scholar] [CrossRef]
  80. Qu, J.; Chen, H.; Zhu, L.; Ambalavanan, N.; Girkin, C.A.; Murphy-Ullrich, J.E.; Downs, J.C.; Zhou, Y. High-Magnitude and/or High-Frequency Mechanical Strain Promotes Peripapillary Scleral Myofibroblast Differentiation. Investig. Opthalmol. Vis. Sci. 2015, 56, 7821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Yan, X.; Tezel, G.; Wax, M.B.; Edward, D.P. Matrix metalloproteinases and tumor necrosis factor alpha in glaucomatous optic nerve head. Arch. Ophthalmol. 2000, 118, 666–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Khaw, P.T.; Occleston, N.L.; Schultz, G.; Grierson, I.; Sherwood, M.B.; Larkin, G. Activation and suppression of fibroblast function. Eye 1994, 8, 188–195. [Google Scholar] [CrossRef] [Green Version]
  83. Daniels, J.T.; Occleston, N.L.; Crowston, J.G.; Cordeiro, M.F.; Alexander, R.A.; Wilkins, M.; Porter, R.; Brown, R.; Khaw, P.T. Understanding and controlling the scarring response: The contribution of histology and microscopy. Microsc Res. Tech. 1998, 42, 317–333. [Google Scholar] [CrossRef]
  84. Skuta, G.L.; Parrish, R.K. Wound healing in glaucoma filtering surgery. Surv. Ophthalmol. 1987, 32, 149–170. [Google Scholar] [CrossRef] [Green Version]
  85. Wu, N.; Chen, L.; Yan, D.; Zhou, M.; Shao, C.; Lu, Y.; Yao, Q.; Sun, H.; Fu, Y. Trehalose attenuates TGF-β1-induced fibrosis of hSCFs by activating autophagy. Mol. Cell. Biochem. 2020, 470, 175–188. [Google Scholar] [CrossRef]
  86. Fuchshofer, R.; Tamm, E.R. The role of TGF-β in the pathogenesis of primary open-angle glaucoma. Cell Tissue Res. 2012, 347, 279–290. [Google Scholar] [CrossRef] [PubMed]
  87. Seet, L.-F.; Su, R.; Barathi, V.A.; Lee, W.S.; Poh, R.; Heng, Y.M.; Manser, E.; Vithana, E.N.; Aung, T.; Weaver, M.; et al. SPARC Deficiency Results in Improved Surgical Survival in a Novel Mouse Model of Glaucoma Filtration Surgery. PLoS ONE 2010, 5, e9415. [Google Scholar] [CrossRef] [Green Version]
  88. Esson, D.W.; Neelakantan, A.; Iyer, S.A.; Blalock, T.D.; Balasubramanian, L.; Grotendorst, G.R.; Schultz, G.S.; Sherwood, M.B. Expression of Connective Tissue Growth Factor after Glaucoma Filtration Surgery in a Rabbit Model. Investig. Opthalmol. Vis. Sci. 2004, 45, 485. [Google Scholar] [CrossRef] [Green Version]
  89. Yuan, H.P.; Li, X.H.; Yang, B.B.; Shao, Z.B.; Yan, L.P. Expression of connective tissue growth factor after trabeculectomy in rabbits. Zhonghua Yan Ke Za Zhi 2009, 45, 168–174. [Google Scholar]
  90. Wang, J.-M.; Hui, N.; Fan, Y.-Z.; Xiong, L.; Sun, N.-X. Filtering bleb area and intraocular pressure following subconjunctival injection of CTGF antibody after glaucoma filtration surgery in rabbits. Int. J. Ophthalmol. 2011, 4, 480–483. [Google Scholar] [CrossRef]
  91. Qiao, H.; Hisatomi, T.; Sonoda, K.H.; Kura, S.; Sassa, Y.; Kinoshita, S.; Nakamura, T.; Sakamoto, T.; Ishibashi, T. The characterisation of hyalocytes: The origin, phenotype, and turnover. Br. J. Ophthalmol. 2005, 89, 513–517. [Google Scholar] [CrossRef]
  92. Bishop, P.N. Structural macromolecules and supramolecular organisation of the vitreous gel. Prog. Retin. Eye Res. 2000, 19, 323–344. [Google Scholar] [CrossRef]
  93. Bishop, P.N.; Crossman, M.V.; McLeod, D.; Ayad, S. Extraction and characterization of the tissue forms of collagen types II and IX from bovine vitreous. Biochem. J. 1994, 299 Pt 2, 497–505. [Google Scholar] [CrossRef] [Green Version]
  94. Bishop, P.N.; Reardon, A.J.; McLeod, D.; Ayad, S. Identification of alternatively spliced variants of type II procollagen in vitreous. Biochem. Biophys. Res. Commun. 1994, 203, 289–295. [Google Scholar] [CrossRef] [PubMed]
  95. Bos, K.J.; Holmes, D.F.; Kadler, K.E.; McLeod, D.; Morris, N.P.; Bishop, P.N. Axial structure of the heterotypic collagen fibrils of vitreous humour and cartilage. J. Mol. Biol. 2001, 306, 1011–1022. [Google Scholar] [CrossRef]
  96. Kielty, C.M.; Whittaker, S.P.; Grant, M.E.; Shuttleworth, C.A. Type VI collagen microfibrils: Evidence for a structural association with hyaluronan. J. Cell Biol. 1992, 118, 979–990. [Google Scholar] [CrossRef] [Green Version]
  97. Foos, R.Y. Posterior vitreous detachment. Trans. Am. Acad. Ophthalmol. Otolaryngol. 1972, 76, 480–497. [Google Scholar] [PubMed]
  98. Matsumoto, B.; Blanks, J.C.; Ryan, S.J. Topographic variations in the rabbit and primate internal limiting membrane. Investig. Ophthalmol. Vis. Sci. 1984, 25, 71–82. [Google Scholar]
  99. Bishop, P.N.; Holmes, D.F.; Kadler, K.E.; McLeod, D.; Bos, K.J. Age-related changes on the surface of vitreous collagen fibrils. Investig. Ophthalmol. Vis. Sci. 2004, 45, 1041–1046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Bos, K.J.; Holmes, D.F.; Meadows, R.S.; Kadler, K.E.; McLeod, D.; Bishop, P.N. Collagen fibril organisation in mammalian vitreous by freeze etch/rotary shadowing electron microscopy. Micron 2001, 32, 301–306. [Google Scholar] [CrossRef]
  101. Ramesh, S.; Bonshek, R.E.; Bishop, P.N. Immunolocalisation of opticin in the human eye. Br. J. Ophthalmol. 2004, 88, 697–702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Hindson, V.J.; Gallagher, J.T.; Halfter, W.; Bishop, P.N. Opticin binds to heparan and chondroitin sulfate proteoglycans. Investig. Ophthalmol. Vis. Sci. 2005, 46, 4417–4423. [Google Scholar] [CrossRef]
  103. Teng, C.C.; Chi, H.H. Vitreous changes and the mechanism of retinal detachment. Am. J. Ophthalmol. 1957, 44, 335–356. [Google Scholar] [CrossRef]
  104. Mitry, D.; Fleck, B.W.; Wright, A.F.; Campbell, H.; Charteris, D.G. Pathogenesis of rhegmatogenous retinal detachment: Predisposing anatomy and cell biology. Retina 2010, 30, 1561–1572. [Google Scholar] [CrossRef] [PubMed]
  105. Streeten, B.W.; Bert, M. The retinal surface in lattice degeneration of the retina. Am. J. Ophthalmol. 1972, 74, 1201–1209. [Google Scholar] [CrossRef]
  106. Straatsma, B.R.; Zeegen, P.D.; Foos, R.Y.; Feman, S.S.; Shabo, A.L. Lattice degeneration of the retina. XXX Edward Jackson Memorial Lecture. Am. J. Ophthalmol. 1974, 77, 619–649. [Google Scholar] [CrossRef]
  107. Wang, J.; McLeod, D.; Henson, D.B.; Bishop, P.N. Age-dependent changes in the basal retinovitreous adhesion. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1793–1800. [Google Scholar] [CrossRef]
  108. Schepens, C.L. Clinical aspects of pathologic changes in the vitreous body. Am. J. Ophthalmol. 1954, 38, 8–21. [Google Scholar] [CrossRef]
  109. Foos, R.Y.; Allen, R.A. Retinal tears and lesser lesions of the peripheral retina in autopsy eyes. Am. J. Ophthalmol. 1967, 64, 643–655. [Google Scholar] [CrossRef]
  110. Hogan, M.J. The vitreous, its structure, and relation to the ciliary body and retina. proctor award lecture. Investig. Ophthalmol. 1963, 2, 418–445. [Google Scholar]
  111. Sohn, E.H.; Strohbehn, A.; Stryjewski, T.; Brodowska, K.; Flamme-Wiese, M.J.; Mullins, R.F.; Eliott, D. Posteriorly inserted vitreous base: Preoperative Characteristics, Intraoperative Findings, and Outcomes After Vitrectomy. Retina 2020, 40, 943–950. [Google Scholar] [CrossRef]
  112. Shukla, D. Correspondence. Retina 2020, 40, e68. [Google Scholar] [CrossRef]
  113. Sohn, E.H.; Mullins, R.F.; Eliott, D. Reply. Retina 2020, 40, e68–e69. [Google Scholar] [CrossRef]
  114. Quinlan, R.; Nilsson, M. Reloading the retina by modifying the glial matrix. Trends Neurosci. 2004, 27, 241–242. [Google Scholar] [CrossRef]
  115. Adler, A.J.; Severin, K.M. Proteins of the bovine interphotoreceptor matrix: Tissues of origin. Exp. Eye Res. 1981, 32, 755–769. [Google Scholar] [CrossRef]
  116. De Smet, M.D.; Gad Elkareem, A.M.; Zwinderman, A.H. The vitreous, the retinal interface in ocular health and disease. Ophthalmologica 2013, 230, 165–178. [Google Scholar] [CrossRef] [PubMed]
  117. Candiello, J.; Cole, G.J.; Halfter, W. Age-dependent changes in the structure, composition and biophysical properties of a human basement membrane. Matrix Biol. 2010, 29, 402–410. [Google Scholar] [CrossRef]
  118. Hausman, R.E. Ocular extracellular matrices in development. Prog. Retin. Eye Res. 2007, 26, 162–188. [Google Scholar] [CrossRef]
  119. Oster, S.F.; Sretavan, D.W. Connecting the eye to the brain: The molecular basis of ganglion cell axon guidance. Br. J. Ophthalmol. 2003, 87, 639–645. [Google Scholar] [CrossRef]
  120. Pires Neto, M.A.; Braga-de-Souza, S.; Lent, R. Extracellular matrix molecules play diverse roles in the growth and guidance of central nervous system axons. Braz. J. Med. Biol Res. 1999, 32, 633–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Ardi, V.C.; Kupriyanova, T.A.; Deryugina, E.I.; Quigley, J.P. Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis. Proc. Natl. Acad. Sci. USA 2007, 104, 20262–20267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Giraudo, E.; Inoue, M.; Hanahan, D. An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervical carcinogenesis. J. Clin. Investig. 2004, 114, 623–633. [Google Scholar] [CrossRef]
  123. Nelissen, I.; Martens, E.; Van den Steen, P.E.; Proost, P.; Ronsse, I.; Opdenakker, G. Gelatinase B/matrix metalloproteinase-9 cleaves interferon-beta and is a target for immunotherapy. Brain A J. Neurol. 2003, 126, 1371–1381. [Google Scholar] [CrossRef] [Green Version]
  124. Vu, T.H.; Shipley, J.M.; Bergers, G.; Berger, J.E.; Helms, J.A.; Hanahan, D.; Shapiro, S.D.; Senior, R.M.; Werb, Z. MMP-9/gelatinase B is a key regulator of growth plate angiogenesis and apoptosis of hypertrophic chondrocytes. Cell 1998, 93, 411–422. [Google Scholar] [CrossRef] [Green Version]
  125. Bishop, P.N. The role of extracellular matrix in retinal vascular development and preretinal neovascularization. Exp. Eye Res. 2015, 133, 30–36. [Google Scholar] [CrossRef]
  126. McLeod, D. A chronic grey matter penumbra, lateral microvascular intussusception and venous peduncular avulsion underlie diabetic vitreous haemorrhage. Br. J. Ophthalmol. 2007, 91, 677–689. [Google Scholar] [CrossRef] [PubMed]
  127. Davis, G.E.; Senger, D.R. Endothelial extracellular matrix: Biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ. Res. 2005, 97, 1093–1107. [Google Scholar] [CrossRef] [Green Version]
  128. Davis, G.E.; Bayless, K.J.; Mavila, A. Molecular basis of endothelial cell morphogenesis in three-dimensional extracellular matrices. Anat. Rec. 2002, 268, 252–275. [Google Scholar] [CrossRef]
  129. Davis, G.E.; Bayless, K.J. An integrin and Rho GTPase-dependent pinocytic vacuole mechanism controls capillary lumen formation in collagen and fibrin matrices. Microcirculation 2003, 10, 27–44. [Google Scholar] [CrossRef]
  130. Heissig, B.; Hattori, K.; Friedrich, M.; Rafii, S.; Werb, Z. Angiogenesis: Vascular remodeling of the extracellular matrix involves metalloproteinases. Curr. Opin. Hematol. 2003, 10, 136–141. [Google Scholar] [CrossRef] [PubMed]
  131. Pepper, M.S. Role of the matrix metalloproteinase and plasminogen activator-plasmin systems in angiogenesis. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 1104–1117. [Google Scholar] [CrossRef] [Green Version]
  132. Bayless, K.J.; Davis, G.E. Sphingosine-1-phosphate markedly induces matrix metalloproteinase and integrin-dependent human endothelial cell invasion and lumen formation in three-dimensional collagen and fibrin matrices. Biochem. Biophys. Res. Commun. 2003, 312, 903–913. [Google Scholar] [CrossRef] [PubMed]
  133. Fassina, G.; Ferrari, N.; Brigati, C.; Benelli, R.; Santi, L.; Noonan, D.M.; Albini, A. Tissue inhibitors of metalloproteases: Regulation and biological activities. Clin. Exp. Metastasis 2000, 18, 111–120. [Google Scholar] [CrossRef]
  134. Yana, I.; Sagara, H.; Takaki, S.; Takatsu, K.; Nakamura, K.; Nakao, K.; Katsuki, M.; Taniguchi, S.; Aoki, T.; Sato, H.; et al. Crosstalk between neovessels and mural cells directs the site-specific expression of MT1-MMP to endothelial tip cells. J. Cell Sci. 2007, 120, 1607–1614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Saunders, W.B.; Bohnsack, B.L.; Faske, J.B.; Anthis, N.J.; Bayless, K.J.; Hirschi, K.K.; Davis, G.E. Coregulation of vascular tube stabilization by endothelial cell TIMP-2 and pericyte TIMP-3. J. Cell Biol. 2006, 175, 179–191. [Google Scholar] [CrossRef] [Green Version]
  136. Ausprunk, D.H.; Folkman, J. Migration and proliferation of endothelial cells in preformed and newly formed blood vessels during tumor angiogenesis. Microvasc. Res. 1977, 14, 53–65. [Google Scholar] [CrossRef]
  137. Akiyama, S.K.; Yamada, S.S.; Chen, W.T.; Yamada, K.M. Analysis of fibronectin receptor function with monoclonal antibodies: Roles in cell adhesion, migration, matrix assembly, and cytoskeletal organization. J. Cell Biol. 1989, 109, 863–875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Hynes, R.O. Integrins: Versatility, modulation, and signaling in cell adhesion. Cell 1992, 69, 11–25. [Google Scholar] [CrossRef]
  139. Giancotti, F.G.; Ruoslahti, E. Integrin signaling. Science 1999, 285, 1028–1032. [Google Scholar] [CrossRef] [PubMed]
  140. Meredith, J.E., Jr.; Schwartz, M.A. Integrins, adhesion and apoptosis. Trends Cell Biol. 1997, 7, 146–150. [Google Scholar] [CrossRef]
  141. Roovers, K.; Assoian, R.K. Integrating the MAP kinase signal into the G1 phase cell cycle machinery. Bioessays 2000, 22, 818–826. [Google Scholar] [CrossRef]
  142. Short, S.M.; Talbott, G.A.; Juliano, R.L. Integrin-mediated signaling events in human endothelial cells. Mol. Biol. Cell 1998, 9, 1969–1980. [Google Scholar] [CrossRef] [Green Version]
  143. Vinals, F.; Pouyssegur, J. Confluence of vascular endothelial cells induces cell cycle exit by inhibiting p42/p44 mitogen-activated protein kinase activity. Mol. Cell Biol. 1999, 19, 2763–2772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Aoudjit, F.; Vuori, K. Matrix attachment regulates Fas-induced apoptosis in endothelial cells: A role for c-flip and implications for anoikis. J. Cell Biol. 2001, 152, 633–643. [Google Scholar] [CrossRef]
  145. Salvesen, G.S. Programmed cell death and the caspases. APMIS 1999, 107, 73–79. [Google Scholar] [CrossRef]
  146. Holderfield, M.T.; Hughes, C.C. Crosstalk between vascular endothelial growth factor, notch, and transforming growth factor-beta in vascular morphogenesis. Circ. Res. 2008, 102, 637–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Fouser, L.; Iruela-Arispe, L.; Bornstein, P.; Sage, E.H. Transcriptional activity of the alpha 1(I)-collagen promoter is correlated with the formation of capillary-like structures by endothelial cells in vitro. J. Biol. Chem. 1991, 266, 18345–18351. [Google Scholar] [CrossRef]
  148. Montesano, R.; Orci, L.; Vassalli, P. In vitro rapid organization of endothelial cells into capillary-like networks is promoted by collagen matrices. J. Cell Biol. 1983, 97, 1648–1652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Sweeney, S.M.; Guy, C.A.; Fields, G.B.; San Antonio, J.D. Defining the domains of type I collagen involved in heparin- binding and endothelial tube formation. Proc. Natl. Acad. Sci. USA 1998, 95, 7275–7280. [Google Scholar] [CrossRef] [Green Version]
  150. Whelan, M.C.; Senger, D.R. Collagen I initiates endothelial cell morphogenesis by inducing actin polymerization through suppression of cyclic AMP and protein kinase A. J. Biol. Chem. 2003, 278, 327–334. [Google Scholar] [CrossRef] [Green Version]
  151. Liu, Y.; Senger, D.R. Matrix-specific activation of Src and Rho initiates capillary morphogenesis of endothelial cells. FASEB J. 2004, 18, 457–468. [Google Scholar] [CrossRef] [PubMed]
  152. Otey, C.A.; Burridge, K. Patterning of the membrane cytoskeleton by the extracellular matrix. Semin Cell Biol 1990, 1, 391–399. [Google Scholar] [PubMed]
  153. Nichols, S.A.; Roberts, B.W.; Richter, D.J.; Fairclough, S.R.; King, N. Origin of metazoan cadherin diversity and the antiquity of the classical cadherin/β-catenin complex. Proc. Natl. Acad. Sci. USA 2012, 109, 13046–13051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Suga, H.; Chen, Z.; de Mendoza, A.; Sebé-Pedrós, A.; Brown, M.W.; Kramer, E.; Carr, M.; Kerner, P.; Vervoort, M.; Sánchez-Pons, N.; et al. The Capsaspora genome reveals a complex unicellular prehistory of animals. Nat. Commun. 2013, 4, 2325. [Google Scholar] [CrossRef] [Green Version]
  155. King, N.; Westbrook, M.J.; Young, S.L.; Kuo, A.; Abedin, M.; Chapman, J.; Fairclough, S.; Hellsten, U.; Isogai, Y.; Letunic, I.; et al. The genome of the choanoflagellate Monosiga brevicollis and the origin of metazoans. Nature 2008, 451, 783–788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Williams, F.; Tew, H.A.; Paul, C.E.; Adams, J.C. The predicted secretomes of Monosiga brevicollis and Capsaspora owczarzaki, close unicellular relatives of metazoans, reveal new insights into the evolution of the metazoan extracellular matrix. Matrix Biol. 2014, 37, 60–68. [Google Scholar] [CrossRef]
  157. Erkenbrack, E.M.; Petsios, E. A Conserved Role for VEGF Signaling in Specification of Homologous Mesenchymal Cell Types Positioned at Spatially Distinct Developmental Addresses in Early Development of Sea Urchins. J. Exp. Zool B Mol. Dev. Evol. 2017, 328, 423–432. [Google Scholar] [CrossRef]
  158. Vega-Macaya, F.; Manieu, C.; Valdivia, M.; Mlodzik, M.; Olguín, P. Establishment of the Muscle-Tendon Junction During Thorax Morphogenesis in Drosophila Requires the Rho-Kinase. Genetics 2016, 204, 1139–1149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Tiozzo, S.; Voskoboynik, A.; Brown, F.D.; De Tomaso, A.W. A conserved role of the VEGF pathway in angiogenesis of an ectodermally-derived vasculature. Dev. Biol. 2008, 315, 243–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Vorotnikova, E.; McIntosh, D.; Dewilde, A.; Zhang, J.; Reing, J.E.; Zhang, L.; Cordero, K.; Bedelbaeva, K.; Gourevitch, D.; Heber-Katz, E.; et al. Extracellular matrix-derived products modulate endothelial and progenitor cell migration and proliferation in vitro and stimulate regenerative healing in vivo. Matrix Biol. 2010, 29, 690–700. [Google Scholar] [CrossRef] [PubMed]
  161. Adams, J.C. Matricellular Proteins: Functional Insights From Non-mammalian Animal Models. Curr. Top. Dev. Biol. 2018, 130, 39–105. [Google Scholar] [CrossRef]
  162. Witjas, F.M.R.; van den Berg, B.M.; van den Berg, C.W.; Engelse, M.A.; Rabelink, T.J. Concise Review: The Endothelial Cell Extracellular Matrix Regulates Tissue Homeostasis and Repair. Stem Cells Transl. Med. 2019, 8, 375–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Casaroli Marano, R.P.; Preissner, K.T.; Vilaro, S. Fibronectin, laminin, vitronectin and their receptors at newly-formed capillaries in proliferative diabetic retinopathy. Exp. Eye Res. 1995, 60, 5–17. [Google Scholar] [CrossRef]
  164. Hosoda, Y.; Okada, M.; Matsumura, M.; Ogino, N.; Honda, Y.; Nagai, Y. Intravitreal neovascular tissue of proliferative diabetic retinopathy: An immunohistochemical study. Ophthalmic Res. 1992, 24, 260–264. [Google Scholar] [CrossRef] [PubMed]
  165. Maatta, M.; Heljasvaara, R.; Pihlajaniemi, T.; Uusitalo, M. Collagen XVIII/endostatin shows a ubiquitous distribution in human ocular tissues and endostatin-containing fragments accumulate in ocular fluid samples. Graefe’s Arch. Clin. Exp. Ophthalmol. Albrecht Graefes Arch. Klin. Exp. Ophthalmol. 2007, 245, 74–81. [Google Scholar] [CrossRef]
  166. Sun, J.; Hopkins, B.D.; Tsujikawa, K.; Perruzzi, C.; Adini, I.; Swerlick, R.; Bornstein, P.; Lawler, J.; Benjamin, L.E. Thrombospondin-1 modulates VEGF-A-mediated Akt signaling and capillary survival in the developing retina. Am. J. Physiol. Heart Circ. Physiol. 2009, 296, H1344–H1351. [Google Scholar] [CrossRef] [Green Version]
  167. Wang, S.; Gottlieb, J.L.; Sorenson, C.M.; Sheibani, N. Modulation of thrombospondin 1 and pigment epithelium-derived factor levels in vitreous fluid of patients with diabetes. Arch. Ophthalmol. 2009, 127, 507–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Le Goff, M.M.; Hindson, V.J.; Jowitt, T.A.; Scott, P.G.; Bishop, P.N. Characterization of opticin and evidence of stable dimerization in solution. J. Biol. Chem. 2003, 278, 45280–45287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Le Goff, M.M.; Sutton, M.J.; Slevin, M.; Latif, A.; Humphries, M.J.; Bishop, P.N. Opticin exerts its anti-angiogenic activity by regulating extracellular matrix adhesiveness. J. Biol. Chem. 2012, 287, 28027–28036. [Google Scholar] [CrossRef] [Green Version]
  170. Tio, L.; Martel-Pelletier, J.; Pelletier, J.P.; Bishop, P.N.; Roughley, P.; Farran, A.; Benito, P.; Monfort, J. Characterization of opticin digestion by proteases involved in osteoarthritis development. Jt. Bone Spine 2014, 81, 137–141. [Google Scholar] [CrossRef] [PubMed]
  171. Kuiper, E.J.; Van Nieuwenhoven, F.A.; de Smet, M.D.; van Meurs, J.C.; Tanck, M.W.; Oliver, N.; Klaassen, I.; Van Noorden, C.J.; Goldschmeding, R.; Schlingemann, R.O. The angio-fibrotic switch of VEGF and CTGF in proliferative diabetic retinopathy. PLoS ONE 2008, 3, e2675. [Google Scholar] [CrossRef]
  172. Van Geest, R.J.; Lesnik-Oberstein, S.Y.; Tan, H.S.; Mura, M.; Goldschmeding, R.; Van Noorden, C.J.; Klaassen, I.; Schlingemann, R.O. A shift in the balance of vascular endothelial growth factor and connective tissue growth factor by bevacizumab causes the angiofibrotic switch in proliferative diabetic retinopathy. Br. J. Ophthalmol. 2012, 96, 587–590. [Google Scholar] [CrossRef]
  173. Sohn, E.H.; He, S.; Kim, L.A.; Salehi-Had, H.; Javaheri, M.; Spee, C.; Dustin, L.; Hinton, D.R.; Eliott, D. Angiofibrotic response to vascular endothelial growth factor inhibition in diabetic retinal detachment: Report no. 1. Arch. Ophthalmol. 2012, 130, 1127–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Jiao, C.; Eliott, D.; Spee, C.; He, S.; Wang, K.; Mullins, R.F.; Hinton, D.R.; Sohn, E.H. Apoptosis and angiofibrosis in diabetic tractional membranes after vascular endothelial growth factor inhibition: Results of a Prospective Trial. Report No. 2. Retina 2019, 39, 265–273. [Google Scholar] [CrossRef]
  175. Kubota, T.; Morita, H.; Tou, N.; Nitta, N.; Tawara, A.; Satoh, H.; Shimajiri, S. Histology of fibrovascular membranes of proliferative diabetic retinopathy after intravitreal injection of bevacizumab. Retina 2010, 30, 468–472. [Google Scholar] [CrossRef]
  176. Nakao, S.; Ishikawa, K.; Yoshida, S.; Kohno, R.; Miyazaki, M.; Enaida, H.; Kono, T.; Ishibashi, T. Altered vascular microenvironment by bevacizumab in diabetic fibrovascular membrane. Retina 2013, 33, 957–963. [Google Scholar] [CrossRef] [PubMed]
  177. Pattwell, D.M.; Stappler, T.; Sheridan, C.; Heimann, H.; Gibran, S.K.; Wong, D.; Hiscott, P. Fibrous membranes in diabetic retinopathy and bevacizumab. Retina 2010, 30, 1012–1016. [Google Scholar] [CrossRef] [PubMed]
  178. Chang, W.; Lajko, M.; Fawzi, A.A. Endothelin-1 is associated with fibrosis in proliferative diabetic retinopathy membranes. PLoS ONE 2018, 13, e0191285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Wong, W.L.; Su, X.; Li, X.; Cheung, C.M.; Klein, R.; Cheng, C.Y.; Wong, T.Y. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: A systematic review and meta-analysis. Lancet Glob. Health 2014, 2, e106–e116. [Google Scholar] [CrossRef] [Green Version]
  180. Lengyel, I.; Tufail, A.; Hosaini, H.A.; Luthert, P.; Bird, A.C.; Jeffery, G. Association of drusen deposition with choroidal intercapillary pillars in the aging human eye. Investig. Ophthalmol. Vis. Sci. 2004, 45, 2886–2892. [Google Scholar] [CrossRef] [Green Version]
  181. Mullins, R.F.; Johnson, M.N.; Faidley, E.A.; Skeie, J.M.; Huang, J. Choriocapillaris vascular dropout related to density of drusen in human eyes with early age-related macular degeneration. Investig. Ophthalmol. Vis. Sci. 2011, 52, 1606–1612. [Google Scholar] [CrossRef] [Green Version]
  182. Zeng, S.; Whitmore, S.S.; Sohn, E.H.; Riker, M.J.; Wiley, L.A.; Scheetz, T.E.; Stone, E.M.; Tucker, B.A.; Mullins, R.F. Molecular response of chorioretinal endothelial cells to complement injury: Implications for macular degeneration. J. Pathol. 2016, 238, 446–456. [Google Scholar] [CrossRef] [Green Version]
  183. Sohn, E.H.; Wang, K.; Thompson, S.; Riker, M.J.; Hoffmann, J.M.; Stone, E.M.; Mullins, R.F. Comparison of drusen and modifying genes in autosomal dominant radial drusen and age-related macular degeneration. Retina 2015, 35, 48–57. [Google Scholar] [CrossRef] [Green Version]
  184. Mullins, R.F.; Khanna, A.; Schoo, D.P.; Tucker, B.A.; Sohn, E.H.; Drack, A.V.; Stone, E.M. Is age-related macular degeneration a microvascular disease? Adv. Exp. Med. Biol. 2014, 801, 283–289. [Google Scholar] [CrossRef]
  185. Whitmore, S.S.; Sohn, E.H.; Chirco, K.R.; Drack, A.V.; Stone, E.M.; Tucker, B.A.; Mullins, R.F. Complement activation and choriocapillaris loss in early AMD: Implications for pathophysiology and therapy. Prog. Retin. Eye Res. 2015, 45, 1–29. [Google Scholar] [CrossRef] [Green Version]
  186. Anderson, D.H.; Radeke, M.J.; Gallo, N.B.; Chapin, E.A.; Johnson, P.T.; Curletti, C.R.; Hancox, L.S.; Hu, J.; Ebright, J.N.; Malek, G.; et al. The pivotal role of the complement system in aging and age-related macular degeneration: Hypothesis re-visited. Prog. Retin. Eye Res. 2010, 29, 95–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Hageman, G.S.; Anderson, D.H.; Johnson, L.V.; Hancox, L.S.; Taiber, A.J.; Hardisty, L.I.; Hageman, J.L.; Stockman, H.A.; Borchardt, J.D.; Gehrs, K.M.; et al. A common haplotype in the complement regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proc. Natl. Acad. Sci. USA 2005, 102, 7227–7232. [Google Scholar] [CrossRef] [Green Version]
  188. Mullins, R.F.; Schoo, D.P.; Sohn, E.H.; Flamme-Wiese, M.J.; Workamelahu, G.; Johnston, R.M.; Wang, K.; Tucker, B.A.; Stone, E.M. The membrane attack complex in aging human choriocapillaris: Relationship to macular degeneration and choroidal thinning. Am. J. Pathol. 2014, 184, 3142–3153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Seth, A.; Cui, J.; To, E.; Kwee, M.; Matsubara, J. Complement-associated deposits in the human retina. Investig. Ophthalmol. Vis. Sci. 2008, 49, 743–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Sohn, E.H.; Flamme-Wiese, M.J.; Whitmore, S.S.; Workalemahu, G.; Marneros, A.G.; Boese, E.A.; Kwon, Y.H.; Wang, K.; Abramoff, M.D.; Tucker, B.A.; et al. Choriocapillaris Degeneration in Geographic Atrophy. Am. J. Pathol. 2019, 189, 1473–1480. [Google Scholar] [CrossRef] [PubMed]
  191. Zarbin, M.A. Current concepts in the pathogenesis of age-related macular degeneration. Arch. Ophthalmol. 2004, 122, 598–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Ambati, J.; Ambati, B.K.; Yoo, S.H.; Ianchulev, S.; Adamis, A.P. Age-related macular degeneration: Etiology, pathogenesis, and therapeutic strategies. Surv. Ophthalmol. 2003, 48, 257–293. [Google Scholar] [CrossRef]
  193. Sohn, E.H.; Patel, P.J.; MacLaren, R.E.; Adatia, F.A.; Pal, B.; Webster, A.R.; Tufail, A. Responsiveness of choroidal neovascular membranes in patients with R345W mutation in fibulin 3 (Doyne honeycomb retinal dystrophy) to anti-vascular endothelial growth factor therapy. Arch. Ophthalmol. 2011, 129, 1626–1628. [Google Scholar] [CrossRef] [Green Version]
  194. Martin, D.F.; Maguire, M.G.; Ying, G.S.; Grunwald, J.E.; Fine, S.L.; Jaffe, G.J. Ranibizumab and bevacizumab for neovascular age-related macular degeneration. N. Engl. J. Med. 2011, 364, 1897–1908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Bakall, B.; Folk, J.C.; Boldt, H.C.; Sohn, E.H.; Stone, E.M.; Russell, S.R.; Mahajan, V.B. Aflibercept therapy for exudative age-related macular degeneration resistant to bevacizumab and ranibizumab. Am. J. Ophthalmol. 2013, 156, 15–22.e11. [Google Scholar] [CrossRef]
  196. Davis, A.S.; Folk, J.C.; Russell, S.R.; Sohn, E.H.; Boldt, H.C.; Stone, E.M.; Mahajan, V.B. Intravitreal bevacizumab for peripapillary choroidal neovascular membranes. Arch. Ophthalmol. 2012, 130, 1073–1075. [Google Scholar] [CrossRef] [Green Version]
  197. Krebs, I.; Glittenberg, C.; Ansari-Shahrezaei, S.; Hagen, S.; Steiner, I.; Binder, S. Non-responders to treatment with antagonists of vascular endothelial growth factor in age-related macular degeneration. Br. J. Ophthalmol. 2013, 97, 1443–1446. [Google Scholar] [CrossRef]
  198. Barthelmes, D.; Walton, R.; Campain, A.E.; Simpson, J.M.; Arnold, J.J.; McAllister, I.L.; Guymer, R.H.; Hunyor, A.P.; Essex, R.W.; Morlet, N.; et al. Outcomes of persistently active neovascular age-related macular degeneration treated with VEGF inhibitors: Observational study data. Br. J. Ophthalmol. 2015, 99, 359–364. [Google Scholar] [CrossRef]
  199. Tozer, K.; Roller, A.B.; Chong, L.P.; Sadda, S.; Folk, J.C.; Mahajan, V.B.; Russell, S.R.; Boldt, H.C.; Sohn, E.H. Combination therapy for neovascular age-related macular degeneration refractory to anti-vascular endothelial growth factor agents. Ophthalmology 2013, 120, 2029–2034. [Google Scholar] [CrossRef]
  200. Keane, P.A.; Liakopoulos, S.; Ongchin, S.C.; Heussen, F.M.; Msutta, S.; Chang, K.T.; Walsh, A.C.; Sadda, S.R. Quantitative subanalysis of optical coherence tomography after treatment with ranibizumab for neovascular age-related macular degeneration. Investig. Ophthalmol. Vis. Sci. 2008, 49, 3115–3120. [Google Scholar] [CrossRef]
  201. Forooghian, F.; Cukras, C.; Meyerle, C.B.; Chew, E.Y.; Wong, W.T. Tachyphylaxis after intravitreal bevacizumab for exudative age-related macular degeneration. Retina 2009, 29, 723–731. [Google Scholar] [CrossRef] [Green Version]
  202. Brown, D.M.; Tuomi, L.; Shapiro, H.; Pier Study, G. Anatomical measures as predictors of visual outcomes in ranibizumab-treated eyes with neovascular age-related macular degeneration. Retina 2013, 33, 23–34. [Google Scholar] [CrossRef] [Green Version]
  203. Schaal, S.; Kaplan, H.J.; Tezel, T.H. Is there tachyphylaxis to intravitreal anti-vascular endothelial growth factor pharmacotherapy in age-related macular degeneration? Ophthalmology 2008, 115, 2199–2205. [Google Scholar] [CrossRef] [PubMed]
  204. Binder, S. Loss of reactivity in intravitreal anti-VEGF therapy: Tachyphylaxis or tolerance? Br. J. Ophthalmol. 2012, 96, 1–2. [Google Scholar] [CrossRef] [Green Version]
  205. Gasperini, J.L.; Fawzi, A.A.; Khondkaryan, A.; Lam, L.; Chong, L.P.; Eliott, D.; Walsh, A.C.; Hwang, J.; Sadda, S.R. Bevacizumab and ranibizumab tachyphylaxis in the treatment of choroidal neovascularisation. Br. J. Ophthalmol. 2012, 96, 14–20. [Google Scholar] [CrossRef]
  206. Zeng, R.; Zhang, X.; Wu, K.; Su, Y.; Wen, F. MMP9 gene polymorphism is not associated with polypoidal choroidal vasculopathy and neovascular age-related macular degeneration in a Chinese Han population. Ophthalmic Genet. 2014, 35, 235–240. [Google Scholar] [CrossRef] [PubMed]
  207. Steen, B.; Sejersen, S.; Berglin, L.; Seregard, S.; Kvanta, A. Matrix metalloproteinases and metalloproteinase inhibitors in choroidal neovascular membranes. Investig. Ophthalmol. Vis. Sci. 1998, 39, 2194–2200. [Google Scholar]
  208. Tatar, O.; Adam, A.; Shinoda, K.; Eckert, T.; Scharioth, G.B.; Klein, M.; Yoeruek, E.; Bartz-Schmidt, K.U.; Grisanti, S. Matrix metalloproteinases in human choroidal neovascular membranes excised following verteporfin photodynamic therapy. Br. J. Ophthalmol. 2007, 91, 1183–1189. [Google Scholar] [CrossRef] [PubMed]
  209. Nussenblatt, R.B.; Ferris, F., 3rd. Age-related macular degeneration and the immune response: Implications for therapy. Am. J. Ophthalmol. 2007, 144, 618–626. [Google Scholar] [CrossRef] [Green Version]
  210. Egeblad, M.; Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer 2002, 2, 161–174. [Google Scholar] [CrossRef]
  211. Overall, C.M.; López-Otín, C. Strategies for MMP inhibition in cancer: Innovations for the post-trial era. Nat. Rev. Cancer 2002, 2, 657–672. [Google Scholar] [CrossRef] [PubMed]
  212. Shapiro, S.D. Matrix metalloproteinase degradation of extracellular matrix: Biological consequences. Curr. Opin. Cell Biol. 1998, 10, 602–608. [Google Scholar] [CrossRef]
  213. Lund, L.R.; Romer, J.; Bugge, T.H.; Nielsen, B.S.; Frandsen, T.L.; Degen, J.L.; Stephens, R.W.; Dano, K. Functional overlap between two classes of matrix-degrading proteases in wound healing. EMBO J. 1999, 18, 4645–4656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Burg-Roderfeld, M.; Roderfeld, M.; Wagner, S.; Henkel, C.; Grotzinger, J.; Roeb, E. MMP-9-hemopexin domain hampers adhesion and migration of colorectal cancer cells. Int. J. Oncol. 2007, 30, 985–992. [Google Scholar] [CrossRef] [PubMed]
  215. Chong, N.H.; Keonin, J.; Luthert, P.J.; Frennesson, C.I.; Weingeist, D.M.; Wolf, R.L.; Mullins, R.F.; Hageman, G.S. Decreased thickness and integrity of the macular elastic layer of Bruch’s membrane correspond to the distribution of lesions associated with age-related macular degeneration. Am. J. Pathol. 2005, 166, 241–251. [Google Scholar] [CrossRef]
  216. Curcio, C.A.; Johnson, M. Structure, function, and pathology of Bruch’s membrane. Retina 2013, 1, 466–481. [Google Scholar]
  217. Johnson, L.V.; Anderson, D.H. Age-related macular degeneration and the extracellular matrix. N. Engl. J. Med. 2004, 351, 320–322. [Google Scholar] [CrossRef] [PubMed]
  218. Ng, E.W.; Adamis, A.P. Targeting angiogenesis, the underlying disorder in neovascular age-related macular degeneration. Can. J. Ophthalmol. 2005, 40, 352–368. [Google Scholar] [CrossRef]
  219. Spraul, C.W.; Lang, G.E.; Grossniklaus, H.E.; Lang, G.K. Histologic and morphometric analysis of the choroid, Bruch’s membrane, and retinal pigment epithelium in postmortem eyes with age-related macular degeneration and histologic examination of surgically excised choroidal neovascular membranes. Surv. Ophthalmol. 1999, 44 (Suppl. 1), S10–S32. [Google Scholar] [CrossRef]
  220. Fiotti, N.; Pedio, M.; Parodi, M.B.; Altamura, N.; Uxa, L.; Guarnieri, G.; Giansante, C.; Ravalico, G. MMP-9 microsatellite polymorphism and susceptibility to exudative form of age-related macular degeneration. Genet. Med. 2005, 7, 272–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Guo, L.; Hussain, A.A.; Limb, G.A.; Marshall, J. Age-dependent variation in metalloproteinase activity of isolated human Bruch’s membrane and choroid. Investig. Ophthalmol. Vis. Sci. 1999, 40, 2676–2682. [Google Scholar]
  222. Kamei, M.; Hollyfield, J.G. TIMP-3 in Bruch’s membrane: Changes during aging and in age-related macular degeneration. Investig. Ophthalmol. Vis. Sci. 1999, 40, 2367–2375. [Google Scholar]
  223. Wissink, S.; van Heerde, E.C.; Schmitz, M.L.; Kalkhoven, E.; van der Burg, B.; Baeuerle, P.A.; van der Saag, P.T. Distinct domains of the RelA NF-kappaB subunit are required for negative cross-talk and direct interaction with the glucocorticoid receptor. J. Biol. Chem. 1997, 272, 22278–22284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Kvanta, A.; Shen, W.Y.; Sarman, S.; Seregard, S.; Steen, B.; Rakoczy, E. Matrix metalloproteinase (MMP) expression in experimental choroidal neovascularization. Curr. Eye Res. 2000, 21, 684–690. [Google Scholar] [CrossRef]
  225. Voigt, A.P.; Mulfaul, K.; Mullin, N.K.; Flamme-Wiese, M.J.; Giacalone, J.C.; Stone, E.M.; Tucker, B.A.; Scheetz, T.E.; Mullins, R.F. Single-cell transcriptomics of the human retinal pigment epithelium and choroid in health and macular degeneration. Proc. Natl. Acad. Sci. USA 2019, 116, 24100–24107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Oshima, Y.; Oshima, S.; Nambu, H.; Kachi, S.; Hackett, S.F.; Melia, M.; Kaleko, M.; Connelly, S.; Esumi, N.; Zack, D.J.; et al. Increased expression of VEGF in retinal pigmented epithelial cells is not sufficient to cause choroidal neovascularization. J. Cell Physiol 2004, 201, 393–400. [Google Scholar] [CrossRef] [PubMed]
  227. Samtani, S.; Amaral, J.; Campos, M.M.; Fariss, R.N.; Becerra, S.P. Doxycycline-mediated inhibition of choroidal neovascularization. Investig. Ophthalmol. Vis. Sci. 2009, 50, 5098–5106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Roychoudhury, J.; Herndon, J.M.; Yin, J.; Apte, R.S.; Ferguson, T.A. Targeting immune privilege to prevent pathogenic neovascularization. Investig. Ophthalmol. Vis. Sci. 2010, 51, 3560–3566. [Google Scholar] [CrossRef] [PubMed]
  229. Schwesinger, C.; Yee, C.; Rohan, R.M.; Joussen, A.M.; Fernandez, A.; Meyer, T.N.; Poulaki, V.; Ma, J.J.; Redmond, T.M.; Liu, S. Intrachoroidal neovascularization in transgenic mice overexpressing vascular endothelial growth factor in the retinal pigment epithelium. Am. J. Pathol. 2001, 158, 1161–1172. [Google Scholar] [CrossRef] [Green Version]
  230. Dawson, D.W.; Volpert, O.V.; Gillis, P.; Crawford, S.E.; Xu, H.; Benedict, W.; Bouck, N.P. Pigment epithelium-derived factor: A potent inhibitor of angiogenesis. Science 1999, 285, 245–248. [Google Scholar] [CrossRef]
  231. Ogata, N.; Wada, M.; Otsuji, T.; Jo, N.; Tombran-Tink, J.; Matsumura, M. Expression of pigment epithelium-derived factor in normal adult rat eye and experimental choroidal neovascularization. Investig. Ophthalmol. Vis. Sci. 2002, 43, 1168–1175. [Google Scholar]
  232. Becerra, S.P. Focus on Molecules: Pigment epithelium-derived factor (PEDF). Exp. Eye Res. 2006, 82, 739–740. [Google Scholar] [CrossRef] [PubMed]
  233. Gehlbach, P.; Demetriades, A.M.; Yamamoto, S.; Deering, T.; Duh, E.J.; Yang, H.S.; Cingolani, C.; Lai, H.; Wei, L.; Campochiaro, P.A. Periocular injection of an adenoviral vector encoding pigment epithelium-derived factor inhibits choroidal neovascularization. Gene Ther. 2003, 10, 637–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Lambert, V.; Munaut, C.; Jost, M.; Noel, A.; Werb, Z.; Foidart, J.M.; Rakic, J.M. Matrix metalloproteinase-9 contributes to choroidal neovascularization. Am. J. Pathol. 2002, 161, 1247–1253. [Google Scholar] [CrossRef] [Green Version]
  235. Lambert, V.; Wielockx, B.; Munaut, C.; Galopin, C.; Jost, M.; Itoh, T.; Werb, Z.; Baker, A.; Libert, C.; Krell, H.W.; et al. MMP-2 and MMP-9 synergize in promoting choroidal neovascularization. FASEB J. 2003, 17, 2290–2292. [Google Scholar] [CrossRef]
  236. Bergers, G.; Brekken, R.; McMahon, G.; Vu, T.H.; Itoh, T.; Tamaki, K.; Tanzawa, K.; Thorpe, P.; Itohara, S.; Werb, Z.; et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2000, 2, 737–744. [Google Scholar] [CrossRef]
  237. Hollborn, M.; Stathopoulos, C.; Steffen, A.; Wiedemann, P.; Kohen, L.; Bringmann, A. Positive feedback regulation between MMP-9 and VEGF in human RPE cells. Investig. Ophthalmol. Vis. Sci. 2007, 48, 4360–4367. [Google Scholar] [CrossRef] [Green Version]
  238. Liutkeviciene, R.; Lesauskaite, V.; Sinkunaite-Marsalkiene, G.; Zaliuniene, D.; Zaliaduonyte-Peksiene, D.; Mizariene, V.; Gustiene, O.; Jasinskas, V.; Jariene, G.; Tamosiunas, A. The Role of Matrix Metalloproteinases Polymorphisms in Age-Related Macular Degeneration. Ophthalmic Genet. 2015, 36, 149–155. [Google Scholar] [CrossRef]
  239. Chau, K.Y.; Sivaprasad, S.; Patel, N.; Donaldson, T.A.; Luthert, P.J.; Chong, N.V. Plasma levels of matrix metalloproteinase-2 and -9 (MMP-2 and MMP-9) in age-related macular degeneration. Eye 2008, 22, 855–859. [Google Scholar] [CrossRef]
  240. Jonas, J.B.; Tao, Y.; Neumaier, M.; Findeisen, P. Cytokine concentration in aqueous humour of eyes with exudative age-related macular degeneration. Acta Ophthalmol. 2012, 90, e381–e388. [Google Scholar] [CrossRef]
  241. Sivaprasad, S.; Webster, A.R.; Egan, C.A.; Bird, A.C.; Tufail, A. Clinical course and treatment outcomes of Sorsby fundus dystrophy. Am. J. Ophthalmol. 2008, 146, 228–234. [Google Scholar] [CrossRef] [PubMed]
  242. Rahman, N.; Georgiou, M.; Khan, K.N.; Michaelides, M. Macular dystrophies: Clinical and imaging features, molecular genetics and therapeutic options. Br. J. Ophthalmol. 2020, 104, 451–460. [Google Scholar] [CrossRef] [PubMed]
  243. Butler, G.S.; Apte, S.S.; Willenbrock, F.; Murphy, G. Human tissue inhibitor of metalloproteinases 3 interacts with both the N-and C-terminal domains of gelatinases A and B: Regulation by polyanions. J. Biol. Chem. 1999, 274, 10846–10851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Sohn, E.H.; Khanna, A.; Tucker, B.A.; Abramoff, M.D.; Stone, E.M.; Mullins, R.F. Structural and biochemical analyses of choroidal thickness in human donor eyes. Investig. Ophthalmol. Vis. Sci. 2014, 55, 1352–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Fritsche, L.G.; Igl, W.; Bailey, J.N.; Grassmann, F.; Sengupta, S.; Bragg-Gresham, J.L.; Burdon, K.P.; Hebbring, S.J.; Wen, C.; Gorski, M.; et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat. Genet. 2016, 48, 134–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Sohn, E.H.; Han, I.C.; Roos, B.R.; Faga, B.; Luse, M.A.; Binkley, E.M.; Boldt, H.C.; Folk, J.C.; Russell, S.R.; Mullins, R.F.; et al. Genetic association between MMP9 and choroidal neovascularization in age-related macular degeneration. Ophthalmol. Sci. 2020, 1, 100002. [Google Scholar] [CrossRef]
  247. Sura, A.A.; Chen, L.; Messinger, J.D.; Swain, T.A.; McGwin, G., Jr.; Freund, K.B.; Curcio, C.A. Measuring the Contributions of Basal Laminar Deposit and Bruch’s Membrane in Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2020, 61, 19. [Google Scholar] [CrossRef]
  248. Mullins, R.F.; McGwin, G., Jr.; Searcey, K.; Clark, M.E.; Kennedy, E.L.; Curcio, C.A.; Stone, E.M.; Owsley, C. The ARMS2 A69S Polymorphism Is Associated with Delayed Rod-Mediated Dark Adaptation in Eyes at Risk for Incident Age-Related Macular Degeneration. Ophthalmology 2019, 126, 591–600. [Google Scholar] [CrossRef]
  249. Karwatowski, W.S.; Jeffries, T.E.; Duance, V.C.; Albon, J.; Bailey, A.J.; Easty, D.L. Preparation of Bruch’s membrane and analysis of the age-related changes in the structural collagens. Br. J. Ophthalmol. 1995, 79, 944–952. [Google Scholar] [CrossRef] [Green Version]
  250. Hussain, A.A.; Lee, Y.; Zhang, J.J.; Marshall, J. Disturbed matrix metalloproteinase activity of Bruch’s membrane in age-related macular degeneration. Investig. Ophthalmol. Vis. Sci. 2011, 52, 4459–4466. [Google Scholar] [CrossRef]
  251. Fariss, R.N.; Apte, S.S.; Olsen, B.R.; Iwata, K.; Milam, A.H. Tissue inhibitor of metalloproteinases-3 is a component of Bruch’s membrane of the eye. Am. J. Pathol. 1997, 150, 323–328. [Google Scholar] [PubMed]
  252. Knupp, C.; Chong, N.H.; Munro, P.M.; Luthert, P.J.; Squire, J.M. Analysis of the collagen VI assemblies associated with Sorsby’s fundus dystrophy. J. Struct. Biol. 2002, 137, 31–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Chen, L.; Miyamura, N.; Ninomiya, Y.; Handa, J.T. Distribution of the collagen IV isoforms in human Bruch’s membrane. Br. J. Ophthalmol. 2003, 87, 212–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Bhutto, I.A.; Uno, K.; Merges, C.; Zhang, L.; McLeod, D.S.; Lutty, G.A. Reduction of endogenous angiogenesis inhibitors in Bruch’s membrane of the submacular region in eyes with age-related macular degeneration. Arch. Ophthalmol. 2008, 126, 670–678. [Google Scholar] [CrossRef] [Green Version]
  255. Haimovici, R.; Gantz, D.L.; Rumelt, S.; Freddo, T.F.; Small, D.M. The lipid composition of drusen, Bruch’s membrane, and sclera by hot stage polarizing light microscopy. Investig. Ophthalmol. Vis. Sci. 2001, 42, 1592–1599. [Google Scholar]
  256. Rudolf, M.; Curcio, C.A. Esterified cholesterol is highly localized to Bruch’s membrane, as revealed by lipid histochemistry in wholemounts of human choroid. J. Histochem. Cytochem. Off. J. Histochem. Soc. 2009, 57, 731–739. [Google Scholar] [CrossRef] [Green Version]
  257. Uno, K.; Bhutto, I.A.; McLeod, D.S.; Merges, C.; Lutty, G.A. Impaired expression of thrombospondin-1 in eyes with age related macular degeneration. Br. J. Ophthalmol. 2006, 90, 48–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Hussain, A.A.; Starita, C.; Hodgetts, A.; Marshall, J. Macromolecular diffusion characteristics of ageing human Bruch’s membrane: Implications for age-related macular degeneration (AMD). Exp. Eye Res. 2010, 90, 703–710. [Google Scholar] [CrossRef]
  259. Chang, J.H.; Spraul, C.W.; Lynn, M.L.; Drack, A.; Grossniklaus, H.E. The two-stage mutation model in retinal hemangioblastoma. Ophthalmic Genet. 1998, 19, 123–130. [Google Scholar] [CrossRef]
  260. Heng, L.Z.; Comyn, O.; Peto, T.; Tadros, C.; Ng, E.; Sivaprasad, S.; Hykin, P.G. Diabetic retinopathy: Pathogenesis, clinical grading, management and future developments. Diabet Med. 2013, 30, 640–650. [Google Scholar] [CrossRef]
  261. Skeie, J.M.; Hernandez, J.; Hinek, A.; Mullins, R.F. Molecular responses of choroidal endothelial cells to elastin derived peptides through the elastin-binding protein (GLB1). Matrix Biol. 2012, 31, 113–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Booij, J.C.; Baas, D.C.; Beisekeeva, J.; Gorgels, T.G.; Bergen, A.A. The dynamic nature of Bruch’s membrane. Prog. Retin. Eye Res. 2010, 29, 1–18. [Google Scholar] [CrossRef] [PubMed]
  263. Curcio, C.A.; Johnson, M.; Rudolf, M.; Huang, J.D. The oil spill in ageing Bruch membrane. Br. J. Ophthalmol. 2011, 95, 1638–1645. [Google Scholar] [CrossRef] [PubMed]
  264. Clark, S.J.; Keenan, T.D.; Fielder, H.L.; Collinson, L.J.; Holley, R.J.; Merry, C.L.; van Kuppevelt, T.H.; Day, A.J.; Bishop, P.N. Mapping the differential distribution of glycosaminoglycans in the adult human retina, choroid, and sclera. Investig. Ophthalmol. Vis. Sci. 2011, 52, 6511–6521. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Corneal stromal wound healing. Stromal wound healing in the cornea is mediated by signaling of transforming growth factor-beta (TGF-β), matrix metalloproteinases (MMPs), and a balance between pro-angiogenic and anti-angiogenic factors. In some cases, corneal neovascularization can occur.
Figure 1. Corneal stromal wound healing. Stromal wound healing in the cornea is mediated by signaling of transforming growth factor-beta (TGF-β), matrix metalloproteinases (MMPs), and a balance between pro-angiogenic and anti-angiogenic factors. In some cases, corneal neovascularization can occur.
Cells 10 00687 g001
Figure 2. Corneal neovascularization with stromal scarring secondary to atopic keratoconjunctivitis.
Figure 2. Corneal neovascularization with stromal scarring secondary to atopic keratoconjunctivitis.
Cells 10 00687 g002
Figure 3. Extracellular fibrosis (arrowhead) and dense guttae (**) in the posterior cornea secondary to Fuchs endothelial corneal dystrophy.
Figure 3. Extracellular fibrosis (arrowhead) and dense guttae (**) in the posterior cornea secondary to Fuchs endothelial corneal dystrophy.
Cells 10 00687 g003
Figure 4. Configuration of the trabecular meshwork. In both the corneoscleral and uveoscleral layers, the trabecular meshwork cells wrap around a core of extracellular matrix components. Note the increasingly large intertrabecular pores between the trabecular meshwork beams in the deeper layers. In the juxtacanalicular layer, the extracellular matrix and the trabecular meshwork cells have a more irregular and interwoven spatial relationship.
Figure 4. Configuration of the trabecular meshwork. In both the corneoscleral and uveoscleral layers, the trabecular meshwork cells wrap around a core of extracellular matrix components. Note the increasingly large intertrabecular pores between the trabecular meshwork beams in the deeper layers. In the juxtacanalicular layer, the extracellular matrix and the trabecular meshwork cells have a more irregular and interwoven spatial relationship.
Cells 10 00687 g004
Figure 5. Overview of cellular and extracellular matrix interactions in glaucomatous tissue remodeling. The interplay of numerous factors, including environmental stressors, enzymatic reactions, growth factors, glycoproteins and proteoglycans as well as cytoskeletal elements all contribute to a feedback loop where outflow facility is disturbed. Red circles indicate negative situational change. Green circle indicates positive situational change. Dark blue diamonds highlight critical signaling factors. Light blue diamonds indicate extracellular matrix glycoproteins and proteoglycans. Red rhombuses indicate negative catalysts. Green rhombus indicates positive catalysts. White circle indicates exogenous factors. ECM: Extracellular matrix; TGF: Transforming growth factor; CTGF: Connective tissue growth factor; α-SMA: Alpha smooth muscle actin; CLANs: Cross-linked actin networks; TIMP-1: Tissue inhibitors of metalloproteinases 1.
Figure 5. Overview of cellular and extracellular matrix interactions in glaucomatous tissue remodeling. The interplay of numerous factors, including environmental stressors, enzymatic reactions, growth factors, glycoproteins and proteoglycans as well as cytoskeletal elements all contribute to a feedback loop where outflow facility is disturbed. Red circles indicate negative situational change. Green circle indicates positive situational change. Dark blue diamonds highlight critical signaling factors. Light blue diamonds indicate extracellular matrix glycoproteins and proteoglycans. Red rhombuses indicate negative catalysts. Green rhombus indicates positive catalysts. White circle indicates exogenous factors. ECM: Extracellular matrix; TGF: Transforming growth factor; CTGF: Connective tissue growth factor; α-SMA: Alpha smooth muscle actin; CLANs: Cross-linked actin networks; TIMP-1: Tissue inhibitors of metalloproteinases 1.
Cells 10 00687 g005
Figure 6. Schematic of vitreous base inserting into the retina. At the pars plana normally (A), when it is posteriorly inserted (B), or at posterior the equator, averaging 7.6 mm posterior to the ora serrata which predisposes to more retinal tears (C). Adapted from Sohn et al. [111].
Figure 6. Schematic of vitreous base inserting into the retina. At the pars plana normally (A), when it is posteriorly inserted (B), or at posterior the equator, averaging 7.6 mm posterior to the ora serrata which predisposes to more retinal tears (C). Adapted from Sohn et al. [111].
Cells 10 00687 g006
Figure 7. Insoluble interphotoreceptor matrix glycoproteins, the gene products for IMPG1 and IMPG2 are distributed in domains surrounding rod and cone photoreceptors. The relative distributions of cone matrix sheaths labeled with peanut agglutinin (red) is depicted compared to rod outer segments labeled with anti-rhodopsin (green).
Figure 7. Insoluble interphotoreceptor matrix glycoproteins, the gene products for IMPG1 and IMPG2 are distributed in domains surrounding rod and cone photoreceptors. The relative distributions of cone matrix sheaths labeled with peanut agglutinin (red) is depicted compared to rod outer segments labeled with anti-rhodopsin (green).
Cells 10 00687 g007
Figure 8. Extracellular matrix mediated endothelial morphogenesis. The diagram illustrates the concept of “fire and ice” representing balance of extracellular matrix-based signaling which dictates endothelial cellular activation and tubulogenesis with endothelial cell stabilization. The complex process is mediated by the interaction between extracellular matrix components (including collagen I, fibrin, fibronectin and laminin) and various integrins. Abbreviations: VEGF: vascular endothelial growth factor; TGF-β: transforming growth factor beta.
Figure 8. Extracellular matrix mediated endothelial morphogenesis. The diagram illustrates the concept of “fire and ice” representing balance of extracellular matrix-based signaling which dictates endothelial cellular activation and tubulogenesis with endothelial cell stabilization. The complex process is mediated by the interaction between extracellular matrix components (including collagen I, fibrin, fibronectin and laminin) and various integrins. Abbreviations: VEGF: vascular endothelial growth factor; TGF-β: transforming growth factor beta.
Cells 10 00687 g008
Figure 9. Representative hematoxylin and eosin (H&E) and immunofluorescence images from four patients’ membranes in a randomized controlled trial. Co-labeling of antibodies for (A) CD31 (Green)-CTGF (Red) and (B) cytokeratin (Green)-VEGF (Red). Note the H&E-stained sections do not correspond precisely to the cytokeratin-labeled sections. While intravitreal bevacizumab did not significantly decrease CTGF (A-top panels) or VEGF (B-top panels) expression in membranes compared to sham group, VEGF was still expressed in membranes of eyes given bevacizumab (B, right panels). Scale bar = 100 μm. Abbreviations: CTGF: connective Tissue Growth Factor; VEGF: vascular endothelial growth factor. Adapted from Jiao et al. [174].
Figure 9. Representative hematoxylin and eosin (H&E) and immunofluorescence images from four patients’ membranes in a randomized controlled trial. Co-labeling of antibodies for (A) CD31 (Green)-CTGF (Red) and (B) cytokeratin (Green)-VEGF (Red). Note the H&E-stained sections do not correspond precisely to the cytokeratin-labeled sections. While intravitreal bevacizumab did not significantly decrease CTGF (A-top panels) or VEGF (B-top panels) expression in membranes compared to sham group, VEGF was still expressed in membranes of eyes given bevacizumab (B, right panels). Scale bar = 100 μm. Abbreviations: CTGF: connective Tissue Growth Factor; VEGF: vascular endothelial growth factor. Adapted from Jiao et al. [174].
Cells 10 00687 g009
Figure 10. Anti-collagen IV labeling in human donor eyes. Drusen (asterisks) associated with aging do not show labeling with antibodies directed against collagen type IV (AC). Laminae within the autosomal dominant radial drusen are immunoreactive with anti-collagen IV antibodies (green fluorescence). Sections were also labeled with DAPI (blue nuclear fluorescence) and were exposed in the rhodamine channel (red autofluorescence of the RPE). Scalebar = 50 μm. Adapted from Sohn et al. [183].
Figure 10. Anti-collagen IV labeling in human donor eyes. Drusen (asterisks) associated with aging do not show labeling with antibodies directed against collagen type IV (AC). Laminae within the autosomal dominant radial drusen are immunoreactive with anti-collagen IV antibodies (green fluorescence). Sections were also labeled with DAPI (blue nuclear fluorescence) and were exposed in the rhodamine channel (red autofluorescence of the RPE). Scalebar = 50 μm. Adapted from Sohn et al. [183].
Cells 10 00687 g010
Figure 11. Transmission electron micrograph depicting the layers of Bruch’s membrane from a human eye. Both the basal laminae of the RPE and choriocapillaris (RPE-BL and CC-BL) are depicted, in addition to inner collagenous zone (ICZ) and outer collagenous zone (OCZ), occupied by fibrillar collagens, as well as the elastic lamina (EL), evident by its thick electron dense bundles. Scale bar = 1 μm.
Figure 11. Transmission electron micrograph depicting the layers of Bruch’s membrane from a human eye. Both the basal laminae of the RPE and choriocapillaris (RPE-BL and CC-BL) are depicted, in addition to inner collagenous zone (ICZ) and outer collagenous zone (OCZ), occupied by fibrillar collagens, as well as the elastic lamina (EL), evident by its thick electron dense bundles. Scale bar = 1 μm.
Cells 10 00687 g011
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pouw, A.E.; Greiner, M.A.; Coussa, R.G.; Jiao, C.; Han, I.C.; Skeie, J.M.; Fingert, J.H.; Mullins, R.F.; Sohn, E.H. Cell–Matrix Interactions in the Eye: From Cornea to Choroid. Cells 2021, 10, 687. https://doi.org/10.3390/cells10030687

AMA Style

Pouw AE, Greiner MA, Coussa RG, Jiao C, Han IC, Skeie JM, Fingert JH, Mullins RF, Sohn EH. Cell–Matrix Interactions in the Eye: From Cornea to Choroid. Cells. 2021; 10(3):687. https://doi.org/10.3390/cells10030687

Chicago/Turabian Style

Pouw, Andrew E., Mark A. Greiner, Razek G. Coussa, Chunhua Jiao, Ian C. Han, Jessica M. Skeie, John H. Fingert, Robert F. Mullins, and Elliott H. Sohn. 2021. "Cell–Matrix Interactions in the Eye: From Cornea to Choroid" Cells 10, no. 3: 687. https://doi.org/10.3390/cells10030687

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop