Next Article in Journal
Possible Role of GnIH as a Novel Link between Hyperphagia-Induced Obesity-Related Metabolic Derangements and Hypogonadism in Male Mice
Next Article in Special Issue
Transcriptomic Analysis Reveals That Granulocyte Colony-Stimulating Factor Trigger a Novel Signaling Pathway (TAF9-P53-TRIAP1-CASP3) to Protect Retinal Ganglion Cells after Ischemic Optic Neuropathy
Previous Article in Journal
Membrane Fusion and SNAREs: Interaction with Ras Proteins
Previous Article in Special Issue
Neuromyelitis Optica Spectrum Disorder: From Basic Research to Clinical Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Remodeling of the Lamina Cribrosa: Mechanisms and Potential Therapeutic Approaches for Glaucoma

by
Ryan G. Strickland
1,
Mary Anne Garner
1,
Alecia K. Gross
1 and
Christopher A. Girkin
2,*
1
Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
2
Department of Ophthalmology and Vision Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(15), 8068; https://doi.org/10.3390/ijms23158068
Submission received: 1 June 2022 / Revised: 18 July 2022 / Accepted: 19 July 2022 / Published: 22 July 2022

Abstract

:
Glaucomatous optic neuropathy is the leading cause of irreversible blindness in the world. The chronic disease is characterized by optic nerve degeneration and vision field loss. The reduction of intraocular pressure remains the only proven glaucoma treatment, but it does not prevent further neurodegeneration. There are three major classes of cells in the human optic nerve head (ONH): lamina cribrosa (LC) cells, glial cells, and scleral fibroblasts. These cells provide support for the LC which is essential to maintain healthy retinal ganglion cell (RGC) axons. All these cells demonstrate responses to glaucomatous conditions through extracellular matrix remodeling. Therefore, investigations into alternative therapies that alter the characteristic remodeling response of the ONH to enhance the survival of RGC axons are prevalent. Understanding major remodeling pathways in the ONH may be key to developing targeted therapies that reduce deleterious remodeling.

1. Introduction

Glaucomatous optic neuropathy (GON) remains the leading cause of irreversible blindness worldwide, and the prevalence is expected to increase in the coming decades [1,2]. Glaucoma is a progressive optic neuropathy which is characterized, in part, by pronounced reorganization of cells in the lamina cribrosa (LC) and peripapillary sclera (ppScl). The variable loading forces imparted on the LC and ppScl by intraocular pressure (IOP), counterbalanced with cerebrospinal fluid (CSF) pressure, result in a region of high strain (tissue stretch) that impacts all ONH cell types and initiates cellular and extracellular matrix (ECM) remodeling. These cellular responses and subsequent ECM remodeling can negatively impact this milieu through which the projecting retinal ganglion cell (RGC) axons must traverse, and this may account for the increased vulnerability to further glaucomatous injury seen in the aged optic nerve or with increasing glaucoma severity (Figure 1).
While IOP lowering remains the only proven treatment, glaucoma can develop and progress even at normal or low levels of IOP. Thus, increasing interest in understanding potential pathways that modulate the pathologic remodeling in the LC and ppScl as a potential approach to develop novel “non-IOP” lowering treatments is emerging, and an abundance of work investigating the mechanisms that underly the ONH remodeling response has been conducted. The aim of this review is to describe the active responses of three major cell populations thought to be most critical to the remodeling response seen in the glaucomatous ONH: LC cells, glial cells, and scleral fibroblasts and to discuss potential therapeutic pathways. While each cell type serves a different purpose, each of these cell populations utilizes similar pathways to respond to the chemical and physical signals presented. Importantly, these responses appear to be consistent between animal models and human tissue culture models of the disease. While therapeutics aims at altering ECM remodeling are a promising potential treatment for glaucoma, it is not the only mechanism that can be exploited clinically.

2. Remodeling Response in Aging and Glaucoma

The ONH contains the LC, which is a thin multilayered, reticular load-bearing connective tissue that allows RGC axons and blood vessels to traverse this region of high strain while being supported by glial cells, LC cells, and a load-bearing collagenous matrix. Its unique structure makes it the “weak point” within the sclero-corneal shell where mechanical strain from changes in internal and external pressures on the globe are focused. The LC inserts and anchors itself into the ppScl which provides substantial support to counteract IOP [3,4,5]. In fact, the ppScl may experience the greatest amount of strain in response to elevated IOP [6,7]. The LC and ppScl also receive counteracting pressure from the post-laminar CSF [8]. Since the LC and overlying peripapillary choroid are perfused via branches of the posterior ciliary arteries that are encased in the ppScl, these vessels are subjected to direct mechanical forces as well. Thus, the classic vascular and mechanical theories of glaucoma pathogenesis are inseparably intertwined with the mechanical behavior of the LC and ppScl directly impacting perfusion and vice versa.
As with any load bearing structure, the amount of deformation (strain) experienced by the LC and ppScl is dependent on the morphology and material properties of these tissues, both of which are altered by both age-related and glaucomatous remodeling. Thus, the mechanocelluar response of the tissues, which is driven by strain, directly modifies the structure and material properties (stiffness) of the LC and ppScl which, in turn, alters the strain driving the remodeling. This dynamic creates a feedforward mechanism that may result in an increasingly pathologic milieu. This mechanism may account for the increased susceptibility to glaucomatous injury seen with aging and with increasing glaucoma severity observed across several prospective glaucoma studies [9,10,11,12,13].
A key pathologic characteristic of the glaucomatous ONH is ECM disorganization [14,15,16]. The generation of new ECM is an important component of the glaucomatous response and reorganization of the existing ECM is vital to understand for the development of new treatments. Animal models and ex vivo testing of human cadaveric tissues has shown that the sclera is known to stiffen with age and glaucoma, driving increased strain to the ONH [17,18]. In the non-human primate model of glaucoma, LC structure is dramatically disturbed, and collagen density is altered differently depending on the collagen subtype [14,19,20].
The molecular mechanisms of ECM reorganization center largely on the transforming growth factor-β (TGF-β) pathway, although other pathways are likely implicated. TGF-β is typically inactivated by latency associated peptide (LAP) and enzymes that cleave LAP can consequently activate TGF-β, allowing it to bind to a TGF-β receptor complex and activate downstream Smads that control transcription (Figure 2) [21]. In turn, this pathway results in the increased production of ECM molecules and proteins [22,23,24]. There are several potential mechanisms for TGF-β activation. Interactions with matrix metalloproteinases (MMPs), integrins, and thrombospondin (TSP) can all trigger TGF-β activation [25,26].
There are a multitude of enzymes in the MMP family with various substrate specificity and all contribute to the degradation of ECM components such as collagen, gelatins, laminin, and more [27,28]. In the glaucomatous ONH, MMP-1, -2, -3, and -14 have all demonstrated upregulation [29]. The TSP family has been implicated in a variety of fibrotic pathologies due to its ability to activate TGF-β, and it has implications in ECM remodeling and in IOP levels in knock-out mouse models [30,31]. However, evidence suggests that expression levels of TSP isoforms may vary depending on disease stage, such as low TSP-4 expression in early glaucoma that increases in late stages of the disease [32]. Lastly, integrin signaling-mediated activation of TGF-β may also be more pertinent to glial cells and vascular endothelial cells in the ONH [33]. In total, the elements of this pathway have multiple different implications on the profibrotic responses of the cell types discussed below. Cellular responses include generation of newly synthesized ECM, ECM editing, cellular migration, and cellular contractility. Overall, there are four cell types involved in ONH remodeling including the lamina cribrosa (LC) cells, which are in contact with the laminar beams, astrocytes and microglia cells within the pores of the LC, and scleral fibroblasts within the ppScl.

3. Lamina Cribrosa Cells

LC cells are typically differentiated from regional astrocytes by the lack of GFAP, constitutive expression of α-smooth muscle actin (α-SMA), alternate shape, and localization among other factors [34,35]. While LC cells typically maintain the supportive laminar beams and ECM through production of collagen, elastin, and fibronectin [35], they are also capable of dynamic reactions to external stimuli that can alter the properties of the ECM. For example, human LC cells in culture exposed to mechanical strain demonstrated altered expression levels of multiple genes that implicate ECM components, cell proliferation, growth factors, and cell surface receptors [36]. In addition to mechanical strain, LC cells can also respond to oxidative stress by upregulating fibrotic genes and production of collagen and α-SMA [37]. Human LC cells cultured in hypoxic conditions also demonstrate increased expression of ECM-related factors such as macrophage migration inhibitory factor and discoidin domain receptor [38,39,40]. This evidence shows that mechanical strain, oxidative stress, and hypoxia, all potentially relevant to the pathogenesis of primary open-angle glaucoma (POAG), cause LC cells to express and secrete collagen as well as other fibrotic molecules.
LC cells are likely to play a critical role in reorganization and remodeling of the LC in response to mechanical activation also through the TGF-β pathway. Specifically, MMP-2 expression and activity are both increased in response to glaucomatous conditions [24,34,41]. MMP and TSP1 expression is upregulated in LC cells responding to mechanical stress [24,36]. Increased expression and secretion of active MMPs that digest ECM components are likely a major component of ECM disorganization in response to strain. Additionally, LC cells use the ECM matrix as a scaffolding and with localized degradation, LC cells may use the detachment to migrate within the ONH which could underlie the ability of the LC to migrate posteriorly within the ONH [42,43]. There is evidence that application of human TGF-β2 also stimulates MMP activity in porcine LC cells which further supports the role of this pathway in another animal model [44]. Unfortunately, rodent ONHs do not contain LC cells and, though it has been noted previously, there remains no published attempts to culture primate LC cells [45].
As previously mentioned, these notable ECM modifying proteins can also function as activators of latent TGF-β. However, there is also evidence of the reverse: active TGF-β and the subsequent Smad transcriptional regulation pathway controls the expression level of its own activating partners. For instance, application of TGF-β1 to cultured human LC cells induced greater expression of TSP [23]. This evidence suggests that the initial triggers of the TGF-β activation initiate a feed-forward mechanism of ECM remodeling in the LC that has deleterious effects on the axons of the RGCs in the region [26]. This feed-forward signaling mechanism has been demonstrated in the other cell types described below.
TGF-β, as well as mechanical and oxidative stress, can also influence aspects of ECM regulation in LC cells through calcium-dependent pathways. For instance, LC cells cultured from glaucomatous human eyes demonstrated dysregulation of calcium, such as high intracellular levels in response to previously mentioned stimuli and a reduced ability to sequester free cytoplasmic calcium [46,47]. Increased levels of cellular calcium can have a variety of effects on signaling pathways due to the dynamic nature of calcium as a second messenger. Of those pathways, the activation of nuclear factor of activated T-cells (NFAT) may be most relevant to LC cells. In short, calcineurin can bind calcium to calmodulin, which can dephosphorylate NFAT. NFAT then complexes with transcription factors to influence transcription of genes including those that modulate the ECM [48]. While inhibition of this pathway may aid in the treatment of glaucoma, it is not fully understood what precedes the loss of calcium regulation in glaucomatous LC cells. However, one potential preceding factor may be the presence of transient receptor potential canonical (TRPC) channels, a class of voltage-independent channels that preferentially respond to calcium ions and are not necessarily dependent on stimuli such as TGF-β or oxidative and mechanical stresses [49,50]. Interestingly, isoforms of TRPCs, such as 1 and 6, are significantly overexpressed in glaucomatous LC cells cultured from human ONHs [51]. The increased presence of these channels may disturb homeostatic levels of calcium in LC cells, thereby inducing dysregulation. Additionally, these channels modulate transcription of ECM components such as TGF-β, α-SMA, collagen, and MMPs likely through the NFAT pathway described previously [49,51]. Furthermore, TRPC-1 and -6, at least in cancerous cells of the central nervous system, also regulate cell migration [49,52,53]. While these mechanisms of migration have not directly been shown in LC cells, it is possible that glaucomatous LC cells overexpressing these TRPCs may initiate signaling cascades that increase the degree to which LC cells migrate within the LC, potentially contributing to posterior LC migration.

4. Glial Cells

The ONH also contains a resident population of glial cells that create the blood–brain barrier (BBB) and myelinate the RGC axons in the post-laminar region. Dormant microglia are also primed for reactionary responses to local insult or disease [29,54]. Astrocytes, like LC cells, demonstrate mechanosensitive properties and are reactive to glaucomatous conditions in humans and other animal models [55,56,57,58]. In the healthy ONH, astrocytes typically support the BBB, but type 1B astrocytes, the dominant subtype in the ONH, can also assist the LC cells in producing the ECM in response to glaucomatous conditions [15,59,60,61]. Furthermore, astrocytes make connections with other astrocytes and LC cells which could aid in coordinating responses to mechanical strain. Actin reorganization of astrocytes in response to elevated pressure can occur within hours of IOP elevation in rodents and reorganization to baseline may happen over the same time scale, or perhaps even days [62,63]. Reactive astrocytes also produce MMPs which could serve a similar purpose as suggested previously; to sever connections to allow cellular displacement as well as rearrangement of the ECM [29]. As discussed above, MMP function ties in closely with TGF-β, and evidence shows that astrocytes utilize the TGF-β pathway in response to glaucoma as well [22,64,65]. Also prominent to the TGF-β pathway is connective tissue growth factor (CCN2; referred to here as CTGF), a significant binding partner of TGF-β that has been shown to affect the TGF-β pathway, and it is required for Smad1 but not Smad3 activation [66]. CTGF is a mediator of ECM synthesis in the anterior segment as well, as demonstrated by a murine model with increased secretion of CTGF resulting in trabecular meshwork (TM) remodeling and increased IOP [67]. Further evidence in mice shows that the astrocytic levels of CTGF in the ONH increases in glaucomatous animals as a result of elevated IOP and stiffness, which agrees with the observation that there are elevated levels of CTGF in glaucomatous ONHs of humans as well [68]. At least in mice, CTGF seems to be predominantly expressed by astrocytes in the ONH [69], but there is reason to suggest that CTGF may affect other cell types such as LC cells [37].
Integrin signaling in astrocytes may also be involved in the LC cell in detecting tissue strain and inducing cellular migration and reattachment [33]. However, these are not the only molecules involved in astrocytic remodeling as myosin light chain kinase has increased expression in response to mechanical strain of astrocytes and is implicated in cellular migration [70]. Moreover, phosphoinositide 3-kinase, protein kinase C, and tyrosine kinase have also be implicated in migration [71]. Astrocytes also detect mechanical strain with TRPCs which may provide early responses to initial IOP increases. For example, in an induced mouse model, reactive astrocytes respond within one hour of an IOP increase, likely mediated by TRPC isoforms sensitive to stretch [72]. As previously mentioned, this TRPC-NFAT pathway can induce transcriptional changes related to the ECM and can influence cellular migration.
Astrocytic responses are not limited to mechanoreceptors as hypoxia can also trigger responses. Hypoxia-inducible factor-1α (HIF-1α) is a transcriptional factor that is upregulated in response to hypoxic conditions and plays a role in cellular metabolism, proliferation, and angiogenesis [73]. The link between the glaucomatous ONH and HIF-1α was first noted by examining human eye post-mortem, but it has been noted in glaucomatous dogs as well [39,74]. These findings have been replicated in induced rodent models of glaucoma, and the evidence indicates that HIF-1α activation is localized to astrocytes of the retina and ONH [75,76]. There is currently no explanation for why HIF-1α responses are localized to astrocytes and not microglia or RGCs in these models. However, it does indicate that global hypoxic conditions of the ONH, at least on these early timescales, cannot explain RGC dysfunction due to ischemia. Alternatively, PACE4, a subtilisin-like protein convertase, is known to increase expression in response to hypoxia, which may also occur due to vascular compression or primary vascular or vasospastic disease [39,77]. PACE4 also displays constitutive expression and activity in glial cells across the retina, but more so in the ONH [78]. This may be an important factor to consider given the evidence that the PACE family interacts with inhibitors of MMPs, tissue inhibitors of matrix metalloproteinases (TIMPs), as well as TGF-β [79].
Astrocytes at the post-laminar, myelination transition zone (MTZ) are also of interest due to the potential posterior shift of the LC that may be signaled by mechanotransduction of the cells. Specifically, galectin-3 (also known as Lgals3 or Mac-2) has been shown to be upregulated and involved in astrocytic phagocytosis of RGC axons [80]. Additionally, recent evidence in an inducible murine model has shown that astrocytes near the MTZ react by projecting longitudinal processes into the axonal bundles of the ONH, rather than encasing the axons, perhaps contributing to phagocytosis [56]. There is also reason to believe that such phagocytotic absorbance of mitochondria localized to the axons may precede RGC degeneration [81,82,83].
Microglia are also present in the ONH and are activated in glaucomatous eyes [84]. Similar to LC cells, activated microglia express both TGF-β and MMPs which are not produced in the microglia of healthy ONHs [29]. While microglia are likely incapable of the secretion of ECM molecules, there is mounting evidence that suggests that microglia across the central nervous system are highly active in the maintenance of the ECM through reorganization using MMPs, TSPs, and other similar proteins [85,86]. These processes may be important in the formation of glial scarring, a deposit of new ECM that may not be beneficial to the RGC axons [87]. While this produces a physical obstruction within the ONH that can damage axons, deposits of proteoglycans such as tenascin, which is produced by astrocytes in a mechanically independent model of glaucoma, may provide some initial protection to the axons [88,89,90]. Furthermore, tenascin is a substrate on which MMPs can act and remodel. Activation of microglia can be dependent on integrin signaling, detection of damaged cells, and growth factors such as TGF-β [33,86,91,92]. While some components of astrocyte activation are necessary for cellular repair and neurotrophic factors, persistent activation can lead to secretion of cytotoxic molecules that are likely further detrimental to the RGC axons [93].
The reactivity of both astrocytes and microglia contributes to the neuroinflammatory conditions of the ONH that may negatively impact the surrounding milieu. This perspective of GON is complex and has generated a rapidly emerging line of work which has recently been adequately reviewed [94,95,96,97,98].

5. Scleral Fibroblasts

The ppScl, or scleral flange, is the portion of sclera immediately surrounding the scleral canal which provides an anchoring point for the LC. It also contains the penetrating branches of the posterior ciliary arteries that perfuse the LC and overlying choroid. The ppScl, as with the remaining sclera, is composed of a dense, collagenous ECM interspersed with resident fibroblasts that maintain the ECM [99]. Similar to the other cell types, the scleral fibroblasts of the ppScl also produce ECM remodeling factors in response to glaucomatous conditions as demonstrated in human tissue and primate, and mouse models [19,100]. A notable characteristic of these cells is that when active and responsive, they differentiate into myofibroblasts consequently expressing α-SMA [101,102,103]. Differentiation of scleral fibroblasts can be caused by a mechanosensitive response to increased pressure and leads to the secretion of ECM materials, such as collagen, and ECM editors, such as MMPs and TIMPs [104,105,106,107]. Myofibroblast differentiation is also partly dependent on Src-kinase pathways as inhibitors of the pathway, such as dasatinib, can restrict the process [108]. Myofibroblast differentiation requires transcriptional changes which can be seen as soon as 30 min after mechanical strain is applied to human tissue culture [109].
Scleral fibroblasts use the collagenous matrix as the point of cell adhesion. This collagenous matrix of the ppScl is morphologically distinct from the rest of the sclera. Specifically, collagen in the ppScl runs in a circumferential pattern around the ONH while collagen in the posterior sclera is arranged in a “basket-woven” pattern [110,111]. Interestingly, this pattern correlates with distribution and alignment of scleral fibroblasts. Fibroblast density increases in proximity to the ONH, and fibroblast projections are highly aligned with collagenous structures [112]. There is also limited evidence to suggest that such fibroblast density gradients may exist in mouse as well [113]. Given this precise alignment of fibroblasts and collagen, these cells likely play a role in the detection of tissue stretch. The reaction of fibroblasts may also differ based on localization as α-SMA expression appears to disrupt fibroblast projection alignment with collagen in the peripheral sclera, but not the ppScl, and fibroblast orientation is most altered when cells detect both strain and TGF-β signaling simultaneously [112]. These synergistic processes likely reinforce chronic glaucomatous remodeling of the ppScl, altering its biomechanical properties.
Properties of scleral fibroblast differentiation and proliferation are partly mediated by both Yes-Associated-Protein (YAP) and Rho-associated protein kinase (ROCK) [114,115]. A notable trait of myofibroblasts is their expression of α-SMA, which may aid in acutely altering scleral stiffness and in cell migration [116]. Furthermore, fibroblast migration is also associated with ROCK and YAP as inhibition of both leads to reduction in rates of migration as well as the contractile abilities associated with α-SMA [114,115]. ROCK inhibitors thus may be a potential treatment that is currently used to increase aqueous outflow in the anterior segment.
Similar to LC cells, TGF-β and Smad-based transcription play a role in fibroblast responses as well. Application of TGF-β to cultured scleral fibroblasts induces higher levels of α-SMA expression and contractility [115] and binding partners of Smad are upregulated in scleral fibroblasts responding to stretch [109]. Additionally, YAP and Smad3 are shown to interact in human scleral fibroblasts which undergo strain [114]. Within an induced mouse model of glaucoma, upregulation in both TSP and integrin expression, both activators of TGF-β, have been demonstrated [104]. Furthermore, an ECM remodeling protein, TGF-β inducible protein (TGF-βip), is known to express in response to TGF-β signaling pathways, especially in collagen rich tissues [117]. There is evidence of the presence and secretion of TGF-βip in human and non-human primate sclera, and TGF-βip has binding properties with integrins at the cell surface in human scleral fibroblasts, which are implicated in stretch detection and in the modulation of the biomechanical properties of the cell [118,119,120]. TGF-βip can also inhibit fibroblast adhesion to collagen, which likely affects remodeling and cellular migration [119]. Taken together, these results suggest that scleral fibroblasts in glaucomatous conditions use similar signaling pathways to LC cells, such as TGF-β, to differentiate, migrate, and induce extensive remodeling of the sclera and ppScl.

6. Discussion

Glaucoma treatment is difficult due to its complex, incompletely understood, pathophysiology, and while IOP lowering is impactful, it does not universally prevent the progression or development of the disease. These approaches to lower IOP focus on reducing the mechanical stress applied to the optic nerve head. Altering the material properties or morphology of the LC and ppScl by manipulation of the processes involved in ONH remodeling has the potential to increase the resilience of the ONH to the stress induced by changing IOP and promote RGC survival. However, it remains unclear what mechanical properties of the sclera and LC are beneficial and what properties are harmful. For example, there have been hypotheses that increased stiffness could resist elevated IOP levels [17]. However, a stiffer scleral may increase the strain experienced by the OHN by directing stress to the weakest point in the eye wall. Multiple scleral stiffening compounds such as glyceraldehyde, glutaraldehyde, and genipin have failed to demonstrate RGC protection in rodent models or tree shrews thus far [121,122,123] and may be harmful to the retina as well [124]. Alternatively, perhaps reducing scleral stiffness could alleviate certain cases of glaucoma through the application of collagenase or other compounds that break down glycosaminoglycans [125,126,127]. Although the evidence is limited, one study showed that rats with experimentally induced glaucoma and subsequently treated with a glycosaminoglycan digesting agent via intravitreal injection demonstrated preservation of RGC dendritic fields [128]. While it is promising that the manipulation of scleral material properties may impact glaucoma development, it is unclear how a collagenous LC may respond to approaches that increase or decrease LC stiffness in terms of RGC survival.
Several therapeutic approaches targeting the cells that create the ECM have been suggested in an attempt to inhibit molecular pathways that trigger ECM secretion (Table 1). Notably, many of the drugs under investigation target some element of the TGF-β pathways referenced previously. For instance, losartan inhibits the G-protein-coupled receptor (GPCR) angiotensin 1 and is a target for the TGF-β ligand [129]. When losartan was administered orally to mice with experimentally induced glaucoma, RGC loss was prevented, likely by inhibiting the degree to which scleral fibroblasts could remodel the ECM [130]. However, a side effect of losartan is decreased blood pressure which could reduce ocular perfusion pressure, a critical risk factor for glaucoma.
Additionally, the competitive antagonist LSKL, which can cross the BBB, may be another candidate for the treatment of glaucoma. LSKL prevents TSP1 from binding to LAP which prevents activation of TGF-β and its downstream pathway. The result is that LSKL administration to rodents leads to reduced fibrosis with minimal side effects [26,256,257]. Despite the potential, there has not been any published evidence that LSKL can ameliorate the glaucomatous ONH and preserve RGCs [30]. CTGF, a primary interactor of TGF-β, is also profibrotic, and inhibition of CTGF in cultured human LC cells using the monoclonal antibody FG-3019 blocked ECM synthesis in these cells [37]. The antibody FG-3019, or pamrevlumab, is currently being evaluated in clinical trials for idiopathic pulmonary fibrosis, but its effectiveness beyond cultured cells in glaucoma has not yet been illustrated [258]. While intriguing, there are also a multitude of other potential therapies or pathway targets of TGF-β that could be beneficial for the treatment of glaucoma [259]. Future therapeutic pathways may further probe the relationship between TGF-β and both bone morphogenic proteins (BMPs) and Wnt signaling; two candidates that may inhibit profibrotic responses [260,261].
Another method to prevent remodeling in the ONH is to prevent the synthesis of collagen by LC cells and astrocytes before it is secreted. The drug tranilast is known to inhibit collagen synthesis and has been shown to work on cultured human LC cells and astrocytes [228]. The inhibition of rho-associated protein kinase (ROCK) has also been shown to prevent scleral fibroblast from exhibiting myofibroblast features, limiting contractility and expression of α-SMA [115]. In fact, ROCK inhibitors, such as K-115 or ripasudil, are currently being investigated in clinical trials for glaucoma treatment [222,262,263,264].
Some common currently available systemic and topical treatment may also potentially impact glaucoma development. Statins have been shown to prevent TGF-β-mediated activation of MMPs through ROCK pathway inhibition in cultured human eye [65]. Additionally, simvastatin has demonstrated a protective effect on RGC survival in a mouse model of retinal ischemia via elevated IOP [240]. Whether or not statins could be effective at treating human cases of glaucoma with little side effects is unknown [265], but administrative database studies and meta-analyses have suggested a potential protective effect, slowing the development of glaucoma [266,267,268]. Lastly, prostaglandin analogues, the most common IOP-lowering treatment for glaucoma, work by upregulation of MMPs via the prostaglandin F receptor. These same pathways that alter the ECM of the iris and ciliary body to lower IOP are also involved in glaucomatous remodeling in the posterior pole [269]. While it is unknown whether these compounds would reach therapeutic levels in the posterior pole with topical administration, methods of delivery could be developed to impact the ONH more directly.
In summary, there is intriguing emerging evidence that manipulation of the mechanical properties of the sclera and/or ONH may provide an alternative treatment for glaucoma that is independent of IOP lowering. However, it is possible that implementing a combination of treatments exploiting several mechanisms, such as IOP lowering, ECM remodeling, and neuroprotection, may provide the most effective treatment for patients. The resident cells of the ONH and ppScl that drive remodeling of these critical load-bearing connective tissues can potentially be recruited to improve the resilience of the optic nerve to glaucomatous injury. It is critical that additional research is conducted to clarify how the material properties in the LC and scleral should be adjusted to a beneficial effect and to elucidate the mechanocellular pathways involved in age-related and glaucomatous remodeling.

Author Contributions

R.G.S.: Conceptualization, writing—original draft preparation, writing—review and editing, investigation. M.A.G.: writing—review and editing, A.K.G.: Conceptualization, review and editing, funding acquisition, supervision. C.A.G.: Conceptualization, writing—review and editing, funding acquisition, supervision. All authors have read and agreed to the published version of the manuscript.

Funding

National Eye Institute: R01EY028284 (Girkin), National Eye Institute: 5R01EY030096 (Gross), EyeSight Foundation of Alabama (Girkin), Research to Prevent Blindness (Girkin).

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.

References

  1. Tham, Y.-C.; Li, X.; Wong, T.Y.; Quigley, H.A.; Aung, T.; Cheng, C.-Y. Global prevalence of glaucoma and projections of glaucoma burden through 2040: A systematic review and meta-analysis. Ophthalmology 2014, 121, 2081–2090. [Google Scholar] [CrossRef] [PubMed]
  2. Quigley, H.A.; Broman, A.T. The number of people with glaucoma worldwide in 2010 and 2020. Br. J. Ophthalmol. 2006, 90, 262–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Downs, J.C.; Girkin, C.A. Lamina cribrosa in glaucoma. Curr. Opin. Ophthalmol. 2017, 28, 113–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sigal, I.A. Interactions between geometry and mechanical properties on the optic nerve head. Investig. Ophthalmol. Vis. Sci. 2009, 50, 2785–2795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Grytz, R.; Fazio, M.A.; Libertiaux, V.; Bruno, L.; Gardiner, S.; Girkin, C.A.; Downs, J.C. Age- and race-related differences in human scleral material properties. Investig. Ophthalmol. Vis. Sci. 2014, 55, 8163–8172. [Google Scholar] [CrossRef] [Green Version]
  6. Coudrillier, B.; Tian, J.; Alexander, S.; Myers, K.M.; Quigley, H.A.; Nguyen, T.D. Biomechanics of the human posterior sclera: Age- and glaucoma-related changes measured using inflation testing. Investig. Ophthalmol. Vis. Sci. 2012, 53, 1714–1728. [Google Scholar] [CrossRef]
  7. Safa, B.N.; Wong, C.A.; Ha, J.; Ethier, C.R. Glaucoma and biomechanics. Curr. Opin. Ophthalmol. 2022, 33, 80–90. [Google Scholar] [CrossRef]
  8. Downs, J.C. Optic nerve head biomechanics in aging and disease. Exp. Eye Res. 2015, 133, 19–29. [Google Scholar] [CrossRef] [Green Version]
  9. AGIS Investigators. The Advanced Glaucoma Intervention Study (AGIS): 12. Baseline risk factors for sustained loss of visual field and visual acuity in patients with advanced glaucoma. Am. J. Ophthalmol. 2002, 134, 499–512. [Google Scholar] [CrossRef]
  10. Leske, M.C.; Heijl, A.; Hussein, M.; Bengtsson, B.; Hyman, L.; Komaroff, E.; Early Manifest Glaucoma Trial Group. Factors for glaucoma progression and the effect of treatment: The Early Manifest Glaucoma Trial. Arch. Ophthalmol. 2003, 121, 48–56. [Google Scholar] [CrossRef]
  11. Musch, D.C.; Gillespie, B.W.; Lichter, P.R.; Niziol, L.M.; Janz, N.K.; CIGTS Study Investigators. Visual field progression in the Collaborative Initial Glaucoma Treatment Study the impact of treatment and other baseline factors. Ophthalmology 2009, 116, 200–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Drance, S.; Anderson, D.R.; Schulzer, M.; Collaborative Normal-Tension Glaucoma Study Group. Risk factors for progression of visual field abnormalities in normal-tension glaucoma. Am. J. Ophthalmol. 2001, 131, 699–708. [Google Scholar] [CrossRef]
  13. Gordon, M.O.; Beiser, J.A.; Brandt, J.D.; Heuer, D.K.; Higginbotham, E.J.; Johnson, C.A.; Keltner, J.L.; Miller, J.P.; Parrish, R.K.; Wilson, M.R.; et al. The Ocular Hypertension Treatment Study: Baseline factors that predict the onset of primary open-angle glaucoma. Arch. Ophthalmol. 2002, 120, 714–720, Discussion 829. [Google Scholar] [CrossRef] [PubMed]
  14. Hernandez, M.R.; Andrzejewska, W.M.; Neufeld, A.H. Changes in the extracellular matrix of the human optic nerve head in primary open-angle glaucoma. Am. J. Ophthalmol. 1990, 109, 180–188. [Google Scholar] [CrossRef]
  15. Hernandez, M.R. Ultrastructural immunocytochemical analysis of elastin in the human lamina cribrosa. Changes in elastic fibers in primary open-angle glaucoma. Investig. Ophthalmol. Vis. Sci. 1992, 33, 2891–2903. [Google Scholar]
  16. Sawaguchi, S.; Yue, B.Y.; Fukuchi, T.; Abe, H.; Suda, K.; Kaiya, T.; Iwata, K. Collagen fibrillar network in the optic nerve head of normal monkey eyes and monkey eyes with laser-induced glaucoma—A scanning electron microscopic study. Curr. Eye Res. 1999, 18, 143–149. [Google Scholar] [CrossRef]
  17. Quigley, H.A. The contribution of the sclera and lamina cribrosa to the pathogenesis of glaucoma: Diagnostic and treatment implications. Prog. Brain Res. 2015, 220, 59–86. [Google Scholar] [CrossRef]
  18. Fazio, M.A.; Grytz, R.; Morris, J.S.; Bruno, L.; Gardiner, S.K.; Girkin, C.A.; Downs, J.C. Age-related changes in human peripapillary scleral strain. Biomech. Model. Mechanobiol. 2014, 13, 551–563. [Google Scholar] [CrossRef] [Green Version]
  19. Quigley, H.A.; Dorman-Pease, M.E.; Brown, A.E. Quantitative study of collagen and elastin of the optic nerve head and sclera in human and experimental monkey glaucoma. Curr. Eye Res. 1991, 10, 877–888. [Google Scholar] [CrossRef]
  20. Fukuchi, T.; Sawaguchi, S.; Hara, H.; Shirakashi, M.; Iwata, K. Extracellular matrix changes of the optic nerve lamina cribrosa in monkey eyes with experimentally chronic glaucoma. Graefe’s Arch. Clin. Exp. Ophthalmol. 1992, 230, 421–427. [Google Scholar] [CrossRef]
  21. Prendes, M.A.; Harris, A.; Wirostko, B.M.; Gerber, A.L.; Siesky, B. The role of transforming growth factor β in glaucoma and the therapeutic implications. Br. J. Ophthalmol. 2013, 97, 680–686. [Google Scholar] [CrossRef] [PubMed]
  22. Zode, G.S.; Sethi, A.; Brun-Zinkernagel, A.-M.; Chang, I.-F.; Clark, A.F.; Wordinger, R.J. Transforming growth factor-β2 increases extracellular matrix proteins in optic nerve head cells via activation of the Smad signaling pathway. Mol. Vis. 2011, 17, 1745–1758. [Google Scholar] [PubMed]
  23. Kirwan, R.P.; Leonard, M.O.; Murphy, M.; Clark, A.F.; O’Brien, C.J. Transforming growth factor-beta-regulated gene transcription and protein expression in human GFAP-negative lamina cribrosa cells. Glia 2005, 52, 309–324. [Google Scholar] [CrossRef] [PubMed]
  24. Kirwan, R.P.; Crean, J.K.; Fenerty, C.H.; Clark, A.F.; O’Brien, C.J. Effect of cyclical mechanical stretch and exogenous transforming growth factor-beta1 on matrix metalloproteinase-2 activity in lamina cribrosa cells from the human optic nerve head. J. Glaucoma 2004, 13, 327–334. [Google Scholar] [CrossRef]
  25. Annes, J.P.; Munger, J.S.; Rifkin, D.B. Making sense of latent TGFbeta activation. J. Cell Sci. 2003, 116, 217–224. [Google Scholar] [CrossRef] [Green Version]
  26. Murphy-Ullrich, J.E.; Downs, J.C. The Thrombospondin1-TGF-β Pathway and Glaucoma. J. Ocul. Pharmacol. Ther. 2015, 31, 371–375. [Google Scholar] [CrossRef]
  27. Murphy, G.; Nagase, H. Progress in matrix metalloproteinase research. Mol. Aspects Med. 2008, 29, 290–308. [Google Scholar] [CrossRef] [Green Version]
  28. 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]
  29. Yuan, L.; Neufeld, A.H. Activated microglia in the human glaucomatous optic nerve head. J. Neurosci. Res. 2001, 64, 523–532. [Google Scholar] [CrossRef]
  30. Murphy-Ullrich, J.E.; Suto, M.J. Thrombospondin-1 regulation of latent TGF-β activation: A therapeutic target for fibrotic disease. Matrix Biol. 2018, 68–69, 28–43. [Google Scholar] [CrossRef]
  31. 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. Ophthalmol. Vis. Sci. 2012, 53, 6708–6717. [Google Scholar] [CrossRef] [PubMed]
  32. Iomdina, E.N.; Tikhomirova, N.K.; Bessmertny, A.M.; Serebryakova, M.V.; Baksheeva, V.E.; Zalevsky, A.O.; Kotelin, V.I.; Kiseleva, O.A.; Kosakyan, S.M.; Zamyatnin, A.A.; et al. Alterations in proteome of human sclera associated with primary open-angle glaucoma involve proteins participating in regulation of the extracellular matrix. Mol. Vis. 2020, 26, 623–640. [Google Scholar] [PubMed]
  33. Morrison, J.C. Integrins in the optic nerve head: Potential roles in glaucomatous optic neuropathy (an American Ophthalmological Society thesis). Trans. Am. Ophthalmol. Soc. 2006, 104, 453–477. [Google Scholar]
  34. 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]
  35. Hernandez, M.R.; Igoe, F.; Neufeld, A.H. Cell culture of the human lamina cribrosa. Investig. Ophthalmol. Vis. Sci. 1988, 29, 78–89. [Google Scholar]
  36. 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] [PubMed]
  37. 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. Ophthalmol. Vis. Sci. 2013, 54, 7836–7848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Kirwan, R.P.; Felice, L.; Clark, A.F.; O’Brien, C.J.; Leonard, M.O. Hypoxia regulated gene transcription in human optic nerve lamina cribrosa cells in culture. Investig. Ophthalmol. Vis. Sci. 2012, 53, 2243–2255. [Google Scholar] [CrossRef] [Green Version]
  39. Tezel, G.; Wax, M.B. Hypoxia-inducible factor 1alpha in the glaucomatous retina and optic nerve head. Arch. Ophthalmol. 2004, 122, 1348–1356. [Google Scholar] [CrossRef] [Green Version]
  40. McElnea, E.M.; Quill, B.; Docherty, N.G.; Irnaten, M.; Siah, W.F.; Clark, A.F.; O’Brien, C.J.; Wallace, D.M. Oxidative stress, mitochondrial dysfunction and calcium overload in human lamina cribrosa cells from glaucoma donors. Mol. Vis. 2011, 17, 1182–1191. [Google Scholar]
  41. 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]
  42. Yang, H.; Williams, G.; Downs, J.C.; Sigal, I.A.; Roberts, M.D.; Thompson, H.; Burgoyne, C.F. Posterior (outward) migration of the lamina cribrosa and early cupping in monkey experimental glaucoma. Investig. Ophthalmol. Vis. Sci. 2011, 52, 7109–7121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yang, H.; Thompson, H.; Roberts, M.D.; Sigal, I.A.; Downs, J.C.; Burgoyne, C.F. Deformation of the early glaucomatous monkey optic nerve head connective tissue after acute IOP elevation in 3-D histomorphometric reconstructions. Investig. Ophthalmol. Vis. Sci. 2011, 52, 345–363. [Google Scholar] [CrossRef] [PubMed]
  44. Liou, J.-J.; Geest, J.P.V. Effect of transforming growth factor beta 2 on matrix metalloproteinase activity in porcine lamina cribrosa cells. Investig. Ophthalmol. Vis. Sci. 2020, 61, 902. [Google Scholar]
  45. Burgoyne, C.F. The non-human primate experimental glaucoma model. Exp. Eye Res. 2015, 141, 57–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Irnaten, M.; Barry, R.C.; Wallace, D.M.; Docherty, N.G.; Quill, B.; Clark, A.F.; O’Brien, C.J. Elevated maxi-K(+) ion channel current in glaucomatous lamina cribrosa cells. Exp. Eye Res. 2013, 115, 224–229. [Google Scholar] [CrossRef]
  47. Irnaten, M.; Zhdanov, A.; Brennan, D.; Crotty, T.; Clark, A.; Papkovsky, D.; O’Brien, C. Activation of the NFAT-Calcium Signaling Pathway in Human Lamina Cribrosa Cells in Glaucoma. Investig. Ophthalmol. Vis. Sci. 2018, 59, 831–842. [Google Scholar] [CrossRef] [Green Version]
  48. Tidu, F.; De Zuani, M.; Jose, S.S.; Bendíčková, K.; Kubala, L.; Caruso, F.; Cavalieri, F.; Forte, G.; Frič, J. NFAT signaling in human mesenchymal stromal cells affects extracellular matrix remodeling and antifungal immune responses. iScience 2021, 24, 102683. [Google Scholar] [CrossRef]
  49. Asghar, M.Y.; Törnquist, K. Transient receptor potential canonical (TRPC) channels as modulators of migration and invasion. Int. J. Mol. Sci. 2020, 21, 1739. [Google Scholar] [CrossRef] [Green Version]
  50. Sharma, S.; Hopkins, C.R. Review of transient receptor potential canonical (TRPC5) channel modulators and diseases. J. Med. Chem. 2019, 62, 7589–7602. [Google Scholar] [CrossRef]
  51. Irnaten, M.; O’Malley, G.; Clark, A.F.; O’Brien, C.J. Transient receptor potential channels TRPC1/TRPC6 regulate lamina cribrosa cell extracellular matrix gene transcription and proliferation. Exp. Eye Res. 2020, 193, 107980. [Google Scholar] [CrossRef] [PubMed]
  52. Chigurupati, S.; Venkataraman, R.; Barrera, D.; Naganathan, A.; Madan, M.; Paul, L.; Pattisapu, J.V.; Kyriazis, G.A.; Sugaya, K.; Bushnev, S.; et al. Receptor channel TRPC6 is a key mediator of Notch-driven glioblastoma growth and invasiveness. Cancer Res. 2010, 70, 418–427. [Google Scholar] [CrossRef] [Green Version]
  53. Bomben, V.C.; Turner, K.L.; Barclay, T.-T.C.; Sontheimer, H. Transient receptor potential canonical channels are essential for chemotactic migration of human malignant gliomas. J. Cell. Physiol. 2011, 226, 1879–1888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. 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]
  55. Bowman, C.L.; Ding, J.P.; Sachs, F.; Sokabe, M. Mechanotransducing ion channels in astrocytes. Brain Res. 1992, 584, 272–286. [Google Scholar] [CrossRef]
  56. Wang, R.; Seifert, P.; Jakobs, T.C. Astrocytes in the optic nerve head of glaucomatous mice display a characteristic reactive phenotype. Investig. Ophthalmol. Vis. Sci. 2017, 58, 924–932. [Google Scholar] [CrossRef]
  57. Pena, J.D.; Agapova, O.; Gabelt, B.T.; Levin, L.A.; Lucarelli, M.J.; Kaufman, P.L.; Hernandez, M.R. Increased elastin expression in astrocytes of the lamina cribrosa in response to elevated intraocular pressure. Investig. Ophthalmol. Vis. Sci. 2001, 42, 2303–2314. [Google Scholar]
  58. Rogers, R.S.; Dharsee, M.; Ackloo, S.; Sivak, J.M.; Flanagan, J.G. Proteomics analyses of human optic nerve head astrocytes following biomechanical strain. Mol. Cell. Proteom. 2012, 11, M111.012302. [Google Scholar] [CrossRef] [Green Version]
  59. Hernandez, M.R.; Wang, N.; Hanley, N.M.; Neufeld, A.H. Localization of collagen types I and IV mRNAs in human optic nerve head by in situ hybridization. Investig. Ophthalmol. Vis. Sci. 1991, 32, 2169–2177. [Google Scholar]
  60. Ye, H.; Yang, J.; Hernandez, M.R. Localization of collagen type III mRNA in normal human optic nerve heads. Exp. Eye Res. 1994, 58, 53–63. [Google Scholar] [CrossRef]
  61. Pena, J.D.; Roy, S.; Hernandez, M.R. Tropoelastin gene expression in optic nerve heads of normal and glaucomatous subjects. Matrix Biol. 1996, 15, 323–330. [Google Scholar] [CrossRef]
  62. Tehrani, S.; Davis, L.; Cepurna, W.O.; Choe, T.E.; Lozano, D.C.; Monfared, A.; Cooper, L.; Cheng, J.; Johnson, E.C.; Morrison, J.C. Astrocyte Structural and Molecular Response to Elevated Intraocular Pressure Occurs Rapidly and Precedes Axonal Tubulin Rearrangement within the Optic Nerve Head in a Rat Model. PLoS ONE 2016, 11, e0167364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Sun, D.; Qu, J.; Jakobs, T.C. Reversible reactivity by optic nerve astrocytes. Glia 2013, 61, 1218–1235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Neumann, C.; Yu, A.; Welge-Lüssen, U.; Lütjen-Drecoll, E.; Birke, M. The effect of TGF-beta2 on elastin, type VI collagen, and components of the proteolytic degradation system in human optic nerve astrocytes. Investig. Ophthalmol. Vis. Sci. 2008, 49, 1464–1472. [Google Scholar] [CrossRef] [PubMed]
  65. Kim, M.-L.; Sung, K.R.; Kwon, J.; Shin, J.A. Statins Suppress TGF-β2-Mediated MMP-2 and MMP-9 Expression and Activation Through RhoA/ROCK Inhibition in Astrocytes of the Human Optic Nerve Head. Investig. Ophthalmol. Vis. Sci. 2020, 61, 29. [Google Scholar] [CrossRef]
  66. Nakerakanti, S.S.; Bujor, A.M.; Trojanowska, M. CCN2 is required for the TGF-β induced activation of Smad1-Erk1/2 signaling network. PLoS ONE 2011, 6, e21911. [Google Scholar] [CrossRef]
  67. Junglas, B.; Kuespert, S.; Seleem, A.A.; Struller, T.; Ullmann, S.; Bösl, M.; Bosserhoff, A.; Köstler, J.; Wagner, R.; Tamm, E.R.; et al. Connective tissue growth factor causes glaucoma by modifying the actin cytoskeleton of the trabecular meshwork. Am. J. Pathol. 2012, 180, 2386–2403. [Google Scholar] [CrossRef]
  68. Dillinger, A.E.; Weber, G.R.; Mayer, M.; Schneider, M.; Göppner, C.; Ohlmann, A.; Shamonin, M.; Monkman, G.J.; Fuchshofer, R. CCN2/CTGF-A Modulator of the Optic Nerve Head Astrocyte. Front. Cell Dev. Biol. 2022, 10, 864433. [Google Scholar] [CrossRef]
  69. Dillinger, A.E.; Kuespert, S.; Froemel, F.; Tamm, E.R.; Fuchshofer, R. CCN2/CTGF promotor activity in the developing and adult mouse eye. Cell Tissue Res. 2021, 384, 625–641. [Google Scholar] [CrossRef]
  70. Miao, H.; Crabb, A.W.; Hernandez, M.R.; Lukas, T.J. Modulation of factors affecting optic nerve head astrocyte migration. Investig. Ophthalmol. Vis. Sci. 2010, 51, 4096–4103. [Google Scholar] [CrossRef]
  71. Tezel, G.; Hernandez, M.R.; Wax, M.B. In vitro evaluation of reactive astrocyte migration, a component of tissue remodeling in glaucomatous optic nerve head. Glia 2001, 34, 178–189. [Google Scholar] [CrossRef] [PubMed]
  72. Choi, H.J.; Sun, D.; Jakobs, T.C. Astrocytes in the optic nerve head express putative mechanosensitive channels. Mol. Vis. 2015, 21, 749–766. [Google Scholar] [PubMed]
  73. Lee, P.; Chandel, N.S.; Simon, M.C. Cellular adaptation to hypoxia through hypoxia inducible factors and beyond. Nat. Rev. Mol. Cell Biol. 2020, 21, 268–283. [Google Scholar] [CrossRef] [PubMed]
  74. Savagian, C.A.; Dubielzig, R.R.; Nork, T.M. Comparison of the distribution of glial fibrillary acidic protein, heat shock protein 60, and hypoxia-inducible factor-1alpha in retinas from glaucomatous and normal canine eyes. Am. J. Vet. Res. 2008, 69, 265–272. [Google Scholar] [CrossRef] [PubMed]
  75. Chidlow, G.; Wood, J.P.M.; Casson, R.J. Investigations into Hypoxia and Oxidative Stress at the Optic Nerve Head in a Rat Model of Glaucoma. Front. Neurosci. 2017, 11, 478. [Google Scholar] [CrossRef] [PubMed]
  76. Ergorul, C.; Ray, A.; Huang, W.; Wang, D.Y.; Ben, Y.; Cantuti-Castelvetri, I.; Grosskreutz, C.L. Hypoxia inducible factor-1α (HIF-1α) and some HIF-1 target genes are elevated in experimental glaucoma. J. Mol. Neurosci. 2010, 42, 183–191. [Google Scholar] [CrossRef] [Green Version]
  77. Egger, M.; Schgoer, W.; Beer, A.G.E.; Jeschke, J.; Leierer, J.; Theurl, M.; Frauscher, S.; Tepper, O.M.; Niederwanger, A.; Ritsch, A.; et al. Hypoxia up-regulates the angiogenic cytokine secretoneurin via an HIF-1alpha- and basic FGF-dependent pathway in muscle cells. FASEB J. 2007, 21, 2906–2917. [Google Scholar] [CrossRef] [Green Version]
  78. Fuller, J.A.; Brun-Zinkernagel, A.-M.; Clark, A.F.; Wordinger, R.J. Subtilisin-like proprotein convertase expression, localization, and activity in the human retina and optic nerve head. Investig. Ophthalmol. Vis. Sci. 2009, 50, 5759–5768. [Google Scholar] [CrossRef] [Green Version]
  79. Nour, N.; Mayer, G.; Mort, J.S.; Salvas, A.; Mbikay, M.; Morrison, C.J.; Overall, C.M.; Seidah, N.G. The cysteine-rich domain of the secreted proprotein convertases PC5A and PACE4 functions as a cell surface anchor and interacts with tissue inhibitors of metalloproteinases. Mol. Biol. Cell 2005, 16, 5215–5226. [Google Scholar] [CrossRef] [Green Version]
  80. Nguyen, J.V.; Soto, I.; Kim, K.-Y.; Bushong, E.A.; Oglesby, E.; Valiente-Soriano, F.J.; Yang, Z.; Davis, C.O.; Bedont, J.L.; Son, J.L.; et al. Myelination transition zone astrocytes are constitutively phagocytic and have synuclein dependent reactivity in glaucoma. Proc. Natl. Acad. Sci. USA 2011, 108, 1176–1181. [Google Scholar] [CrossRef] [Green Version]
  81. Cooper, M.L.; Crish, S.D.; Inman, D.M.; Horner, P.J.; Calkins, D.J. Early astrocyte redistribution in the optic nerve precedes axonopathy in the DBA/2J mouse model of glaucoma. Exp. Eye Res. 2016, 150, 22–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Davis, C.O.; Kim, K.-Y.; Bushong, E.A.; Mills, E.A.; Boassa, D.; Shih, T.; Kinebuchi, M.; Phan, S.; Zhou, Y.; Bihlmeyer, N.A.; et al. Transcellular degradation of axonal mitochondria. Proc. Natl. Acad. Sci. USA 2014, 111, 9633–9638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Muench, N.A.; Patel, S.; Maes, M.E.; Donahue, R.J.; Ikeda, A.; Nickells, R.W. The influence of mitochondrial dynamics and function on retinal ganglion cell susceptibility in optic nerve disease. Cells 2021, 10, 1593. [Google Scholar] [CrossRef] [PubMed]
  84. Neufeld, A.H. Microglia in the optic nerve head and the region of parapapillary chorioretinal atrophy in glaucoma. Arch. Ophthalmol. 1999, 117, 1050–1056. [Google Scholar] [CrossRef] [Green Version]
  85. Crapser, J.D.; Arreola, M.A.; Tsourmas, K.I.; Green, K.N. Microglia as hackers of the matrix: Sculpting synapses and the extracellular space. Cell. Mol. Immunol. 2021, 18, 2472–2488. [Google Scholar] [CrossRef] [PubMed]
  86. Nayak, D.; Roth, T.L.; McGavern, D.B. Microglia development and function. Annu. Rev. Immunol. 2014, 32, 367–402. [Google Scholar] [CrossRef] [Green Version]
  87. Hirsch, S.; Bähr, M. Immunocytochemical characterization of reactive optic nerve astrocytes and meningeal cells. Glia 1999, 26, 36–46. [Google Scholar] [CrossRef]
  88. Pena, J.D.; Varela, H.J.; Ricard, C.S.; Hernandez, M.R. Enhanced tenascin expression associated with reactive astrocytes in human optic nerve heads with primary open angle glaucoma. Exp. Eye Res. 1999, 68, 29–40. [Google Scholar] [CrossRef]
  89. Pena, J.D.; Taylor, A.W.; Ricard, C.S.; Vidal, I.; Hernandez, M.R. Transforming growth factor beta isoforms in human optic nerve heads. Br. J. Ophthalmol. 1999, 83, 209–218. [Google Scholar] [CrossRef] [Green Version]
  90. Reinehr, S.; Reinhard, J.; Wiemann, S.; Stute, G.; Kuehn, S.; Woestmann, J.; Dick, H.B.; Faissner, A.; Joachim, S.C. Early remodelling of the extracellular matrix proteins tenascin-C and phosphacan in retina and optic nerve of an experimental autoimmune glaucoma model. J. Cell. Mol. Med. 2016, 20, 2122–2137. [Google Scholar] [CrossRef] [Green Version]
  91. Hou, L.; Bao, X.; Zang, C.; Yang, H.; Sun, F.; Che, Y.; Wu, X.; Li, S.; Zhang, D.; Wang, Q. Integrin CD11b mediates α-synuclein-induced activation of NADPH oxidase through a Rho-dependent pathway. Redox Biol. 2018, 14, 600–608. [Google Scholar] [CrossRef] [PubMed]
  92. Hanisch, U.-K.; Kettenmann, H. Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 2007, 10, 1387–1394. [Google Scholar] [CrossRef] [PubMed]
  93. García-Bermúdez, M.Y.; Freude, K.K.; Mouhammad, Z.A.; van Wijngaarden, P.; Martin, K.K.; Kolko, M. Glial cells in glaucoma: Friends, foes, and potential therapeutic targets. Front. Neurol. 2021, 12, 624983. [Google Scholar] [CrossRef] [PubMed]
  94. Rolle, T.; Ponzetto, A.; Malinverni, L. The role of neuroinflammation in glaucoma: An update on molecular mechanisms and new therapeutic options. Front. Neurol. 2020, 11, 612422. [Google Scholar] [CrossRef]
  95. Tezel, G. Molecular regulation of neuroinflammation in glaucoma: Current knowledge and the ongoing search for new treatment targets. Prog. Retin. Eye Res. 2022, 87, 100998. [Google Scholar] [CrossRef]
  96. Mac Nair, C.E.; Nickells, R.W. Neuroinflammation in glaucoma and optic nerve damage. Prog. Mol. Biol. Transl. Sci. 2015, 134, 343–363. [Google Scholar] [CrossRef]
  97. Soto, I.; Howell, G.R. The complex role of neuroinflammation in glaucoma. Cold Spring Harb. Perspect. Med. 2014, 4, a017269. [Google Scholar] [CrossRef]
  98. Williams, P.A.; Marsh-Armstrong, N.; Howell, G.R.; Lasker/IRRF Initiative on Astrocytes and Glaucomatous Neurodegeneration Participants. Neuroinflammation in glaucoma: A new opportunity. Exp. Eye Res. 2017, 157, 20–27. [Google Scholar] [CrossRef] [Green Version]
  99. Watson, P.G.; Young, R.D. Scleral structure, organisation and disease. A review. Exp. Eye Res. 2004, 78, 609–623. [Google Scholar] [CrossRef]
  100. Cone-Kimball, E.; Nguyen, C.; Oglesby, E.N.; Pease, M.E.; Steinhart, M.R.; Quigley, H.A. Scleral structural alterations associated with chronic experimental intraocular pressure elevation in mice. Mol. Vis. 2013, 19, 2023–2039. [Google Scholar]
  101. Hinz, B.; Phan, S.H.; Thannickal, V.J.; Prunotto, M.; Desmoulière, A.; Varga, J.; De Wever, O.; Mareel, M.; Gabbiani, G. Recent developments in myofibroblast biology: Paradigms for connective tissue remodeling. Am. J. Pathol. 2012, 180, 1340–1355. [Google Scholar] [CrossRef] [PubMed]
  102. Hinz, B.; Phan, S.H.; Thannickal, V.J.; Galli, A.; Bochaton-Piallat, M.-L.; Gabbiani, G. The myofibroblast: One function, multiple origins. Am. J. Pathol. 2007, 170, 1807–1816. [Google Scholar] [CrossRef] [PubMed]
  103. Hinz, B. Myofibroblasts. Exp. Eye Res. 2016, 142, 56–70. [Google Scholar] [CrossRef] [PubMed]
  104. Oglesby, E.N.; Tezel, G.; Cone-Kimball, E.; Steinhart, M.R.; Jefferys, J.; Pease, M.E.; Quigley, H.A. Scleral fibroblast response to experimental glaucoma in mice. Mol. Vis. 2016, 22, 82–99. [Google Scholar]
  105. Shelton, L.; Rada, J.S. Effects of cyclic mechanical stretch on extracellular matrix synthesis by human scleral fibroblasts. Exp. Eye Res. 2007, 84, 314–322. [Google Scholar] [CrossRef] [Green Version]
  106. Fujikura, H.; Seko, Y.; Tokoro, T.; Mochizuki, M.; Shimokawa, H. Involvement of mechanical stretch in the gelatinolytic activity of the fibrous sclera of chicks, in vitro. JPN J. Ophthalmol. 2002, 46, 24–30. [Google Scholar] [CrossRef]
  107. Yamaoka, A.; Matsuo, T.; Shiraga, F.; Ohtsuki, H. TIMP-1 production by human scleral fibroblast decreases in response to cyclic mechanical stretching. Ophthalmic Res. 2001, 33, 98–101. [Google Scholar] [CrossRef]
  108. Chow, A.; McCrea, L.; Kimball, E.; Schaub, J.; Quigley, H.; Pitha, I. Dasatinib inhibits peripapillary scleral myofibroblast differentiation. Exp. Eye Res. 2020, 194, 107999. [Google Scholar] [CrossRef]
  109. Cui, W.; Bryant, M.R.; Sweet, P.M.; McDonnell, P.J. Changes in gene expression in response to mechanical strain in human scleral fibroblasts. Exp. Eye Res. 2004, 78, 275–284. [Google Scholar] [CrossRef]
  110. Coudrillier, B.; Pijanka, J.; Jefferys, J.; Sorensen, T.; Quigley, H.A.; Boote, C.; Nguyen, T.D. Collagen structure and mechanical properties of the human sclera: Analysis for the effects of age. J. Biomech. Eng. 2015, 137, 041006. [Google Scholar] [CrossRef] [Green Version]
  111. Voorhees, A.P.; Jan, N.-J.; Hua, Y.; Yang, B.; Sigal, I.A. Peripapillary sclera architecture revisited: A tangential fiber model and its biomechanical implications. Acta Biomater. 2018, 79, 113–122. [Google Scholar] [CrossRef] [PubMed]
  112. Szeto, J.; Chow, A.; McCrea, L.; Mozzer, A.; Nguyen, T.D.; Quigley, H.A.; Pitha, I. Regional differences and physiologic behaviors in peripapillary scleral fibroblasts. Investig. Ophthalmol. Vis. Sci. 2021, 62, 27. [Google Scholar] [CrossRef] [PubMed]
  113. Wu, H.; Chen, W.; Zhao, F.; Zhou, Q.; Reinach, P.S.; Deng, L.; Ma, L.; Luo, S.; Srinivasalu, N.; Pan, M.; et al. Scleral hypoxia is a target for myopia control. Proc. Natl. Acad. Sci. USA 2018, 115, E7091–E7100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Hu, D.; Jiang, J.; Ding, B.; Xue, K.; Sun, X.; Qian, S. Mechanical strain regulates myofibroblast differentiation of human scleral fibroblasts by YAP. Front. Physiol. 2021, 12, 712509. [Google Scholar] [CrossRef]
  115. Pitha, I.; Oglesby, E.; Chow, A.; Kimball, E.; Pease, M.E.; Schaub, J.; Quigley, H. Rho-Kinase Inhibition Reduces Myofibroblast Differentiation and Proliferation of Scleral Fibroblasts Induced by Transforming Growth Factor β and Experimental Glaucoma. Transl. Vis. Sci. Technol. 2018, 7, 6. [Google Scholar] [CrossRef] [Green Version]
  116. 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. Ophthalmol. Vis. Sci. 2015, 56, 7821–7830. [Google Scholar] [CrossRef] [Green Version]
  117. Skonier, J.; Neubauer, M.; Madisen, L.; Bennett, K.; Plowman, G.D.; Purchio, A.F. cDNA cloning and sequence analysis of beta ig-h3, a novel gene induced in a human adenocarcinoma cell line after treatment with transforming growth factor-beta. DNA Cell Biol. 1992, 11, 511–522. [Google Scholar] [CrossRef]
  118. Shelton, L.; Troilo, D.; Lerner, M.R.; Gusev, Y.; Brackett, D.J.; Rada, J.S. Microarray analysis of choroid/RPE gene expression in marmoset eyes undergoing changes in ocular growth and refraction. Mol. Vis. 2008, 14, 1465–1479. [Google Scholar]
  119. Shelton, L.; Rada, J.A.S. Inhibition of human scleral fibroblast cell attachment to collagen type I by TGFBIp. Investig. Ophthalmol. Vis. Sci. 2009, 50, 3542–3552. [Google Scholar] [CrossRef] [Green Version]
  120. Hu, S.; Cui, D.; Yang, X.; Hu, J.; Wan, W.; Zeng, J. The crucial role of collagen-binding integrins in maintaining the mechanical properties of human scleral fibroblasts-seeded collagen matrix. Mol. Vis. 2011, 17, 1334–1342. [Google Scholar]
  121. Kimball, E.C.; Nguyen, C.; Steinhart, M.R.; Nguyen, T.D.; Pease, M.E.; Oglesby, E.N.; Oveson, B.C.; Quigley, H.A. Experimental scleral cross-linking increases glaucoma damage in a mouse model. Exp. Eye Res. 2014, 128, 129–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Hannon, B.G.; Schwaner, S.A.; Boazak, E.M.; Gerberich, B.G.; Winger, E.J.; Prausnitz, M.R.; Ethier, C.R. Sustained scleral stiffening in rats after a single genipin treatment. J. R. Soc. Interface 2019, 16, 20190427. [Google Scholar] [CrossRef] [PubMed]
  123. Coudrillier, B.; Campbell, I.C.; Read, A.T.; Geraldes, D.M.; Vo, N.T.; Feola, A.; Mulvihill, J.; Albon, J.; Abel, R.L.; Ethier, C.R. Effects of peripapillary scleral stiffening on the deformation of the lamina cribrosa. Investig. Ophthalmol. Vis. Sci. 2016, 57, 2666–2677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Hamdaoui, M.E.; Levy, A.M.; Stuber, A.B.; Girkin, C.A.; Kraft, T.W.; Samuels, B.C.; Grytz, R. Scleral crosslinking using genipin can compromise retinal structure and function in tree shrews. Exp. Eye Res. 2022, 219, 109039. [Google Scholar] [CrossRef]
  125. Murienne, B.J.; Jefferys, J.L.; Quigley, H.A.; Nguyen, T.D. The effects of glycosaminoglycan degradation on the mechanical behavior of the posterior porcine sclera. Acta Biomater. 2015, 12, 195–206. [Google Scholar] [CrossRef] [Green Version]
  126. Spoerl, E.; Boehm, A.G.; Pillunat, L.E. The influence of various substances on the biomechanical behavior of lamina cribrosa and peripapillary sclera. Investig. Ophthalmol. Vis. Sci. 2005, 46, 1286–1290. [Google Scholar] [CrossRef] [Green Version]
  127. Hatami-Marbini, H.; Pachenari, M. Tensile Viscoelastic Properties of the Sclera after Glycosaminoglycan Depletion. Curr. Eye Res. 2021, 46, 1299–1308. [Google Scholar] [CrossRef]
  128. Tribble, J.R.; Williams, P.A.; Caterson, B.; Sengpiel, F.; Morgan, J.E. Digestion of the glycosaminoglycan extracellular matrix by chondroitinase ABC supports retinal ganglion cell dendritic preservation in a rodent model of experimental glaucoma. Mol. Brain 2018, 11, 69. [Google Scholar] [CrossRef]
  129. Ehanire, T.; Ren, L.; Bond, J.; Medina, M.; Li, G.; Bashirov, L.; Chen, L.; Kokosis, G.; Ibrahim, M.; Selim, A.; et al. Angiotensin II stimulates canonical TGF-β signaling pathway through angiotensin type 1 receptor to induce granulation tissue contraction. J. Mol. Med. 2015, 93, 289–302. [Google Scholar] [CrossRef] [Green Version]
  130. Quigley, H.A.; Pitha, I.F.; Welsbie, D.S.; Nguyen, C.; Steinhart, M.R.; Nguyen, T.D.; Pease, M.E.; Oglesby, E.N.; Berlinicke, C.A.; Mitchell, K.L.; et al. Losartan treatment protects retinal ganglion cells and alters scleral remodeling in experimental glaucoma. PLoS ONE 2015, 10, e0141137. [Google Scholar] [CrossRef] [Green Version]
  131. Ota, T.; Murata, H.; Sugimoto, E.; Aihara, M.; Araie, M. Prostaglandin analogues and mouse intraocular pressure: Effects of tafluprost, latanoprost, travoprost, and unoprostone, considering 24-hour variation. Investig. Ophthalmol. Vis. Sci. 2005, 46, 2006–2011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Park, H.Y.L.; Kim, J.H.; Lee, D.E.; Lee, J.H.; Park, C.K. Changes of the Retina and Intrinsic Survival Signals in a Rat Model of Glaucoma following Brinzolamide and Travoprost Treatments. Ophthalmic Res. 2011, 46, 208–217. [Google Scholar] [CrossRef] [PubMed]
  133. Kurashima, H.; Watabe, H.; Sato, N.; Abe, S.; Ishida, N.; Yoshitomi, T. Effects of prostaglandin F(2α) analogues on endothelin-1-induced impairment of rabbit ocular blood flow: Comparison among tafluprost, travoprost, and latanoprost. Exp. Eye Res. 2010, 91, 853–859. [Google Scholar] [CrossRef] [PubMed]
  134. Gagliuso, D.J.; Wang, R.-F.; Mittag, T.W.; Podos, S.M. Additivity of bimatoprost or travoprost to latanoprost in glaucomatous monkey eyes. Arch. Ophthalmol. 2004, 122, 1342–1347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Netland, P.A.; Landry, T.; Sullivan, E.K.; Andrew, R.; Silver, L.; Weiner, A.; Mallick, S.; Dickerson, J.; Bergamini, M.V.; Robertson, S.M.; et al. Travoprost compared with latanoprost and timolol in patients with open-angle glaucoma or ocular hypertension. Am. J. Ophthalmol. 2001, 132, 472–484. [Google Scholar] [CrossRef]
  136. Hellberg, M.R.; Sallee, V.L.; McLaughlin, M.A.; Sharif, N.A.; Desantis, L.; Dean, T.R.; Zinke, P.W. Preclinical efficacy of travoprost, a potent and selective FP prostaglandin receptor agonist. J. Ocul. Pharmacol. Ther. 2001, 17, 421–432. [Google Scholar] [CrossRef]
  137. Carvalho, A.B.; Laus, J.L.; Costa, V.P.; Barros, P.S.M.; Silveira, P.R. Effects of travoprost 0.004% compared with latanoprost 0.005% on the intraocular pressure of normal dogs. Vet. Ophthalmol. 2006, 9, 121–125. [Google Scholar] [CrossRef]
  138. Schnichels, S.; Hurst, J.; de Vries, J.W.; Ullah, S.; Gruszka, A.; Kwak, M.; Löscher, M.; Dammeier, S.; Bartz-Schmidt, K.-U.; Spitzer, M.S.; et al. Self-assembled DNA nanoparticles loaded with travoprost for glaucoma-treatment. Nanomedicine 2020, 29, 102260. [Google Scholar] [CrossRef]
  139. Hernández, M.; Urcola, J.H.; Vecino, E. Retinal ganglion cell neuroprotection in a rat model of glaucoma following brimonidine, latanoprost or combined treatments. Exp. Eye Res. 2008, 86, 798–806. [Google Scholar] [CrossRef]
  140. McDonald, J.E.; Kiland, J.A.; Kaufman, P.L.; Bentley, E.; Ellinwood, N.M.; McLellan, G.J. Effect of topical latanoprost 0.005% on intraocular pressure and pupil diameter in normal and glaucomatous cats. Vet. Ophthalmol. 2016, 19, 13–23. [Google Scholar] [CrossRef]
  141. El-Nimri, N.W.; Wildsoet, C.F. Effects of topical latanoprost on intraocular pressure and myopia progression in young guinea pigs. Investig. Ophthalmol. Vis. Sci. 2018, 59, 2644–2651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Ling, Y.; Hu, Z.; Meng, Q.; Fang, P.; Liu, H. Bimatoprost increases mechanosensitivity of trigeminal ganglion neurons innervating the inner walls of rat anterior chambers via activation of TRPA1. Investig. Ophthalmol. Vis. Sci. 2016, 57, 567–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Crowston, J.G.; Lindsey, J.D.; Morris, C.A.; Wheeler, L.; Medeiros, F.A.; Weinreb, R.N. Effect of bimatoprost on intraocular pressure in prostaglandin FP receptor knockout mice. Investig. Ophthalmol. Vis. Sci. 2005, 46, 4571–4577. [Google Scholar] [CrossRef] [PubMed]
  144. Chen, J.; Dinh, T.; Woodward, D.F.; Holland, M.; Yuan, Y.-D.; Lin, T.-H.; Wheeler, L.A. Bimatoprost: Mechanism of ocular surface hyperemia associated with topical therapy. Cardiovasc. Drug Rev. 2005, 23, 231–246. [Google Scholar] [CrossRef] [PubMed]
  145. Stamer, W.D.; Piwnica, D.; Jolas, T.; Carling, R.W.; Cornell, C.L.; Fliri, H.; Martos, J.; Pettit, S.N.; Wang, J.W.; Woodward, D.F. Cellular basis for bimatoprost effects on human conventional outflow. Investig. Ophthalmol. Vis. Sci. 2010, 51, 5176–5181. [Google Scholar] [CrossRef]
  146. Bartoe, J.T.; Davidson, H.J.; Horton, M.T.; Jung, Y.; Brightman, A.H. The effects of bimatoprost and unoprostone isopropyl on the intraocular pressure of normal cats. Vet. Ophthalmol. 2005, 8, 247–252. [Google Scholar] [CrossRef]
  147. Ogundele, A.B.; Earnest, D.; McLaughlin, M.A. In vivo comparative study of ocular vasodilation, a relative indicator of hyperemia, in guinea pigs following treatment with bimatoprost ophthalmic solutions 0.01% and 0.03%. Clin. Ophthalmol. 2010, 4, 649–652. [Google Scholar] [CrossRef] [Green Version]
  148. Lee, S.S.; Burke, J.; Shen, J.; Almazan, A.; Orilla, W.; Hughes, P.; Zhang, J.; Li, H.; Struble, C.; Miller, P.E.; et al. Bimatoprost sustained-release intracameral implant reduces episcleral venous pressure in dogs. Vet. Ophthalmol. 2018, 21, 376–381. [Google Scholar] [CrossRef] [Green Version]
  149. Fukano, Y.; Kawazu, K. Disposition and metabolism of a novel prostanoid antiglaucoma medication, tafluprost, following ocular administration to rats. Drug Metab. Dispos. 2009, 37, 1622–1634. [Google Scholar] [CrossRef] [Green Version]
  150. Kanamori, A.; Naka, M.; Fukuda, M.; Nakamura, M.; Negi, A. Tafluprost protects rat retinal ganglion cells from apoptosis in vitro and in vivo. Graefes Arch. Clin. Exp. Ophthalmol. 2009, 247, 1353–1360. [Google Scholar] [CrossRef]
  151. Mayama, C.; Ishii, K.; Saeki, T.; Ota, T.; Tomidokoro, A.; Araie, M. Effects of topical phenylephrine and tafluprost on optic nerve head circulation in monkeys with unilateral experimental glaucoma. Investig. Ophthalmol. Vis. Sci. 2010, 51, 4117–4124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Izumi, N.; Nagaoka, T.; Sato, E.; Mori, F.; Takahashi, A.; Sogawa, K.; Yoshida, A. Short-term effects of topical tafluprost on retinal blood flow in cats. J. Ocul. Pharmacol. Ther. 2008, 24, 521–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Liu, Y.; Mao, W. Tafluprost once daily for treatment of elevated intraocular pressure in patients with open-angle glaucoma. Clin. Ophthalmol. 2013, 7, 7–14. [Google Scholar] [CrossRef] [Green Version]
  154. Kwak, J.; Kang, S.; Lee, E.R.; Park, S.; Park, S.; Park, E.; Lim, J.; Seo, K. Effect of preservative-free tafluprost on intraocular pressure, pupil diameter, and anterior segment structures in normal canine eyes. Vet. Ophthalmol. 2017, 20, 34–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Arfaee, F.; Armin, A. A comparison between the effect of topical tafluprost and latanoprost on intraocular pressure in healthy male guinea pigs. J. Exotic Pet Med. 2021, 39, 91–95. [Google Scholar] [CrossRef]
  156. Fuchshofer, R.; Kuespert, S.; Junglas, B.; Tamm, E.R. The prostaglandin f2α analog fluprostenol attenuates the fibrotic effects of connective tissue growth factor on human trabecular meshwork cells. J. Ocul. Pharmacol. Ther. 2014, 30, 237–245. [Google Scholar] [CrossRef] [PubMed]
  157. Saeki, T.; Tsuruga, H.; Aihara, M.; Araie, M.; Rittenhouse, K. Dose-Response Profile of PF-03187207 (PF-207) and Peak IOP Lowering Response Following Single Topical Administration to FP Receptor Knockout Mice vs. Wild Type Mice. Investig. Ophthalmol. Vis. Sci. 2009, 50, 4064. [Google Scholar]
  158. Krauss, A.H.P.; Impagnatiello, F.; Toris, C.B.; Gale, D.C.; Prasanna, G.; Borghi, V.; Chiroli, V.; Chong, W.K.M.; Carreiro, S.T.; Ongini, E. Ocular hypotensive activity of BOL-303259-X, a nitric oxide donating prostaglandin F2α agonist, in preclinical models. Exp. Eye Res. 2011, 93, 250–255. [Google Scholar] [CrossRef]
  159. Mehran, N.A.; Sinha, S.; Razeghinejad, R. New glaucoma medications: Latanoprostene bunod, netarsudil, and fixed combination netarsudil-latanoprost. Eye 2020, 34, 72–88. [Google Scholar] [CrossRef]
  160. Liu, H.K.; Chiou, G.C.; Garg, L.C. Ocular hypotensive effects of timolol in cat eyes. Arch. Ophthalmol. 1980, 98, 1467–1469. [Google Scholar] [CrossRef]
  161. Watanabe, K.; Chiou, G.C. Action mechanism of timolol to lower the intraocular pressure in rabbits. Ophthalmic Res. 1983, 15, 160–167. [Google Scholar] [CrossRef] [PubMed]
  162. Schuettauf, F.; Quinto, K.; Naskar, R.; Zurakowski, D. Effects of anti-glaucoma medications on ganglion cell survival: The DBA/2J mouse model. Vision Res. 2002, 42, 2333–2337. [Google Scholar] [CrossRef] [Green Version]
  163. Goto, W.; Ota, T.; Morikawa, N.; Otori, Y.; Hara, H.; Kawazu, K.; Miyawaki, N.; Tano, Y. Protective effects of timolol against the neuronal damage induced by glutamate and ischemia in the rat retina. Brain Res. 2002, 958, 10–19. [Google Scholar] [CrossRef]
  164. Watson, P.; Stjernschantz, J. A six-month, randomized, double-masked study comparing latanoprost with timolol in open-angle glaucoma and ocular hypertension. The Latanoprost Study Group. Ophthalmology 1996, 103, 126–137. [Google Scholar] [CrossRef]
  165. Bartels, S.P. Aqueous humor flow measured with fluorophotometry in timolol-treated primates. Investig. Ophthalmol. Vis. Sci. 1988, 29, 1498–1504. [Google Scholar]
  166. Yu, D.Y.; Su, E.N.; Cringle, S.J.; Alder, V.A.; Yu, P.K.; Desantis, L. Effect of betaxolol, timolol and nimodipine on human and pig retinal arterioles. Exp. Eye Res. 1998, 67, 73–81. [Google Scholar] [CrossRef] [PubMed]
  167. Smith, L.N.; Miller, P.E.; Felchle, L.M. Effects of topical administration of latanoprost, timolol, or a combination of latanoprost and timolol on intraocular pressure, pupil size, and heart rate in clinically normal dogs. Am. J. Vet. Res. 2010, 71, 1055–1061. [Google Scholar] [CrossRef]
  168. Millar, J.C.; Clark, A.F.; Pang, I.-H. Assessment of aqueous humor dynamics in the mouse by a novel method of constant-flow infusion. Investig. Ophthalmol. Vis. Sci. 2011, 52, 685–694. [Google Scholar] [CrossRef]
  169. Wood, J.P.; DeSantis, L.; Chao, H.M.; Osborne, N.N. Topically applied betaxolol attenuates ischaemia-induced effects to the rat retina and stimulates BDNF mRNA. Exp. Eye Res. 2001, 72, 79–86. [Google Scholar] [CrossRef]
  170. Osborne, N.N.; DeSantis, L.; Bae, J.H.; Ugarte, M.; Wood, J.P.; Nash, M.S.; Chidlow, G. Topically applied betaxolol attenuates NMDA-induced toxicity to ganglion cells and the effects of ischaemia to the retina. Exp. Eye Res. 1999, 69, 331–342. [Google Scholar] [CrossRef]
  171. Uji, Y.; Kuze, M.; Matubara, H.; Doi, M.; Sasoh, M. Effects of the beta1-selective adrenergic antagonist betaxolol on electroretinography in the perfused cat eye. Doc. Ophthalmol. 2003, 106, 37–41. [Google Scholar] [CrossRef] [PubMed]
  172. Holló, G.; Whitson, J.T.; Faulkner, R.; McCue, B.; Curtis, M.; Wieland, H.; Chastain, J.; Sanders, M.; DeSantis, L.; Przydryga, J.; et al. Concentrations of betaxolol in ocular tissues of patients with glaucoma and normal monkeys after 1 month of topical ocular administration. Investig. Ophthalmol. Vis. Sci. 2006, 47, 235–240. [Google Scholar] [CrossRef] [PubMed]
  173. Tamaki, Y.; Araie, M.; Tomita, K.; Nagahara, M. Effect of topical betaxolol on tissue circulation in the human optic nerve head. J. Ocul. Pharmacol. Ther. 1999, 15, 313–321. [Google Scholar] [CrossRef] [PubMed]
  174. Miller, P.E.; Schmidt, G.M.; Vainisi, S.J.; Swanson, J.F.; Herrmann, M.K. The efficacy of topical prophylactic antiglaucoma therapy in primary closed angle glaucoma in dogs: A multicenter clinical trial. J. Am. Anim. Hosp. Assoc. 2000, 36, 431–438. [Google Scholar] [CrossRef]
  175. Goldenberg-Cohen, N.; Dadon-Bar-El, S.; Hasanreisoglu, M.; Avraham-Lubin, B.C.R.; Dratviman-Storobinsky, O.; Cohen, Y.; Weinberger, D. Possible neuroprotective effect of brimonidine in a mouse model of ischaemic optic neuropathy. Clin. Exp. Ophthalmol. 2009, 37, 718–729. [Google Scholar] [CrossRef]
  176. Acheampong, A.A.; Small, D.; Baumgarten, V.; Welty, D.; Tang-Liu, D. Formulation effects on ocular absorption of brimonidine in rabbit eyes. J. Ocul. Pharmacol. Ther. 2002, 18, 325–337. [Google Scholar] [CrossRef]
  177. Gelatt, K.N.; MacKay, E.O. Effect of single and multiple doses of 0.2% brimonidine tartrate in the glaucomatous Beagle. Vet. Ophthalmol. 2002, 5, 253–262. [Google Scholar] [CrossRef]
  178. Burke, J.; Schwartz, M. Preclinical evaluation of brimonidine. Surv. Ophthalmol. 1996, 41, S9–S18. [Google Scholar] [CrossRef]
  179. Schnichels, S.; Hurst, J.; de Vries, J.W.; Ullah, S.; Frößl, K.; Gruszka, A.; Löscher, M.; Bartz-Schmidt, K.-U.; Spitzer, M.S.; Herrmann, A. Improved Treatment Options for Glaucoma with Brimonidine-Loaded Lipid DNA Nanoparticles. ACS Appl. Mater. Interfaces 2021, 13, 9445–9456. [Google Scholar] [CrossRef]
  180. Tamhane, M.; Luu, K.T.; Attar, M. Ocular pharmacokinetics of brimonidine drug delivery system in monkeys and translational modeling for selection of dose and frequency in clinical trials. J. Pharmacol. Exp. Ther. 2021, 378, 207–214. [Google Scholar] [CrossRef]
  181. Toris, C.B.; Gleason, M.L.; Camras, C.B.; Yablonski, M.E. Effects of brimonidine on aqueous humor dynamics in human eyes. Arch. Ophthalmol. 1995, 113, 1514–1517. [Google Scholar] [CrossRef] [PubMed]
  182. Liu, Y.; Wang, Y.; Lv, H.; Jiang, X.; Zhang, M.; Li, X. α-adrenergic agonist brimonidine control of experimentally induced myopia in guinea pigs: A pilot study. Mol. Vis. 2017, 23, 785–798. [Google Scholar] [PubMed]
  183. Morrison, J.C.; Nylander, K.B.; Lauer, A.K.; Cepurna, W.O.; Johnson, E. Glaucoma drops control intraocular pressure and protect optic nerves in a rat model of glaucoma. Investig. Ophthalmol. Vis. Sci. 1998, 39, 526–531. [Google Scholar]
  184. Gabelt, B.T.; Robinson, J.C.; Hubbard, W.C.; Peterson, C.M.; Debink, N.; Wadhwa, A.; Kaufman, P.L. Apraclonidine and brimonidine effects on anterior ocular and cardiovascular physiology in normal and sympathectomized monkeys. Exp. Eye Res. 1994, 59, 633–644. [Google Scholar] [CrossRef] [PubMed]
  185. Toris, C.B.; Tafoya, M.E.; Camras, C.B.; Yablonski, M.E. Effects of apraclonidine on aqueous humor dynamics in human eyes. Ophthalmology 1995, 102, 456–461. [Google Scholar] [CrossRef]
  186. Orgül, S.; Bacon, D.R.; Van Buskirk, E.M.; Cioffi, G.A. Optic nerve vasomotor effects of topical apraclonidine hydrochloride. Br. J. Ophthalmol. 1996, 80, 82–84. [Google Scholar] [CrossRef] [Green Version]
  187. Miller, P.E.; Nelson, M.J.; Rhaesa, S.L. Effects of topical administration of 0.5% apraclonidine on intraocular pressure, pupil size, and heart rate in clinically normal dogs. Am. J. Vet. Res. 1996, 57, 79–82. [Google Scholar]
  188. Miller, P.E.; Rhaesa, S.L. Effects of topical administration of 0.5% apraclonidine on intraocular pressure, pupil size, and heart rate in clinically normal cats. Am. J. Vet. Res. 1996, 57, 83–86. [Google Scholar]
  189. Li, T.; Wang, Y.; Chen, J.; Gao, X.; Pan, S.; Su, Y.; Zhou, X. Co-delivery of brinzolamide and miRNA-124 by biodegradable nanoparticles as a strategy for glaucoma therapy. Drug Deliv. 2020, 27, 410–421. [Google Scholar] [CrossRef] [Green Version]
  190. Desantis, L. Preclinical overview of brinzolamide1. Surv. Ophthalmol. 2000, 44, S119–S129. [Google Scholar] [CrossRef]
  191. Li, N.; Shi, H.-M.; Cong, L.; Lu, Z.-Z.; Ye, W.; Zhang, Y.-Y. Outflow facility efficacy of five drugs in enucleated porcine eyes by a method of constant-pressure perfusion. Int. J. Clin. Exp. Med. 2015, 8, 7184–7191. [Google Scholar] [PubMed]
  192. Di, Y.; Luo, X.-M.; Qiao, T.; Lu, N. Intraocular pressure with rebound tonometry and effects of topical intraocular pressure reducing medications in guinea pigs. Int. J. Ophthalmol. 2017, 10, 186–190. [Google Scholar] [CrossRef] [PubMed]
  193. Toris, C.B.; Zhan, G.-L.; McLaughlin, M.A. Effects of brinzolamide on aqueous humor dynamics in monkeys and rabbits. J. Ocul. Pharmacol. Ther. 2003, 19, 397–404. [Google Scholar] [CrossRef] [PubMed]
  194. Cvetkovic, R.S.; Perry, C.M. Brinzolamide: A review of its use in the management of primary open-angle glaucoma and ocular hypertension. Drugs Aging 2003, 20, 919–947. [Google Scholar] [CrossRef] [PubMed]
  195. Ingram, C.J.; Brubaker, R.F. Effect of brinzolamide and dorzolamide on aqueous humor flow in human eyes. Am. J. Ophthalmol. 1999, 128, 292–296. [Google Scholar] [CrossRef]
  196. Chandra, S.; Muir, E.R.; Deo, K.; Kiel, J.W.; Duong, T.Q. Effects of dorzolamide on retinal and choroidal blood flow in the DBA/2J mouse model of glaucoma. Investig. Ophthalmol. Vis. Sci. 2016, 57, 826–831. [Google Scholar] [CrossRef] [Green Version]
  197. Pitha, I.; Kimball, E.C.; Oglesby, E.N.; Pease, M.E.; Fu, J.; Schaub, J.; Kim, Y.-C.; Hu, Q.; Hanes, J.; Quigley, H.A. Sustained Dorzolamide Release Prevents Axonal and Retinal Ganglion Cell Loss in a Rat Model of IOP-Glaucoma. Transl. Vis. Sci. Technol. 2018, 7, 13. [Google Scholar] [CrossRef] [Green Version]
  198. Percicot, C.L.; Schnell, C.R.; Debon, C.; Hariton, C. Continuous intraocular pressure measurement by telemetry in alpha-chymotrypsin-induced glaucoma model in the rabbit: Effects of timolol, dorzolamide, and epinephrine. J. Pharmacol. Toxicol. Methods 1996, 36, 223–228. [Google Scholar] [CrossRef]
  199. Stefánsson, E.; Jensen, P.K.; Eysteinsson, T.; Bang, K.; Kiilgaard, J.F.; Dollerup, J.; Scherfig, E.; la Cour, M. Optic Nerve Oxygen Tension in Pigs and the Effect of Carbonic Anhydrase Inhibitors. Investig. Ophthalmol. Vis. Sci. 1999, 40, 2756–2761. [Google Scholar]
  200. Dietrich, U.M.; Chandler, M.J.; Cooper, T.; Vidyashankar, A.; Chen, G. Effects of topical 2% dorzolamide hydrochloride alone and in combination with 0.5% timolol maleate on intraocular pressure in normal feline eyes. Vet. Ophthalmol. 2007, 10, 95–100. [Google Scholar] [CrossRef]
  201. Gelatt, K.N.; MacKay, E.O. Changes in intraocular pressure associated with topical dorzolamide and oral methazolamide in glaucomatous dogs. Vet. Ophthalmol. 2001, 4, 61–67. [Google Scholar] [CrossRef] [PubMed]
  202. Wang, R.F.; Serle, J.B.; Gagliuso, D.J.; Podos, S.M. Comparison of the ocular hypotensive effect of brimonidine, dorzolamide, latanoprost, or artificial tears added to timolol in glaucomatous monkey eyes. J. Glaucoma 2000, 9, 458–462. [Google Scholar] [CrossRef]
  203. Larsson, L.I.; Alm, A. Aqueous humor flow in human eyes treated with dorzolamide and different doses of acetazolamide. Arch. Ophthalmol. 1998, 116, 19–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Avila, M.Y.; Stone, R.A.; Civan, M.M. Knockout of A3 Adenosine Receptors Reduces Mouse Intraocular Pressure. Investig. Ophthalmol. Vis. Sci. 2002, 43, 3021–3026. [Google Scholar]
  205. Findl, O.; Hansen, R.M.; Fulton, A.B. The effects of acetazolamide on the electroretinographic responses in rats. Investig. Ophthalmol. Vis. Sci. 1995, 36, 1019–1026. [Google Scholar]
  206. Kaur, I.P.; Singh, M.; Kanwar, M. Formulation and evaluation of ophthalmic preparations of acetazolamide. Int. J. Pharm. 2000, 199, 119–127. [Google Scholar] [CrossRef]
  207. Maren, T.H. Ion secretion into the posterior aqueous humor of dogs and monkeys. Exp. Eye Res. 1977, 25, 245–247. [Google Scholar] [CrossRef]
  208. Macri, F.J.; Dixon, R.L.; Rall, D.P. Aqueous humor turnover rates in the cat. I. Effect of acetazolamide. Investig. Ophthalmol. 1965, 4, 927–934. [Google Scholar]
  209. Fridriksdóttir, H.; Loftsson, T.; Stefánsson, E. Formulation and testing of methazolamide cyclodextrin eye drop solutions. J. Control. Release 1997, 44, 95–99. [Google Scholar] [CrossRef]
  210. Guđmundsdóttir, E.; Stefánsson, E.; Bjarnadóttir, G.; Sigurjónsdóttir, J.F.; Guđmundsdóttir, G.; Masson, M.; Loftsson, T. Methazolamide 1% in Cyclodextrin Solution Lowers IOP in Human Ocular Hypertension. Investig. Ophthalmol. Vis. Sci. 2000, 41, 3552–3554. [Google Scholar]
  211. Li, G.; Mukherjee, D.; Navarro, I.; Ashpole, N.E.; Sherwood, J.M.; Chang, J.; Overby, D.R.; Yuan, F.; Gonzalez, P.; Kopczynski, C.C.; et al. Visualization of conventional outflow tissue responses to netarsudil in living mouse eyes. Eur. J. Pharmacol. 2016, 787, 20–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Ren, R.; Li, G.; Le, T.D.; Kopczynski, C.; Stamer, W.D.; Gong, H. Netarsudil increases outflow facility in human eyes through multiple mechanisms. Investig. Ophthalmol. Vis. Sci. 2016, 57, 6197–6209. [Google Scholar] [CrossRef] [PubMed]
  213. Leary, K.A.; Lin, K.-T.; Steibel, J.P.; Harman, C.D.; Komáromy, A.M. Safety and efficacy of topically administered netarsudil (RhopressaTM) in normal and glaucomatous dogs with ADAMTS10-open-angle glaucoma (ADAMTS10-OAG). Vet. Ophthalmol. 2021, 24, 75–86. [Google Scholar] [CrossRef] [PubMed]
  214. Kitaoka, Y.; Sase, K.; Tsukahara, C.; Fujita, N.; Arizono, I.; Kogo, J.; Tokuda, N.; Takagi, H. Axonal Protection by Netarsudil, a ROCK Inhibitor, Is Linked to an AMPK-Autophagy Pathway in TNF-Induced Optic Nerve Degeneration. Investig. Ophthalmol. Vis. Sci. 2022, 63, 4. [Google Scholar] [CrossRef] [PubMed]
  215. Lin, C.-W.; Sherman, B.; Moore, L.A.; Laethem, C.L.; Lu, D.-W.; Pattabiraman, P.P.; Rao, P.V.; deLong, M.A.; Kopczynski, C.C. Discovery and preclinical development of netarsudil, a novel ocular hypotensive agent for the treatment of glaucoma. J. Ocul. Pharmacol. Ther. 2018, 34, 40–51. [Google Scholar] [CrossRef]
  216. Isobe, T.; Kasai, T.; Kawai, H. Ocular penetration and pharmacokinetics of ripasudil following topical administration to rabbits. J. Ocul. Pharmacol. Ther. 2016, 32, 405–414. [Google Scholar] [CrossRef]
  217. Kamiya, T.; Omae, T.; Nakabayashi, S.; Takahashi, K.; Tanner, A.; Yoshida, A. Effect of Rho Kinase Inhibitor Ripasudil (K-115) on Isolated Porcine Retinal Arterioles. J. Ocul. Pharmacol. Ther. 2021, 37, 104–111. [Google Scholar] [CrossRef]
  218. Nakabayashi, S.; Kawai, M.; Yoshioka, T.; Song, Y.-S.; Tani, T.; Yoshida, A.; Nagaoka, T. Effect of intravitreal Rho kinase inhibitor ripasudil (K-115) on feline retinal microcirculation. Exp. Eye Res. 2015, 139, 132–135. [Google Scholar] [CrossRef]
  219. Nishijima, E.; Namekata, K.; Kimura, A.; Guo, X.; Harada, C.; Noro, T.; Nakano, T.; Harada, T. Topical ripasudil stimulates neuroprotection and axon regeneration in adult mice following optic nerve injury. Sci. Rep. 2020, 10, 15709. [Google Scholar] [CrossRef]
  220. Wada, Y.; Higashide, T.; Nagata, A.; Sugiyama, K. Effects of ripasudil, a rho kinase inhibitor, on blood flow in the optic nerve head of normal rats. Graefe’s Arch. Clin. Exp. Ophthalmol. 2019, 257, 303–311. [Google Scholar] [CrossRef]
  221. Inoue, T.; Tanihara, H. Ripasudil hydrochloride hydrate: Targeting Rho kinase in the treatment of glaucoma. Expert Opin. Pharmacother. 2017, 18, 1669–1673. [Google Scholar] [CrossRef] [PubMed]
  222. Yamamoto, K.; Maruyama, K.; Himori, N.; Omodaka, K.; Yokoyama, Y.; Shiga, Y.; Morin, R.; Nakazawa, T. The novel Rho kinase (ROCK) inhibitor K-115: A new candidate drug for neuroprotective treatment in glaucoma. Investig. Ophthalmol. Vis. Sci. 2014, 55, 7126–7136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Song, H.; Gao, D. Fasudil, a Rho-associated protein kinase inhibitor, attenuates retinal ischemia and reperfusion injury in rats. Int. J. Mol. Med. 2011, 28, 193–198. [Google Scholar] [CrossRef] [Green Version]
  224. Khallaf, A.M.; El-Moslemany, R.M.; Ahmed, M.F.; Morsi, M.H.; Khalafallah, N.M. Exploring a Novel Fasudil-Phospholipid Complex Formulated as Liposomal Thermosensitive in situ Gel for Glaucoma. Int. J. Nanomed. 2022, 17, 163–181. [Google Scholar] [CrossRef] [PubMed]
  225. Ichikawa, M.; Yoshida, J.; Saito, K.; Sagawa, H.; Tokita, Y.; Watanabe, M. Differential effects of two ROCK inhibitors, Fasudil and Y-27632, on optic nerve regeneration in adult cats. Brain Res. 2008, 1201, 23–33. [Google Scholar] [CrossRef]
  226. Pakravan, M.; Beni, A.N.; Ghahari, E.; Varshochian, R.; Yazdani, S.; Esfandiari, H.; Ahmadieh, H. The Ocular Hypotensive Efficacy of Topical Fasudil, a Rho-Associated Protein Kinase Inhibitor, in Patients with End-Stage Glaucoma. Am. J. Ther. 2016, 24, e676–e680. [Google Scholar] [CrossRef]
  227. Da, B.; Cao, Y.; Wei, H.; Chen, Z.; Shui, Y.; Li, Z. Antagonistic effects of tranilast on proliferation and collagen synthesis induced by TGF-beta2 in cultured human trabecular meshwork cells. J. Huazhong Univ. Sci. Technol. Med. Sci. 2004, 24, 490–496. [Google Scholar] [CrossRef]
  228. Cao, Y.; Hu, Y.; Li, J.; Shui, Y.; Da, B.; Wei, H. Effect of Tranilast on Collagen Synthesis and TGF–Beta2 Expression of Cultured Human Lamina Cribrosa Astrocytes. Investig. Ophthalmol. Vis. Sci. 2006, 47, 1544. [Google Scholar]
  229. Spitzer, M.S.; Sat, M.; Schramm, C.; Schnichels, S.; Schultheiss, M.; Yoeruek, E.; Dzhelebov, D.; Szurman, P. Biocompatibility and antifibrotic effect of UV-cross-linked hyaluronate as a release-system for tranilast after trabeculectomy in a rabbit model—A pilot study. Curr. Eye Res. 2012, 37, 463–470. [Google Scholar] [CrossRef]
  230. Pfeiffer, N.; Voykov, B.; Renieri, G.; Bell, K.; Richter, P.; Weigel, M.; Thieme, H.; Wilhelm, B.; Lorenz, K.; Feindor, M.; et al. First-in-human phase I study of ISTH0036, an antisense oligonucleotide selectively targeting transforming growth factor beta 2 (TGF-β2), in subjects with open-angle glaucoma undergoing glaucoma filtration surgery. PLoS ONE 2017, 12, e0188899. [Google Scholar] [CrossRef] [Green Version]
  231. Hasenbach, K.; Van Bergen, T.; Vandewalle, E.; De Groef, L.; Van Hove, I.; Moons, L.; Stalmans, I.; Fettes, P.; Leo, E.; Wosikowski, K.; et al. Potent and selective antisense oligonucleotides targeting the transforming growth factor beta (TGF-β) isoforms in advanced glaucoma: A preclinical evaluation. MAIO 2016, 1, 20–28. [Google Scholar] [CrossRef]
  232. Nakamura, H.; Siddiqui, S.S.; Shen, X.; Malik, A.B.; Pulido, J.S.; Kumar, N.M.; Yue, B.Y.J.T. RNA interference targeting transforming growth factor-beta type II receptor suppresses ocular inflammation and fibrosis. Mol. Vis. 2004, 10, 703–711. [Google Scholar] [PubMed]
  233. Mead, A.L.; Wong, T.T.L.; Cordeiro, M.F.; Anderson, I.K.; Khaw, P.T. Evaluation of Anti-TGF-β2 Antibody as a New Postoperative Anti-scarring Agent in Glaucoma Surgery. Investig. Ophthalmol. Vis. Sci. 2003, 44, 3394. [Google Scholar] [CrossRef] [Green Version]
  234. CAT-152 0102 Trabeculectomy Study Group; Khaw, P.; Grehn, F.; Holló, G.; Overton, B.; Wilson, R.; Vogel, R.; Smith, Z. A phase III study of subconjunctival human anti-transforming growth factor beta(2) monoclonal antibody (CAT-152) to prevent scarring after first-time trabeculectomy. Ophthalmology 2007, 114, 1822–1830. [Google Scholar] [CrossRef] [PubMed]
  235. Shan, S.-W.; Do, C.-W.; Lam, T.C.; Li, H.-L.; Stamer, W.D.; To, C.-H. Thrombospondin-1 mediates Rho-kinase inhibitor-induced increase in outflow-facility. J. Cell. Physiol. 2021, 236, 8226–8238. [Google Scholar] [CrossRef]
  236. Chen, W.-S.; Cao, Z.; Krishnan, C.; Panjwani, N. Verteporfin without light stimulation inhibits YAP activation in trabecular meshwork cells: Implications for glaucoma treatment. Biochem. Biophys. Res. Commun. 2015, 466, 221–225. [Google Scholar] [CrossRef]
  237. Matsubara, A.; Nakazawa, T.; Husain, D.; Iliaki, E.; Connolly, E.; Michaud, N.A.; Gragoudas, E.S.; Miller, J.W. Investigating the effect of ciliary body photodynamic therapy in a glaucoma mouse model. Investig. Ophthalmol. Vis. Sci. 2006, 47, 2498–2507. [Google Scholar] [CrossRef]
  238. Parodi, M.B.; Iacono, P. Photodynamic therapy with verteporfin for anterior segment neovascularizations in neovascular glaucoma. Am. J. Ophthalmol. 2004, 138, 157–158. [Google Scholar] [CrossRef]
  239. Ko, M.-L.; Chen, C.-F.; Peng, P.-H.; Peng, Y.-H. Simvastatin upregulates Bcl-2 expression and protects retinal neurons from early ischemia/reperfusion injury in the rat retina. Exp. Eye Res. 2011, 93, 580–585. [Google Scholar] [CrossRef]
  240. Krempler, K.; Schmeer, C.W.; Isenmann, S.; Witte, O.W.; Löwel, S. Simvastatin improves retinal ganglion cell survival and spatial vision after acute retinal ischemia/reperfusion in mice. Investig. Ophthalmol. Vis. Sci. 2011, 52, 2606–2618. [Google Scholar] [CrossRef]
  241. Nagaoka, T.; Takahashi, A.; Sato, E.; Izumi, N.; Hein, T.W.; Kuo, L.; Yoshida, A. Effect of systemic administration of simvastatin on retinal circulation. Arch. Ophthalmol. 2006, 124, 665–670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Nagaoka, T.; Hein, T.W.; Yoshida, A.; Kuo, L. Simvastatin elicits dilation of isolated porcine retinal arterioles: Role of nitric oxide and mevalonate-rho kinase pathways. Investig. Ophthalmol. Vis. Sci. 2007, 48, 825–832. [Google Scholar] [CrossRef] [PubMed]
  243. Kim, M.-L.; Sung, K.R.; Shin, J.A.; Young Yoon, J.; Jang, J. Statins reduce TGF-beta2-modulation of the extracellular matrix in cultured astrocytes of the human optic nerve head. Exp. Eye Res. 2017, 164, 55–63. [Google Scholar] [CrossRef]
  244. Kim, M.; Shin, J.; Sung, K. Statins regulate MMP-2 and MMP-9 secretion and activation in human ONH astrocytes. Investig. Ophthalmol. Vis. Sci. 2018, 59, 6145. [Google Scholar]
  245. Villarreal, G.; Chatterjee, A.; Oh, S.S.; Oh, D.-J.; Rhee, D.J. Pharmacological regulation of SPARC by lovastatin in human trabecular meshwork cells. Investig. Ophthalmol. Vis. Sci. 2014, 55, 1657–1665. [Google Scholar] [CrossRef] [Green Version]
  246. Park, J.-H.; Yoo, C.; Kim, Y.Y. Effect of Lovastatin on Wound-Healing Modulation After Glaucoma Filtration Surgery in a Rabbit Model. Investig. Ophthalmol. Vis. Sci. 2016, 57, 1871–1877. [Google Scholar] [CrossRef] [Green Version]
  247. Song, X.-Y.; Chen, Y.-Y.; Liu, W.-T.; Cong, L.; Zhang, J.-L.; Zhang, Y.; Zhang, Y.-Y. Atorvastatin reduces IOP in ocular hypertension in vivo and suppresses ECM in trabecular meshwork perhaps via FGD4. Int. J. Mol. Med. 2022, 49, 76. [Google Scholar] [CrossRef]
  248. Kim, M.-L.; Sung, K.R.; Kwon, J.; Choi, G.W.; Shin, J.A. Neuroprotective effect of statins in a rat model of chronic ocular hypertension. Int. J. Mol. Sci. 2021, 22, 12500. [Google Scholar] [CrossRef]
  249. Cong, L.; Fu, S.; Zhang, J.; Zhao, J.; Zhang, Y. Effects of atorvastatin on porcine aqueous humour outflow and trabecular meshwork cells. Exp. Ther. Med. 2018, 15, 210–216. [Google Scholar] [CrossRef] [Green Version]
  250. Agarwal, R.; Krasilnikova, A.; Mohamed, S.N.L. Topical losartan reduces IOP by altering TM morphology in rats with steroid-induced ocular hypertension. Indian J. Physiol. 2018, 62, 238–248. [Google Scholar]
  251. Shah, G.B.; Sharma, S.; Mehta, A.A.; Goyal, R.K. Oculohypotensive effect of angiotensin-converting enzyme inhibitors in acute and chronic models of glaucoma. J. Cardiovasc. Pharmacol. 2000, 36, 169–175. [Google Scholar] [CrossRef] [PubMed]
  252. Costagliola, C.; Verolino, M.; De Rosa, M.L.; Iaccarino, G.; Ciancaglini, M.; Mastropasqua, L. Effect of oral losartan potassium administration on intraocular pressure in normotensive and glaucomatous human subjects. Exp. Eye Res. 2000, 71, 167–171. [Google Scholar] [CrossRef] [PubMed]
  253. Sawaguchi, S.; Yue, B.Y.; Yeh, P.; Tso, M.O. Effects of intracameral injection of chondroitinase ABC in vivo. Arch. Ophthalmol. 1992, 110, 110–117. [Google Scholar] [CrossRef] [PubMed]
  254. Murienne, B.J.; Chen, M.L.; Quigley, H.A.; Nguyen, T.D. The contribution of glycosaminoglycans to the mechanical behaviour of the posterior human sclera. J. R. Soc. Interface 2016, 13, 20160367. [Google Scholar] [CrossRef]
  255. Kirihara, T.; Shimazaki, A.; Nakamura, M.; Miyawaki, N. Ocular hypotensive efficacy of Src-family tyrosine kinase inhibitors via different cellular actions from Rock inhibitors. Exp. Eye Res. 2014, 119, 97–105. [Google Scholar] [CrossRef]
  256. Belmadani, S.; Bernal, J.; Wei, C.-C.; Pallero, M.A.; Dell’italia, L.; Murphy-Ullrich, J.E.; Berecek, K.H. A thrombospondin-1 antagonist of transforming growth factor-beta activation blocks cardiomyopathy in rats with diabetes and elevated angiotensin II. Am. J. Pathol. 2007, 171, 777–789. [Google Scholar] [CrossRef] [Green Version]
  257. Lu, A.; Miao, M.; Schoeb, T.R.; Agarwal, A.; Murphy-Ullrich, J.E. Blockade of TSP1-dependent TGF-β activity reduces renal injury and proteinuria in a murine model of diabetic nephropathy. Am. J. Pathol. 2011, 178, 2573–2586. [Google Scholar] [CrossRef] [Green Version]
  258. Leask, A. Breathe, breathe in the air: The anti-CCN2 antibody pamrevlumab (FG-3019) completes a successful phase II clinical trial for idiopathic pulmonary fibrosis. J. Cell Commun. Signal. 2019, 13, 441–442. [Google Scholar] [CrossRef]
  259. Wang, J.; Harris, A.; Prendes, M.A.; Alshawa, L.; Gross, J.C.; Wentz, S.M.; Rao, A.B.; Kim, N.J.; Synder, A.; Siesky, B. Targeting Transforming Growth Factor-β Signaling in Primary Open-Angle Glaucoma. J. Glaucoma 2017, 26, 390–395. [Google Scholar] [CrossRef]
  260. Webber, H.C.; Bermudez, J.Y.; Sethi, A.; Clark, A.F.; Mao, W. Crosstalk between TGFβ and Wnt signaling pathways in the human trabecular meshwork. Exp. Eye Res. 2016, 148, 97–102. [Google Scholar] [CrossRef] [Green Version]
  261. Wordinger, R.J.; Sharma, T.; Clark, A.F. The role of TGF-β2 and bone morphogenetic proteins in the trabecular meshwork and glaucoma. J. Ocul. Pharmacol. Ther. 2014, 30, 154–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Tanihara, H.; Inoue, T.; Yamamoto, T.; Kuwayama, Y.; Abe, H.; Suganami, H.; Araie, M.; K-115 Clinical Study Group. Intra-ocular pressure-lowering effects of a Rho kinase inhibitor, ripasudil (K-115), over 24 hours in primary open-angle glaucoma and ocular hypertension: A randomized, open-label, crossover study. Acta Ophthalmol. 2015, 93, e254–e260. [Google Scholar] [CrossRef]
  263. Tanihara, H.; Inoue, T.; Yamamoto, T.; Kuwayama, Y.; Abe, H.; Araie, M.; K-115 Clinical Study Group. Phase 2 randomized clinical study of a Rho kinase inhibitor, K-115, in primary open-angle glaucoma and ocular hypertension. Am. J. Ophthalmol. 2013, 156, 731–736. [Google Scholar] [CrossRef] [PubMed]
  264. Sato, S.; Hirooka, K.; Nitta, E.; Ukegawa, K.; Tsujikawa, A. Additive intraocular pressure lowering effects of the rho kinase inhibitor, ripasudil in glaucoma patients not able to obtain adequate control after other maximal tolerated medical therapy. Adv. Ther. 2016, 33, 1628–1634. [Google Scholar] [CrossRef]
  265. Pokrovskaya, O.; Wallace, D.; O’Brien, C. The emerging role of statins in glaucoma pathological mechanisms and therapeutics. Open J. Ophthalmol. 2014, 4, 124–138. [Google Scholar] [CrossRef] [Green Version]
  266. McCann, P.; Hogg, R.E.; Fallis, R.; Azuara-Blanco, A. The Effect of Statins on Intraocular Pressure and on the Incidence and Progression of Glaucoma: A Systematic Review and Meta-Analysis. Investig. Ophthalmol. Vis. Sci. 2016, 57, 2729–2748. [Google Scholar] [CrossRef] [Green Version]
  267. Yuan, Y.; Xiong, R.; Wu, Y.; Ha, J.; Wang, W.; Han, X.; He, M. Associations of statin use with the onset and progression of open-angle glaucoma: A systematic review and meta-analysis. EClinicalMedicine 2022, 46, 101364. [Google Scholar] [CrossRef]
  268. McGwin, G.; McNeal, S.; Owsley, C.; Girkin, C.; Epstein, D.; Lee, P.P. Statins and other cholesterol-lowering medications and the presence of glaucoma. Arch. Ophthalmol. 2004, 122, 822–826. [Google Scholar] [CrossRef] [Green Version]
  269. Weinreb, R.N. Enhancement of scleral macromolecular permeability with prostaglandins. Trans. Am. Ophthalmol. Soc. 2001, 99, 319–343. [Google Scholar]
Figure 1. Mechanotransduction and optic nerve head remodeling. (A) Healthy optic nerve head (ONH) anatomy detailing key regions. (B,C) Increased intraocular pressure (IOP, blue arrows) is counterbalanced by optic nerve sheath pressure (ONSP, open arrows) resulting in tissue strain in the optic nerve head (ONH). This can damage axons directly (red) and activates cellular mechanotransduction that drives remodeling of the lamina cribrosa (LC) and peripapillary scleral (ppScl). (C) This remodeling alters the material properties and tissue architecture that modulates the stain that drives further remodeling. (D) This creates a negative feedback loop (−) that increases the vulnerability of the RGC axons to further glaucomatous injury. Deformation of any mechanical structure under load (strain) is determined by the loading forces (stress) along with its architecture and material properties.
Figure 1. Mechanotransduction and optic nerve head remodeling. (A) Healthy optic nerve head (ONH) anatomy detailing key regions. (B,C) Increased intraocular pressure (IOP, blue arrows) is counterbalanced by optic nerve sheath pressure (ONSP, open arrows) resulting in tissue strain in the optic nerve head (ONH). This can damage axons directly (red) and activates cellular mechanotransduction that drives remodeling of the lamina cribrosa (LC) and peripapillary scleral (ppScl). (C) This remodeling alters the material properties and tissue architecture that modulates the stain that drives further remodeling. (D) This creates a negative feedback loop (−) that increases the vulnerability of the RGC axons to further glaucomatous injury. Deformation of any mechanical structure under load (strain) is determined by the loading forces (stress) along with its architecture and material properties.
Ijms 23 08068 g001
Figure 2. Canonical and non-canonical TGF-β pathways of importance and notable interactions with CTGF and integrin signaling. All pathways ultimately lead to alterations in ECM remodeling responses when activated. Adapted from “Canonical and Non-canonical TGFb Pathways in EMT”, by BioRender.com (2022). Available online: https://app.biorender.com/biorender-templates, accessed on 7 June 2022.
Figure 2. Canonical and non-canonical TGF-β pathways of importance and notable interactions with CTGF and integrin signaling. All pathways ultimately lead to alterations in ECM remodeling responses when activated. Adapted from “Canonical and Non-canonical TGFb Pathways in EMT”, by BioRender.com (2022). Available online: https://app.biorender.com/biorender-templates, accessed on 7 June 2022.
Ijms 23 08068 g002
Table 1. Potential therapeutic targets to alter glaucomatous remodeling.
Table 1. Potential therapeutic targets to alter glaucomatous remodeling.
Mechanism of ActionDrug(s)Impact on Optic Nerve RemodelingModels TestedReferences
Prostaglandin F receptor agonistBimatoprost, Latanoprost, Fluprostenol, Tafluprost, TravoprostUpregulation of MMP-1, -3, -9Mouse, Rat, Rabbit, Guinea Pig, Cat, Dog, Pig, Primate, Human[131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156]
Hybrid prostaglandin F receptor agonist and nitric oxide donatorLatanoprostene bunodUpregulation of MMPs and decrease cell contractilityMouse, Rabbit, Dog, Primate, Human[157,158,159]
β-adrenoceptor antagonistBetaxolol, TimololIncreased blood flow velocityMouse, Rat, Rabbit, Cat, Dog, Pig, Primate, Human[160,161,162,163,164,165,166,167,168,169,170,171,172,173,174]
α2-adrenergic agonistApraclonidine, BrimonidineAnti-apoptotic; RGC survival signalMouse, Rat, Guinea Pig, Rabbit, Cat, Dog, Pig, Primate, Human[139,175,176,177,178,179,180,181,182,183,184,185,186,187,188]
Carbonic anhydrase inhibitorAcetazolamide, Brinzolamide, Dorzolamide, MethazolamideIncreased blood flow and oxygen tensionMouse, Rat, Guinea Pig, Rabbit, Dog, Pig, Primate, Human[189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210]
ROCK InhibitorFasudil, Netarsudil, RipasudilInhibits contractility and migration of fibroblasts; inhibits production of ECM; inhibits cell death pathwaysMouse, Rat, Rabbit, Dog, Primate, Human[211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226]
Inhibits secretion of TGF-βTranilastPrevents TGF-β mediated fibrotic responses by nearby cellsRabbit, Human culture[227,228,229]
Inhibit transcription of TGF-βISTH0036, TbetaRII (RNAi)Decreased levels of TGF-β expressionMouse, Human Culture, Human[230,231,232]
Direct immunosuppression of TGF-βLerdelimumabTargeted inactivation of TGF-β to prevent receptor bindingRabbit, Human[233,234]
Inhibit TSP1 binding to LAPLSKLInhibits TSP1 mediated activation of latent TGF-βMouse[235]
Direct immunosuppression of CTGFPamrevlumabInhibits CTGF interaction with TGF-βHuman Culture[37]
Reduce YAP and CTGF expressionVerteporfin (without light activation)Reduces cell contractility via YAP; reduces CTGF interaction with TGF-βMouse, Human Culture, Human[236,237,238]
Increased nitric oxide productionAtorvastatin, Lovastatin, SimvastatinInhibit RhoA/ROCK pathway and reduce levels of MMP-2 and -9, decrease cell contractilityMouse, Rat, Rabbit, Dog, Pig, Human Culture[65,239,240,241,242,243,244,245,246,247,248,249]
Angiotensin 1 receptor (AT1R) inhibitorLosartanInhibits Smad2 phosphorylationMice, Rat, Rabbit, Human[130,250,251,252]
Glycosaminoglycan degrading enzymeChondroitinase ABCWeakens ECM (reduces stiffness)Rat, Pig, Primate, Human Culture[125,128,253,254]
Inhibit myosin light chain phosphorylationSrc-family tyrosine kinase (SFK) inhibitors (PP2)Alters cell adhesion, reduces cell contractility, and permeability of cell layersRabbit, Human Culture[108,255]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Strickland, R.G.; Garner, M.A.; Gross, A.K.; Girkin, C.A. Remodeling of the Lamina Cribrosa: Mechanisms and Potential Therapeutic Approaches for Glaucoma. Int. J. Mol. Sci. 2022, 23, 8068. https://doi.org/10.3390/ijms23158068

AMA Style

Strickland RG, Garner MA, Gross AK, Girkin CA. Remodeling of the Lamina Cribrosa: Mechanisms and Potential Therapeutic Approaches for Glaucoma. International Journal of Molecular Sciences. 2022; 23(15):8068. https://doi.org/10.3390/ijms23158068

Chicago/Turabian Style

Strickland, Ryan G., Mary Anne Garner, Alecia K. Gross, and Christopher A. Girkin. 2022. "Remodeling of the Lamina Cribrosa: Mechanisms and Potential Therapeutic Approaches for Glaucoma" International Journal of Molecular Sciences 23, no. 15: 8068. https://doi.org/10.3390/ijms23158068

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