Abstract
Primary open-angle glaucoma (POAG) is a leading cause of blindness worldwide, and intraocular pressure (IOP) is an important modifiable risk factor. IOP is a function of aqueous humor production and aqueous humor outflow, and it is thought that prolonged IOP elevation leads to optic nerve damage over time. Within the trabecular meshwork (TM), the eye's primary drainage system for aqueous humor, matricellular proteins generally allow cells to modulate their attachments with and alter the characteristics of their surrounding extracellular matrix (ECM). It is now well established that ECM turnover in the TM affects outflow facility, and matricellular proteins are emerging as significant players in IOP regulation. The formalized study of matricellular proteins in TM has gained increased attention. Secreted protein acidic and rich in cysteine (SPARC), myocilin, connective tissue growth factor (CTGF), and thrombospondin-1 and -2 (TSP-1 and -2) have been localized to the TM, and a growing body of evidence suggests that these matricellular proteins play an important role in IOP regulation and possibly the pathophysiology of POAG. As evidence continues to emerge, these proteins are now seen as potential therapeutic targets. Further study is warranted to assess their utility in treating glaucoma in humans.
Introduction
Patients with primary open-angle glaucoma (POAG) exhibit relatively elevated intraocular pressure (IOP), which is thought to be due to increased resistance to aqueous humor outflow. The trabecular meshwork (TM) represents the anatomic location with the highest resistance to outflow (Fig. 1).1–7 Across the conventional pathway, 3 major pathways govern the regulation of outflow resistance: (1) the transcellular route through vacuoles in the inner wall endothelial cells of Schlemm's canal8–12; (2) the paracellular route passing between Schlemm's canal inner wall endothelial cells13–15; and (3) through extracellular matrix (ECM) turnover16–22 in the juxtacanalicular region (JCT). To reach either the transcellular or paracellular pathways, aqueous humor must first flow through the ECM of the JCT region. Across the unconventional or uveoscleral pathway, outflow resistance in the ciliary body (CB) is most likely regulated by ECM turnover in the stroma23–30 and by cellular tone in the CB smooth muscle.31,32 Thus, ECM modification and turnover is crucial to regulating resistance to outflow in both the conventional and unconventional pathways. It is not yet fully understood how ECM equilibrium is regulated in outflow tissues, and there is renewed interest in the degree to which global and segmental modification of these tissues may play a role in altering outflow facility.33,34
FIG. 1.

Schematic diagram showing key ocular structures involved in aqueous humor inflow and outflow (adapted, with permission, from Tomarev 2001, Nature Med. 7, 294–295; Copyright © 2001 Nature Publishing Group). (A) Secreted by the ciliary body (CB), aqueous humor passes from the posterior chamber (PC) to the anterior chamber (AC) via the pupil. Aqueous humor outflow through the conventional outflow pathway involves the trabecular meshwork (TM) and juxtacanalicular (JCT) region, Schlemm's canal (SC), and the episcleral venous system. (B) An enlargement of the TM, JCT, and SC from (A).
Matricellular proteins are nonstructural secreted glycoproteins that aide in cellular communication with and control over surrounding ECM. This expanding family of proteins includes the prototypical SPARC (secreted protein, acidic and rich in cysteine), also known as osteonectin and BM-40, myocilin, connective tissue growth factor (CTGF), and other CCN family members, thrombospondins-1 and −2 (TSP-1 and −2), tenascins-C and–X, SC1/hevin, and osteopontin.35–38 Galectin family proteins perform similar functions, meeting several matricellular protein criteria, but they lack a high degree of structural similarity to the proteins above. Matricellular proteins are generally characterized by their importance in regulating interactions between structural proteins and signaling molecules such as cytokines and matrix proteases.
Due to the vital role that ECM turnover plays in managing outflow facility, matricellular proteins are of great interest. A growing body of evidence suggests that some matricellular proteins may serve as promising therapeutic targets. This article represents a comprehensive update on recent progress in the rapidly developing field, expanding upon an initial review of matricellular proteins in the TM from several years ago.39
Refining Our Understanding
Matricellular proteins in the TM
Secreted protein, acidic and rich in cysteine also known as osteonectin or BM-40
The prototypical matricellular protein SPARC was first studied in bone in 1981.40 Mammalian, amphibian, and avian SPARC demonstrate greater than 70% amino acid sequence homology.41 Wherever and whenever there is a significant degree of ECM turnover, SPARC is likely to be highly expressed, playing notable roles during both embryogenesis and tissue repair.42,43 In a number of conditions involving aberrant tissue remodeling—such as hepatic fibrosis,44,45 cholangitis,46 renal interstitial fibrosis,47 systemic sclerosis,48 osteoporosis,49 and certain carcinomas50—elevated SPARC expression levels have been noted. Through its interactions with integrin-linked kinase, SPARC has been shown to enhance fibronectin-mediated ECM assembly.51,52 SPARC suppression has been shown to decrease the expression of multiple collagens and tissue inhibitor of metalloproteinase (TIMP)-3.53,54 Mice harboring single gene deletions of Sparc exhibit accelerated dermal wound closure, decreased levels of dermal collagen, elevated adiposity, decreased bone mineral density, early-onset cataractogenesis, and a suppressed reaction to implanted synthetic materials.55–61 In addition, SPARC-null mice exhibit decreased laminin and collagen (types I and IV) deposition within renal tissue, decreasing the risk for damage consistent with diabetic nephropathy.62 Taken together, these studies of tissues demonstrating suppressed SPARC levels suggest that this particular matricellular protein normally acts to promote ECM deposition.
SPARC induces changes in matrix metalloproteinases (MMPs) in various human tissues and cell types. For example, in malignant human breast cancer (MDA-MB-231, BT549) and glioblastoma-astrocytoma cell lines (U87MG), SPARC increases MMP-2 activation by decreasing TIMP-2 and upregulating membrane-bound 1-MMP (MT1-MMP or MMP-14), the principle inhibitor and activator of MMP-2, respectively, presumably affecting the tumor's tissue invasiveness.63,64 In monocytes, SPARC upregulates MMP-1 and gelatinase B (MMP-9) via prostaglandin synthase-dependent signaling, thereby facilitating leukocytic movement through ECM.65
In humans, SPARC localizes to a variety of ocular tissues and its levels can act as a marker for processes such as corneal wound repair and cataractogenesis.66–68 Corneal epithelial cells, retinal pigment epithelium, and lens epithelial cells have all been demonstrated to produce SPARC.68–70 Within lens tissue, tight regulation of SPARC levels is required to maintain lens transparency given that both increased SPARC production71 and complete absence of SPARC60,72 are associated with cataractogenesis. SPARC is also produced by both TM and CB smooth muscle cells73,74 and aqueous and vitreous humor have been noted to contain significant SPARC levels.68 Immunofluorescence studies of ex vivo human anterior segment tissues reveal that SPARC is distributed throughout the TM and JCT region. SPARC is one of the most highly expressed genes in primary cultured human TM cells, and its levels are elevated in response to mechanical stretch, suggesting that SPARC plays an important role in normal TM function.75,76
Interest in SPARC as a regulator of ECM homeostasis in the TM continues to grow. Our laboratory has shown that SPARC-null mice exhibit 15%–20% lower IOP compared with their wild-type counterparts77,78 and that these mice demonstrate a more uniform pattern of aqueous humor outflow (Fig. 2).34 SPARC overexpression in human TM cells increases IOP in an ex vivo perfusion chamber model, likely by increasing the production of various ECM components (Fig. 3).79 Upstream regulation of SPARC is complex and may involve a combination of microRNA (miRNA) elements,80,81 pro-fibrotic cytokines,82,83 isoprenoid precursors,84 AMP-activated protein kinase (AMPK),85 and even dynamic responses to TM stiffness.86 transforming growth factor (TGF)-β2 cytokine levels are significantly elevated in eyes with POAG,87,88 and treatment with activated TGF-β2 upregulates SPARC in cultured human TM cells (Fig. 4).83 Of note, preliminary evidence suggests that the TGF-β2-mediated ocular hypertension observed in wild-type mice is not as pronounced in their SPARC-null counterparts.89 TM cells cultured on synthetic hydrogels mimicking the stiffness found in glaucomatous meshwork show significantly greater SPARC and myocilin mRNA expression than those cultured on hydrogels that approximate the relatively reduced stiffness of normal meshwork (Fig. 5).86
FIG. 2.
(A, B) Representative pair of matched WT and SPARC-null (KO) mouse eyes (adapted, with permission, from Swaminathan et al.,34 Copyright © 2013 Association for Research in Vision and Ophthalmology). Fluorescence microscopy was complete en face through the corneal side. Tracer bead distribution in the TM of wild-type (WT) eyes was heterogeneous, with large sections having little staining. Greater fluorescence in a more homogeneous pattern was seen in SPARC KO eyes, reflecting more uniform outflow. Gray portions of these images are from the projection of the brightfield lamp on the tissue and medium. Central portions of these images were removed due to iris autofluorescence. (C, D) Representative confocal microscopy of frontal sections of WT and KO high-tracer sections, respectively. Sclera, TM, SC, CB, collector channel (CC), and episcleral vein (EV) are labeled. Tracer is strongly present throughout the TM. Tracer can also be noted within SC. (E) En face image of a high-tracer section demonstrating microbeads within the TM and along the walls of EVs. (F) Percentage effective filtration length (PEFL) values in WT and KO eyes with SEM error bars. KO PEFL was significantly higher (asterisk) than WT PEFL (n=11 pairs, P<0.005).
FIG. 3.
Representative immunolabeling of ECM proteins after infection with adenovirus overexpressing human SPARC versus vector only control (adapted, with permission, from Oh et al.,79 Copyright © 2013 Association for Research in Vision and Ophthalmology). Collagen I (A), collagen IV (B), and fibronectin (D) were increased, but collagen VI (C) and laminin (E) were not changed. Collagen I was increased throughout the TM with some increase in the JCT region. Collagen IV was detected more prominently in the JCT region and under the outer wall of SC. Fibronectin was increased throughout the TM, but prominently within the JCT region. The secondary staining (F) was shown as a negative control. Scale bar: 30 μm; C, control, S+, SPARC+; magnification=× 100.
FIG. 4.
Analysis of SPARC levels following TGF-β2 treatment (2 ng/mL) in cultured TM cells, n=6 (adapted, with permission, from Kang et al.,83 Copyright © 2013 Association for Research in Vision and Ophthalmology). SPARC mRNA was analyzed by qPCR and normalized with β-actin. SPARC protein levels in conditioned media were analyzed by immunoblotting and calculation of integrated band intensity. Representative immunoblot shown.
FIG. 5.

(A) Substratum stiffness impacts SPARC mRNA and cellular response to treatment with the actin cytoskeletal disruptor Latrunculin-B (Lat-B) in human TM cells (adapted, with permission, from Thomasy et al.,86 Copyright © 2012 Association for Research in Vision and Ophthalmology). Cells grown on stiff hydrogels and treated with DMSO had significantly different SPARC mRNA expression in comparison with DMSO-treated cells on tissue culture polystyrene (TCP; >1 GPa) or more compliant hydrogels. Cells grown on hydrogels and treated with Lat-B had significantly less SPARC mRNA expression versus treatment with DMSO. In contrast, cells grown on TCP and treated with Lat-B had significantly greater SPARC mRNA expression versus treatment with DMSO. Data are mean±SEM (*P<0.05 for hydrogel versus TCP; a,b,cP<0.05 between the different hydrogels; †P<0.05 for DMSO versus Lat-B). (B) Substratum stiffness modulates expression of SPARC protein and the impact of Lat-B treatment on SPARC protein expression. TM cells on 75 kPa hydrogels (mimicking the stiffness of glaucomatous TM) have markedly greater SPARC expression than on the much stiffer (>1 GPa) substrates. Treatment with 2 μM Lat-B decreases SPARC protein expression on the 75 kPa hydrogels. Exposure time for each image was 3.0 s. Scale bar, 50 μm.
Myocilin
Myocilin—a secreted protein found in human TM and thought to be involved in ECM regulation90—has demonstrated de-adhesive properties that have earned it classification as a matricellular protein.91,92 The gene that encodes myocilin was one of the first to be linked to POAG,93 and myocilin's C-terminal domain interacts with that of SPARC.94 Previous studies have suggested that most disease-causing myocilin gene mutations interfere with myocilin protein production93 or proper folding and secretion,95–97 giving rise to an “intracellular sequestration” model of disease pathogenesis. However, one report found that approximately twenty percent of disease-causing mutants yielded protein products that were still successfully secreted and recovered from culture media.97 Furthermore, half of those involved mutations in the olfactomedin-like domain near the C-terminal region, which harbors a demonstrated interaction site for SPARC protein.94 It is therefore reasonable to theorize that altered ECM in the JCT as a result of mutated myocilin may in some cases derive from abnormal interactions with SPARC or other matricellular proteins and may partially explain the pathophysiology of increased resistance to aqueous humor outflow. Taken together, these studies further strengthen the notion that SPARC, like myocilin, may be a key regulatory node that governs ECM homeostasis in the TM and IOP.
Connective tissue growth factor
The CCN family of 6 multifunctional matricellular proteins derives its name from 3 prototypical members named in order of discovery (Cyr61 or CCN1, CTGF or CCN2, Nov or CCN3), and has been found to be involved in a variety of processes such as angiogenesis, adhesion, migration, tissue repair, and fibrosis.98–100 A unifying feature of this family is the presence of 4 discrete functional domains, including (1) an insulin-like growth factor binding protein-like module (IGFBP), (2) a von Willebrand factor type C repeat module (VWC), (3) a TSP type 1 repeat, and (4) a cysteine-knot-containing module. One of the first 2 family members to be discovered,101 CTGF (also known as CCN2) is by far the most studied and the most well understood.99
Knockout mutations of the gene that encodes CTGF are fatal in mice, and initial work revealed that the protein is crucial for bone and soft tissue development.102 Studies in human dermal fibroblasts revealed that CTGF may potentiate a cell's ability to spread on fibronectin, a process required for adhesion during tissue development and wound healing.103 Interestingly, CTGF was also found to play a critical role in regulating fibrosis through its interactions with TGF-β signaling.104 Given its profibrotic tendencies, it became a protein of interest in surgical models, demonstrating increased expression following glaucoma filtration surgery in rabbits and being associated with an elevated risk of bleb failure.105,106
The putative role of CTGF in human glaucoma pathophysiology was first identified in pseudoexfoliation syndrome,107,108 and attention soon focused on how CTGF regulates TM matrix deposition,109,110 cellular tone,111 and aqueous outflow facility.112,113 In one promising recent study, the prostaglandin analog fluprostenol was found to mitigate CTGF-induced fibrogenesis in cultured human TM cells.114 Pharmacological targeting of CTGF therefore shows great therapeutic potential and remains an active area of research. Further work elucidating the potential role of other CCN family members is required.
Thrombospondins
First reported as a thrombin-sensitive protein, TSP—also known as glycoprotein G or GPG—was first described in 1971.115 Subsequent investigation has led to the adoption of nomenclature that identifies this protein as TSP-1,116,117 one member of a family of proteins that are now classified into subgroups A (TSP-1 and TSP-2) and B (TSP-3, TSP-4, and TSP-5).118–120 The C-terminal regions of TSPs demonstrate shared calcium-, ECM-, and cell membrane-binding properties. Subgroup A TSPs, which all contain an antiangiogenic domain at the N-terminal region, have been found in both nonocular and ocular tissues.119,121 Subgroup B TSPs, which do not clearly belong to the matricellular family, lack antiangiogenic regions and have variable N-terminal domains.119 Although they are not well characterized, it is believed that TSP-3 may be involved in bone growth plate maintenance122; TSP-4 may be involved in stretch-mediated contractility enhancement within cardiac muscle tissue123 and spinal sensitization in models of neuropathic pain.124,125 TSP-5, also known as cartilage oligomeric matrix protein, is found in cartilaginous tissue and is extensively reviewed elsewhere.126–128
As they more clearly belong to the matricellular family, TSP-1 and TSP-2 are reviewed here. In vitro studies show that these TSPs, like SPARC, regulate cellular tone, shape, adhesion, proliferation, and apoptosis.129,130 Subgroup A TSPs also play important roles in tissue formation and remodeling.38,129,131 For example, both TSP-1 and TSP-2 have been shown to interact with MMPs, with TSP-1 inhibiting MMP-3-dependent activation of pro-MMP-9 and thrombin-induced activation of pro-MMP-2,132 and TSP-2 forming a complex with and internalizing MMP-2.133 Under physiologic conditions, TSP-1 is also capable of binding and activating the pro-fibrotic cytokine TGF-β1,134,135 in addition to binding ECM components such as collagen types I-V, laminin, and fibronectin.136–139 Its role both in physiologic and aberrant tissue repair and fibrosis is reviewed elsewhere.140 TGF-β1 treatment has been shown to upregulate TSP-1 expression, suggesting the possibility of a reciprocal relationship.141
Mice harboring single gene deletions of either Thbs1 or Thbs2 have yielded information regarding the 2 protein's functional similarities and differences. TSP-1-null mice demonstrate phenotypes that are generally consistent with states of decreased TGF-β1 activity,142,143 with TSP-1-null and TGF-β1-null mice exhibiting histopathological similarities in at least 9 organ systems.142 For example, the 2 strains both suffer from reduced dermal matrix, hyperplasia of various epithelial cells, and pulmonary inflammation. Further strengthening the link, cultured TSP-1-null cell isolates exhibit one eighth the amount of active TGF-β1 of their wild-type counterparts, and actively replacing TSP-1 metabolites restores TGF-β1 levels and bronchial epithelial histology in the TSP-1-null cultured cells and mice, respectively.142 Despite the TSP subgroup A sequence homology, TSP-2-null mice demonstrate phenotypic characteristics that are distinct from their TSP-1-null counterparts. The former demonstrate frail skin, tendon laxity, disordered collagen fibrillogenesis, accelerated wound healing, increased bone density, increased vascular density, and a distinct bleeding diathesis.119,144,145 TSP-1 and TSP-2 also differ in their temporal and spatial expression in knockout mouse models of wound healing.146 Interestingly, one member of the related ADAMTS (a disintegrin and metalloproteinase with TSP type 1 motifs) family may play a role in the pathogenesis of ectopia lentis,147,148 while another appears to regulate outflow facility in ex vivo perfused human anterior segments.149
Subgroup A TSPs are expressed throughout a variety of ocular tissues and appear to play a role in several pathologies involving angiogenesis including diabetic retinopathy, age-related macular degeneration, corneal wound repair, pseudoexfoliation syndrome, and uveal melanoma.121,150,151 TSP-1 and TSP-2 are both expressed in human TM,75,121 but TSP-1 localizes to the TM and JCT regions, whereas TSP-2 predominates in the uveal meshwork.121 In one study, TSP-1 exhibited more intense immunoreactivity in one-third of patients with POAG,152 making it of particular interest. Of note, both TGF-β1 and TGF-β2 upregulate TSP-1 in human TM cells,152–154 and treatment with TGF-β2 treatment generally favors ECM accumulation and an increase in resistance to aqueous outflow.153,155–158 Furthermore, TSP-1—like SPARC—may play a role in TM cellular tone, as it has been shown to be antagonized by the actin cytoskeletal disruptor latrunculin A.
We have begun preliminary investigations into the ocular phenotypes of TSP-1-null and TSP-2-null mice, given the demonstrated importance of the subgroup A TSPs in matrix remodeling throughout the body and, in particular, within the TM. The average IOPs of TSP-1-null and TSP-2-null mice are approximately 10% and 7% lower than that of their corresponding wild-type mice, respectively, while maintaining similar central corneal thickness (CCT) and iridocorneal angle architecture.159 These findings were confirmed in a study summarizing IOP changes in all matricellular null strains, which also demonstrated that CCT need not be measured to evaluate IOP differences in these strains (Table 1).78 Interestingly, aqueous humor turnover rates—as measured by fluorophotometric techniques—are enhanced in both TSP-1- and TSP-2-null mice, suggesting that both may play roles in regulating aqueous outflow facility.
Table 1.
Average Mouse IOP±SEM of Matricellular Knockout Strains and Wild-Type Counterparts
| Strain | WT (n) | KO (n) | % Difference | P-value |
|---|---|---|---|---|
| SPARC | 19.9±0.25 (55) | 16.9±0.24 (59) | −15.08a | <0.0001 |
| TSP-1 | 15.7±0.17 (72) | 14.4±0.25 (41) | −8.28a | <0.0001 |
| TSP-2 | 18.1±0.20 (60) | 16.9±0.36 (32) | −6.63a | <0.0001 |
| Tenascin-C | 17.8±0.25 (60) | 18.1±0.19 (60) | +1.69 | 0.290 |
| Tenascin-X | 17.8±0.23 (60) | 17.5±0.17 (74) | −1.69 | 0.201 |
| Hevin | 16.7±0.29 (59) | 16.3±0.22 (76) | −2.40 | 0.26 |
| Osteopontin | 17.3±0.21 (81) | 17.5±0.29 (49) | +1.16 | 0.57 |
Tenascin-C and tenascin-X
The first in its family to be characterized, Tenascin-C was initially described in the stroma of gliomas and in tendons.160,161 Since then, the vertebrate tenascin family has grown to include 3 additional members. Each of them is comprised of linked subunits that each contain EGF-like and heptad repeats, fibronectin type III domains, and a C-terminal globular domain shared with the fibrinogen family.36,162 Because tenascin-R has thus far only been described in the central nervous system163 and tenascin-W has yet to be well characterized,164 tenascin-C and tenascin-X are the only 2 family members currently classified as matricellular proteins.38,130 Generally, tenascins are involved in tissue remodeling and are highly expressed during development and processes involving injury and wound repair. Although they both demonstrate cellular adhesion-modulating properties and the ability to affect cellular tone, migration, and growth, tenascin-C and tenascin-X exhibit significant differences in their organ distribution and overall function.
Tenascin-C is primarily expressed in tendons, ligaments, cartilage, and the ECM of tumor stroma and blood vessels.160,161,165–167 Like its fellow matricellular proteins SPARC and TSP-1, tenascin-C regulates cellular adhesion and migration in a tissue-dependent manner.168 One mechanism by which this occurs may include interference with integrin-mediated cellular interaction with fibronectin.169,170 At sites of inflammation, tenascin-C is found to be expressed relatively early,171,172 and it appears to be upregulated by TGF-β2 and other anti-inflammatory cytokines173–176 Enhanced expression of tenascin-C has also been observed in response to mechanical stretch,177,178 for example, in models of cardiomyocyte damage and regeneration.179,180 Mice deficient in tenascin-C have abnormal neurochemistry and demonstrate abnormal dermal and corneal would healing with reduced fibronectin.181–184 These phenotypes presumably result from aberrant cell migration and adhesion and disruption of ECM formation.
In healthy mammalian adults, tenascin-C is expressed at the limbus, at the scleromuscular junctions, and in the JCT region of the TM.185–187 It has also been shown to be expressed during normal corneal development.188 In disease states, the protein is found in the vitreous of patients with vitreoretinopathy and proliferative diabetic retinopathy189,190—likely contributing to disease progression191—and in the optic nerve head of rat glaucoma models.192 Within the TM, tenascin-C is specifically present in sheaths and sheath-derived plaque cores186 and co-localizes with fibronectin in vitro.193 Mechanical stress of human TM leads to upregulation and alternative splicing of tenascin-C.76,194 Initial evidence from cultured human TM cells also suggested that tenascin-C may contribute to Rho GTPase-mediated resistance to aqueous humor drainage.195 One recent study in perfused ex vivo human and porcine anterior chambers has confirmed its predominance in the JCT region and revealed a potential role in the physiologic maintenance and remodeling of TM but found that its RNA interference-based suppression did not affect outflow facility significantly.196 Furthermore, tenascin-C-null mice exhibit IOPs and CCTs similar to their wild-type counterparts, supporting the notion that the protein—while likely to be involved in dynamic TM maintenance—may not significantly contribute to IOP regulation.196
Tenascin-X is generally found in the connective tissue of cardiac, skeletal muscle, and skin, and its distribution is often distinct from that of tenascin-C.197 Patients harboring mutations and deletions of tenascin-X exhibit phenotypic similarities with those who have Ehler–Danlos syndrome, including hyperelastic skin, increased joint and vascular laxity, and abnormal wound healing.198–201 Tenascin-X is believed to modulate cellular adhesion and collagen stiffness.202,203 Mice deficient in tenascin-X, like their human counterparts, exhibit skin hyperextensibility and fragility, with histologically normal-appearing collagen fibrils that are less densely packed.204 This mouse strain is now recognized as a suitable model for Ehlers–Danlos syndrome.205 Dermal fibroblasts lacking the gene that encodes tenascin-X demonstrate normal levels of collagen production in vitro but poor collagen deposition in the cell-associated matrix. Although it is not completely understood how tenascin-X mediates ECM deposition, it is thought that its binding to decorin may be crucial, since deficiency of this proteoglycan is also associated with Ehlers–Danlos-like phenotypes.162,206
In ocular tissues, tenascin-X is expressed both during development and in the normal adult cornea, corneoscleral junction, and limbal and conjunctival blood vessels.207 Of note, the region encompassing its gene has been implicated in one recent genome-wide association study of age-related macular degeneration.208 Tenascin-X-null mice are now well characterized,186,209–211 and similar to tenascin-C-null mice, they exhibit IOPs and CCTs comparable to their wild-type counterparts, making it less likely that deficiency of this protein significantly contributes to IOP regulation.196 For this reason, SPARC, TSP-1, and TSP-2 appear to be more promising with regard to potential therapeutic intervention.
Hevin
Hevin was originally identified from among other synaptic junction glycoproteins in the rat brain and was found to share a high degree of homology with SPARC.212 Like SPARC, hevin demonstrates anti-adhesive properties,213 which may potentially be mediated by antagonizing fibronectin-mediated adhesion.214 Furthermore, it has been shown to modulate decorin and collagen fibrillogenesis and to bind collagen I.215,216 Hevin has been shown to help mediate the progression of foreign body-induced inflammation while SPARC directs the formation of a collagenous capsule.51,52 This suggests that hevin may have similar pro-fibrotic functions as its matricellular family member SPARC.
Hevin has been found to be expressed in murine ocular tissue.217 It was originally perceived as a potential key player in regulation of ECM turnover within the TM and JCT, in part due to its notable C-terminal interaction with myocilin,218 something it shares in common with SPARC.94 In human 293-T cells, Li et al. showed that hevin interacts with myocilin and that myocilin causes accumulation of hevin.218 Of note, patients with myocilin-associated POAG—which demonstrates an over 90% penetrance in heterozygous individuals—generally present with high IOP.219 Since our original review of matricellular proteins, we have demonstrated that both human and murine TM tissue contain detectable levels of hevin protein.220 Hevin-null mice exhibited similar IOPs and CCTs to their wild-type counterparts, with similar aqueous humor turnover rates. Furthermore, in cultured human TM cells, TGF-β2 did not induce hevin; this is in contrast to SPARC, which exhibits a reproducible dose-dependent induction.220 Taken together, these results diminish the likelihood that hevin plays a critical role in glaucoma pathophysiology by altering resistance to aqueous outflow.
Osteopontin
Originally called sialoprotein,221 osteopontin owes its present name to its potential role as a “bridge” (from the French, pont) between cells and hydroxyapatite.221,222 Osteopontin is now widely considered a member of the matricellular protein family, owning to its observed role in regulating ECM turnover in various tissues.38,130 It still, however, has a structural role in bone,223 as evidenced by its various binding sites for integrin, hydroxyapatite, and calcium.224,225 In addition to these active domains, it harbors bindings sites for heparin, putative thrombin cleavage sites, and an N-terminal region that is both chemotactic for neutrophils and associated with carotid plaque formation.224–227 Osteopontin's diverse effects—including during development, bone resorption and calcification, wound healing, mucosal protection, and immunological responses, all of which are reviewed elsewhere223,228—have been conceptually grouped into 2 broad functions: regulation of acute and chronic inflammation, and regulation of macrophage-derived cells such as osteoclasts.229 These putative functions are more or less confirmed by phenotypic traits found in osteopontin-deficient mice, including reduced bone resorption, increased dystrophic calcification, altered sympathetic control of bone mass, impaired periprosthetic osteolysis, abnormal matrix reorganization following tissue injury, and reduced T-cell-mediated immunity.230–235
Within the eye, osteopontin is involved in corneal and choroidal wound healing, lens epithelial injury, autoimmune uveitis, and proliferative diabetic retinopathy.236–242 Using cDNA microarrays, TM cells have been shown to express osteopontin.243 Given that TM cells have phagocytic activity that may alter outflow facility, osteopontin may be involved in this process. Interestingly, the relative concentrations of osteopontin in the aqueous humor of patients with pseudoexfoliation syndrome is lower than those found in control patients.244 Additionally, osteopontin exhibits an age-dependent increase in aqueous humor and abnormal levels are a risk factor associated with degeneration of the optic nerve in D2J mice.245 We found that osteopontin is present in human TM and exhibits lower concentrations in the aqueous humor of POAG patients.246 There was, however, no significant IOP difference between osteopontin-null mice and their wild-type counterparts. Further investigation is warranted to investigate whether manipulation of osteopontin levels affect outflow facility.
Lectins and galectins
Lectins represent a family of proteins—each containing a subtype-specific carbohydrate recognition domain (CRD)—that are neither catalytic enzymes nor immunoglobulins.247,248 Lectins that specifically bind β-galactosides are now termed galectins, having previously been known as S-type lectins. Galectins are thought to be involved with cell adhesion, migration, and survival.249,250 Thus far, 15 galectins have been identified and are subdivided according to structural characteristics: (1) Monovalent (prototypic) galectins contain a single CRD; (2) bivalent galectins contain tandem-repeats; and (3) chimeric galectins contain a single CRD and a unique N-terminal region rich in proline and glycine. Galectins, which are nonstructural components of the ECM that direct cell-to-cell and cell-to-matrix interactions, are now widely accepted as matricellular proteins.249–251 The viability, fertility, and relatively normal appearance of mice with genetic deletions of single galectins suggests that there is a certain degree of redundancy within the family.252,253
To date, galectin-1 and galectin-3 are the most well described in humans, although progress is being made on all fronts.254 Galectin-1 is the prototypical galectin and is capable of binding numerous carbohydrate residues255,256 and fibronectin and laminin,257,258 mediating both intra- and intermolecular cross-linking. TGF-β2 increases galectin-1 expression in endothelial cells,255,256 and the effects of galectin-1 on cell adhesion are dose-dependent259 while its effects on cell growth are biphasic.260,261 The only chimeric galectin identified thus far,262 galectin-3 binds to a variety of ECM components such as fibronectin, laminin, and collagen IV.263,264 Of note, galectin-3 has also been shown to be either a substrate or an upstream regulator for MMP-1, MMP-2, and MMP-9. These MMPs are important regulators of ECM turnover during pathologic tissue remodeling events, such as those that occur during modulation of the tumor microenvironment or in response to cardiomyocyte damage in heart failure patients.265–270 With regard to cellular proliferation in neoplastic and autoimmune disease, galectin-1 and galectin-3 often promote antagonistic agendas, with galectin-1 mediating apoptosis and galectin-3 mediating cell survival.250,271,272
Galectin family proteins have been implicated in a variety of ocular disorders, including inflammatory conditions of the conjunctivita and cornea, proliferative vitreoretinopathy, and diabetic retinopathy.273–278 Numerous studies suggest that galectins are important in the maintenance of a healthy ocular surface at rest and after injury,279,280 with galectins-1 and −9 specifically playing roles in the response to bacterial and viral pathogens that damage the corneal surface.281–283 The healthy human lens284 also expresses galectin-3, and a galectin-like molecule called GRIFIN (galectin-related inter-fiber protein).285,286 In depth proteomic analysis of normal human CB tissue—which participates in aqueous humor production and aqueous humor outflow through the unconventional pathway—has revealed expression of galectin-1.287 Additionally, there is now evidence that suggests that galectin-3 may play a role in neuroprotection against glaucomatous damage.288,289
Galectins—specifically galectins-1, -3, and -8—may be involved in the TM.290–292 Fautsch et al. have confirmed the presence of galectins-1 and -3 RNA and protein using PCR, western blot analysis, and immunohistochemistry.290 Both galectins were observed to stain throughout the TM, including the uveal, limbal, and JCT regions, and the inner and outer walls of Schlemm's canal. Of note, the authors found no visible difference in the galectin-1 or galectin-3 labeling distributions of glaucomatous versus normal TM tissues. Diskin et al. found that TM cells express and adhere to galectin-8-coated wells but do not adhere to galectin-1 or −3-coated wells, which was supported by their discovery that TM cell β1-integrins carry predominantly α2-3-sialylated glycans, which exhibit high affinity for galectin-8 specifically. These studies provided the foundation for a recent investigation that revealed that galectin-8 promotes cytoskeletal rearrangement in TM cells through activation of Rho signaling, which presents the intriguing possibility that galectin-8 may indeed participate in the regulation of aqueous outflow facility.292 Mice with deletions of the genes that encode galectin-1 and -3 have been thoroughly characterized since being engineered over 2 decades ago,252,253,293 but galectin-8-null mice have not been widely studied or described. Future studies aimed at assessing outflow facility and IOP in galectin-null mice may provide insight into the functional importance of galectin family proteins. Of the matricellular proteins reviewed in this article (Table 1),78 galectins present the greatest number of unanswered questions in this regard.
Conclusions
Matricellular proteins represent an intriguing family of nonstructural proteins that affect ECM turnover within the TM, just as they do in nonocular tissues. Some of these proteins antagonize fibronectin binding, thereby interfering with cellular adhesion. All of them share a certain pro-fibrotic theme, interacting with cytokines such as TGF-β2, whose link to glaucoma pathogenesis is now widely accepted. Over the past 5 years, significant strides have been made in our understanding of the role of these matricellular proteins in the promotion of ECM deposition and regulation of outflow facility within the TM. With recent studies into SPARC, myocilin, CTGF, TSP-1, TSP-2, tenascin-C, tenascin-X, hevin, osteopontin, and galectins, it is clear that several of these proteins show promise as possible therapeutic targets for glaucoma. SPARC, myocilin, CTGF, TSP-1, TSP-2, and galectins show the greatest promise thus far and warrant further study.
Acknowledgments
Howard Hughes Medical Institute Research Training Fellowship for Medical Students (A.C.), National Eye Institute EY 019654-01 (D.J.R.) and EY 014104 (MEEI Vision-Core Grant).
Author Disclosure Statement
No competing financial interests exist.
References
- 1.Bill A.Editorial: the drainage of aqueous humor. Invest. Ophthalmol. 14:1–3, 1975 [PubMed] [Google Scholar]
- 2.Bill A., and Phillips C.I.Uveoscleral drainage of aqueous humour in human eyes. Exp. Eye Res. 12:275–281, 1971 [DOI] [PubMed] [Google Scholar]
- 3.Grant W.M.Further studies on facility of flow through the trabecular meshwork. AMA Arch. Ophthalmol. 60:523–533, 1958 [DOI] [PubMed] [Google Scholar]
- 4.Maepea O., and Bill A.The pressures in the episcleral veins, schlemm's canal and the trabecular meshwork in monkeys: effects of changes in intraocular pressure. Exp. Eye Res. 49:645–663, 1989 [DOI] [PubMed] [Google Scholar]
- 5.Moses R.A., Grodzki W.J., Jr., Etheridge E.L., and Wilson C.D.Schlemm's canal: the effect of intraocular pressure. Invest. Ophthalmol. Vis. Sci. 20:61–68, 1981 [PubMed] [Google Scholar]
- 6.Pederson J.E., Gaasterland D.E., and MacLellan H.M.Uveoscleral aqueous outflow in the rhesus monkey: importance of uveal reabsorption. Invest. Ophthalmol. Vis. Sci. 16:1008–1017, 1977 [PubMed] [Google Scholar]
- 7.Seiler T., and Wollensak J.The resistance of the trabecular meshwork to aqueous humor outflow. Graefes Arch. Clin. Exp. Ophthalmol. 223:88–91, 1985 [DOI] [PubMed] [Google Scholar]
- 8.Holmberg A.The fine structure of the inner wall of schlemm's canal. Arch. Ophthalmol. 62:956–958, 1959 [Google Scholar]
- 9.Holmberg A.Schlemm's canal and the trabecular meshwork: an electron microscopic study of the normal structure in man and monkey (cercopithecus aethiops). Doc. Ophthalmol. 19:339–373, 1965 [Google Scholar]
- 10.Inomata H., Bill A., and Smelser G.K.Aqueous humor pathways through the trabecular meshwork and into schlemm's canal in the cynomolgus monkey (macaca irus). An electron microscopic study. Am. J. Ophthalmol. 73:760–789, 1972 [DOI] [PubMed] [Google Scholar]
- 11.Lee W.R., Grierson I., and McMenamin P.G.The morphological response of the primate outflow system to changes in pressure and flow. In: Lutjen-Drecholl E., ed. Basic Aspects of Glaucoma Research. Stuttgart: Schattauer Verlag; 1982 [Google Scholar]
- 12.Tripathi R.C.Mechanism of the aqueous outflow across the trabecular wall of schlemm's canal. Exp. Eye Res. 11:116–121, 1971 [DOI] [PubMed] [Google Scholar]
- 13.Epstein D.L., Freddo T.F., Bassett-Chu S., Chung M., and Karageuzian L.Influence of ethacrynic acid on outflow facility in the monkey and calf eye. Invest. Ophthalmol. Vis. Sci. 28:2067–2075, 1987 [PubMed] [Google Scholar]
- 14.Epstein D.L., and Rohen J.W.Morphology of the trabecular meshwork and inner-wall endothelium after cationized ferritin perfusion in the monkey eye. Invest. Ophthalmol. Vis. Sci. 32:160–171, 1991 [PubMed] [Google Scholar]
- 15.Erickson-Lamy K., Schroeder A., and Epstein D.L.Ethacrynic acid induces reversible shape and cytoskeletal changes in cultured cells. Invest. Ophthalmol. Vis. Sci. 33:2631–2640, 1992 [PubMed] [Google Scholar]
- 16.Barany E.H., and Scotchbrook S.Influence of testicular hyaluronidase on the resistance to flow through the angle of the anterior chamber. Acta Physiol. Scand. 30:240–248, 1954 [DOI] [PubMed] [Google Scholar]
- 17.Barany E.H., and Woodin A.M.Hyaluronic acid and hyaluronidase in the aqueous humour and the angle of the anterior chamber. Acta Physiol. Scand. 33:257–290, 1955 [DOI] [PubMed] [Google Scholar]
- 18.Bradley J.M., Vranka J., Colvis C.M., et al. Effect of matrix metalloproteinases activity on outflow in perfused human organ culture. Invest. Ophthalmol. Vis. Sci. 39:2649–2658, 1998 [PubMed] [Google Scholar]
- 19.Ethier C.R., Kamm R.D., Palaszewski B.A., Johnson M.C., and Richardson T.M.Calculations of flow resistance in the juxtacanalicular meshwork. Invest. Ophthalmol. Vis. Sci. 27:1741–1750, 1986 [PubMed] [Google Scholar]
- 20.Gum G.G., Samuelson D.A., and Gelatt K.N.Effect of hyaluronidase on aqueous outflow resistance in normotensive and glaucomatous eyes of dogs. Am. J. Vet. Res. 53:767–770, 1992 [PubMed] [Google Scholar]
- 21.Knepper P.A., Farbman A.I., and Telser A.G.Exogenous hyaluronidases and degradation of hyaluronic acid in the rabbit eye. Invest. Ophthalmol. Vis. Sci. 25:286–293, 1984 [PubMed] [Google Scholar]
- 22.Pang I.H., Fleenor D.L., Hellberg P.E., Stropki K., McCartney M.D., and Clark A.F.Aqueous outflow-enhancing effect of tert-butylhydroquinone: involvement of AP-1 activation and MMP-3 expression. Invest. Ophthalmol. Vis. Sci. 44:3502–3510, 2003 [DOI] [PubMed] [Google Scholar]
- 23.Anthony T.L., Lindsey J.D., and Weinreb R.N.Latanoprost's effects on TIMP-1 and TIMP-2 expression in human ciliary muscle cells. Invest. Ophthalmol. Vis. Sci. 43:3705–3711, 2002 [PubMed] [Google Scholar]
- 24.el-Shabrawi Y., Eckhardt M., Berghold A., et al. Synthesis pattern of matrix metalloproteinases (MMPs) and inhibitors (TIMPs) in human explant organ cultures after treatment with latanoprost and dexamethasone. Eye (Lond) 14:375–383, 2000 [DOI] [PubMed] [Google Scholar]
- 25.Lindsey J.D., Kashiwagi K., Boyle D., Kashiwagi F., Firestein G.S., and Weinreb R.N.Prostaglandins increase proMMP-1 and proMMP-3 secretion by human ciliary smooth muscle cells. Curr. Eye Res. 15:869–875, 1996 [DOI] [PubMed] [Google Scholar]
- 26.Lutjen-Drecoll E., and Tamm E.Morphological study of the anterior segment of cynomolgus monkey eyes following treatment with prostaglandin F2 alpha. Exp. Eye Res. 47:761–769, 1988 [DOI] [PubMed] [Google Scholar]
- 27.Lutjen-Drecoll E., and Tamm E.The effects of ocular hypotensive doses of PGF2 alpha-isopropylester on anterior segment morphology. Prog. Clin. Biol. Res. 312:437–446, 1989 [PubMed] [Google Scholar]
- 28.Ocklind A.Effect of latanoprost on the extracellular matrix of the ciliary muscle. A study on cultured cells and tissue sections. Exp. Eye Res. 67:179–191, 1998 [DOI] [PubMed] [Google Scholar]
- 29.Weinreb R.N., Kashiwagi K., Kashiwagi F., Tsukahara S., and Lindsey J.D.Prostaglandins increase matrix metalloproteinase release from human ciliary smooth muscle cells. Invest. Ophthalmol. Vis. Sci. 38:2772–2780, 1997 [PubMed] [Google Scholar]
- 30.Weinreb R.N., and Lindsey J.D.Metalloproteinase gene transcription in human ciliary muscle cells with latanoprost. Invest. Ophthalmol. Vis. Sci. 43:716–722, 2002 [PubMed] [Google Scholar]
- 31.Toris C.B., Zhan G.L., Zhao J., Camras C.B., and Yablonski M.E.Potential mechanism for the additivity of pilocarpine and latanoprost. Am. J. Ophthalmol. 131:722–728, 2001 [DOI] [PubMed] [Google Scholar]
- 32.Nilsson S.F., Drecoll E., Lutjen-Drecoll E., et al. The prostanoid EP2 receptor agonist butaprost increases uveoscleral outflow in the cynomolgus monkey. Invest. Ophthalmol. Vis. Sci. 47:4042–4049, 2006 [DOI] [PubMed] [Google Scholar]
- 33.Keller K.E., Bradley J.M., Vranka J.A., and Acott T.S.Segmental versican expression in the trabecular meshwork and involvement in outflow facility. Invest. Ophthalmol. Vis. Sci. 52:5049–5057, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Swaminathan S.S., Oh D.J., Kang M.H., et al. Secreted protein acidic and rich in cysteine (SPARC)-null mice exhibit more uniform outflow. Invest. Ophthalmol. Vis. Sci. 54:2035–2047, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sage E.H., and Bornstein P.Extracellular proteins that modulate cell-matrix interactions. SPARC, tenascin, and thrombospondin. J. Biol. Chem. 266:14831–14834, 1991 [PubMed] [Google Scholar]
- 36.Chiquet-Ehrismann R.Tenascins, a growing family of extracellular matrix proteins. Experientia 51:853–862, 1995 [DOI] [PubMed] [Google Scholar]
- 37.Brekken R.A., Sullivan M.M., Workman G., et al. Expression and characterization of murine hevin (SC1), a member of the SPARC family of matricellular proteins. J. Histochem. Cytochem. 52:735–748, 2004 [DOI] [PubMed] [Google Scholar]
- 38.Bornstein P., and Sage E.H.Matricellular proteins: extracellular modulators of cell function. Curr. Opin. Cell Biol. 14:608–616, 2002 [DOI] [PubMed] [Google Scholar]
- 39.Rhee D.J., Haddadin R.I., Kang M.H., and Oh D.J.Matricellular proteins in the trabecular meshwork. Exp. Eye Res. 88:694–703, 2009 [DOI] [PubMed] [Google Scholar]
- 40.Termine J.D., Kleinman H.K., Whitson S.W., Conn K.M., McGarvey M.L., and Martin G.R.Osteonectin, a bone-specific protein linking mineral to collagen. Cell 26:99–105, 1981 [DOI] [PubMed] [Google Scholar]
- 41.Yan Q., and Sage E.H.SPARC, a matricellular glycoprotein with important biological functions. J. Histochem. Cytochem. 47:1495–1506, 1999 [DOI] [PubMed] [Google Scholar]
- 42.Brekken R.A., and Sage E.H.SPARC, a matricellular protein: at the crossroads of cell-matrix communication. Matrix Biol. 19:816–827, 2001 [DOI] [PubMed] [Google Scholar]
- 43.Sage H., Vernon R.B., Decker J., Funk S., and Iruela-Arispe M.L.Distribution of the calcium-binding protein SPARC in tissues of embryonic and adult mice. J. Histochem. Cytochem. 37:819–829, 1989 [DOI] [PubMed] [Google Scholar]
- 44.Frizell E., Liu S.L., Abraham A., et al. Expression of SPARC in normal and fibrotic livers. Hepatology 21:847–854, 1995 [PubMed] [Google Scholar]
- 45.Blazejewski S., Le Bail B., Boussarie L., et al. Osteonectin (SPARC) expression in human liver and in cultured human liver myofibroblasts. Am. J. Pathol. 151:651–657, 1997 [PMC free article] [PubMed] [Google Scholar]
- 46.Nakken K.E., Nygard S., Haaland T., et al. Multiple inflammatory-, tissue remodelling- and fibrosis genes are differentially transcribed in the livers of Abcb4 (-/-) mice harbouring chronic cholangitis. Scand. J. Gastroenterol. 42:1245–1255, 2007 [DOI] [PubMed] [Google Scholar]
- 47.Pichler R.H., Hugo C., Shankland S.J., et al. SPARC is expressed in renal interstitial fibrosis and in renal vascular injury. Kidney Int. 50:1978–1989, 1996 [DOI] [PubMed] [Google Scholar]
- 48.Allanore Y., Wipff J., Kahan A., and Boileau C.Genetic basis for systemic sclerosis. Joint Bone Spine 74:577–583, 2007 [DOI] [PubMed] [Google Scholar]
- 49.Delany A.M., McMahon D.J., Powell J.S., Greenberg D.A., and Kurland E.S.Osteonectin/SPARC polymorphisms in caucasian men with idiopathic osteoporosis. Osteoporos. Int. 19:969–978, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Framson P.E., and Sage E.H.SPARC and tumor growth: where the seed meets the soil? J. Cell Biochem. 92:679–690, 2004 [DOI] [PubMed] [Google Scholar]
- 51.Barker T.H., Baneyx G., Cardo-Vila M., et al. SPARC regulates extracellular matrix organization through its modulation of integrin-linked kinase activity. J. Biol. Chem. 280:36483–36493, 2005 [DOI] [PubMed] [Google Scholar]
- 52.Barker T.H., Framson P., Puolakkainen P.A., Reed M., Funk S.E., and Sage E.H.Matricellular homologs in the foreign body response: hevin suppresses inflammation, but hevin and SPARC together diminish angiogenesis. Am. J. Pathol. 166:923–933, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhou X., Tan F.K., Guo X., and Arnett F.C.Attenuation of collagen production with small interfering RNA of SPARC in cultured fibroblasts from the skin of patients with scleroderma. Arthritis Rheum. 54:2626–2631, 2006 [DOI] [PubMed] [Google Scholar]
- 54.Zhou X.D., Xiong M.M., Tan F.K., Guo X.J., and Arnett F.C.SPARC, an upstream regulator of connective tissue growth factor in response to transforming growth factor beta stimulation. Arthritis Rheum. 54:3885–3889, 2006 [DOI] [PubMed] [Google Scholar]
- 55.Bassuk J.A., Birkebak T., Rothmier J.D., et al. Disruption of the sparc locus in mice alters the differentiation of lenticular epithelial cells and leads to cataract formation. Exp. Eye Res. 68:321–331, 1999 [DOI] [PubMed] [Google Scholar]
- 56.Bradshaw A.D., Reed M.J., and Sage E.H.SPARC-null mice exhibit accelerated cutaneous wound closure. J. Histochem. Cytochem. 50:1–10, 2002 [DOI] [PubMed] [Google Scholar]
- 57.Bradshaw A.D., Graves D.C., Motamed K., and Sage E.H.SPARC-null mice exhibit increased adiposity without significant differences in overall body weight. Proc. Natl. Acad. Sci. U.S.A. 100:6045–6050, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bradshaw A.D., Puolakkainen P., Dasgupta J., Davidson J.M., Wight T.N., and Helene Sage E.SPARC-null mice display abnormalities in the dermis characterized by decreased collagen fibril diameter and reduced tensile strength. J. Invest. Dermatol. 120:949–955, 2003 [DOI] [PubMed] [Google Scholar]
- 59.Delany A.M., Amling M., Priemel M., Howe C., Baron R., and Canalis E.Osteopenia and decreased bone formation in osteonectin-deficient mice. J. Clin. Invest. 105:915–923, 2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Norose K., Clark J.I., Syed N.A., et al. SPARC deficiency leads to early-onset cataractogenesis. Invest. Ophthalmol. Vis. Sci. 39:2674–2680, 1998 [PubMed] [Google Scholar]
- 61.Puolakkainen P., Bradshaw A.D., Kyriakides T.R., et al. Compromised production of extracellular matrix in mice lacking secreted protein, acidic and rich in cysteine (SPARC) leads to a reduced foreign body reaction to implanted biomaterials. Am. J. Pathol. 162:627–635, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Taneda S., Pippin J.W., Sage E.H., et al. Amelioration of diabetic nephropathy in SPARC-null mice. J. Am. Soc. Nephrol. 14:968–980, 2003 [DOI] [PubMed] [Google Scholar]
- 63.Gilles C., Bassuk J.A., Pulyaeva H., Sage E.H., Foidart J.M., and Thompson E.W.SPARC/osteonectin induces matrix metalloproteinase 2 activation in human breast cancer cell lines. Cancer Res. 58:5529–5536, 1998 [PubMed] [Google Scholar]
- 64.McClung H.M., Thomas S.L., Osenkowski P., et al. SPARC upregulates MT1-MMP expression, MMP-2 activation, and the secretion and cleavage of galectin-3 in U87MG glioma cells. Neurosci. Lett. 419:172–177, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Shankavaram U.T., DeWitt D.L., Funk S.E., Sage E.H., and Wahl L.M.Regulation of human monocyte matrix metalloproteinases by SPARC. J. Cell Physiol. 173:327–334, 1997 [DOI] [PubMed] [Google Scholar]
- 66.Berryhill B.L., Kane B., Stramer B.M., Fini M.E., and Hassell J.R.Increased SPARC accumulation during corneal repair. Exp. Eye Res. 77:85–92, 2003 [DOI] [PubMed] [Google Scholar]
- 67.Kantorow M., Huang Q., Yang X.J., et al. Increased expression of osteonectin/SPARC mRNA and protein in age-related human cataracts and spatial expression in the normal human lens. Mol. Vis. 6:24–29, 2000 [PMC free article] [PubMed] [Google Scholar]
- 68.Yan Q., Clark J.I., and Sage E.H.Expression and characterization of SPARC in human lens and in the aqueous and vitreous humors. Exp. Eye Res. 71:81–90, 2000 [DOI] [PubMed] [Google Scholar]
- 69.Mishima H., Hibino T., Hara H., Murakami J., and Otori T.SPARC from corneal epithelial cells modulates collagen contraction by keratocytes. Invest. Ophthalmol. Vis. Sci. 39:2547–2553, 1998 [PubMed] [Google Scholar]
- 70.Magee R.M., Hagan S., Hiscott P.S., et al. Synthesis of osteonectin by human retinal pigment epithelial cells is modulated by cell density. Invest. Ophthalmol. Vis. Sci. 41:2707–2711, 2000 [PubMed] [Google Scholar]
- 71.Kantorow M., Horwitz J., and Carper D.Up-regulation of osteonectin/SPARC in age-related cataractous human lens epithelia. Mol. Vis. 4:17, 1998 [PubMed] [Google Scholar]
- 72.Gilmour D.T., Lyon G.J., Carlton M.B., et al. Mice deficient for the secreted glycoprotein SPARC/osteonectin/BM40 develop normally but show severe age-onset cataract formation and disruption of the lens. Embo J. 17:1860–1870, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gilbert R.E., Cox A.J., Kelly D.J., et al. Localization of secreted protein acidic and rich in cysteine (SPARC) expression in the rat eye. Connect. Tissue Res. 40:295–303, 1999 [DOI] [PubMed] [Google Scholar]
- 74.Rhee D.J., Fariss R.N., Brekken R., Sage E.H., and Russell P.The matricellular protein SPARC is expressed in human trabecular meshwork. Exp. Eye Res. 77:601–607, 2003 [DOI] [PubMed] [Google Scholar]
- 75.Tomarev S.I., Wistow G., Raymond V., Dubois S., and Malyukova I.Gene expression profile of the human trabecular meshwork: NEIBank sequence tag analysis. Invest. Ophthalmol. Vis. Sci. 44:2588–2596, 2003 [DOI] [PubMed] [Google Scholar]
- 76.Vittal V., Rose A., Gregory K.E., Kelley M.J., and Acott T.S.Changes in gene expression by trabecular meshwork cells in response to mechanical stretching. Invest. Ophthalmol. Vis. Sci. 46:2857–2868, 2005 [DOI] [PubMed] [Google Scholar]
- 77.Haddadin R.I., Oh D.J., Kang M.H., et al. SPARC-null mice exhibit lower intraocular pressures. Invest. Ophthalmol. Vis. Sci. 50:3771–3777, 2009 [DOI] [PubMed] [Google Scholar]
- 78.Chatterjee A., Oh D.J., Kang M.H., and Rhee D.J.Central corneal thickness does not correlate with TonoLab-measured IOP in several mouse strains with single transgenic mutations of matricellular proteins. Exp. Eye Res. 115C:106–112, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Oh D.J., Kang M.H., Ooi Y.H., Choi K.R., Sage E.H., and Rhee D.J.Overexpression of SPARC in human trabecular meshwork increases intraocular pressure and alters extracellular matrix. Invest. Ophthalmol. Vis. Sci. 54:3309–3319, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Villarreal G., Jr., Oh D.J., Kang M.H., and Rhee D.J.Coordinated regulation of extracellular matrix synthesis by the microRNA-29 family in the trabecular meshwork. Invest. Ophthalmol. Vis. Sci. 52:3391–3397, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Luna C., Li G., Qiu J., Epstein D.L., and Gonzalez P.Role of miR-29b on the regulation of the extracellular matrix in human trabecular meshwork cells under chronic oxidative stress. Mol. Vis. 15:2488–2497, 2009 [PMC free article] [PubMed] [Google Scholar]
- 82.Yu A.L., Birke K., Moriniere J., and Welge-Lussen U.TGF-{beta}2 induces senescence-associated changes in human trabecular meshwork cells. Invest. Ophthalmol. Vis. Sci. 51:5718–5723, 2010 [DOI] [PubMed] [Google Scholar]
- 83.Kang M.H., Oh D.J., Kang J.H., and Rhee D.J.Regulation of SPARC by transforming growth factor beta2 in human trabecular meshwork. Invest. Ophthalmol. Vis. Sci. 54:2523–2532, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Villarreal G., Jr., Chatterjee A., Oh S.S., Oh D.J., and Rhee D.J.Pharmacological regulation of SPARC by lovastatin in human trabecular meshwork cells. Invest. Ophthalmol. Vis. Sci. 55:1657–1665, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Chatterjee A., Villarreal G., Jr., Oh D.J., Kang M.H., and Rhee D.J.AMP-activated protein kinase regulates intraocular pressure, extracellular matrix, and cytoskeleton in trabecular meshwork. Invest. Ophthalmol. Vis. Sci. 55:3127–3139, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Thomasy S.M., Wood J.A., Kass P.H., Murphy C.J., and Russell P.Substratum stiffness and latrunculin B regulate matrix gene and protein expression in human trabecular meshwork cells. Invest. Ophthalmol. Vis. Sci. 53:952–958, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Picht G., Welge-Luessen U., Grehn F., and Lutjen-Drecoll E.Transforming growth factor beta 2 levels in the aqueous humor in different types of glaucoma and the relation to filtering bleb development. Graefes Arch. Clin. Exp. Ophthalmol. 239:199–207, 2001 [DOI] [PubMed] [Google Scholar]
- 88.Tripathi R.C., Li J., Chan W.F., and Tripathi B.J.Aqueous humor in glaucomatous eyes contains an increased level of TGF-beta 2. Exp. Eye Res. 59:723–727, 1994 [DOI] [PubMed] [Google Scholar]
- 89.Swaminathan S.S., Oh D.J., Kang M.H., Shepard A.R., Pang I, and Rhee D.J.TGFβ2-mediated ocular hypertension is attenuated in SPARC-null mice. IOVS (In press) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Tawara A., Okada Y., Kubota T., et al. Immunohistochemical localization of MYOC/TIGR protein in the trabecular tissue of normal and glaucomatous eyes. Curr. Eye Res. 21:934–943, 2000 [DOI] [PubMed] [Google Scholar]
- 91.Peters D.M., Herbert K., Biddick B., and Peterson J.A.Myocilin binding to hep II domain of fibronectin inhibits cell spreading and incorporation of paxillin into focal adhesions. Exp. Cell Res. 303:218–228, 2005 [DOI] [PubMed] [Google Scholar]
- 92.Wentz-Hunter K., Shen X., Okazaki K., Tanihara H., and Yue B.Y.Overexpression of myocilin in cultured human trabecular meshwork cells. Exp. Cell Res. 297:39–48, 2004 [DOI] [PubMed] [Google Scholar]
- 93.Stone E.M., Fingert J.H., Alward W.L., et al. Identification of a gene that causes primary open angle glaucoma. Science 275:668–670, 1997 [DOI] [PubMed] [Google Scholar]
- 94.Aroca-Aguilar J.D., Sanchez-Sanchez F., Ghosh S., Fernandez-Navarro A., Coca-Prados M., and Escribano J.Interaction of recombinant myocilin with the matricellular protein SPARC: functional implications. Invest. Ophthalmol. Vis. Sci. 52:179–189, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Aroca-Aguilar J.D., Sanchez-Sanchez F., Ghosh S., Coca-Prados M., and Escribano J.Myocilin mutations causing glaucoma inhibit the intracellular endoproteolytic cleavage of myocilin between amino acids Arg226 and Ile227. J. Biol. Chem. 280:21043–21051, 2005 [DOI] [PubMed] [Google Scholar]
- 96.Vollrath D., and Liu Y.Temperature sensitive secretion of mutant myocilins. Exp. Eye Res. 82:1030–1036, 2006 [DOI] [PubMed] [Google Scholar]
- 97.Gobeil S., Letartre L., and Raymond V.Functional analysis of the glaucoma-causing TIGR/myocilin protein: integrity of amino-terminal coiled-coil regions and olfactomedin homology domain is essential for extracellular adhesion and secretion. Exp. Eye Res. 82:1017–1029, 2006 [DOI] [PubMed] [Google Scholar]
- 98.Perbal B.CCN proteins: multifunctional signalling regulators. Lancet 363:62–64, 2004 [DOI] [PubMed] [Google Scholar]
- 99.Leask A., and Abraham D.J.All in the CCN family: essential matricellular signaling modulators emerge from the bunker. J. Cell. Sci. 119:4803–4810, 2006 [DOI] [PubMed] [Google Scholar]
- 100.Holbourn K.P., Acharya K.R., and Perbal B.The CCN family of proteins: structure-function relationships. Trends Biochem. Sci. 33:461–473, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Brigstock D.R.The connective tissue growth factor/cysteine-rich 61/nephroblastoma overexpressed (CCN) family. Endocr. Rev. 20:189–206, 1999 [DOI] [PubMed] [Google Scholar]
- 102.Ivkovic S., Yoon B.S., Popoff S.N., et al. Connective tissue growth factor coordinates chondrogenesis and angiogenesis during skeletal development. Development 130:2779–2791, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Chen Y., Abraham D.J., Shi-Wen X., et al. CCN2 (connective tissue growth factor) promotes fibroblast adhesion to fibronectin. Mol. Biol. Cell 15:5635–5646, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Abreu J.G., Ketpura N.I., Reversade B., and De Robertis E.M.Connective-tissue growth factor (CTGF) modulates cell signalling by BMP and TGF-beta. Nat. Cell Biol. 4:599–604, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Esson D.W., Neelakantan A., Iyer S.A., et al. Expression of connective tissue growth factor after glaucoma filtration surgery in a rabbit model. Invest. Ophthalmol. Vis. Sci. 45:485–491, 2004 [DOI] [PubMed] [Google Scholar]
- 106.Wang J.M., Hui N., Fan Y.Z., Xiong L., and Sun N.X.Filtering bleb area and intraocular pressure following subconjunctival injection of CTGF antibody after glaucoma filtration surgery in rabbits. Int. J. Ophthalmol. 4:480–483, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ho S.L., Dogar G.F., Wang J., et al. Elevated aqueous humour tissue inhibitor of matrix metalloproteinase-1 and connective tissue growth factor in pseudoexfoliation syndrome. Br. J. Ophthalmol. 89:169–173, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Browne J.G., Ho S.L., Kane R., et al. Connective tissue growth factor is increased in pseudoexfoliation glaucoma. Invest. Ophthalmol. Vis. Sci. 52:3660–3666, 2011 [DOI] [PubMed] [Google Scholar]
- 109.Junglas B., Yu A.H., Welge-Lussen U., Tamm E.R., and Fuchshofer R.Connective tissue growth factor induces extracellular matrix deposition in human trabecular meshwork cells. Exp. Eye Res. 88:1065–1075, 2009 [DOI] [PubMed] [Google Scholar]
- 110.Wallace D.M., Clark A.F., Lipson K.E., Andrews D., Crean J.K., and O'Brien C.J.Anti-connective tissue growth factor antibody treatment reduces extracellular matrix production in trabecular meshwork and lamina cribrosa cells. Invest. Ophthalmol. Vis. Sci. 54:7836–7848, 2013 [DOI] [PubMed] [Google Scholar]
- 111.Junglas B., Kuespert S., Seleem A.A., et al. Connective tissue growth factor causes glaucoma by modifying the actin cytoskeleton of the trabecular meshwork. Am. J. Pathol. 180:2386–2403, 2012 [DOI] [PubMed] [Google Scholar]
- 112.Iyer P., Maddala R., Pattabiraman P.P., and Rao P.V.Connective tissue growth factor-mediated upregulation of neuromedin U expression in trabecular meshwork cells and its role in homeostasis of aqueous humor outflow. Invest. Ophthalmol. Vis. Sci. 53:4952–4962, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Su Y., Cheng J., Liu H., Wang F., and Zhao S.Adenovirus conducted connective tissue growth factor on extracellular matrix in trabecular meshwork and its role on aqueous humor outflow facility. Mol. Biol. Rep. 40:6091–6096, 2013 [DOI] [PubMed] [Google Scholar]
- 114.Fuchshofer R., Kuespert S., Junglas B., and Tamm E.R.The prostaglandin F analog fluprostenol attenuates the fibrotic effects of connective tissue growth factor on human trabecular meshwork cells. J. Ocul. Pharmacol. Ther. 30:237–245, 2014 [DOI] [PubMed] [Google Scholar]
- 115.Baenziger N.L., Brodie G.N., and Majerus P.W.A thrombin-sensitive protein of human platelet membranes. Proc. Natl. Acad. Sci. U.S.A. 68:240–243, 1971 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Baenziger N.L., Brodie G.N., and Majerus P.W.Isolation and properties of a thrombin-sensitive protein of human platelets. J. Biol. Chem. 247:2723–2731, 1972 [PubMed] [Google Scholar]
- 117.McPherson J., Sage H., and Bornstein P.Isolation and characterization of a glycoprotein secreted by aortic endothelial cells in culture. apparent identity with platelet thrombospondin. J. Biol. Chem. 256:11330–11336, 1981 [PubMed] [Google Scholar]
- 118.Adams J., and Lawler J.Extracellular matrix: the thrombospondin family. Curr. Biol. 3:188–190, 1993 [DOI] [PubMed] [Google Scholar]
- 119.Adams J.C., and Lawler J.The thrombospondins. Int. J. Biochem. Cell Biol. 36:961–968, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Adams J.C., and Lawler J.The thrombospondins. Cold Spring Harb Perspect. Biol. 3:a009712, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Hiscott P., Paraoan L., Choudhary A., Ordonez J.L., Al-Khaier A., and Armstrong D.J.Thrombospondin 1, thrombospondin 2 and the eye. Prog. Retin. Eye Res. 25:1–18, 2006 [DOI] [PubMed] [Google Scholar]
- 122.Hankenson K.D., Sweetwyne M.T., Shitaye H., and Posey K.L.Thrombospondins and novel TSR-containing proteins, R-spondins, regulate bone formation and remodeling. Curr. Osteoporos Rep. 8:68–76, 2010 [DOI] [PubMed] [Google Scholar]
- 123.Cingolani O.H., Kirk J.A., Seo K., et al. Thrombospondin-4 is required for stretch-mediated contractility augmentation in cardiac muscle. Circ. Res. 109:1410–1414, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kim D.S., Li K.W., Boroujerdi A., et al. Thrombospondin-4 contributes to spinal sensitization and neuropathic pain states. J. Neurosci. 32:8977–8987, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Zeng J., Kim D., Li K.W., et al. Thrombospondin-4 contributes to spinal cord injury-induced changes in nociception. Eur. J. Pain 17: 1458–1464, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Unger S., and Hecht J.T.Pseudoachondroplasia and multiple epiphyseal dysplasia: new etiologic developments. Am. J. Med. Genet. 106:244–250, 2001 [PubMed] [Google Scholar]
- 127.Posey K.L., and Hecht J.T.The role of cartilage oligomeric matrix protein (COMP) in skeletal disease. Curr. Drug Targets 9:869–877, 2008 [DOI] [PubMed] [Google Scholar]
- 128.Tseng S., Reddi A.H., and Di Cesare P.E.Cartilage oligomeric matrix protein (COMP): a biomarker of arthritis. Biomark Insights 4:33–44, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Adams J.C.Thrombospondins: multifunctional regulators of cell interactions. Annu. Rev. Cell Dev. Biol. 17:25–51, 2001 [DOI] [PubMed] [Google Scholar]
- 130.Alford A.I., and Hankenson K.D.Matricellular proteins: extracellular modulators of bone development, remodeling, and regeneration. Bone 38:749–757, 2006 [DOI] [PubMed] [Google Scholar]
- 131.Bornstein P.Thrombospondins as matricellular modulators of cell function. J. Clin. Invest. 107:929–934, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Bein K., and Simons M.Thrombospondin type 1 repeats interact with matrix metalloproteinase 2. regulation of metalloproteinase activity. J. Biol. Chem. 275:32167–32173, 2000 [DOI] [PubMed] [Google Scholar]
- 133.Yang Z., Strickland D.K., and Bornstein P.Extracellular matrix metalloproteinase 2 levels are regulated by the low density lipoprotein-related scavenger receptor and thrombospondin 2. J. Biol. Chem. 276:8403–8408, 2001 [DOI] [PubMed] [Google Scholar]
- 134.Murphy-Ullrich J.E., Schultz-Cherry S., and Hook M.Transforming growth factor-beta complexes with thrombospondin. Mol. Biol. Cell 3:181–188, 1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Schultz-Cherry S., and Murphy-Ullrich J.E.Thrombospondin causes activation of latent transforming growth factor-beta secreted by endothelial cells by a novel mechanism. J. Cell Biol. 122:923–932, 1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Cockburn C.G., and Barnes M.J.Characterization of thrombospondin binding to collagen (type I) fibres: role of collagen telopeptides. Matrix 11:168–176, 1991 [DOI] [PubMed] [Google Scholar]
- 137.Galvin N.J., Vance P.M., Dixit V.M., Fink B., and Frazier W.A.Interaction of human thrombospondin with types I-V collagen: direct binding and electron microscopy. J. Cell Biol. 104:1413–1422, 1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Mumby S.M., Raugi G.J., and Bornstein P.Interactions of thrombospondin with extracellular matrix proteins: selective binding to type V collagen. J. Cell Biol. 98:646–652, 1984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sipes J.M., Guo N., Negre E., Vogel T., Krutzsch H.C., and Roberts D.D.Inhibition of fibronectin binding and fibronectin-mediated cell adhesion to collagen by a peptide from the second type I repeat of thrombospondin. J. Cell Biol. 121:469–477, 1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Sweetwyne M.T., and Murphy-Ullrich J.E.Thrombospondin1 in tissue repair and fibrosis: TGF-beta-dependent and independent mechanisms. Matrix Biol. 31:178–186, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Penttinen R.P., Kobayashi S., and Bornstein P.Transforming growth factor beta increases mRNA for matrix proteins both in the presence and in the absence of changes in mRNA stability. Proc. Natl. Acad. Sci. U.S.A. 85:1105–1108, 1988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Crawford S.E., Stellmach V., Murphy-Ullrich J.E., et al. Thrombospondin-1 is a major activator of TGF-beta1 in vivo. Cell 93:1159–1170, 1998 [DOI] [PubMed] [Google Scholar]
- 143.Fitchev P.P., Wcislak S.M., Lee C., et al. Thrombospondin-1 regulates the normal prostate in vivo through angiogenesis and TGF-beta activation. Lab. Invest. 90:1078–1090, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Hankenson K.D., Bain S.D., Kyriakides T.R., Smith E.A., Goldstein S.A., and Bornstein P.Increased marrow-derived osteoprogenitor cells and endosteal bone formation in mice lacking thrombospondin 2. J. Bone Miner. Res. 15:851–862, 2000 [DOI] [PubMed] [Google Scholar]
- 145.Kyriakides T.R., Zhu Y.H., Smith L.T., et al. Mice that lack thrombospondin 2 display connective tissue abnormalities that are associated with disordered collagen fibrillogenesis, an increased vascular density, and a bleeding diathesis. J. Cell Biol. 140:419–430, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Agah A., Kyriakides T.R., Lawler J., and Bornstein P.The lack of thrombospondin-1 (TSP1) dictates the course of wound healing in double-TSP1/TSP2-null mice. Am. J. Pathol. 161:831–839, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Christensen A.E., Fiskerstrand T., Knappskog P.M., Boman H., and Rodahl E.A novel ADAMTSL4 mutation in autosomal recessive ectopia lentis et pupillae. Invest. Ophthalmol. Vis. Sci. 51:6369–6373, 2010 [DOI] [PubMed] [Google Scholar]
- 148.Greene V.B., Stoetzel C., Pelletier V., et al. Confirmation of ADAMTSL4 mutations for autosomal recessive isolated bilateral ectopia lentis. Ophthalmic Genet. 31:47–51, 2010 [DOI] [PubMed] [Google Scholar]
- 149.Keller K.E., Bradley J.M., and Acott T.S.Differential effects of ADAMTS-1, -4, and -5 in the trabecular meshwork. Invest. Ophthalmol. Vis. Sci. 50:5769–5777, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Hiscott P., Schlotzer-Schrehardt U., and Naumann G.O.Unexpected expression of thrombospondin 1 by corneal and iris fibroblasts in the pseudoexfoliation syndrome. Hum. Pathol. 27:1255–1258, 1996 [DOI] [PubMed] [Google Scholar]
- 151.Wang S., Neekhra A., Albert D.M., Sorenson C.M., and Sheibani N.Suppression of thrombospondin-1 expression during uveal melanoma progression and its potential therapeutic utility. Arch. Ophthalmol. 130:336–341, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Flugel-Koch C., Ohlmann A., Fuchshofer R., Welge-Lussen U., and Tamm E.R.Thrombospondin-1 in the trabecular meshwork: localization in normal and glaucomatous eyes, and induction by TGF-beta1 and dexamethasone in vitro. Exp. Eye Res. 79:649–663, 2004 [DOI] [PubMed] [Google Scholar]
- 153.Fleenor D.L., Shepard A.R., Hellberg P.E., Jacobson N., Pang I.H., and Clark A.F.TGFbeta2-induced changes in human trabecular meshwork: implications for intraocular pressure. Invest. Ophthalmol. Vis. Sci. 47:226–234, 2006 [DOI] [PubMed] [Google Scholar]
- 154.Fuchshofer R., Yu A.H., Welge-Lussen U., and Tamm E.R.Bone morphogenetic protein-7 is an antagonist of transforming growth factor-beta2 in human trabecular meshwork cells. Invest. Ophthalmol. Vis. Sci. 48:715–726, 2007 [DOI] [PubMed] [Google Scholar]
- 155.Fuchshofer R., Welge-Lussen U., and Lutjen-Drecoll E.The effect of TGF-beta2 on human trabecular meshwork extracellular proteolytic system. Exp. Eye Res. 77:757–765, 2003 [DOI] [PubMed] [Google Scholar]
- 156.Gottanka J., Chan D., Eichhorn M., Lutjen-Drecoll E., and Ethier C.R.Effects of TGF-beta2 in perfused human eyes. Invest. Ophthalmol. Vis. Sci. 45:153–158, 2004 [DOI] [PubMed] [Google Scholar]
- 157.Ochiai Y., and Ochiai H.Higher concentration of transforming growth factor-beta in aqueous humor of glaucomatous eyes and diabetic eyes. Jpn. J. Ophthalmol. 46:249–253, 2002 [DOI] [PubMed] [Google Scholar]
- 158.Zhao X., Ramsey K.E., Stephan D.A., and Russell P.Gene and protein expression changes in human trabecular meshwork cells treated with transforming growth factor-beta. Invest. Ophthalmol. Vis. Sci. 45:4023–4034, 2004 [DOI] [PubMed] [Google Scholar]
- 159.Haddadin R.I., Oh D.J., Kang M.H., et al. Thrombospondin-1 (TSP1)-null and TSP2-null mice exhibit lower intraocular pressures. Invest. Ophthalmol. Vis. Sci. 53:6708–6717, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Bourdon M.A., Wikstrand C.J., Furthmayr H., Matthews T.J., and Bigner D.D.Human glioma-mesenchymal extracellular matrix antigen defined by monoclonal antibody. Cancer Res. 43:2796–2805, 1983 [PubMed] [Google Scholar]
- 161.Chiquet M., and Fambrough D.M.Chick myotendinous antigen. II. A novel extracellular glycoprotein complex consisting of large disulfide-linked subunits. J. Cell Biol. 98:1937–1946, 1984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Chiquet-Ehrismann R., and Chiquet M.Tenascins: regulation and putative functions during pathological stress. J. Pathol. 200:488–499, 2003 [DOI] [PubMed] [Google Scholar]
- 163.Jakovcevski I., Miljkovic D., Schachner M., and Andjus P.R.Tenascins and inflammation in disorders of the nervous system. Amino Acids 44:1115–1127, 2013 [DOI] [PubMed] [Google Scholar]
- 164.Brellier F., Martina E., Chiquet M., et al. The adhesion modulating properties of tenascin-W. Int. J. Biol. Sci. 8:187–194, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Grogan S.P., Chen X., Sovani S., et al. Influence of cartilage extracellular matrix molecules on cell phenotype and neocartilage formation. Tissue Eng. Part A 20: 264–274, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Katoh D., Nagaharu K., Shimojo N., et al. Binding of alphavbeta1 and alphavbeta6 integrins to tenascin-C induces epithelial-mesenchymal transition-like change of breast cancer cells. Oncogenesis 2:e65, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Brosicke N., van Landeghem F.K., Scheffler B., and Faissner A.Tenascin-C is expressed by human glioma in vivo and shows a strong association with tumor blood vessels. Cell Tissue Res. 354:409–430, 2013 [DOI] [PubMed] [Google Scholar]
- 168.Orend G., and Chiquet-Ehrismann R.Adhesion modulation by antiadhesive molecules of the extracellular matrix. Exp. Cell Res. 261:104–110, 2000 [DOI] [PubMed] [Google Scholar]
- 169.Chiquet-Ehrismann R., Kalla P., Pearson C.A., Beck K., and Chiquet M.Tenascin interferes with fibronectin action. Cell 53:383–390, 1988 [DOI] [PubMed] [Google Scholar]
- 170.De Laporte, L., Rice J.J., Tortelli F., and Hubbell J.A.Tenascin C promiscuously binds growth factors via its fifth fibronectin type III-like domain. PLoS One 8:e62076, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Feugate J.E., Wong L., Li Q.J., and Martins-Green M.The CXC chemokine cCAF stimulates precocious deposition of ECM molecules by wound fibroblasts, accelerating development of granulation tissue. BMC Cell Biol. 3:13, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Hou Y., Lin H., Zhu L., et al. LPS increases the incidence of collagen-induced arthritis in mice through induction of protease HTRA1 expression. Arthritis Rheum. 65:2835–2846, 2013 [DOI] [PubMed] [Google Scholar]
- 173.Makhluf H.A., Stepniakowska J., Hoffman S., Smith E., LeRoy E.C., and Trojanowska M.IL-4 upregulates tenascin synthesis in scleroderma and healthy skin fibroblasts. J. Invest. Dermatol. 107:856–859, 1996 [DOI] [PubMed] [Google Scholar]
- 174.Pearson C.A., Pearson D., Shibahara S., Hofsteenge J., and Chiquet-Ehrismann R.Tenascin: CDNA cloning and induction by TGF-beta. Embo J. 7:2977–2982, 1988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Fitch P.M., Howie S.E., and Wallace W.A.Oxidative damage and TGF-beta differentially induce lung epithelial cell sonic hedgehog and tenascin-C expression: implications for the regulation of lung remodelling in idiopathic interstitial lung disease. Int. J. Exp. Pathol. 92:8–17, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Chablais F., and Jazwinska A.The regenerative capacity of the zebrafish heart is dependent on TGFbeta signaling. Development 139:1921–1930, 2012 [DOI] [PubMed] [Google Scholar]
- 177.Fluck M., Chiquet M., Schmutz S., Mayet-Sornay M.H., and Desplanches D.Reloading of atrophied rat soleus muscle induces tenascin-C expression around damaged muscle fibers. Am. J. Physiol. Regul. Integr. Comp. Physiol. 284:R792–R801, 2003 [DOI] [PubMed] [Google Scholar]
- 178.Jarvinen T.A., Jozsa L., Kannus P., et al. Mechanical loading regulates tenascin-C expression in the osteotendinous junction. J. Cell Sci. 112Pt 18:3157–3166, 1999 [DOI] [PubMed] [Google Scholar]
- 179.Mercer S., Odelberg S.J., and Simon H.G.A dynamic spatiotemporal extracellular matrix facilitates epicardial-mediated vertebrate heart regeneration. Dev. Biol. 382:457–469, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Franz M., Berndt A., Neri D., et al. Matrix metalloproteinase-9, tissue inhibitor of metalloproteinase-1, B tenascin-C and ED-A fibronectin in dilated cardiomyopathy: potential impact on disease progression and patients' prognosis. Int. J. Cardiol. 168:5344–5351, 2013 [DOI] [PubMed] [Google Scholar]
- 181.Forsberg E., Hirsch E., Frohlich L., et al. Skin wounds and severed nerves heal normally in mice lacking tenascin-C. Proc. Natl. Acad. Sci. U.S.A. 93:6594–6599, 1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Fukamauchi F., Mataga N., Wang Y.J., Sato S., Youshiki A., and Kusakabe M.Abnormal behavior and neurotransmissions of tenascin gene knockout mouse. Biochem. Biophys. Res. Commun. 221:151–156, 1996 [DOI] [PubMed] [Google Scholar]
- 183.Mackie E.J., and Tucker R.P.The tenascin-C knockout revisited. J. Cell. Sci. 112:3847–3853, 1999 [DOI] [PubMed] [Google Scholar]
- 184.Matsuda A., Yoshiki A., Tagawa Y., Matsuda H., and Kusakabe M.Corneal wound healing in tenascin knockout mouse. Invest. Ophthalmol. Vis. Sci. 40:1071–1080, 1999 [PubMed] [Google Scholar]
- 185.Jaggi G.P., Laeng H.R., Muntener M., and Killer H.E.The anatomy of the muscle insertion (scleromuscular junction) of the lateral and medial rectus muscle in humans. Invest. Ophthalmol. Vis. Sci. 46:2258–2263, 2005 [DOI] [PubMed] [Google Scholar]
- 186.Ueda J., Wentz-Hunter K., and Yue B.Y.Distribution of myocilin and extracellular matrix components in the juxtacanalicular tissue of human eyes. Invest. Ophthalmol. Vis. Sci. 43:1068–1076, 2002 [PubMed] [Google Scholar]
- 187.Yeung A.M., Schlotzer-Schrehardt U., Kulkarni B., Tint N.L., Hopkinson A., and Dua H.S.Limbal epithelial crypt: a model for corneal epithelial maintenance and novel limbal regional variations. Arch. Ophthalmol. 126:665–669, 2008 [DOI] [PubMed] [Google Scholar]
- 188.Spurlin J.W., 3rd, and Lwigale P.Wounded embryonic corneas exhibit non-fibrotic regeneration and complete innervation. Invest. Ophthalmol. Vis. Sci. 54:6334–6344, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Mitamura Y., Takeuchi S., Ohtsuka K., Matsuda A., Hiraiwa N., and Kusakabe M.Tenascin-C levels in the vitreous of patients with proliferative diabetic retinopathy. Diabetes Care 25:1899, 2002 [DOI] [PubMed] [Google Scholar]
- 190.Mitamura Y., Takeuchi S., Ohtsuka K., et al. Tenascin-C levels in the vitreous of patients with proliferative vitreoretinopathy. Ophthalmologica 217:422–425, 2003 [DOI] [PubMed] [Google Scholar]
- 191.Palenski T.L., Sorenson C.M., and Sheibani N.Inflammatory cytokine-specific alterations in retinal endothelial cell function. Microvasc. Res. 89:57–69, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Johnson E.C., Jia L., Cepurna W.O., Doser T.A., and Morrison J.C.Global changes in optic nerve head gene expression after exposure to elevated intraocular pressure in a rat glaucoma model. Invest. Ophthalmol. Vis. Sci. 48:3161–3177, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Keller K.E., Bradley J.M., Kelley M.J., and Acott T.S.Effects of modifiers of glycosaminoglycan biosynthesis on outflow facility in perfusion culture. Invest. Ophthalmol. Vis. Sci. 49:2495–2505, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Keller K.E., Kelley M.J., and Acott T.S.Extracellular matrix gene alternative splicing by trabecular meshwork cells in response to mechanical stretching. Invest. Ophthalmol. Vis. Sci. 48:1164–1172, 2007 [DOI] [PubMed] [Google Scholar]
- 195.Pattabiraman P.P., and Rao P.V.Mechanistic basis of rho GTPase-induced extracellular matrix synthesis in trabecular meshwork cells. Am. J. Physiol. Cell. Physiol. 298:C749–C763, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Keller K.E., Vranka J.A., Haddadin R.I., et al. The effects of tenascin C knockdown on trabecular meshwork outflow resistance. Invest. Ophthalmol. Vis. Sci. 54:5613–5623, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Matsumoto K., Saga Y., Ikemura T., Sakakura T., and Chiquet-Ehrismann R.The distribution of tenascin-X is distinct and often reciprocal to that of tenascin-C. J. Cell Biol. 125:483–493, 1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Burch G.H., Gong Y., Liu W., et al. Tenascin-X deficiency is associated with ehlers-danlos syndrome. Nat. Genet. 17:104–108, 1997 [DOI] [PubMed] [Google Scholar]
- 199.Schalkwijk J., Zweers M.C., Steijlen P.M., et al. A recessive form of the ehlers-danlos syndrome caused by tenascin-X deficiency. N. Engl. J. Med. 345:1167–1175, 2001 [DOI] [PubMed] [Google Scholar]
- 200.Penisson-Besnier I., Allamand V., Beurrier P., et al. Compound heterozygous mutations of the TNXB gene cause primary myopathy. Neuromuscul. Disord. 23:664–669, 2013 [DOI] [PubMed] [Google Scholar]
- 201.Merke D.P., Chen W., Morissette R., et al. Tenascin-X haploinsufficiency associated with ehlers-danlos syndrome in patients with congenital adrenal hyperplasia. J. Clin. Endocrinol. Metab. 98:E379–E387, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Lethias C., Elefteriou F., Parsiegla G., Exposito J.Y., and Garrone R.Identification and characterization of a conformational heparin-binding site involving two fibronectin type III modules of bovine tenascin-X. J. Biol. Chem. 276:16432–16438, 2001 [DOI] [PubMed] [Google Scholar]
- 203.Margaron Y., Bostan L., Exposito J.Y., et al. Tenascin-X increases the stiffness of collagen gels without affecting fibrillogenesis. Biophys. Chem. 147:87–91, 2010 [DOI] [PubMed] [Google Scholar]
- 204.Mao J.R., Taylor G., Dean W.B., et al. Tenascin-X deficiency mimics ehlers-danlos syndrome in mice through alteration of collagen deposition. Nat. Genet. 30:421–425, 2002 [DOI] [PubMed] [Google Scholar]
- 205.Voermans N.C., Verrijp K., Eshuis L., et al. Mild muscular features in tenascin-X knockout mice, a model of ehlers-danlos syndrome. Connect. Tissue Res. 52:422–432, 2011 [DOI] [PubMed] [Google Scholar]
- 206.Elefteriou F., Exposito J.Y., Garrone R., and Lethias C.Binding of tenascin-X to decorin. FEBS Lett. 495:44–47, 2001 [DOI] [PubMed] [Google Scholar]
- 207.Tuori A., Uusitalo H., Thornell L.E., Yoshida T., and Virtanen I.The expression of tenascin-X in developing and adult rat and human eye. Histochem. J. 31:245–252, 1999 [DOI] [PubMed] [Google Scholar]
- 208.Cipriani V., Leung H.T., Plagnol V., et al. Genome-wide association study of age-related macular degeneration identifies associated variants in the TNXB-FKBPL-NOTCH4 region of chromosome 6p21.3. Hum. Mol. Genet. 21:4138–4150, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Tanihara H., Ohira A., Takahashi M., Honda Y., and Suzuki S.Localization and possible gene expression of proteoglycan decorin in the trabecular meshwork. Curr. Eye Res. 14:727–730, 1995 [DOI] [PubMed] [Google Scholar]
- 210.Ueda J., and Yue B.Y.Distribution of myocilin and extracellular matrix components in the corneoscleral meshwork of human eyes. Invest. Ophthalmol. Vis. Sci. 44:4772–4779, 2003 [DOI] [PubMed] [Google Scholar]
- 211.Wirtz M.K., Bradley J.M., Xu H., et al. Proteoglycan expression by human trabecular meshworks. Curr. Eye Res. 16:412–421, 1997 [DOI] [PubMed] [Google Scholar]
- 212.Johnston I.G., Paladino T., Gurd J.W., and Brown I.R.Molecular cloning of SC1: a putative brain extracellular matrix glycoprotein showing partial similarity to osteonectin/BM40/SPARC. Neuron 4:165–176, 1990 [DOI] [PubMed] [Google Scholar]
- 213.Nelson P.S., Plymate S.R., Wang K., et al. Hevin, an antiadhesive extracellular matrix protein, is down-regulated in metastatic prostate adenocarcinoma. Cancer Res. 58:232–236, 1998 [PubMed] [Google Scholar]
- 214.Girard J.P., and Springer T.A.Modulation of endothelial cell adhesion by hevin, an acidic protein associated with high endothelial venules. J. Biol. Chem. 271:4511–4517, 1996 [DOI] [PubMed] [Google Scholar]
- 215.Hambrock H.O., Nitsche D.P., Hansen U., et al. SC1/hevin. an extracellular calcium-modulated protein that binds collagen I. J. Biol. Chem. 278:11351–11358, 2003 [DOI] [PubMed] [Google Scholar]
- 216.Sullivan M.M., Barker T.H., Funk S.E., et al. Matricellular hevin regulates decorin production and collagen assembly. J. Biol. Chem. 281:27621–27632, 2006 [DOI] [PubMed] [Google Scholar]
- 217.Soderling J.A., Reed M.J., Corsa A., and Sage E.H.Cloning and expression of murine SC1, a gene product homologous to SPARC. J. Histochem. Cytochem. 45:823–835, 1997 [DOI] [PubMed] [Google Scholar]
- 218.Li Y., Aroca-Aguilar J.D., Ghosh S., Sanchez-Sanchez F., Escribano J., and Coca-Prados M.Interaction of myocilin with the C-terminal region of hevin. Biochem. Biophys. Res. Commun. 339:797–804, 2006 [DOI] [PubMed] [Google Scholar]
- 219.Graul T.A., Kwon Y.H., Zimmerman M.B., et al. A case-control comparison of the clinical characteristics of glaucoma and ocular hypertensive patients with and without the myocilin Gln368Stop mutation. Am. J. Ophthalmol. 134:884–890, 2002 [DOI] [PubMed] [Google Scholar]
- 220.Kang M.H., Oh D.J., and Rhee D.J.Effect of hevin deletion in mice and characterization in trabecular meshwork. Invest. Ophthalmol. Vis. Sci. 52:2187–2193, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Senger D.R., Wirth D.F., and Hynes R.O.Transformed mammalian cells secrete specific proteins and phosphoproteins. Cell 16:885–893, 1979 [DOI] [PubMed] [Google Scholar]
- 222.Oldberg A., Franzen A., and Heinegard D.Cloning and sequence analysis of rat bone sialoprotein (osteopontin) cDNA reveals an arg-gly-asp cell-binding sequence. Proc. Natl. Acad. Sci. U.S.A. 83:8819–8823, 1986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Sodek J., Ganss B., and McKee M.D.Osteopontin. Crit. Rev. Oral Biol. Med. 11:279–303, 2000 [DOI] [PubMed] [Google Scholar]
- 224.Denhardt D.T., and Guo X.Osteopontin: a protein with diverse functions. Faseb J. 7:1475–1482, 1993 [PubMed] [Google Scholar]
- 225.O'Regan A., and Berman J.S.Osteopontin: a key cytokine in cell-mediated and granulomatous inflammation. Int. J. Exp. Pathol. 81:373–390, 2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Nishimichi N., Hayashita-Kinoh H., Chen C., Matsuda H., Sheppard D., and Yokosaki Y.Osteopontin undergoes polymerization in vivo and gains chemotactic activity for neutrophils mediated by integrin alpha9beta1. J. Biol. Chem. 286:11170–11178, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Wolak T., Sion-Vardi N., Novack V., et al. N-terminal rather than full-length osteopontin or its C-terminal fragment is associated with carotid-plaque inflammation in hypertensive patients. Am. J. Hypertens. 26:326–333, 2013 [DOI] [PubMed] [Google Scholar]
- 228.Sodek J., Batista Da Silva A.P., and Zohar R.Osteopontin and mucosal protection. J. Dent. Res. 85:404–415, 2006 [DOI] [PubMed] [Google Scholar]
- 229.Giachelli C.M., and Steitz S.Osteopontin: a versatile regulator of inflammation and biomineralization. Matrix Biol. 19:615–622, 2000 [DOI] [PubMed] [Google Scholar]
- 230.Liaw L., Birk D.E., Ballas C.B., Whitsitt J.S., Davidson J.M., and Hogan B.L.Altered wound healing in mice lacking a functional osteopontin gene (spp1). J. Clin. Invest. 101:1468–1478, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Rittling S.R., and Denhardt D.T.Osteopontin function in pathology: lessons from osteopontin-deficient mice. Exp. Nephrol. 7:103–113, 1999 [DOI] [PubMed] [Google Scholar]
- 232.Yoshitake H., Rittling S.R., Denhardt D.T., and Noda M.Osteopontin-deficient mice are resistant to ovariectomy-induced bone resorption. Proc. Natl. Acad. Sci. U.S.A. 96:8156–8160, 1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Nagao M., Feinstein T.N., Ezura Y., et al. Sympathetic control of bone mass regulated by osteopontin. Proc. Natl. Acad. Sci. U.S.A. 108:17767–17772, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Morishita M., Ono N., Miyai K., et al. Osteopontin deficiency enhances parathyroid hormone/parathyroid hormone related peptide receptor (PPR) signaling-induced alteration in tooth formation and odontoblastic morphology. Tissue Cell 43:196–200, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Shimizu S., Okuda N., Kato N., et al. Osteopontin deficiency impairs wear debris-induced osteolysis via regulation of cytokine secretion from murine macrophages. Arthritis Rheum. 62:1329–1337, 2010 [DOI] [PubMed] [Google Scholar]
- 236.Kase S., Yokoi M., Saito W., et al. Increased osteopontin levels in the vitreous of patients with diabetic retinopathy. Ophthalmic Res. 39:143–147, 2007 [DOI] [PubMed] [Google Scholar]
- 237.Miyazaki K., Okada Y., Yamanaka O., et al. Corneal wound healing in an osteopontin-deficient mouse. Invest. Ophthalmol. Vis. Sci. 49:1367–1375, 2008 [DOI] [PubMed] [Google Scholar]
- 238.Saika S., Miyamoto T., Ishida I., Ohnishi Y., and Ooshima A.Osteopontin: a component of matrix in capsular opacification and subcapsular cataract. Invest. Ophthalmol. Vis. Sci. 44:1622–1628, 2003 [DOI] [PubMed] [Google Scholar]
- 239.Saika S., Sumioka T., Okada Y., et al. Wakayama symposium: modulation of wound healing response in the corneal stroma by osteopontin and tenascin-C. Ocul. Surf. 11:12–15, 2013 [DOI] [PubMed] [Google Scholar]
- 240.Fujita N., Fujita S., Ogata N., et al. Endogenous osteopontin involvement in laser-induced choroidal neovascularization in mice. Invest. Ophthalmol. Vis. Sci. 52:9310–9315, 2011 [DOI] [PubMed] [Google Scholar]
- 241.Abu El-Asrar A.M., Imtiaz Nawaz, M., Kangave D., Siddiquei M.M., and Geboes K.Osteopontin and other regulators of angiogenesis and fibrogenesis in the vitreous from patients with proliferative vitreoretinal disorders. Mediators Inflamm. 2012:493043, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Deeg C.A., Eberhardt C., Hofmaier F., Amann B., and Hauck S.M.Osteopontin and fibronectin levels are decreased in vitreous of autoimmune uveitis and retinal expression of both proteins indicates ECM re-modeling. PLoS One 6:e27674, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Keller K.E., Aga M., Bradley J.M., Kelley M.J., and Acott T.S.Extracellular matrix turnover and outflow resistance. Exp. Eye Res. 88:676–682, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Taube A.B., Hardenborg E., Wetterhall M., et al. Proteins in aqueous humor from cataract patients with and without pseudoexfoliation syndrome. Eur. J. Mass. Spectrom. (Chichester, Eng) 18:531–541, 2012 [DOI] [PubMed] [Google Scholar]
- 245.Birke M.T., Neumann C., Birke K., Kremers J., and Scholz M.Changes of osteopontin in the aqueous humor of the DBA2/J glaucoma model correlated with optic nerve and RGC degenerations. Invest. Ophthalmol. Vis. Sci. 51:5759–5767, 2010 [DOI] [PubMed] [Google Scholar]
- 246.Chowdhury U.R., Jea S.Y., Oh D.J., Rhee D.J., and Fautsch M.P.Expression profile of the matricellular protein osteopontin in primary open-angle glaucoma and the normal human eye. Invest. Ophthalmol. Vis. Sci. 52:6443–6451, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Gabius H.J.Animal lectins. Eur. J. Biochem. 243:543–576, 1997 [DOI] [PubMed] [Google Scholar]
- 248.von der Lieth, C., Siebert H., Kozar T., et al. Lectin ligands: new insights into their conformations and their dynamic behavior and the discovery of conformer selection by lectins. Acta Anat. (Basel) 161:91–109, 1998 [DOI] [PubMed] [Google Scholar]
- 249.Elola M.T., Wolfenstein-Todel C., Troncoso M.F., Vasta G.R., and Rabinovich G.A.Galectins: matricellular glycan-binding proteins linking cell adhesion, migration, and survival. Cell Mol. Life Sci. 64:1679–1700, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Rabinovich G.A., Riera C.M., Landa C.A., and Sotomayor C.E.Galectins: a key intersection between glycobiology and immunology. Braz. J. Med. Biol. Res. 32:383–393, 1999 [DOI] [PubMed] [Google Scholar]
- 251.Zick Y., Eisenstein M., Goren R.A., Hadari Y.R., Levy Y., and Ronen D.Role of galectin-8 as a modulator of cell adhesion and cell growth. Glycoconj. J. 19:517–526, 2004 [DOI] [PubMed] [Google Scholar]
- 252.Poirier F., and Robertson E.J.Normal development of mice carrying a null mutation in the gene encoding the L14 S-type lectin. Development 119:1229–1236, 1993 [DOI] [PubMed] [Google Scholar]
- 253.Colnot C., Fowlis D., Ripoche M.A., Bouchaert I., and Poirier F.Embryonic implantation in galectin 1/galectin 3 double mutant mice. Dev. Dyn. 211:306–313, 1998 [DOI] [PubMed] [Google Scholar]
- 254.Gruson D., and Ko G.Galectins testing: new promises for the diagnosis and risk stratification of chronic diseases? Clin. Biochem. 45:719–726, 2012 [DOI] [PubMed] [Google Scholar]
- 255.Baum L.G., Seilhamer J.J., Pang M., Levine W.B., Beynon D., and Berliner J.A.Synthesis of an endogeneous lectin, galectin-1, by human endothelial cells is up-regulated by endothelial cell activation. Glycoconj. J. 12:63–68, 1995 [DOI] [PubMed] [Google Scholar]
- 256.Perillo N.L., Marcus M.E., and Baum L.G.Galectins: versatile modulators of cell adhesion, cell proliferation, and cell death. J. Mol. Med. (Berl) 76:402–412, 1998 [DOI] [PubMed] [Google Scholar]
- 257.Ozeki Y., Matsui T., Yamamoto Y., Funahashi M., Hamako J., and Titani K.Tissue fibronectin is an endogenous ligand for galectin-1. Glycobiology 5:255–261, 1995 [DOI] [PubMed] [Google Scholar]
- 258.Zhou Q., and Cummings R.D.The S-type lectin from calf heart tissue binds selectively to the carbohydrate chains of laminin. Arch. Biochem. Biophys. 281:27–35, 1990 [DOI] [PubMed] [Google Scholar]
- 259.van den Brule F.A., Buicu C., Baldet M., et al. Galectin-1 modulates human melanoma cell adhesion to laminin. Biochem. Biophys. Res. Commun. 209:760–767, 1995 [DOI] [PubMed] [Google Scholar]
- 260.Adams L., Scott G.K., and Weinberg C.S.Biphasic modulation of cell growth by recombinant human galectin-1. Biochim. Biophys. Acta 1312:137–144, 1996 [DOI] [PubMed] [Google Scholar]
- 261.Scott K., and Weinberg C.Galectin-1: a bifunctional regulator of cellular proliferation. Glycoconj. J. 19:467–477, 2004 [DOI] [PubMed] [Google Scholar]
- 262.Vasta G.R., Ahmed H., Bianchet M.A., Fernandez-Robledo J.A., and Amzel L.M.Diversity in recognition of glycans by F-type lectins and galectins: molecular, structural, and biophysical aspects. Ann. N.Y. Acad. Sci. 1253:E14–E26, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Matarrese P., Fusco O., Tinari N., et al. Galectin-3 overexpression protects from apoptosis by improving cell adhesion properties. Int. J. Cancer 85:545–554, 2000 [PubMed] [Google Scholar]
- 264.Ohannesian D.W., Lotan D., Thomas P., et al. Carcinoembryonic antigen and other glycoconjugates act as ligands for galectin-3 in human colon carcinoma cells. Cancer Res. 55:2191–2199, 1995 [PubMed] [Google Scholar]
- 265.Nangia-Makker P., Raz T., Tait L., Hogan V., Fridman R., and Raz A.Galectin-3 cleavage: a novel surrogate marker for matrix metalloproteinase activity in growing breast cancers. Cancer Res. 67:11760–11768, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Ortega N., Behonick D.J., Colnot C., Cooper D.N., and Werb Z.Galectin-3 is a downstream regulator of matrix metalloproteinase-9 function during endochondral bone formation. Mol. Biol. Cell 16:3028–3039, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Wang Y.G., Kim S.J., Baek J.H., Lee H.W., Jeong S.Y., and Chun K.H.Galectin-3 increases the motility of mouse melanoma cells by regulating matrix metalloproteinase-1 expression. Exp. Mol. Med. 44:387–393, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Kim S.J., Shin J.Y., Lee K.D., et al. Galectin-3 facilitates cell motility in gastric cancer by up-regulating protease-activated receptor-1 (PAR-1) and matrix metalloproteinase-1 (MMP-1). PLoS One 6:e25103, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Lin Y.H., Lin L.Y., Wu Y.W., et al. The relationship between serum galectin-3 and serum markers of cardiac extracellular matrix turnover in heart failure patients. Clin. Chim. Acta 409:96–99, 2009 [DOI] [PubMed] [Google Scholar]
- 270.Santiago-Gomez A., Barrasa J.I., Olmo N., et al. 4F2hc-silencing impairs tumorigenicity of HeLa cells via modulation of galectin-3 and beta-catenin signaling, and MMP-2 expression. Biochim. Biophys. Acta 1833:2045–2056, 2013 [DOI] [PubMed] [Google Scholar]
- 271.Astorgues-Xerri L., Riveiro M.E., Tijeras-Raballand A., et al. Unraveling galectin-1 as a novel therapeutic target for cancer. Cancer Treat. Rev. 40:307–319, 2014 [DOI] [PubMed] [Google Scholar]
- 272.Fermin Lee, A., Chen H.Y., Wan L., et al. Galectin-3 modulates Th17 responses by regulating dendritic cell cytokines. Am. J. Pathol. 183:1209–1222, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Alge C.S., Priglinger S.G., Kook D., et al. Galectin-1 influences migration of retinal pigment epithelial cells. Invest. Ophthalmol. Vis. Sci. 47:415–426, 2006 [DOI] [PubMed] [Google Scholar]
- 274.Canning P., Glenn J.V., Hsu D.K., Liu F.T., Gardiner T.A., and Stitt A.W.Inhibition of advanced glycation and absence of galectin-3 prevent blood-retinal barrier dysfunction during short-term diabetes. Exp. Diabetes Res. 2007:51837, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Fukushima A., Sumi T., Fukuda K., et al. Roles of galectin-9 in the development of experimental allergic conjunctivitis in mice. Int. Arch. Allergy Immunol. 146:36–43, 2008 [DOI] [PubMed] [Google Scholar]
- 276.Wang Z., Handa J.T., Green W.R., Stark W.J., Weinberg R.S., and Jun A.S.Advanced glycation end products and receptors in fuchs' dystrophy corneas undergoing descemet's stripping with endothelial keratoplasty. Ophthalmology 114:1453–1460, 2007 [DOI] [PubMed] [Google Scholar]
- 277.Jin Q.H., Lou Y.F., Li T.L., Chen H.H., Liu Q., and He X.J.Serum galectin-3: a risk factor for vascular complications in type 2 diabetes mellitus. Chin. Med. J. (Engl) 126:2109–2115, 2013 [PubMed] [Google Scholar]
- 278.Shimmura-Tomita M., Wang M., Taniguchi H., Akiba H., Yagita H., and Hori J.Galectin-9-mediated protection from allo-specific T cells as a mechanism of immune privilege of corneal allografts. PLoS One 8:e63620, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Argueso P.Glycobiology of the ocular surface: mucins and lectins. Jpn. J. Ophthalmol. 57:150–155, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Saravanan C., Cao Z., Head S.R., and Panjwani N.Detection of differentially expressed wound-healing-related glycogenes in galectin-3-deficient mice. Invest. Ophthalmol. Vis. Sci. 50:5690–5696, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Suryawanshi A., Cao Z., Thitiprasert T., Zaidi T.S., and Panjwani N.Galectin-1-mediated suppression of pseudomonas aeruginosa-induced corneal immunopathology. J. Immunol. 190:6397–6409, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Rajasagi N.K., Suryawanshi A., Sehrawat S., et al. Galectin-1 reduces the severity of herpes simplex virus-induced ocular immunopathological lesions. J. Immunol. 188:4631–4643, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.J Reddy P.B., Schreiber T.H., Rajasagi N.K., et al. TNFRSF25 agonistic antibody and galectin-9 combination therapy controls herpes simplex virus-induced immunoinflammatory lesions. J. Virol. 86:10606–10620, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Dahm R., Bramke S., Dawczynski J., Nagaraj R.H., and Kasper M.Developmental aspects of galectin-3 expression in the lens. Histochem. Cell Biol. 119:219–226, 2003 [DOI] [PubMed] [Google Scholar]
- 285.Ogden A.T., Nunes I., Ko K., et al. GRIFIN, a novel lens-specific protein related to the galectin family. J. Biol. Chem. 273:28889–28896, 1998 [DOI] [PubMed] [Google Scholar]
- 286.Barton K.A., Hsu C.D., and Petrash J.M.Interactions between small heat shock protein alpha-crystallin and galectin-related interfiber protein (GRIFIN) in the ocular lens. Biochemistry 48:3956–3966, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Goel R., Murthy K.R., Srikanth S.M., et al. Characterizing the normal proteome of human ciliary body. Clin. Proteomics 10:9, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Nguyen J.V., Soto I., Kim K.Y., et al. Myelination transition zone astrocytes are constitutively phagocytic and have synuclein dependent reactivity in glaucoma. Proc. Natl. Acad. Sci. U.S.A. 108:1176–1181, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Tezel G., Yang X., Luo C., et al. Oxidative stress and the regulation of complement activation in human glaucoma. Invest. Ophthalmol. Vis. Sci. 51:5071–5082, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Fautsch M.P., Silva A.O., and Johnson D.H.Carbohydrate binding proteins galectin-1 and galectin-3 in human trabecular meshwork. Exp. Eye Res. 77:11–16, 2003 [DOI] [PubMed] [Google Scholar]
- 291.Diskin S., Cao Z., Leffler H., and Panjwani N.The role of integrin glycosylation in galectin-8-mediated trabecular meshwork cell adhesion and spreading. Glycobiology 19:29–37, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Diskin S., Chen W.S., Cao Z., et al. Galectin-8 promotes cytoskeletal rearrangement in trabecular meshwork cells through activation of rho signaling. PLoS One 7:e44400, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Colnot C., Ripoche M.A., Milon G., Montagutelli X., Crocker P.R., and Poirier F.Maintenance of granulocyte numbers during acute peritonitis is defective in galectin-3-null mutant mice. Immunology 94:290–296, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]



