Abstract
Tears are highly concentrated in proteins relative to other biofluids, and a notable fraction of tear proteins are proteases and protease inhibitors. These components are present in a delicate equilibrium that maintains ocular surface homeostasis in response to physiological and temporal cues. Dysregulation of the activity of protease and protease inhibitors in tears occurs in ocular surface diseases including dry eye and infection, and ocular surface conditions including wound healing following refractive surgery and contact lens wear. Measurement of these changes can provide general information regarding ocular surface health and, increasingly, has the potential to give specific clues regarding disease diagnosis and guidance for treatment. Here we review three major categories of tear proteases (matrix metalloproteinases, cathepsins, plasminogen activators) and their endogenous inhibitors (tissue inhibitor of metalloproteinases, cystatins, plasminogen activator inhibitors), and the changes in these factors associated with dry eye, infection and allergy, refractive surgery, and contact lenses. We highlight suggestions for development of these and other protease/protease inhibitor biomarkers in this promising field.
Keywords: Matrix metalloproteinase, cathepsin, cystatin, plasminogen activator, dry eye, ocular surface
1. Tears as a Source of Biomarkers
1.1. Tear Production
The ocular surface is covered by a thin fluid named the tear film, which provides the avascular cornea with nutrients, promotes oxygen uptake, provides lubrication for comfort, provides light refraction to the eye, and protects against infection.1 Closest to the ocular surface is a hydrophilic glycocalyx, consisting of cell associated mucins and galectins produced by corneal and conjunctival epithelial cells.2, 3 The mucoaqueous layer contains fluid, proteins and electrolytes secreted primarily by main and accessory lacrimal glands as well as soluble mucins.1 Covering but integrated with the mucoaqeuos layer, there is a thin layer of lipids secreted from the meibomian glands in the eyelid.4 The complex composition of this biofluid is critical to its vital role in ocular surface homeostasis.
The lacrimal gland producing the aqueous component is the principal source of tear proteins. Secretion from the lacrimal gland contributes constitutively to produce basal tears, and provides regulated secretion of proteins and fluid in response to stimulation by corneal sensory nerve fibers, so-called reflex tears.5 The composition and protein content differs between basal and reflex tears.6, 7 The lipid content of reflex tears is reduced,8 while osmolality falls within the same range in both basal and reflex tears.9 Other physiological factors that affect tear composition include prolonged closure of the eyelids, diurnal and seasonal variations.10–13
The total protein concentration of basal tears is high, ranging from 6–11 mg/mL in humans.14 Although the bulk of the tear protein concentration is comprised of just a few proteins (lysozyme, lactoferrin, secretory IgA, serum albumin, lipocalin and lipophilin), close to 1800 other proteins have been identified at concentrations ranging from pg/mL to mg/mL levels.15 A notable fraction of tear proteins are proteases and protease inhibitors, present in a delicate equilibrium which is affected by both local and systemic conditions.16 Proteases, also known as peptidases or proteinases, are enzymes that break down other proteins through enzymatic cleavage of peptide bonds.17 Based on the location of their preferred sites of action within the cleaved target, proteases can be further classified into serine, cysteine, threonine, aspartic, glutamic, asparagine and metallo- proteases. In this review, we sought to analyze what had been reported in the clinical literature on the role of three physiologically important families of tear proteases and their inhibitors in ocular surface disease: matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs); cathepsins and cystatins; and plasminogen activator (PA) and plasminogen activator inhibitor (PAI). These proteins are intimately involved with ocular surface physiology.18–20 Dissociating events associated with their chronic dysregulation from the natural fluctuations in their activity that occur during physiological processes remains a central challenge. We have further highlighted those with the greatest biomarker potential for specific ocular surface diseases versus those more reflective of generalized inflammatory processes.
For this review, we utilized the MEDLINE/PUBMED database from 1957–2019 with general keywords and disease terms as follows in different combinations: tears; tear film; lacrimal fluid; tear fluid; protease; protease inhibitor; MMP; matrix metalloproteinase; tissue inhibitor of metalloproteinase; TIMP; cathepsin; cystatin; stefin; plasminogen activator; plasminogen activator inhibitor; dry eye; contact lens; wound healing; LASIK; PRK; refractive surgery; allergic eye; keratitis; conjunctivitis; keratoconus; and ocular Graft-versus-Host Disease. Our analysis revealed a shortage of review articles in our chosen focus area, although we identified several excellent reviews focused on roles of specific proteases in certain diseases, including MMP-9 in dry eye syndrome and MMPs in keratoconus.21–23 Review articles focusing on tear biomarkers of ocular and systematic diseases were also identified; however their focus on proteases was limited.24–27 In this review, we have not included articles utilizing in vitro systems or animal models, unless these same articles also included a clinical study component. We have also not included articles which highlight other types of tear proteases as putative biomarkers because of a relative lack of critical mass in the literature.
Biofluid samples or so-called “liquid biopsies” are a popular source in the quest for new biomarkers for both local and systemic diseases, as they can be assessed relatively non-invasively, and follow up samples may be obtained to monitor disease progression or responsiveness to treatment. Tears are less complex with regards to the number of different proteins found relative to serum or plasma. In 2014, the plasma protein database contained 10,546 proteins detected in serum/plasma, of which 3784 were reported in two or more studies.28 Tears, in contrast, have 1800 identified species.15 Tear proteins are also more concentrated (6–11 mg/ml)14 relative to whole saliva (1 mg/mL).29 Saliva sampling is further complicated by oral flora, which produce proteases and other proteins that may be unrelated to underlying disease.30, 31 Although, our focus is the relationship between tear proteases, their inhibitors and ocular surface diseases, other studies have revealed that tear composition can shed insights into systemic disease.26, 32–35
Tears may be collected using different techniques, including collection with blunt capillaries held in the lateral tear meniscus or with a tear collection strip with or without topical anesthesia for basal and reflex tears, respectively. Less commonly-used methods include collection with polyester rods, or flush techniques where tears are collected after the addition of an eye drop. 36, 37 Collection using glass capillaries is slow and it can be difficult to obtain sufficient sample volume in subjects with low tear production. Furthermore, patients may feel uncomfortable having a pointed object close to the eye. It is also important to avoid contact with the eyelids and conjunctiva to avoid potential contamination, and reflex tearing (if basal tears are desired). Tear collection using collection strips is generally well accepted by patients, but contamination with epithelial cells may occur.38 Furthermore, recovery from the strip may vary by protein and may also be affected by the elution method used.39 It has long been known that the tear collection method impacts composition and the relative abundance of specific proteins.40–42 One recent example of this was reported in a study comparing collection methods for the analysis of ocular mucins. Collection with glass capillaries, generally considered to represent basal tears, yielded the highest relative concentration of MUC16. In contrast, Schirmer’s strip tear collection without anesthesia, generally considered as reflex tears, yielded the highest MUC5AC concentration.43 A review by Rentka el al. discusses the advantages and disadvantages of the most commonly-used tear collection methods based on the subsequent analysis.44 Unfortunately, there are insufficient studies on the impact of the collection methods on the activity and abundance of the specific proteases and inhibitors discussed in the specific ocular surface conditions of interest here. As the collection method and post-collection processing of the sample may impact the tear composition and results, standardized methodologies would be desirable comparison across studies. As specific proteins are affected differently, such standards may still need to differ depending on the protein of interest.
1.2. Major proteases and protease inhibitors in tears
Historically, proteases in tears were identified based on their enzymatic activity towards specific substrates.45 With the development of antibody-based detection methods including Western blotting and ELISA, and later the development of high throughput mass spectrometry techniques, the identification and quantification of proteases in tears is now often based on detection of protein structure and peptide sequence. The activity of secreted proteases in biofluids is tightly regulated by four principal mechanisms: 1. Gene expression; 2. Environmental factors including pH; 3. Zymogen activation (many proteases are synthesized in inactive pro-enzyme forms requiring post-translational activation); and 4. Inhibition by endogenous inhibitors.46 Therefore, it may be important to measure the quantity and activity of the protease, and/or the quantity of potential inhibitors, if known, to understand aspects of dysregulation. As an example, in studying the cysteine protease, cathepsin S, and its endogenous inhibitor cystatin C, we have shown that the enzymatic activity of cathepsin S is highly increased in the tears of Sjögren’s Syndrome (SS) patients compared to patients with other autoimmune diseases and patients with other forms of dry eye.47, 48 Interestingly, this elevation in activity may be primarily due to the decreased abundance of its natural inhibitor, cystatin C, in tears of this SS patient population.47 Findings on the enzymatic activity of cathepsin S in tears of SS patients, however, when viewed in isolation would appear to be inconsistent with findings by others that elevations in tear cathepsin S protein in SS patients have not been seen in proteomics studies.49, 50
1.2.1. Matrix Metalloproteinases (MMPs) and Tissue Inhibitor of Metalloproteinases (TIMPs)
MMPs are a family of neutral zinc proteases involved in angiogenesis, inflammation, wound repair and tissue remodeling,51 that traditionally have been divided into gelatinases (MMP-2,-9), collagenases (MMP-1, -8, -13), stromelysins (MMP-3, -10– 11) and matrilysins (MMP-7, -26).52, 53 They are synthesized as pro-enzymes and activated by other proteases through a process called the cysteine switch.54 These proteases include: other MMPs, for example MMP-3 can activate proMMP-1, proMMP-8, and proMMP-13;55–57 plasmin, which activates proMMP-1, proMMP-3, proMMP-9, and proMMP-13;58, 59 and cathepsins, for example, Cathepsin K activates MMP-9.60 MMPs are secreted or attached to the outer cell membrane. MMP primary activity is thus against proteins within secretory pathways, membrane proteins, or proteins in the extracellular space/body fluids.46 MMP activity is further modulated by endogenous inhibitors called tissue inhibitor of metalloproteinases (TIMPs).61, 62 The most recognized role of MMPs is in ECM degradation and turnover, however MMPs have non-matrix substrates and can modulate the ECM microenvironment and stimulate cell proliferation through direct regulation of cytokines, growth factors and other proteases.63, 64 MMP-1, MMP-2, MMP-3 and MMP-9 all participate in repair of the corneal epithelium and stroma.65–67 Increased tear MMP activity was first described in patients with failed corneal grafts, but has later been shown in many ocular surface conditions.68
TIMPs are the endogenous inhibitors of MMPs. The four homologous TIMPs (TIMP1, -2, -3 and -4) can inhibit all MMPs, albeit with different efficacy.46 The inhibition is upheld by interaction with the MMP active site. However, TIMPs can regulate ECM turnover through indirect mechanisms, including stimulation through tyrosine kinase and MAP kinase pathways and promotion of cell apoptosis.69 TIMP binding to MMPs does not always result in inhibition, as MMP-2-TIMP-2 interaction results in activation of the enzyme. Although, MMPs have been extensively studied in tears, TIMP and MMP interactions have not been as well-investigated.70
Another regulator of MMP-9 is neutrophil gelatinase-associated lipocalin (NGAL), also called lipocalin-2, that forms a complex with MMP-9 and preserves its activity by protecting it from degradation.71 Although, NGAL has been shown to be present in excess of MMP-9 in tears, and its levels in tears are subject to diurnal variations, it has not been extensively studied in the disease conditions discussed here.10
Levels of MMPs in tears are most commonly measured using either single or multiplex ELISA approaches. However, there are inconsistencies in the ELISA assays used as some are directed towards the pro-form, while others are directed to the active form or to both.72–75 MMP activity is traditionally measured using gelatinase zymography for MMP-2 and MMP-9, or collagenase zymography for MMP-1, MMP-8 and MMP-13. Although zymography only provides semi-quantitative activity measurements, these assays provide additional information about the forms of MMP present.76 For instance, for MMP-9 assayed by zymography, the pro-form can be observed at 92 kD, the active form at 85 kD and the MMP-9/NGAL complex at 135 kD.10 Reverse zymography can also be used to study the endogenous activity of TIMPs.77
1.2.2. Cathepsins and Cystatins
Cathepsins are a diverse group of proteases which include serine cathepsins (A and G), aspartic cathepsins (D and E), and lysosomal cysteine cathepsins (B, C, F, H, K, L, O, S, V, X, W).78 Initially considered as lysosomal proteases, cathepsins are increasingly recognized as key players in the extracellular space. The cysteine cathepsins have collagenase and elastase activity and participate in ECM remodeling.79 These cathepsins can also serve as signaling molecules through activation of protease-activated receptors and processing of cytokines and chemokines.79 One of the first studies of cathepsins in tears identified cathepsin C- and cathepsin B-like activity in healthy human tears.45 However, their role in the tears and ocular surface has not been systematically investigated beyond cathepsin S.19, 47, 48, 80–83
Cystatins. Cystatins have long been known as constituents of human tears and are thought to mediate the activity of cysteine proteases.84 They are categorized into two subgroups: Type I are cytosolic while Type II are found intracellularly and extracellularly.85 Cystatins are associated with cell proliferation, cell migration and cell differentiation.86 Normally, cystatins of the type II category, especially cystatin C, B, S, SN, and SA, are detected in human tears.16
1.2.3. Plasminogen Activators (PAs) and Plasminogen Activator Inhibitors (PAI)
Plasminogen activators (PAs) are serine proteases that convert plasminogen into plasmin, a serine protease identified in the mammalian cornea in 1967.87, 88 Ocular PAs prevent fibrin deposition following inflammation,88, 89 stimulate leucocyte migration and activation of collagenases,90, 91 maintain aqueous humor fluidity;92 and regulate angiogenesis.91 Urokinase-type PA (uPA) and tissue-type PA (tPA) are the main types, with both present in human tears.87, 93 Each PA has different roles, with tPA involved in fibrinolytic processes and uPA involved in cell migration and tissue remodeling.94–96 The lacrimal gland, conjunctival blood vessels and corneal epithelium are sources of tPA, while uPA originates from corneal and conjunctival epithelia.89, 97 uPA and tPA have different activity levels in tears from healthy individuals, with tPA activity higher than uPA activity.93
Plasminogen activator inhibitors (PAI) are serpin-class anti-proteases that inhibit both uPA and tPA. PAI-1 mainly inhibits tPA, while PAI-2 is an effective inhibitor of uPA. PAI-1 and PAI-2 may be present in human tears when either cornea or conjunctiva are affected by disease.98
2. Tear Proteases and Protease Inhibitors in Ocular Surface Diseases
2.1. Dry Eye Disease
Dry eye disease (DED) is a multifactorial disease resulting in discomfort, visual disturbance, and tear film instability and is associated with an increase in tear osmolarity and ocular surface inflammation.99 Dryness is a downstream consequence of this inflammatory cycle, which is associated with increased production of many proteases in ocular surface tissues and tears.100 Balance between protease and protease inhibitors plays an important role in ocular surface homeostasis in DED.101
DED patients can be stratified into those with aqueous tear-deficient dry eye (ADDE) or evaporative dry eye (EDE). Aqueous tear deficiency is caused by insufficient lacrimal gland tear secretion secondary to lacrimal gland destruction or dysfunction, leading to tear hyperosmolarity that can stimulate proinflammatory cascades.99 Lacrimal gland dysfunction in ADDE leads to production of proinflammatory mediators which are delivered to the ocular surface and into tears.99 Aqueous tear deficiency can be subdivided into 2 categories, Sjögren’s syndrome (SS)-associated and non-SS associated ADDE. SS is a systemic autoimmune disease that leads to dryness of many mucosal surfaces, including the eyes and mouth. A majority of SS patients are women while the spectrum of clinical symptoms includes ADDE, interstitial lung disease, renal failure, cryoglobulinaemic vasculitis, infections due to compromised mucosal surfaces, cardiovascular disease and development of B-cell lymphoma.102, 103 Non-SS ADDE is associated with obstruction of the lacrimal gland duct or with age-related DED and occurs in otherwise healthy individuals without autoimmune disease.99
Evaporative DED is characterized by premature evaporation of the tear film in the setting of normal lacrimal gland secretion. Blepharitis is a primary cause of EDE, and can also be categorized into 2 subclasses, anterior and posterior blepharitis.104 Meibomian gland dysfunction (MGD), often used as a synonym for posterior blepharitis, is associated with altered lipid composition in the meibum, and leads to reduced nonpolar lipids in tears and increased tear evaporation.105 These categories are summarized in Figure 1.
Figure 1. Dry eyes disease classification.
Dry eyes diseases (DED) are categorized into evaporative dry eye (EDE) and aqueous deficient dry eye (ADDE). EDE is mainly caused by meibomian gland dysfunction (MGD)/blepharitis. ADDE is primarily based on defective secretion by the lacrimal gland (LG). ADDE is further subdivided into Sjögren’s syndrome (SS) related and non-Sjögren’s syndrome related (non-SS). Image created with BioRender.
The diagnostic criteria for both ADDE and EDE are complicated: symptoms overlap and diagnosis is based on patient symptoms obtained from questionnaires such as the Ocular Surface Disease Index (OSDI) and objective tests such as tear film break-up time (TBUT), corneal fluorescein staining and tear secretion tests.106 Frequently, objective tests do not correlate with patient-reported symptoms, complicating workup of DED.107 Proteases and their inhibitors have the potential for serving as biomarkers for DED because of the correlation of protease dysregulation in the tear film with DED symptoms and pathology.108
2.1.1. MMPs and TIMPs
Changes in MMP levels have been extensively studied in DED. In a study of 46 newly diagnosed DED patients with OSDI levels >20 and exhibiting at least one of several additional indicators of disease, compared to 18 healthy controls, tear film MMP-9 activity was increased, with levels correlated with DED severity as defined by the DEWS criteria.109 Tears from 9 SS ADDE patients also exhibited increased MMP-9 abundance and activity in conjunction with accumulation of the proinflammatory form of IL-1 (IL-1α and mature IL-1β) and decreased inactive IL-1β, relative to 10 healthy control subjects. 110 This finding linked elevated MMP-9 to activation of IL-1β. Levels of MMP-9, MMP-3 (an activator of MMP-9) and TIMP-1 were increased in tears of 15 ocular rosacea patients who met clinical criteria for EDE and MGD, relative to 8 healthy controls.111 The concurrent elevation of TIMP-1 in these ocular rosacea patient tears suggested MMP-9 activation in this case might be related to MMP-3 accumulation.111 Another study from this group in 13 ocular rosacea patients meeting clinical criteria for EDE and MGD versus 13 healthy controls showed that elevated MMP-9 activity in ocular rosacea patients tears, as measured by gelatin zymography, was correlated with high tear IL-1α, an effector of MMP-9 production.112 Levels of pro-MMP-9 measured by ELISA were significantly elevated in tears of 12 blepharitis patients with diagnosis based on clinical symptoms and medical history compared to tears of 18 healthy controls, although the criteria for DED diagnosis in this study were not as well-defined as in other studies.113
The InflammaDry (Rapid Pathogen Screening, Sarasota, FL) is an immunoassay that measures expression of pro-MMP-9 and MMP-9 in human tears with a detection threshold of 40 ng/ml.22 If total MMP-9 expression is ≥ 40 ng/ml, the kit shows a positive result with 2 lines (one blue and the other one red). Schargus et al directly compared MMP-9 concentrations in tears measured by ELISA versus InflammaDry in 20 elderly patients aged ≥ 60 years and previously undiagnosed with DED.114 Of these patients, most met clinical criteria for mild to moderate DED based on OSDI, Schirmer’s test values, corneal staining and TBUT.114 Only 3 of the participants showed positive values for MMP-9 by InflammaDry (> 40 ng/mL) while the other 17 participants with negative InflammaDry test values showed mean ELISA MMP-9 value of 7.1 ng/ml.114 InflammaDry results in this study were correlated with tear MMP-9 levels measured by ELISA, with no false positives detected, suggesting that the InflammaDry test results were accurate in this small cohort of human tears.114 However, the test distinguished only DED patients with high MMP-9 levels in this study.
In a prospective multicenter clinical trial conducted in 143 DED patients meeting clinical criteria of OSDI >7, Schirmer’s test <10 mm, TBUT <10 sec and detectable keratoconjunctival staining versus 63 healthy controls, this assay showed 85% sensitivity and 94% specificity.115 In addition, a positive MMP-9 signal measured by the InflammaDry was correlated with DED in 47 patients meeting 3 out of 4 of the following clinical diagnostic criteria: OSDI>12 (P = 0.001), TBUT≤5 (P < 0.013), Schirmer’s test ≤10 (P < 0.001) and corneal staining >1 (P = 0.007), relative to 54 healthy controls.116 This study also linked meibomian gland obstruction and increased pathological features of meibomian gland secretion with a positive MMP-9 signal with the InflammaDry.116 While analysis of MMP-9 levels appears to have diagnostic utility in DED workup, particularly in identifying patients with significant underlying inflammation, this test is unable to distinguish between different DED states.
2.1.2. Cathepsins and Cystatins
Cathepsin S is globally involved in initiating inflammatory responses. It is an established effector of lysosomal protein degradation and ECM degradation, functions essential for cellular homeostasis and differentiation.117 In addition, cathepsin S mediates major histocompatibility complex (MHC) class II-antigen presentation by cleavage of invariant chain.118, 119
In two studies utilizing two independent cohorts of primary and secondary SS patients, respectively, tear film cathepsin S activity was significantly higher in SS patients relative to those with non-SS DED, other autoimmune diseases without DED, or healthy controls.47, 48 Increased tear film cathepsin S activity may affect the tear quality in SS patients since it promotes the degradation of other tear proteins such as cystatin C, lactoferrin, and secretory IgA.47 Although cathepsin S may discriminate between SS and non-SS ADDE, the possibility of utilizing it to distinguish between non-SS ADDE and EDE has not been evaluated.
Cystatin C is an endogenous cathepsin S inhibitor which interferes with antigen processing by regulating cathepsin S, and which also interacts with numerous inflammatory mediators to regulate inflammatory processes at the molecular level.85 Decreased tear cystatin C, as measured by ELISA, was found in SS patient tears in one of the two studies evaluating identifying increased cathepsin S activity, which compared 33 primary and secondary SS patients diagnosed based on clinical signs and symptoms and Ro/SSA autoantigen seropositivity, to 35 patients with non-SS ADDE and EDE, 33 with rheumatoid arthritis, 31 with other autoimmune diseases, and 24 healthy controls.47 The ratio of cathepsin S to cystatin C contributing to elevated enzymatic activity of cathepsin S may reflect the severity of ocular surface inflammation in SS, more so than actual cathepsin S protein.47
2.2. Infection and Allergy
The tear film and conjunctiva are the eye’s physical barrier against insults such as allergens, chemicals and infectious agents. The conjunctiva is the most immunologically-active component of the ocular surface and consists of two layers: epithelium and substantia propria. The epithelium is stratified with several layers of epithelial cells, and is invested with goblet cells, Langerhans cells and lymphocytes. The substantia propria also contains multiple immune cell including mast cells, eosinophil and basophils. Upon exposure to irritants, the eyelids and tear film provide a first line of defense through the blink reflex and increased tear flow. The conjunctiva contributes anti-microbial and anti-infectious factors to the ocular surface and tears, while inflammatory activators are also released to regulate immune responses.120 Both corneal (keratitis) and conjunctival (conjunctivitis) inflammation are caused by allergic or infectious triggers and in both cases, tear proteases are altered.
2.2.1. MMPs and TIMPs
Changes in MMPs are linked to conjunctivitis and keratitis, due to their role in remodeling of ECM and their modulation of inflammation. Vernal keratoconjunctivitis (VKC), a chronic allergic eye disease, leads to conjunctival tissue remodeling due to excessive ECM deposition, subepithelial fibrosis, chronic inflammatory cell infiltration and epithelial thickening and corneal erosion.73 A small study utilized gelatin zymography to evaluate MMPs in tears of 6 individuals with active VKC, 14 with active allergic conjunctivitis and 6 healthy non-allergic volunteers. Tears of VKC patients contained both pro- and active MMP-2 and MMP-9, while healthy patient tears contained only pro-MMPs and only a subset of patients with allergic conjunctivitis showed active MMPs in tears.121 In a study of 16 patients with active VKC who were free of treatment for 3 days, increased MMP-1 and MMP-9 levels in tears were detected by ELISA in conjunction with increased TIMP-1, relative to 10 age-matched healthy control individuals. However, the increase of MMP-1 and MMP-9 exceeded TIMP-1 increases, causing elevated MMP-1/TIMP-1 and MMP-9/TIMP-1 ratios.73 In another study of 24 patients with active VKC, VKC patient tears showed a higher frequency of detection of MMP-1, MMP-2, MMP-3, MMP-8, MMP-9 and MMP-10 compared to tears of 12 healthy control individuals.122 Further, in a study of 26 patients with atopic blepharoconjunctivitis versus 26 healthy control individuals, MMP-8 concentration was significantly elevated in tears of patients with atopic blepharoconjunctivitis.123 This protein was mainly found localized in inflammatory cells within in the conjunctiva.123 MMP-8 protein concentration and activity was increased in tears of 26 consecutive patients with non-allergic eosinophilic conjunctivitis compared to 26 asymptomatic controls. This elevation was also correlated with increased conjunctival neutrophils (ρ=0.66, P=0.0002).124
Herpes stromal keratitis (HSK) is a blinding disease caused by herpes simplex or zoster virus infection. Tissue injury and vision loss results from host immune responses, rather than viral infection. The balance between MMPs and TIMPs may have a role in necrotizing HSK. A small study by Smith et al. measuring tear MMPs by zymography in 4 HSK patients and 10 healthy controls showed in the HSK patient tears that MMP-9 activity was ~4.5-fold higher.125
A study evaluating MMP-9 activity in tears of 25 patients with fungal keratitis, 7 patients with bacterial keratitis and 3 with viral keratitis relative to 10 healthy control subjects found that 72% of fungal keratitis patients had detectable tear MMP-9 activity, 71.4% of patients with bacterial keratitis had detectable tear MMP-9 activity and 33.3% of patients with viral keratitis had detectable tear MMP-9 activity compared to tears from healthy controls.126
2.2.2. Cathepsins and Cystatins
Analysis of reflex tears of patients with Fusarium keratitis found differential expression of cystatins. In this study, multiple samples were collected from 35 male individuals aged 20–60 years at early (within 7 days), intermediate (7–14 days) and late (after 14 days) stages of infection, relative to tears from 20 healthy individuals. In each case, 5 samples from each category were subjected to 2D-difference gel electrophoresis (2D-DIGE) and subsequent proteomics identification of species of interest. Tear cystatin SA was significantly reduced in early to late stages of infection in patients with Fusarium keratitis.127 In another study utilizing pooled reflex tear samples from 16 Fusarium keratitis patients and 16 Aspergillus keratitis patients, versus 24 healthy control individuals, analyzed by two-dimensional electrophoresis and MALDI-TOF mass spectrometry, several cystatin precursors were decreased in tears from both Aspergillus and Fusarium keratitis patient tears.128
2.2.3. PAs and PAIs
Changes in levels of PAs are linked to keratitis and conjunctivitis. 10 of 12 patients with superficial keratitis showed elevated uPA activity in tears relative to activity levels in 18 healthy control subjects.129 In a study of 20 patients with active VKC compared to 19 age-matched healthy controls, tear uPA and tPA levels were significantly increased in VKC patient tears, although PAI-1 was undetectable in tears of both populations. Tear plasminogen activity in a subset of 10 VKC patients (4.7%±3.4%) was also greater than an equivalent number of tear samples selected from healthy controls (0.65%±0.83%), while immunohistochemistry evaluations showed that uPA and uPA receptors (uPAR) were elevated in conjunctival epithelium and stroma of VKC patients relative to levels in healthy controls. Overall, tear tPA levels were correlated with the severity of corneal injury (ρ=0.466, P < 0.04) in VKC.130
2.2. Refractive Corneal Surgery
Corneal wound healing involves multiple steps such as inflammation, myofibroblast differentiation, ECM deposition and scar formation. The corneal epithelium, stroma and endothelium each may be affected and react differently to injury.131–133 In deep corneal injuries where basement membrane is disrupted and wounding extends into the stroma, a fibrotic response can be activated which can lead to corneal scarring and opacification. Maintaining the appropriate balance of growth factors, cytokines, proteases and ECM signaling is essential to restore corneal integrity and maintain homeostasis. With increasing numbers of refractive surgeries performed, corneal wound healing has become a clinically-significant issue. Thus, we included this topic in the list of ocular surface conditions linked to protease dysregulation. Laser in situ keratomileusis (LASIK) is the most commonly performed vision correction surgery in the United States. Although generally successful, LASIK can generate complications including abnormal wound healing, flap detachment, and ectasia at a frequency of ~2%.134 Photorefractive keratectomy (PRK) is another popular laser refractive surgery option. PRK differs from LASIK by ablating the corneal surface without creating a flap.135 A main complication of PRK is corneal subepithelial haze, and PRK patients are more likely to suffer from corneal light scattering than LASIK patients.136
2.3.1. MMPs and TIMPs
During corneal wound healing, many MMPs are upregulated to regulate cell migration by facilitating ECM degradation and modifying cell adhesion. Due to their intensive involvement, their utility as potential biomarkers for surgical wound healing has been proposed. A study comparing tear inflammatory mediators in 62 myopic patients undergoing treatment with orthokeratology (OK) contact lens or LASIK treatment found that of the 30 patients who had undergone LASIK, significant elevations of MMP-9 as measured by ELISA were detected in tears relative to levels in 30 healthy control subjects at a mean window of 10.8 months post-LASIK.137 In the affected eyes of 12 patients with LASIK ectasia, significant elevations in tear MMP-9 and decreases in TIMP-1 as measured by ELISA were found, compared to tears from the affected eyes of 25 patients with uncomplicated LASIK surgery or the tears from eyes of 25 healthy controls.138 The relationship between elevated MMP activity and LASIK complications may be complicated by individual patient predisposition to ectasia. For example, patients with keratoconus where MMP dysregulation is linked to disease pathogenesis are at higher risk for development of post-LASIK ectasia.139
DED associated with LASIK surgery is one of its most common complications, attributable to neurotrophic effects, damage to goblet cells, altered corneal shape, and tear film instability following surgery.140 The TFOS DEWSII report also identified surgical induced iatrogenic dry eye as an entity.141 A study evaluated multiple aspects of ocular surface integrity following bilateral LASIK surgery in a patient population that was rigorously prescreened and excluded from surgery if indications for DED and corneal abnormalities were detected. In these LASIK patients, the OSDI score was significantly increased and TBUT decreased in conjunction with a 10-fold increase in tear MMP-9 levels measured by sandwich ELISA, compared to age-matched healthy controls.142 Study participant tears were also assayed using the InflammaDry test in parallel. Despite measurement of significantly elevated MMP-9 levels by ELISA, only half of the patients showed a positive signal with InflammaDry. The InflammaDry assay may thus have a threshold of detection too high to detect clinically-significant increases in MMP-9 post-LASIK.
MMPs also appear to participate in corneal wound healing after PRK.143–145 Tears of 24 patients and 21 healthy control subjects were collected one day post-PRK for evaluation of MMP-8, and in parallel, tears of 13 patients and 10 healthy control subjects were collected one day post-PRK for analysis of MMP-8-activating membrane type-1-MMP.146 In post-PRK patients, tear fluid flow-corrected active and latent MMP-8, together with MMP-8-activating membrane type-1-MMP (MT1-MMP) levels were significantly elevated. These results suggest an increase in MMP-8 synthesis and activation in tear fluid in response to PRK. However, a study limitation was that MT1-MMP and MMP-8 were not measured simultaneously in the tear fluid of the same patients, and the role of TIMPs were not elucidated. Finally, a six-month long follow-up of tear MMP-9 and chondroitin sulfate levels (which regulate MMP expression and activity) in a single 20-year old patient post-PRK utilizing microcapillary tube collection showed that MMP-9 activity was elevated 1-day post-surgery and decreased after day 4.147 At the same time, a form of chondroitin sulfate dramatically increased within a month, decreased from months 1–3 and increased again after 3 months. While the study was limited to a single patient, these temporal changes suggest complex interactions between MMPs and other factors during sustained corneal wound healing. With global significance of MMP upregulation in LASIK and PRK remaining unclear, MMP expression profiles may still serve as indicators for treatment response and identify patients at risk for complications such as ectasia.
2.3.2. Cathepsins and Cystatins
Zhou et al. used iTRAQ quantitative proteomics to assess tear protein changes after LASIK surgery. Tears were collected from the affected and contralateral eyes of 22 patients at 1 week and 3 months after the procedure. Of 824 proteins quantified, the cystatin SN precursor increased by two-fold, while cathepsin B was downregulated by 0.64-fold one week following surgery.148 Further exploration of these changes, including the actual activity of the affected cathepsin B protein, under these conditions is warranted.
2.3.3. PAs and PAIs
PA activity is altered in individuals with corneal ulcers, epithelial defects, recurrent erosions and chemical burns.129 PA activity results in generation of plasmin, an effector protease, involved in fibrinolysis, ECM degradation, cell migration, tissue remodeling, wound healing, angiogenesis and inflammation.149 In a study of 16 patients with myopia treated with PRK, analysis of pre- and post-operative tears revealed a significant increase in flow-corrected plasmin excretion rate in tears (median 40.0 μIU/min range 13.3 to 222.8 μIU/min vs. median 1.6 to 41.5 μIU/min) and decrease in tear plasmin activity (median 0.65 IU/l range 0.6 to 1.49 IU/I vs. median 1.29 IU/I range 0.6 to 6.9 IU/I) in the first 2 days post-PRK. 150 Both factors were restored to baseline by day 7. In a study comprising 86 patients undergoing PRK for correction of myopia at two different ablation depths, who were randomly assigned to three different treatment groups post-PRK, Brart et al. evaluated the effects of acute treatment with the plasmin inhibitor, aprontinin versus sustained treatment with 0.1% fluorometholone or no treatment for up to a year post-surgery. No beneficial effects on refractive outcomes were seen with aprontinin and a greater haze occurred in treatments correcting more than 6.00 diopters.151 In a study of 42 patients experiencing bilateral PRK surgery with an interval of 1–2 weeks between surgeries and monitored for uPA activity in tears collected with glass capillaries, uPA activity was decreased immediately post-surgery, increased dramatically on day 3, and returned to baseline by day 5.152 However, in patients with abnormal corneal healing and haze development, uPA activity remained low through the third postoperative day.152 Many studies have reported on similar uPA activity patterns in rabbits after PRK, and explored the relationship between tear uPA and corneal haze development in rabbit models.153, 154
PAI levels may also be affected by PRK and LASIK. Levels of PAI-1 and PAI-2 in tears were assessed before and after surgeries in 46 eyes of 38 patients receiving PRK (8 patients with bilateral PRK) and 13 eyes of 8 patients with LASIK (5 patients with bilateral LASIK).155 Only PAI-2 was upregulated in patient tears immediately post-PRK and LASIK, while returning to basal levels within a day. However, no correlation was observed between corneal haze and increased PAI-2 levels in this study.
Proteases including MMPs and PAs, as well as protease inhibitors such as cystatins are globally associated with wound healing and tissue remodeling processes. Abnormalities in tears are reported post-LASIK, with both TBUT and basal tear secretion decreasing after surgery. Surgical injuries to the epithelium increase tear cytokines and tear hyperosmolarity, which in turn trigger stress-activated kinases, other proinflammatory cytokines, MMPs and ultimately induce ocular surface inflammation.156 DED post-LASIK may drive some elevations in tear MMP-9. However, another study found similar tear MMP-9 levels in patients with LASIK-induced DED and asymptomatic patients post-LASIK,142 suggesting that some elevated tear MMP-9 may also be due to prolonged corneal wound healing associated with LASIK.
2.4. Contact Lenses
Contact lenses (CL) contribute greatly to risk of ocular infection, DED and other ocular complications.157–159 In a large study of 393 CL wearers, 213 glasses wearers and 287 individuals without refractive correction, DED incidence as determined by a self-reported questionnaire was significantly higher in the CL group than in glasses wearers and those without refractive correction.160 A cross-sectional study of 730 subjects utilizing a self-reported questionnaire identified the most common reasons for discontinuing CL wear as CL discomfort and dryness.161 Symptoms of lens-related dryness result from increased tear film thinning by evaporation or tear film dispersion, resulting in increased tear film osmolarity.162 Increased osmolarity associated with CL affects the expression of inflammatory mediators in the ocular surface.163 In addition, microbial keratitis, frequently associated with CL wear, can alter tear proteins.158 A large clinical multi-site study of 500 full-time hydrogel CL wearers showed that 55% of CL wearers displayed corneal staining while 8% of this group had severe ocular surface damage.164 Corneal damage from CL can also alter the tear proteome. Some of the changes in tear protease and protease inhibitor composition in CL wearers are discussed below, although the significant incidence of DED and infection in CL wearers means that many CL wearers conform to patterns already described.
2.4.1. MMPs and TIMPs
As noted already, MMP-9 elevations are linked to DED, infection and other ocular surface conditions. In a study of 20 myopic patients wearing rigid gas-permeable CL and with good CL tolerance versus 20 myopic non-CL wearers, increased tear MMP-9 as measured by ELISA was detected in capillary tears in CL wearers relative to those without CL wear.165 Orthokeratology (OK) uses reverse geometry CL to temporarily correct corneal curvature in myopia by having the patient wear the CL overnight.166 In 32 patients utilizing OK for 12 months, higher tear MMP-9 as measured by ELISA was seen compared to equal numbers of healthy age- and sex-matched individuals without CL wear.167 The increased MMP-9 was associated with the degree of myopia to be corrected and also the presence of corneal fluorescein staining, suggesting that it may participate in the tissue redistribution and epithelial cell migration induced by OK.167 The mechanisms associated with corneal erosion in CL wear and the possible relationship(s) to changes in MMP-9 levels or its activity are still unclear.159, 168, 169 Interpretation of data is complicated by diurnal variations in MMP-9 levels10 and inclusion of study cohorts with both extended wear versus daily wear CL.170 However, increased MMP-9/TIMP-1 ratios may predispose CL-wearing individuals to corneal erosions.170
2.4.2. Cathepsins and Cystatins
Several studies have shown cystatin abnormalities in tears of CL wearers. In a study of 13 soft CL wearers, 13 rigid CL wearers and 13 individuals without CL, cystatin SN analyzed from tears collected on Schirmer’s strips by ProteinChip SELDI-TOF (surface-enhanced laser desorption and ionization in time of flight mass spectrometry) was upregulated in rigid gas permeable CL wearers while, it was downregulated in soft CL wearers after 1 year of use relative to individuals without CL.162 In a study of 8 rigid gas permeable CL wearers, 9 soft CL wearers and 11 individuals without CL, Cystatin D, measured from tears collected with Schirmer’s strips, was decreased in both rigid gas permeable and soft CL wearers with 1 year of use compared to individuals without CL.171 It also remained decreased even after subjects stopped using CL for 4–5 days. In this study, subjects wearing rigid gas permeable CL had higher tear cystatin C than soft CL wearers.171 Collectively, these data suggest that soft CL wearers may have decreased cystatin inhibitor capacity in tears relative to those wearing rigid CL.
2.5. Other Ocular Surface Diseases
Keratoconus is a corneal ectasia associated with progressive thinning of the corneal stroma linked to degradation of corneal collagen.172 Keratoconus leads to irregular astigmatism due to loss of corneal shape from increased curvature of the cornea, resulting in loss of visual acuity.172 The cause of keratoconus is largely unknown, but it is of multifactorial etiology.173–179 Dysregulation of MMPs is of great interest in keratoconus pathophysiology. In a study of 34 subjects with diseases of the anterior segment (7 patients with keratoconus with atopy, 7 keratoconus patients without atopy, 7 patients with non-herpetic keratitis, 4 patients with herpetic keratitis, 4 patients with SS ADDE and 5 patients with non-SS ADDE) and 11 healthy control subjects, Smith et al125 showed that detectable activity levels of MMP-9 and an unidentified MMP of MW 116K measured by zymography were present in the tear film of all study groups. Furthermore, MMP-9 activity levels were elevated in all patient groups relative to healthy controls except keratoconus patients without atopy. Another study reported increased gelatinase (19 patients) and collagenase (15 patients) in tears of untreated keratoconus patients compared to healthy controls (17 and 16 healthy controls, respectively). However, the gelatinase (13 patients) and collagenase activity (11 patients) in patient receiving collagen cross-linking treatments were not significantly different from either untreated keratoconus patients or controls.180 Recent studies have focused on measurement of levels of pro-MMP and active-MMP and/or total MMP protein levels in tears, rather than actual protease activity. Using a proteomics approach in a study population of 20 healthy controls who were CL wearers, 18 subjects with keratoconus wearing CLs, and six subjects with keratoconus (non-lens wearers), Pannebaker et al. found increased levels of MMP-1 in keratoconus patient tears compared to healthy controls, with the increase independent of CL wearing.181 Similarly, in the same study discussed above reporting collagenase and gelatinase activity in keratoconus tears, Balasubramanian found that tear levels of MMP-1 were increased in keratoconus patients using an antibody array approach180, while these levels were reduced with collagen cross-linking treatment.
Studies have shown mixed results regarding whether other MMPs such as MMP-3 and MMP-7 are affected in tears of keratoconus patients.180–182 MMP-9 changes are the best characterized in keratoconus, with elevated levels confirmed by several independent studies using activity and antibody based approaches.125, 183–185 However, neither the Pannebaker nor Balasubramian studies referenced above detected differences in MMP-9 tear levels between keratoconus patients and healthy controls.180, 181 Several reviews have been published specifically on MMPs in keratoconus.23, 186 Beyond MMPs, in a study of 36 keratoconus patients and 18 healthy controls, Balasubramanian et al showed a threefold increase in the abundance of tear cathepsin B and a two-fold decrease in cystatin S and cystatin SN using proteomics analysis of capillary tears of keratoconus patients compared to healthy controls.187 Additional reviews in the keratoconus area reflect interest in other proteases/protease inhibitors that are potentially dysregulated.23, 186, 188, 189 Clinically, the diagnostic utility of protease dysregulation in keratoconus may have greatest value by identifying patients at risk of post-LASIK ectasia prior to their development of the characteristic thinning and curvature of the cornea.
Graft-versus-host disease (GVHD) is a systemic complication that can be a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT).190 GVHD can affect the skin, gastrointestinal system, liver, lung, oral mucosa, and eyes.191 40% - 60% of allo-HSCT recipients and 90% of patient with chronic GVHD develop ocular GVHD.190 Donor-derived CD4+ and CD8+ T cells are primary effectors of ocular GVHD pathogenesis.192 DED symptoms are hallmarks of ocular GVHD, which is associated with inflammatory damage and ECM degradation of the tissues of the ocular surface.192 Therefore, the proteases and protease inhibitors which are found in DED patient tears are also altered in ocular GVHD patient tears. A study of 20 allo-HSCT patients identified 8 with ocular GVHD based on Schirmer’s test values ≤ 5 mm and use of treatment for keratoconjunctivitis sicca. In tears of these patients, MMP-9 levels measured by ELISA were increased compared to levels in non-ocular GVHD patients and healthy controls.193 Another study from Arafat. et al found that MMP-9 and MMP-8 levels were increased and TIMP-1 decreased, as measured by multiplex ELISA, in tears of 14 ocular GVHD (defined based on OSDI >23, Schirmer’s test ≤10 mm, and corneal fluorescein staining ≥4) compared to tears of healthy controls.194 A study comparing tears of 10 patients with ocular GVHD (defined based on the severity of their ocular impairment, threat to visual acuity, need for treatment, and undefined parameters measuring corneal staining, Schirmer’s test, OSDI) versus GVHD patients without ocular symptoms found significant decreases in cystatin S and cystatin SN in tears of ocular GVHD patients.195
3. Discussion
Tear biomarkers have potential use in diagnostics, prognostics, pharmacodynamic response, patient stratification and risk evaluation. Proteases and protease inhibitors are attractive candidates as potential tear biomarkers, due to their abundance in tears, sensitivity for detection in assays and relationship to disease pathophysiology. The aqueous component of the tear film is rich in proteases and protease inhibitors. These proteins are produced actively by tissues contributing to aqueous tears to maintain ocular surface homeostasis, and in response to challenges like dryness, infection and wounding. Proteases in tears have potent actions on the cornea including activation of signaling pathways, cleavage of proinflammatory cytokines and other tear proteins, and remodeling of the underlying stroma. These actions occur physiologically, but when they are prolonged or dysregulated, they may reflect disease pathogenesis. In turn, the proteases produced in corneal and conjunctival tissue in response to wounding and inflammation can be detected in tears as indications of homeostatic and/or pathogenic processes on the ocular surface. The relationship between tear composition and ocular surface integrity is interdependent. Tear protease dysfunction can disrupt ocular surface integrity, expose and sensitize pain-sensing nociceptors, and induce immune responses through T-cell recruitment to the ocular surface. Conversely, proteins upregulated in the cornea in ocular surface disease may directly contribute to alterations in tear composition. Figure 2A and B demonstrate the intricate interrelationships between tear composition and corneal integrity by illustrating the changes associated with two ocular surface conditions, DED and corneal wound healing, respectively.
Figure 2. Proteases and protease inhibitors in the tear film and the cycle of homeostasis/pathogenesis.
Proteases and protease inhibitors maintain ocular surface and tear film homeostasis. A. Dry Eye Disease (DED). Various conditions induce elevated tear osmolarity, production of proinflammatory cytokines and proteases in tears. High tear osmolarity and cathepsin S may increase the expression of proinflammatory cytokines and MMP-9 in corneal epithelium. Elevated tear cytokines may further upregulate proinflammatory cytokines, cathepsin S and MMP-9 which (1) cleave epithelial junctional complexes, accelerate apical corneal epithelial detachment, expose subapical epithelium and; (2) activate sensing nociceptors. Inflammatory cytokines and MMP-9 facilitate the activation of dendritic cells and their migration to the lymph nodes (afferent arm). In lymph nodes, dendritic cells present antigen to CD4+ T cells, which can be recruited to the ocular surface (efferent arm). Cytokines produced by CD4+ T cells feedback to affect epithelial cell survival and differentiation. B. Corneal Wound Healing. Corneal conditions such as wounding through refractive surgery give rise to alterations in tear composition. MMP-2, MMP-9 and uPA are upregulated in corneal epithelium and contribute to epithelial migration by (1) cleaving epithelial junctional complexes and (2) degradation of ECM. Cytokines in the epithelium also facilitate wound healing by inducing apoptosis in epithelial cells. IL-1α and TGF-β secreted by the epithelium activate fibroblasts (keratocytes) and trans-differentiation of myofibroblasts. (3) Myofibroblasts secret ECM and MMPs, including MMP-1 and MMP-3, which further facilitate stromal healing and ECM remodeling. (4) Plasmin and PAs modulate normal wound healing processes through ECM remodeling, fibrin degradation and pro-MMP activation. Corneal wounding is accompanied by inflammation, upregulation of proinflammatory cytokines and immune cell recruitment leading to increased tear uPA, MMP-8 and MMP-9. Image created with BioRender.
Table 1 summarizes the common changes in the families of proteases and protease inhibitors of focus in this review in tears across multiple ocular surface diseases, refractive surgery or CL use. As highlighted, a general trend in upregulated tear MMPs is observed across conditions sharing an inflammatory component. MMP-9, in particular, is elevated in DED, infectious conditions, allergic eyes, after refractive surgery and in CL users. Pflugfelder’s group also reported increased tear MMP-9 in patients with dysfunctional tear syndrome.109 Therefore, MMP-9 abundance and/or activity alone is not a stand-alone diagnostic tool due to its lack of specificity in ocular surface diseases. However, findings of elevated MMP activities or their prolonged expression in tears may serve as a general tool to indicate delayed or complicated wound healing, identify unresolved infectious processes, reinforce a DED diagnosis, or identify at-risk patients prior to LASIK.
Table 1:
Changes in tear proteases and protease inhibitors associated with ocular surface diseases and conditions.
| Diseases/Conditions | Proteases/ Protease Inhibitors | |||
|---|---|---|---|---|
| Common | Possibly Distinct | |||
| Protein | Activity | Protein | Activity | |
| Aqueous tear-Deficient Dry Eye Disease | MMP-9115,116↗ | MMP-9109↗ | ||
| Sjögren’s Syndrome Aqueous tear-Deficient Dry Eye Disease | MMP-9110 ↗ | MMP-9110↗ | Cystatin C47↘ | Cathepsin S47,48↗ |
| Evaporative Dry Eye Disease | MMP-9111,113,116↗ MMP-3111↗ TIMP-1111 ↗ |
MMP-9112↗ | ||
| Herpes stromal keratitis | MMP-9125↗ | |||
| Bacterial Keratitis | MMP-9126↗ | |||
| Fungal Keratitis | Cystatin S 128↘ Cystatin SN128 ↘ Cystatin SA127↘ |
MMP-9126 ↗ | ||
| Vernal keratoconjunctivitis | MMP-173 ↗ MMP-973,122↗ TIMP-173↗ |
MMP-2121↗ MMP-9121↗ |
uPA130 ↗ tPA130 ↗ |
Plasminogen130↗ |
| Atopic Blepharoconjunctivitis | MMP-8123↗ | MMP-8123↗ | ||
| Non-allergic Eosinophilic Conjunctivitis | MMP-8124↗ | MMP-8124↗ | ||
|
Laser In Situ Keratomileusis Surgery |
MMP-9137↗ PAI-2158↗↘ Cystatin SN148↗ In LASIK ectasia patients MMP-9138↗ TIMP-1138↘ In LASIK induced iatrogenic dry eye patients MMP-9142↗ |
Cathepsin B148↘ | ||
|
Photorefractive Keratectomy Surgery |
MMP-8146↗ PAI-2155 ↗↘ |
MMP-9147↗↘ | MT1-MMP146↗ | uPA152↘→ (Corneal haze) |
| Contact lens | MMP-9165,167↗ Cystatin SN162↗ (Rigid CL) Cystatin SN162↘ (Soft CL) Cystatin D171↘ Cystatin C171↗ (Rigid > Soft CL) |
|||
| Keratoconus | MMP-1181↗ MMP-9183–185↗ Cystatin S187↘ Cystatin SN187↘ |
MMP-9125↗ | Cathepsin B187↗ | |
| Graft-versus-host disease | MMP-8194↗ MMP-9193,194↗ TIMP-1194↘ Cystatin S195↘ Cystatin SN195↘ |
|||
Factors which may change more specifically in particular disease states may have greater utility as potential diagnostic biomarkers and to guide clinicians toward a particular treatment course. Proteases and protease inhibitors that may have more specificity in their association with individual ocular surface diseases or conditions are also highlighted in Table 1. The particular combination of CTSS and its endogenous inhibitor, cystatin C, shows the most distinctive change in protease and inhibitor composition seen thus far in the study of DED. However, more work is needed to evaluate whether this marker can be specific in distinguishing SS patients from those with EDE and non-SS ADDE. Likewise, uPA and tPA appear to be consistently and significantly elevated in VKC, but there is a lack of information on the corresponding PAIs that may be important in modulating this activity, and on PA/PAI ratios. Decreased Cathepsin B protein showed specificity in LASIK patients in correlating with rapidity of recovery from the procedure in a single study which utilized proteomics analysis, but without any corresponding evaluation of enzyme activity levels which would be a critical next step. Decreased uPA activity has repeatedly been associated with corneal haze, but better rigor in the association of its activity with PAI inhibitor levels may be needed to fully understand the ramifications and potential usefulness of this change. It is finally important to note that changes in MMPs have been explored in tears for several decades, and the apparent initial specificity seen with some of the “unique” protease/protease inhibitors highlighted may not be upheld if they are more extensively explored in tears of patients with different ocular disease states. Still, like MMPs, they may be further developed as powerful tools to investigate the extent of inflammation, identify delayed wound healing, and/or aid in predicting prognosis or guiding treatment.
We have mentioned throughout the review the general challenges that must also be addressed in advancing any of these protease/protease inhibitor biomarkers to clinical use. To the individual limitations discussed above, we must add adoption of a uniform tear collection modality for a particular protease of interest, and the concurrent assay of protease activity and abundance and/or its protease and protease inhibitor abundance. As well, in many disease areas, more rigorous patient inclusion and exclusion criteria will improve comparison of results across studies. Study inclusion criteria vary the most in the DED literature in studies evaluating ocular surface conditions including a component of DED (ocular GVHD, DED associated with CL wear, DED associated with LASIK) and are less variable for conditions such as keratoconus and ocular infection.
Both general and specific protease/protease inhibitor combinations are potentially powerful and easily accessible pharmacodynamic indicators of response to therapies. These can thus be of significant value in improving treatments for ocular surface diseases. Some studies have followed MMP-9 in human tears to monitor ocular surface disease in response to treatment, as a general inflammatory marker. MMP-9 in tears of 16 MGD patients with posterior blepharitis tears was reduced after 1% of topical azithromycin for 4 weeks, retaining a 70% reduction compared to pretreatment even after the drug was stopped for 4 weeks.196 MMP-9 in DED patients tear was also reduced with 0.05% cyclosporine treatment.197 Also, MMP-9 levels in tears were used to evaluate the efficacy of succinylated collagen bandage lenses on several corneal healing conditions including corneal ulcers, recurrent corneal erosions, DED, and corneal lesions in a limited patient cohort. 198 After succinylated collagen bandage lenses treatment, patients in all groups showed significant reduction of pain, redness, irritation, foreign body sensation, and increased watering. Tear MMP-2 and MMP-9 were reduced after 7 days of treatment and further decreased to minimal levels after 21 days of treatment.198 Finally, a large study of 94 keratoconus patients found that tear levels of MMP-9 were decreased in parallel with improved corneal curvature after 6 months of treatment with 0.05% cyclosporine.185 These results collectively illustrate the potential for using tear MMP-9 as a general pharmacodynamic indicator of ocular surface inflammation. This utility may be similarly employed for other protease/protease inhibitor combinations in Table 1.
In conclusion, some changes in protease and protease inhibitor composition, such as MMP-9 and its effectors, already provide important information on ocular surface recovery from injury and/or its state of inflammation. Other changes in proteases and inhibitors such as cathepsin S/cystatin C or uPA/PAI may eventually represent more specific biomarkers which can improve patient stratification into disease subgroups, as in DED, or reflect a higher risk of adverse outcomes following PRK, respectively. Continued research in this area in expanded patient cohorts is critical in bringing forward the best candidates for clinical use, as well as developing accessible clinical tests for rapid measurement of protease/protease inhibitor levels and/or protease activity for biomarkers of interest.
Acknowledgments
Support and Disclosures: This work was supported by NIH RO1 grants EY011386 and EY026635 to SHA. There are no disclosures.
Abbreviations
- ADDE
Aqueous tear-deficient Dry Eye
- APC
Antigen Presenting Cell
- CL
Contact Lens
- DED
Dry eye disease
- ECM
Extracellular Matrix
- EDE
Evaporative Dry Eye
- GVHD
Graft versus host disease
- HSK
Herpes Stromal Keratitis
- LASIK
Laser in Situ Keratomileusis
- MGD
Meibomian gland disease
- MMP
Matrix Metalloproteinase
- NGAL
Neutrophil Gelatinase-Associated Lipocalin
- Non-SS
Non Sjögren’s Syndrome
- OK
Orthokeratology
- OSDI
Ocular Surface Disease Index
- PA
Plasminogen activator
- PAI
Plasminogen Activator Inhibitor
- PRK
Photorefractive keratectomy
- PUK
Peripheral ulcerative keratitis
- SCBL
Succinylated collagen bandage lenses
- SS
Sjögren’s Syndrome
- TIMP
Tissue Inhibitor of Metalloproteinases
- TBUT
Tear Break-up Time
- tPA
tissue-type PA
- uPA
Urokinase-type PA
- uPAR
Urokinase-type PA Receptor
- VKC
Vernal keratoconjunctivitis
4. References
- 1.Willcox MDP, Argueso P, Georgiev GA, et al. TFOS DEWS II Tear Film Report. Ocul Surf 2017;15:366–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ablamowicz AF, Nichols JJ. Ocular Surface Membrane-Associated Mucins. Ocul Surf 2016;14:331–341. [DOI] [PubMed] [Google Scholar]
- 3.Argueso P, Guzman-Aranguez A, Mantelli F, et al. Association of cell surface mucins with galectin-3 contributes to the ocular surface epithelial barrier. J Biol Chem 2009;284:23037–23045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Pucker AD, Nichols JJ. Analysis of meibum and tear lipids. Ocul Surf 2012;10:230–250. [DOI] [PubMed] [Google Scholar]
- 5.Acosta MC, Peral A, Luna C, et al. Tear secretion induced by selective stimulation of corneal and conjunctival sensory nerve fibers. Invest Ophthalmol Vis Sci 2004;45:2333–2336. [DOI] [PubMed] [Google Scholar]
- 6.Perumal N, Funke S, Wolters D, et al. Characterization of human reflex tear proteome reveals high expression of lacrimal proline-rich protein 4 (PRR4). Proteomics 2015;15:3370–3381. [DOI] [PubMed] [Google Scholar]
- 7.Murube J Basal, reflex, and psycho-emotional tears. Ocul Surf 2009;7:60–66. [DOI] [PubMed] [Google Scholar]
- 8.Rohit A, Stapleton F, Brown SH, et al. Comparison of tear lipid profile among basal, reflex, and flush tear samples. Optom Vis Sci 2014;91:1391–1395. [DOI] [PubMed] [Google Scholar]
- 9.Abusharha AA, AlShehri TM, Hakami AY, et al. Analysis of basal and reflex human tear osmolarity in normal subjects: assessment of tear osmolarity. Ther Adv Ophthalmol 2018;10:2515841418794886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Markoulli M, Papas E, Cole N, et al. The diurnal variation of matrix metalloproteinase-9 and its associated factors in human tears. Invest Ophthalmol Vis Sci 2012;53:1479–1484. [DOI] [PubMed] [Google Scholar]
- 11.Runstrom G, Mann A, Tighe B. The fall and rise of tear albumin levels: a multifactorial phenomenon. Ocul Surf 2013;11:165–180. [DOI] [PubMed] [Google Scholar]
- 12.Sack RA, Beaton AR, Sathe S. Diurnal variations in angiostatin in human tear fluid: a possible role in prevention of corneal neovascularization. Curr Eye Res 1999;18:186–193. [DOI] [PubMed] [Google Scholar]
- 13.Uchino E, Sonoda S, Nakao K, et al. Alteration of tear cytokine balance by eye closure: analysis by multicytokine assay. Graefes Arch Clin Exp Ophthalmol 2006;244:747–749. [DOI] [PubMed] [Google Scholar]
- 14.Zhou L, Beuerman RW. The power of tears: how tear proteomics research could revolutionize the clinic. Expert Rev Proteomics 2017;14:189–191. [DOI] [PubMed] [Google Scholar]
- 15.Zhou L, Zhao SZ, Koh SK, et al. In-depth analysis of the human tear proteome. J Proteomics 2012;75:3877–3885. [DOI] [PubMed] [Google Scholar]
- 16.de Souza GA, Godoy LM, Mann M. Identification of 491 proteins in the tear fluid proteome reveals a large number of proteases and protease inhibitors. Genome Biol 2006;7:R72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Rawlings N, Salvesen G. Handbook of Proteolytic Enzymes. 3rd ed: Elsevier; 2013. [Google Scholar]
- 18.Sakimoto T, Sawa M. Metalloproteinases in corneal diseases: degradation and processing. Cornea 2012;31 Suppl 1:S50–56. [DOI] [PubMed] [Google Scholar]
- 19.Klinngam W, Fu R, Janga SR, et al. Cathepsin S Alters the Expression of Pro-Inflammatory Cytokines and MMP-9, Partially through Protease-Activated Receptor-2, in Human Corneal Epithelial Cells. Int J Mol Sci 2018;19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kinoshita S, Adachi W, Sotozono C, et al. Characteristics of the human ocular surface epithelium. Prog Retin Eye Res 2001;20:639–673. [DOI] [PubMed] [Google Scholar]
- 21.Pflugfelder S, Fang B, De Paiva C. Matrix metalloproteinase-9 in the pathophysiology and diagnosis of dry eye syndrome. Metalloproteinases Med 2017;Volume 4:37–46. [Google Scholar]
- 22.Lanza NL, Valenzuela F, Perez VL, et al. The Matrix Metalloproteinase 9 Point-of-Care Test in Dry Eye. Ocul Surf 2016;14:189–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.di Martino E, Ali M, Inglehearn CF. Matrix metalloproteinases in keratoconus - Too much of a good thing? Exp Eye Res 2019;182:137–143. [DOI] [PubMed] [Google Scholar]
- 24.Di Zazzo A, Micera A, De Piano M, et al. Tears and ocular surface disorders: Usefulness of biomarkers. J Cell Physiol 2019;234:9982–9993. [DOI] [PubMed] [Google Scholar]
- 25.Hagan S, Martin E, Enriquez-de-Salamanca A. Tear fluid biomarkers in ocular and systemic disease: potential use for predictive, preventive and personalised medicine. EPMA J 2016;7:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.von Thun Und Hohenstein-Blaul N, Funke S, Grus FH. Tears as a source of biomarkers for ocular and systemic diseases. Exp Eye Res 2013;117:126–137. [DOI] [PubMed] [Google Scholar]
- 27.Zhou L, Beuerman RW. Tear analysis in ocular surface diseases. Prog Retin Eye Res 2012;31:527–550. [DOI] [PubMed] [Google Scholar]
- 28.Nanjappa V, Thomas JK, Marimuthu A, et al. Plasma Proteome Database as a resource for proteomics research: 2014 update. Nucleic Acids Res 2014;42:D959–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shaila M, Pai GP, Shetty P. Salivary protein concentration, flow rate, buffer capacity and pH estimation: A comparative study among young and elderly subjects, both normal and with gingivitis and periodontitis. J Indian Soc Periodontol 2013;17:42–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Dewhirst FE, Chen T, Izard J, et al. The human oral microbiome. J Bacteriol 2010;192:5002–5017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Helmerhorst EJ, Oppenheim FG. Saliva: a dynamic proteome. J Dent Res 2007;86:680–693. [DOI] [PubMed] [Google Scholar]
- 32.Börger M, Funke S, Bähr M, et al. Biomarker sources for Parkinson’s disease: Time to shed tears? Basal Ganglia 2015;5:63–69. [Google Scholar]
- 33.Hamm-Alvarez SF, Lew MF, Feigenbaum D, et al. Tear Proteins as Possible Biomarkers for Parkinson’s Disease. Invest Ophthalmol Vis Sci 2018;58:4909. [Google Scholar]
- 34.Kallo G, Emri M, Varga Z, et al. Changes in the Chemical Barrier Composition of Tears in Alzheimer’s Disease Reveal Potential Tear Diagnostic Biomarkers. PLoS One 2016;11:e0158000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Aass C, Norheim I, Eriksen EF, et al. Comparative proteomic analysis of tear fluid in Graves’ disease with and without orbitopathy. Clin Endocrinol (Oxf) 2016;85:805–812. [DOI] [PubMed] [Google Scholar]
- 36.Jones DT, Monroy D, Pflugfelder SC. A novel method of tear collection: comparison of glass capillary micropipettes with porous polyester rods. Cornea 1997;16:450–458. [PubMed] [Google Scholar]
- 37.Markoulli M, Papas E, Petznick A, et al. Validation of the flush method as an alternative to basal or reflex tear collection. Curr Eye Res 2011;36:198–207. [DOI] [PubMed] [Google Scholar]
- 38.Quah JH, Tong L, Barbier S. Patient acceptability of tear collection in the primary healthcare setting. Optom Vis Sci 2014;91:452–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Denisin AK, Karns K, Herr AE. Post-collection processing of Schirmer strip-collected human tear fluid impacts protein content. Analyst 2012;137:5088–5096. [DOI] [PubMed] [Google Scholar]
- 40.Copeland JR, Lamberts DW, Holly FJ. Investigation of the accuracy of tear lysozyme determination by the Quantiplate method. Invest Ophthalmol Vis Sci 1982;22:103–110. [PubMed] [Google Scholar]
- 41.Tuft SJ, Dart JK. The measurement of IgE in tear fluid: a comparison of collection by sponge or capillary. Acta Ophthalmol 1989;67:301–305. [DOI] [PubMed] [Google Scholar]
- 42.Stuchell RN, Feldman JJ, Farris RL, et al. The effect of collection technique on tear composition. Invest Ophthalmol Vis Sci 1984;25:374–377. [PubMed] [Google Scholar]
- 43.Ablamowicz AF, Nichols JJ. Concentrations of MUC16 and MUC5AC using three tear collection methods. Mol Vis 2017;23:529–537. [PMC free article] [PubMed] [Google Scholar]
- 44.Rentka A, Koroskenyi K, Harsfalvi J, et al. Evaluation of commonly used tear sampling methods and their relevance in subsequent biochemical analysis. Ann Clin Biochem 2017;54:521–529. [DOI] [PubMed] [Google Scholar]
- 45.Tsung PK, Holly FJ. Protease activities in human tears. Curr Eye Res 1981;1:351–355. [DOI] [PubMed] [Google Scholar]
- 46.Parks WC, Wilson CL, Lopez-Boado YS. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol 2004;4:617–629. [DOI] [PubMed] [Google Scholar]
- 47.Edman MC, Janga SR, Meng Z, et al. Increased Cathepsin S activity associated with decreased protease inhibitory capacity contributes to altered tear proteins in Sjogren’s Syndrome patients. Sci Rep 2018;8:11044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hamm-Alvarez SF, Janga SR, Edman MC, et al. Tear cathepsin S as a candidate biomarker for Sjogren’s syndrome. Arthritis Rheumatol 2014;66:1872–1881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Versura P, Bavelloni A, Grillini M, et al. Diagnostic performance of a tear protein panel in early dry eye. Mol Vis 2013;19:1247–1257. [PMC free article] [PubMed] [Google Scholar]
- 50.Aqrawi LA, Galtung HK, Vestad B, et al. Identification of potential saliva and tear biomarkers in primary Sjogren’s syndrome, utilising the extraction of extracellular vesicles and proteomics analysis. Arthritis Res Ther 2017;19:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sambursky R, O’Brien TP. MMP-9 and the perioperative management of LASIK surgery. Curr Opin Ophthalmol 2011;22:294–303. [DOI] [PubMed] [Google Scholar]
- 52.Kim HS, Shang T, Chen Z, et al. TGF-beta1 stimulates production of gelatinase (MMP-9), collagenases (MMP-1, -13) and stromelysins (MMP-3, -10, -11) by human corneal epithelial cells. Exp Eye Res 2004;79:263–274. [DOI] [PubMed] [Google Scholar]
- 53.Gordon GM, Ledee DR, Feuer WJ, et al. Cytokines and signaling pathways regulating matrix metalloproteinase-9 (MMP-9) expression in corneal epithelial cells. J Cell Physiol 2009;221:402–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Van Wart HE, Birkedal-Hansen H. The cysteine switch: a principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metalloproteinase gene family. Proc Natl Acad Sci U S A 1990;87:5578–5582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Murphy G, Cockett MI, Stephens PE, et al. Stromelysin is an activator of procollagenase. A study with natural and recombinant enzymes. Biochem J 1987;248:265–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Knauper V, Wilhelm SM, Seperack PK, et al. Direct activation of human neutrophil procollagenase by recombinant stromelysin. Biochem J 1993;295 ( Pt 2):581–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Knauper V, Lopez-Otin C, Smith B, et al. Biochemical characterization of human collagenase-3. J Biol Chem 1996;271:1544–1550. [DOI] [PubMed] [Google Scholar]
- 58.Eeckhout Y, Vaes G. Further studies on the activation of procollagenase, the latent precursor of bone collagenase. Effects of lysosomal cathepsin B, plasmin and kallikrein, and spontaneous activation. Biochem J 1977;166:21–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Werb Z, Mainardi CL, Vater CA, et al. Endogenous activation of latent collagenase by rheumatoid synovial cells. Evidence for a role of plasminogen activator. N Engl J Med 1977;296:1017–1023. [DOI] [PubMed] [Google Scholar]
- 60.Christensen J, Shastri VP. Matrix-metalloproteinase-9 is cleaved and activated by cathepsin K. BMC Res Notes 2015;8:322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gordon GM, Austin JS, Sklar AL, et al. Comprehensive gene expression profiling and functional analysis of matrix metalloproteinases and TIMPs, and identification of ADAM-10 gene expression, in a corneal model of epithelial resurfacing. J Cell Physiol 2011;226:1461–1470. [DOI] [PubMed] [Google Scholar]
- 62.Kenney MC, Chwa M, Alba A, et al. Localization of TIMP-1, TIMP-2, TIMP-3, gelatinase A and gelatinase B in pathological human corneas. Curr Eye Res 1998;17:238–246. [DOI] [PubMed] [Google Scholar]
- 63.Vu TH, Werb Z. Matrix metalloproteinases: effectors of development and normal physiology. Genes Dev 2000;14:2123–2133. [DOI] [PubMed] [Google Scholar]
- 64.Mott JD, Werb Z. Regulation of matrix biology by matrix metalloproteinases. Curr Opin Cell Biol 2004;16:558–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Daniels JT, Limb GA, Saarialho-Kere U, et al. Human corneal epithelial cells require MMP-1 for HGF-mediated migration on collagen I. Invest Ophthalmol Vis Sci 2003;44:1048–1055. [DOI] [PubMed] [Google Scholar]
- 66.Matsubara M, Girard MT, Kublin CL, et al. Differential roles for two gelatinolytic enzymes of the matrix metalloproteinase family in the remodelling cornea. Dev Biol 1991;147:425–439. [DOI] [PubMed] [Google Scholar]
- 67.Girard MT, Matsubara M, Kublin C, et al. Stromal fibroblasts synthesize collagenase and stromelysin during long-term tissue remodeling. J Cell Sci 1993;104 ( Pt 4):1001–1011. [DOI] [PubMed] [Google Scholar]
- 68.Barro CD, Romanet JP, Fdili A, et al. Gelatinase concentration in tears of corneal-grafted patients. Curr Eye Res 1998;17:174–182. [DOI] [PubMed] [Google Scholar]
- 69.Baker AH, Edwards DR, Murphy G. Metalloproteinase inhibitors: biological actions and therapeutic opportunities. J Cell Sci 2002;115:3719–3727. [DOI] [PubMed] [Google Scholar]
- 70.Qi JH, Ebrahem Q, Moore N, et al. A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat Med 2003;9:407–415. [DOI] [PubMed] [Google Scholar]
- 71.Yan L, Borregaard N, Kjeldsen L, et al. The high molecular weight urinary matrix metalloproteinase (MMP) activity is a complex of gelatinase B/MMP-9 and neutrophil gelatinase-associated lipocalin (NGAL). Modulation of MMP-9 activity by NGAL. J Biol Chem 2001;276:37258–37265. [DOI] [PubMed] [Google Scholar]
- 72.Acera A, Vecino E, Duran JA. Tear MMP-9 levels as a marker of ocular surface inflammation in conjunctivochalasis. Invest Ophthalmol Vis Sci 2013;54:8285–8291. [DOI] [PubMed] [Google Scholar]
- 73.Leonardi A, Brun P, Abatangelo G, et al. Tear levels and activity of matrix metalloproteinase (MMP)-1 and MMP-9 in vernal keratoconjunctivitis. Invest Ophthalmol Vis Sci 2003;44:3052–3058. [DOI] [PubMed] [Google Scholar]
- 74.Ghavami S, Hashemi M, de Serres FJ, et al. Trypsin inhibitory capacity in vernal keratoconjunctivitis. Invest Ophthalmol Vis Sci 2007;48:264–269. [DOI] [PubMed] [Google Scholar]
- 75.Symeonidis C, Papakonstantinou E, Galli A, et al. Matrix metalloproteinase (MMP-2, -9) and tissue inhibitor (TIMP-1, -2) activity in tear samples of pediatric type 1 diabetic patients: MMPs in tear samples from type 1 diabetes. Graefes Arch Clin Exp Ophthalmol 2013;251:741–749. [DOI] [PubMed] [Google Scholar]
- 76.Toth M, Fridman R. Assessment of Gelatinases (MMP-2 and MMP-9 by Gelatin Zymography. Methods Mol Med 2001;57:163–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ren Z, Chen J, Khalil RA. Zymography as a Research Tool in the Study of Matrix Metalloproteinase Inhibitors. Methods Mol Biol 2017;1626:79–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Fonovic M, Turk B. Cysteine cathepsins and extracellular matrix degradation. Biochim Biophys Acta 2014;1840:2560–2570. [DOI] [PubMed] [Google Scholar]
- 79.Vidak E, Javorsek U, Vizovisek M, et al. Cysteine Cathepsins and their Extracellular Roles: Shaping the Microenvironment. Cells 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Janga SR, Shah M, Ju Y, et al. Longitudinal analysis of tear cathepsin S activity levels in male non-obese diabetic mice suggests its potential as an early stage biomarker of Sjogren’s Syndrome. Biomarkers 2019;24:91–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ju Y, Janga SR, Klinngam W, et al. NOD and NOR mice exhibit comparable development of lacrimal gland secretory dysfunction but NOD mice have more severe autoimmune dacryoadenitis. Exp Eye Res 2018;176:243–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Li X, Wu K, Edman M, et al. Increased expression of cathepsins and obesity-induced proinflammatory cytokines in lacrimal glands of male NOD mouse. Invest Ophthalmol Vis Sci 2010;51:5019–5029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Meng Z, Klinngam W, Edman MC, et al. Interferon-gamma treatment in vitro elicits some of the changes in cathepsin S and antigen presentation characteristic of lacrimal glands and corneas from the NOD mouse model of Sjogren’s Syndrome. PLoS One 2017;12:e0184781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Barka T, Asbell PA, van der Noen H, et al. Cystatins in human tear fluid. Curr Eye Res 1991;10:25–34. [DOI] [PubMed] [Google Scholar]
- 85.Zi M, Xu Y. Involvement of cystatin C in immunity and apoptosis. Immunol Lett 2018;196:80–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Dikovskaya MA, Trunov AN, Chernykh VV, et al. Cystatin C and lactoferrin concentrations in biological fluids as possible prognostic factors in eye tumor development. Int J Circumpolar Health 2013;72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mayer M. Biochemical and biological aspects of the plasminogen activation system. Clin Biochem 1990;23:197–211. [DOI] [PubMed] [Google Scholar]
- 88.Pandolfi M, Astrup T. A histochemical study of the fibrinolytic activity. Cornea, conjunctiva, and lacrimal gland. Arch Ophthalmol 1967;77:258–264. [DOI] [PubMed] [Google Scholar]
- 89.Lantz E, Andersson A. Release of fibrinolytic activators from the cornea and conjunctiva. Graefes Arch Clin Exp Ophthalmol 1982;219:263–267. [DOI] [PubMed] [Google Scholar]
- 90.Sugioka K, Kodama A, Yoshida K, et al. The roles of urokinase-type plasminogen activator in leukocyte infiltration and inflammatory responses in mice corneas treated with lipopolysaccharide. Invest Ophthalmol Vis Sci 2014;55:5338–5350. [DOI] [PubMed] [Google Scholar]
- 91.Berman M, Leary R, Gage J. Evidence for a role of the plasminogen activator--plasmin system in corneal ulceration. Invest Ophthalmol Vis Sci 1980;19:1204–1221. [PubMed] [Google Scholar]
- 92.Tripathi RC, Park JK, Tripathi BJ, et al. Tissue plasminogen activator in human aqueous humor and its possible therapeutic significance. Am J Ophthalmol 1988;106:719–722. [DOI] [PubMed] [Google Scholar]
- 93.Hayashi K, Sueishi K. Fibrinolytic activity and species of plasminogen activator in human tears. Exp Eye Res 1988;46:131–137. [DOI] [PubMed] [Google Scholar]
- 94.Crippa MP. Urokinase-type plasminogen activator. Int J Biochem Cell Biol 2007;39:690–694. [DOI] [PubMed] [Google Scholar]
- 95.Gebbink MF. Tissue-type plasminogen activator-mediated plasminogen activation and contact activation, implications in and beyond haemostasis. J Thromb Haemost 2011;9 Suppl 1:174–181. [DOI] [PubMed] [Google Scholar]
- 96.Lantz E, Pandolfi M. Fibrinolysis in cornea and conjunctiva: evidence of two types of activators. Graefes Arch Clin Exp Ophthalmol 1986;224:393–396. [DOI] [PubMed] [Google Scholar]
- 97.Thorig L, Wijngaards G, Van Haeringen NJ. Immunological characterization and possible origin of plasminogen activator in human tear fluid. Ophthalmic Res 1983;15:268–276. [DOI] [PubMed] [Google Scholar]
- 98.Tozser J, Berta A. Plasminogen-Activator Inhibitors in Human Tears. Acta Ophthalmol 1991;69:426–431. [DOI] [PubMed] [Google Scholar]
- 99.Lemp MA. The Definition and Classification of Dry Eye Disease: Report of the Definition and Classification Subcommittee of the International Dry Eye Workshop (2007). Ocul Surf 2007;5:75–92. [DOI] [PubMed] [Google Scholar]
- 100.Stern ME, Pflugfelder SC. Inflammation in dry eye. Ocul Surf 2004;2:124–130. [DOI] [PubMed] [Google Scholar]
- 101.Pescosolido N, Barbato A, Pascarella A, et al. Role of Protease-Inhibitors in Ocular Diseases. Molecules 2014;19:20557–20569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Brito-Zeron P, Kostov B, Solans R, et al. Systemic activity and mortality in primary Sjogren syndrome: predicting survival using the EULAR-SS Disease Activity Index (ESSDAI) in 1045 patients. Ann Rheum Dis 2016;75:348–355. [DOI] [PubMed] [Google Scholar]
- 103.Brito-Zeron P, Baldini C, Bootsma H, et al. Sjogren syndrome. Nat Rev Dis Primers 2016;2:16047. [DOI] [PubMed] [Google Scholar]
- 104.Foulks GN, Bron AJ. Meibomian gland dysfunction: a clinical scheme for description, diagnosis, classification, and grading. Ocul Surf 2003;1:107–126. [DOI] [PubMed] [Google Scholar]
- 105.Tong L, Zhou L, Beuerman RW, et al. Association of tear proteins with Meibomian gland disease and dry eye symptoms. Br J Ophthalmol 2011;95:848–852. [DOI] [PubMed] [Google Scholar]
- 106.Soria J, Duran JA, Etxebarria J, et al. Tear proteome and protein network analyses reveal a novel pentamarker panel for tear film characterization in dry eye and meibomian gland dysfunction. J Proteomics 2013;78:94–112. [DOI] [PubMed] [Google Scholar]
- 107.Bartlett JD, Keith MS, Sudharshan L, et al. Associations between signs and symptoms of dry eye disease: a systematic review. Clin Ophthalmol 2015;9:1719–1730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.D’Souza S, Tong L. Practical issues concerning tear protein assays in dry eye. Eye Vis (Lond) 2014;1:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Chotikavanich S, de Paiva CS, Li de Q, et al. Production and activity of matrix metalloproteinase-9 on the ocular surface increase in dysfunctional tear syndrome. Invest Ophthalmol Vis Sci 2009;50:3203–3209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Solomon A, Dursun D, Liu Z, et al. Pro- and anti-iflammatory forms of Interleukin-1 in the tear fluid and conjunctiva of Patients with dry-eye disease. Invest Ophthalmol Vis Sci 2001;42:2283–2292. [PubMed] [Google Scholar]
- 111.Sobrin L, Liu Z, Monroy DC, et al. Regulation of MMP-9 activity in human tear fluid and corneal epithelial cuture supernatant. Invest Ophthalmol Vis Sci 2000;41:1703–1709. [PubMed] [Google Scholar]
- 112.Afonso A A., Sobrin L, Monroy D, et al. Tear fluid gelatinase B activity correlates with IL-1α concentration and fluorescein clearance in ocular rosacea. Invest Ophthalmol Vis Sci 1999;40:2507–2512. [PubMed] [Google Scholar]
- 113.Acera A, Rocha G, Vecino E, et al. Inflammatory markers in the tears of patients with ocular surface disease. Ophthalmic Res 2008;40:315–321. [DOI] [PubMed] [Google Scholar]
- 114.M. S, Ivanova S, Kakkassery V, et al. Correlation of tear film osmolarity and 2 different MMP-9 test with common dry eye tests in a cohort of non-dry eye patients. Cornea 2015;34:739–744. [DOI] [PubMed] [Google Scholar]
- 115.Sambursky R, Davitt WF 3rd, Latkany R, et al. Sensitivity and specificity of a point-of-care matrix metalloproteinase 9 immunoassay for diagnosing inflammation related to dry eye. JAMA Ophthalmol 2013;131:24–28. [DOI] [PubMed] [Google Scholar]
- 116.Messmer EM, von Lindenfels V, Garbe A, et al. Matrix Metalloproteinase 9 Testing in Dry Eye Disease Using a Commercially Available Point-of-Care Immunoassay. Ophthalmology 2016;123:2300–2308. [DOI] [PubMed] [Google Scholar]
- 117.Stoka V, Turk V, Turk B. Lysosomal cathepsins and their regulation in aging and neurodegeneration. Ageing Res Rev 2016;32:22–37. [DOI] [PubMed] [Google Scholar]
- 118.Shi GP, Villadangos JA, Dranoff G, et al. Cathepsin S required for normal MHC class II peptide loading and germinal center development. Immunity 1999;10:197–206. [DOI] [PubMed] [Google Scholar]
- 119.Chang W, Wu H, Yeh C, et al. Lysosomal cysteine proteinase cathepsin S as a potential target for anti-cancer therapy. J Cancer Mol 2007;3:5–14. [Google Scholar]
- 120.Bielory L Allergic and immunologic disorders of the eye. Part I: immunology of the eye. J Allergy Clin Immunol 2000;106:805–816. [DOI] [PubMed] [Google Scholar]
- 121.Kumagai N, Yamamoto K, Fukuda K, et al. Active matrix metalloproteinases in the tear fluid of individuals with vernal keratoconjunctivitis. J Allergy Clin Immunol 2002;110:489–491. [DOI] [PubMed] [Google Scholar]
- 122.Leonardi A, Sathe S, Bortolotti M, et al. Cytokines, matrix metalloproteases, angiogenic and growth factors in tears of normal subjects and vernal keratoconjunctivitis patients. Allergy 2009;64:710–717. [DOI] [PubMed] [Google Scholar]
- 123.Maatta M, Kari O, Tervahartiala T, et al. Elevated expression and activation of matrix metalloproteinase 8 in tear fluid in atopic blepharoconjunctivitis. Cornea 2008;27:297–301. [DOI] [PubMed] [Google Scholar]
- 124.Kari O, Maatta M, Tervahartiala T, et al. Tear fluid concentration of mmp-8 is elevated in non-allergic eosinophilic conjunctivitis and correlates with conjunctival inflammatory cell infiltration. Graefes Arch Clin Exp Ophthalmol 2009;247:681–686. [DOI] [PubMed] [Google Scholar]
- 125.Smith VA, Rishmawi H, Hussein H, et al. Tear film MMP accumulation and corneal disease. Br J Ophthalmol 2001;85:147–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Singh A, Maurya OP, Jagannadhan MV, et al. Matrix metalloproteinases (MMP-2 and MMP-9) activity in corneal ulcer and ocular surface disorders determined by gelatin zymography. J Ocul Biol Dis Infor 2012;5:31–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ananthi S, Venkatesh Prajna N, Lalitha P, et al. Pathogen induced changes in the protein profile of human tears from Fusarium keratitis patients. PLoS One 2013;8:e53018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ananthi S, Chitra T, Bini R, et al. Comparative analysis of the tear protein profile in mycotic keratitis patients. Mol Vis 2008;14:500–507. [PMC free article] [PubMed] [Google Scholar]
- 129.Berta A, Tozser J, Holly FJ. Determination of plasminogen activator activities in normal and pathological human tears. The significance of tear plasminogen activators in the inflammatory and traumatic lesions of the cornea and the conjunctiva. Acta Ophthalmol 1990;68:508–514. [DOI] [PubMed] [Google Scholar]
- 130.Leonardi A, Brun P, Sartori MT, et al. Urokinase plasminogen activator, uPa receptor, and its inhibitor in vernal keratoconjunctivitis. Invest Ophthalmol Vis Sci 2005;46:1364–1370. [DOI] [PubMed] [Google Scholar]
- 131.Bukowiecki A, Hos D, Cursiefen C, et al. Wound-Healing Studies in Cornea and Skin: Parallels, Differences and Opportunities. Int J Mol Sci 2017;18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zieske JD. Extracellular matrix and wound healing. Curr Opin Ophthalmol 2001;12:237–241. [DOI] [PubMed] [Google Scholar]
- 133.Ljubimov AV, Saghizadeh M. Progress in corneal wound healing. Prog Retin Eye Res 2015;49:17–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Nettune GR, Pflugfelder SC. Post-LASIK tear dysfunction and dysesthesia. Ocul Surf 2010;8:135–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Munnerlyn CR, Koons SJ, Marshall J. Photorefractive keratectomy: a technique for laser refractive surgery. J Cataract Refract Surg 1988;14:46–52. [DOI] [PubMed] [Google Scholar]
- 136.Jain S, Khoury JM, Chamon W, et al. Corneal light scattering after laser in situ keratomileusis and photorefractive keratectomy. Am J Ophthalmol 1995;120:532–534. [DOI] [PubMed] [Google Scholar]
- 137.Gonzalez-Perez J, Villa-Collar C, Gonzalez-Meijome JM, et al. Long-term changes in corneal structure and tear inflammatory mediators after orthokeratology and LASIK. Invest Ophthalmol Vis Sci 2012;53:5301–5311. [DOI] [PubMed] [Google Scholar]
- 138.Elmohamady MN, Abdelghaffar W, Salem TI. Tear Martix Metalloproteinase-9 and Tissue Inhibitor of Metalloproteinase-1 in Post-Lasik Ectasia. Int Ophthalmol 2019;39:631–637. [DOI] [PubMed] [Google Scholar]
- 139.Giri P, Azar DT. Risk profiles of ectasia after keratorefractive surgery. Curr Opin Ophthalmol 2017;28:337–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Konomi K, Chen LL, Tarko RS, et al. Preoperative characteristics and a potential mechanism of chronic dry eye after LASIK. Invest Ophthalmol Vis Sci 2008;49:168–174. [DOI] [PubMed] [Google Scholar]
- 141.Gomes JAP, Azar DT, Baudouin C, et al. TFOS DEWS II iatrogenic report. Ocul Surf 2017;15:511–538. [DOI] [PubMed] [Google Scholar]
- 142.Chan TC, Ye C, Chan KP, et al. Evaluation of point-of-care test for elevated tear matrix metalloproteinase 9 in post-LASIK dry eyes. Br J Ophthalmol 2016;100:1188–1191. [DOI] [PubMed] [Google Scholar]
- 143.Azar DT, Hahn TW, Jain S, et al. Matrix metalloproteinases are expressed during wound healing after excimer laser keratectomy. Cornea 1996;15:18–24. [PubMed] [Google Scholar]
- 144.Azar DT, Pluznik D, Jain S, et al. Gelatinase B and A expression after laser in situ keratomileusis and photorefractive keratectomy. Arch Ophthalmol 1998;116:1206–1208. [DOI] [PubMed] [Google Scholar]
- 145.Mulholland B, Tuft SJ, Khaw PT. Matrix metalloproteinase distribution during early corneal wound healing. Eye (Lond) 2005;19:584–588. [DOI] [PubMed] [Google Scholar]
- 146.Holopainen JM, Moilanen JA, Sorsa T, et al. Activation of matrix metalloproteinase-8 by membrane type 1-MMP and their expression in human tears after photorefractive keratectomy. Invest Ophthalmol Vis Sci 2003;44:2550–2556. [DOI] [PubMed] [Google Scholar]
- 147.Mutoh T, Nishio M, Matsumoto Y, et al. Photorefractive keratectomy: measuring the matrix metalloproteinase activity and chondroitin sulfate concentration in tear fluid. Clin Ophthalmol 2010;4:1015–1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.D’Souza S, Petznick A, Tong L, et al. Comparative analysis of two femtosecond LASIK platforms using iTRAQ quantitative proteomics. Invest Ophthalmol Vis Sci 2014;55:3396–3402. [DOI] [PubMed] [Google Scholar]
- 149.Law RH, Abu-Ssaydeh D, Whisstock JC. New insights into the structure and function of the plasminogen/plasmin system. Curr Opin Struct Biol 2013;23:836–841. [DOI] [PubMed] [Google Scholar]
- 150.Tervo T, Virtanen T, Honkanen N, et al. Tear fluid plasmin activity after excimer laser photorefractive keratectomy. Invest Ophthalmol Vis Sci 1994;35:3045–3050. [PubMed] [Google Scholar]
- 151.O’Brart DP, Lohmann CP, Klonos G, et al. The effects of topical corticosteroids and plasmin inhibitors on refractive outcome, haze, and visual performance after photorefractive keratectomy. A prospective, randomized, observer-masked study. Ophthalmology 1994;101:1565–1574. [DOI] [PubMed] [Google Scholar]
- 152.Csutak A, Tozser J, Bekesi L, et al. Plasminogen activator activity in tears after excimer laser photorefractive keratectomy. Invest Ophthalmol Vis Sci 2000;41:3743–3747. [PubMed] [Google Scholar]
- 153.Csutak A, Silver DM, Tozser J, et al. Plasminogen activator activity and inhibition in rabbit tears after photorefractive keratectomy. Exp Eye Res 2003;77:675–680. [DOI] [PubMed] [Google Scholar]
- 154.Csutak A, Silver DM, Tozser J, et al. Urokinase-type plasminogen activator to prevent haze after photorefractive keratectomy, and pregnancy as a risk factor for haze in rabbits. Invest Ophthalmol Vis Sci 2004;45:1329–1333. [DOI] [PubMed] [Google Scholar]
- 155.Csutak A, Silver DM, Tozser J, et al. Plasminogen activator inhibitor in human tears after laser refractive surgery. J Cataract Refract Surg 2008;34:897–901. [DOI] [PubMed] [Google Scholar]
- 156.Pflugfelder SC, de Paiva CS, Tong L, et al. Stress-activated protein kinase signaling pathways in dry eye and ocular surface disease. Ocul Surf 2005;3:S154–157. [DOI] [PubMed] [Google Scholar]
- 157.Farris RL. The dry eye: its mechanisms and therapy, with evidence that contact lens is a cause. CLAO J 1986;12:234–246. [PubMed] [Google Scholar]
- 158.Boost M, Cho P, Wang Z. Disturbing the balance: effect of contact lens use on the ocular proteome and microbiome. Clin Exp Optom 2017;100:459–472. [DOI] [PubMed] [Google Scholar]
- 159.Lin MC, Yeh TN. Mechanical complications induced by silicone hydrogel contact lenses. Eye Contact Lens 2013;39:115–124. [DOI] [PubMed] [Google Scholar]
- 160.Nichols JJ, Ziegler C, Mitchell GL, et al. Self-reported dry eye disease across refractive modalities. Invest Ophthalmol Vis Sci 2005;46:1911–1914. [DOI] [PubMed] [Google Scholar]
- 161.Richdale K, Sinnott LT, Skadahl E, et al. Frequency of and factors associated with contact lens dissafaction and discontinuation. Cornea 2007;26:168–174. [DOI] [PubMed] [Google Scholar]
- 162.Kramann C, Boehm N, Lorenz K, et al. Effect of contact lenses on the protein composition in tear film: a ProteinChip study. Graefes Arch Clin Exp Ophthalmol 2011;249:233–243. [DOI] [PubMed] [Google Scholar]
- 163.Nichols JJ, Green-Church KB. Mass spectrometry-based proteomic analyses in contact lens-related dry eye. Cornea 2009;28:1109–1117. [DOI] [PubMed] [Google Scholar]
- 164.Nichols KK, Mitchell GL, Simon KM, et al. Corneal staining in hydrogel lens wearers. Optom Vis Sci 2002;79:20–30. [DOI] [PubMed] [Google Scholar]
- 165.Lema I, Duran JA, Ruiz C, et al. Inflammatory response to contact lenses in patients with keratoconus compared with myopic subjects. Cornea 2008;27:758–763. [DOI] [PubMed] [Google Scholar]
- 166.Mountford J, Pesudovs K. An analysis of the astigmatic changes induced by accelerated orthokeratology. Clin Exp Optom 2002;85:284–293. [DOI] [PubMed] [Google Scholar]
- 167.Gonzalez-Perez J, Villa-Collar C, Sobrino Moreiras T, et al. Tear film inflammatory mediators during continuous wear of contact lenses and corneal refractive therapy. Br J Ophthalmol 2012;96:1092–1098. [DOI] [PubMed] [Google Scholar]
- 168.Markoulli M, Papas E, Cole N, et al. Corneal erosions in contact lens wear. Cont Lens Anterior Eye 2012;35:2–8. [DOI] [PubMed] [Google Scholar]
- 169.Garrana RM, Zieske JD, Assouline M, et al. Matrix metalloproteinases in epithelia from human recurrent corneal erosion. Invest Ophthalmol Vis Sci 1999;40:1266–1270. [PubMed] [Google Scholar]
- 170.Markoulli M, Papas E, Cole N, et al. Effect of contact lens wear on the diurnal profile of matrix metalloproteinase 9 in tears. Optom Vis Sci 2013;90:419–429. [DOI] [PubMed] [Google Scholar]
- 171.Manicam C, Perumal N, Wasielica-Poslednik J, et al. Proteomics Unravels the Regulatory Mechanisms in Human Tears Following Acute Renouncement of Contact Lens Use: A Comparison between Hard and Soft Lenses. Sci Rep 2018;8:11526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Mas Tur V, MacGregor C, Jayaswal R, et al. A review of keratoconus: Diagnosis, pathophysiology, and genetics. Surv Ophthalmol 2017;62:770–783. [DOI] [PubMed] [Google Scholar]
- 173.Burdon KP, Vincent AL. Insights into keratoconus from a genetic perspective. Clin Exp Optom 2013;96:146–154. [DOI] [PubMed] [Google Scholar]
- 174.Foster CS, Calonge M. Atopic keratoconjunctivitis. Ophthalmology 1990;97:992–1000. [DOI] [PubMed] [Google Scholar]
- 175.Georgiou T, Funnell CL, Cassels-Brown A, et al. Influence of ethnic origin on the incidence of keratoconus and associated atopic disease in Asians and white patients. Eye (Lond) 2004;18:379–383. [DOI] [PubMed] [Google Scholar]
- 176.Loukovitis E, Sfakianakis K, Syrmakesi P, et al. Genetic Aspects of Keratoconus: A Literature Review Exploring Potential Genetic Contributions and Possible Genetic Relationships with Comorbidities. Ophthalmol Ther 2018;7:263–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Macsai MS, Varley GA, Krachmer JH. Development of keratoconus after contact lens wear. Patient characteristics. Arch Ophthalmol 1990;108:534–538. [DOI] [PubMed] [Google Scholar]
- 178.McMonnies CW. Mechanisms of rubbing-related corneal trauma in keratoconus. Cornea 2009;28:607–615. [DOI] [PubMed] [Google Scholar]
- 179.Pearson AR, Soneji B, Sarvananthan N, et al. Does ethnic origin influence the incidence or severity of keratoconus? Eye (Lond) 2000;14 ( Pt 4):625–628. [DOI] [PubMed] [Google Scholar]
- 180.Balasubramanian SA, Mohan S, Pye DC, et al. Proteases, proteolysis and inflammatory molecules in the tears of people with keratoconus. Acta Ophthalmol 2012;90:e303–309. [DOI] [PubMed] [Google Scholar]
- 181.Pannebaker C, Chandler HL, Nichols JJ. Tear proteomics in keratoconus. Mol Vis 2010;16:1949–1957. [PMC free article] [PubMed] [Google Scholar]
- 182.Prochazka M, Serreze DV, Frankel WN, et al. NOR/Lt mice: MHC-matched diabetes-resistant control strain for NOD mice. Diabetes 1992;41:98–106. [DOI] [PubMed] [Google Scholar]
- 183.Lema I, Duran JA. Inflammatory molecules in the tears of patients with keratoconus. Ophthalmology 2005;112:654–659. [DOI] [PubMed] [Google Scholar]
- 184.Lema I, Sobrino T, Duran JA, et al. Subclinical keratoconus and inflammatory molecules from tears. Br J Ophthalmol 2009;93:820–824. [DOI] [PubMed] [Google Scholar]
- 185.Shetty R, Ghosh A, Lim RR, et al. Elevated expression of matrix metalloproteinase-9 and inflammatory cytokines in keratoconus patients is inhibited by cyclosporine A. Invest Ophthalmol Vis Sci 2015;56:738–750. [DOI] [PubMed] [Google Scholar]
- 186.Collier SA. Is the corneal degradation in keratoconus caused by matrix-metalloproteinases? Clin Exp Ophthalmol 2001;29:340–344. [DOI] [PubMed] [Google Scholar]
- 187.Balasubramanian SA, Wasinger VC, Pye DC, et al. Preliminary identification of differentially expressed tear proteins in keratoconus. Mol Vis 2013;19:2124–2134. [PMC free article] [PubMed] [Google Scholar]
- 188.Balasubramanian SA, Pye DC, Willcox MD. Are proteinases the reason for keratoconus? Curr Eye Res 2010;35:185–191. [DOI] [PubMed] [Google Scholar]
- 189.Ionescu C, Corbu CG, Tanase C, et al. Inflammatory Biomarkers Profile as Microenvironmental Expression in Keratoconus. Dis Markers 2016;2016:1243819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Shikari H, Antin JH, Dana R. Ocular graft-versus-host disease: a review. Surv Ophthalmol 2013;58:233–251. [DOI] [PubMed] [Google Scholar]
- 191.Kim SK. Ocular graft vs. host disease. Ocul Surf 2005;3:S177–179. [DOI] [PubMed] [Google Scholar]
- 192.Giannaccare G, Pellegrini M, Bernabei F, et al. Ocular surface system alterations in ocular graft-versus-host disease: all the pieces of the complex puzzle. Graefes Arch Clin Exp Ophthalmol 2019;257:1341–1351. [DOI] [PubMed] [Google Scholar]
- 193.Nair S, Vanathi M, Mahapatra M, et al. Tear inflammatory mediators and protein in eyes of post allogenic hematopoeitic stem cell transplant patients. Ocul Surf 2018;16:352–367. [DOI] [PubMed] [Google Scholar]
- 194.Arafat SN, Robert MC, Abud T, et al. Elevated Neutrophil Elastase in Tears of Ocular Graft-Versus-Host Disease Patients. Am J Ophthalmol 2017;176:46–52. [DOI] [PubMed] [Google Scholar]
- 195.Gerber-Hollbach N, Plattner K, O’Leary OE, et al. Tear Film Proteomics Reveal Important Differences Between Patients With and Without Ocular GvHD After Allogeneic Hematopoietic Cell Transplantation. Invest Ophthalmol Vis Sci 2018;59:3521–3530. [DOI] [PubMed] [Google Scholar]
- 196.Zhang L, Su Z, Zhang Z, et al. Effects of azithromycin on gene expression profiles of proinflammatory and anti-inflammatory mediators in the eyelid margin and conjunctiva of patients with meibomian gland disease. JAMA Ophthalmol 2015;133:1117–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Park JY, Kim BG, Kim JS, et al. Matrix Metalloproteinase 9 Point-of-Care Immunoassay Result Predicts Response to Topical Cyclosporine Treatment in Dry Eye Disease. Transl Vis Sci Technol 2018;7:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Hadassah J, Bhuvaneshwari N, Rao U, et al. Evaluation of succinylated collagen bandage lenses in corneal healing by the expression of matrix metalloproteinases (MMP-2 and MMP-9) in tear fluid. Ophthalmic Res 2009;42:64–72. [DOI] [PubMed] [Google Scholar]


